You are on page 1of 139

Interaction of Corynebacterium diphtheriae

with host cells


(Interaktion von Corynebacterium diphtheriae mit Wirtszellen)

Der Naturwissenschaftlichen Fakultät


der Friedrich-Alexander-Universität Erlangen-Nürnberg
zur
Erlangung des Doktorgrades Dr. rer. nat.

vorgelegt von
Lisa Ott

aus Nürnberg
Als Dissertation genehmigt von der Naturwissen-
schaftlichen Fakultät der Friedrich-Alexander-Universität
Erlangen-Nürnberg

Tag der mündlichen Prüfung: 19.12.2012

Vorsitzender
der Promotionskomission: Prof. Dr. Johannes Barth

Erstberichterstatter: Prof. Dr. Andreas Burkovski

Zweitberichterstatter: Prof. Dr. Uwe Sonnewald


Table of contents

Table of contents

1 Introduction............................................................................................................4
1.1 The genus Corynebacterium .................................................................................................... 4
1.2 Pathogenicity of Corynebacterium diphtheriae ...................................................................... 7
1.2.1 Adhesion properties of C. diphtheriae ............................................................................. 7
1.2.2 Factors involved in the invasion process of C. diphtheriae ........................................ 12
1.2.3 C. diphtheriae and host immune response .................................................................... 15
1.2.4 Phagocytosis and induction of apoptosis ..................................................................... 17

2 Aim of the work .................................................................................................... 23

3 Publications ......................................................................................................... 24
3.1 Corynebacterium diphtheriae invasion-associated protein (DIP1281) is involved in cell
surface organization, adhesion and internalization in epithelial cells (Ott et al, 2010a) ............. 24
3.2 Strain-specific differences in pili formation and the interaction of Corynebacterium
diphtheriae with host cells (Ott et al, 2010b).................................................................................... 34
3.3 The complete genome sequence of Corynebacterium pseudotuberculosis FRC41
isolated from a 12-year-old girl with necrotizing lymphadenitis reveals insights into gene-
regulatory networks contributing to virulence (Trost et al, 2010b) ............................................... 47
3.4 Impact of improved potassium accumulation on pH homeostasis, membrane potential
adjustment and survival of Corynebacterium glutamicum (Ochrombel et al, 2011) ................... 65
3.5 Evaluation of invertebrate infection models for pathogenic corynebacteria (Ott et al,
2012a) ................................................................................................................................................... 73
3.6 Contour and persistence length of Corynebacterium diphtheriae pili by atomic force
microscopy (Rheinlaender et al, 2012) ............................................................................................. 83
3.7 Induction  of  the  NFĸB  signal  transduction  pathway  in  response  to  Corynebacterium
diphtheriae infection (Ott et al, 2012b) ............................................................................................. 94

4 Summary/Zusammenfassung........................................................................... 122
4.1 Summary ................................................................................................................................. 122
4.2 Zusammenfassung ................................................................................................................ 124

5 References ......................................................................................................... 126

6 Curriculum Vitae ................................................................................................ 135

7 Danksagung ....................................................................................................... 139

3
Introduction

1 Introduction

1.1 The genus Corynebacterium

Corynebacteria belong to the class of Actinobacteria (high G+C Gram-positive bacteria) and

comprise a collection of morphologically similar, irregular- or clubbed-shaped microorganisms

(Ventura et al, 2007; Zhi et al, 2009). Since 1970, corynebacteria are dedicated to the CMN

group (Corynebacterium, Mycobacterium, Nocardia), which is based on the molecular

similarity of cell wall components. Figure 1 shows the typical v-shaped cell dimers, which

occur during cell division of corynebacteria. This is also called snapping division (Barksdale,

1970).

Fig. 1. Differential staining of infection of non-phagocytic epithelial cells with non-


toxigenic C. diphtheriae. (A) extracellular C. diphtheriae are depicted in green; (B)
internalized bacteria are indicated in dark red; adherent bacteria in light red; (C) Detroit 562
cells appear in blue; extracellular bacteria in orange; intracellular bacteria in light red. Scale
bars: 20 µm (Ott et al, 2010b).

Almost all members of the genus are characterized by complex cell envelope architecture: in

addition to cytoplasmic membrane and peptidoglycan layer, an outer lipid layer is observable,

which is covalently linked to arabinogalactan and mainly composed of mycolic acids. In some

cases the outer membrane is additionally covered by an ordered S- (surface-) layer of proteins

(Bayan et al, 2003) or complex protein mixtures are found (Marienfeld et al, 1997) (Hermann

et al, 2000) (Hansmeier et al, 2007; Hansmeier et al, 2006a; Hansmeier et al, 2006b). Figure 2

presents the mycobacterial outer cell envelope, which is similar to that of corynebacteria.

4
Introduction

Fig. 2. Cryo-electron microscopy of the mycobacterial cell envelope. (a) The


mycobacterial outer membrane is visualized by cryosectioning and (b) cryo-electron
tomography. The periplasmic space between the outer membrane and the cytoplasmic
membrane contains the layers of the arabinogalactan-peptidoglycan polymer (indicated in blue
in the 3-D representation). Scale bars: 50 nm (Niederweis et al, 2010).

To date, the genus Corynebacterium comprises 88 species (Bernard, 2012), including soil

bacteria with biotechnological importance (Corynebacterium glutamicum), commensals found

on skin of humans and animals as well as pathogens (Corynebacterium diphtheriae,

Corynebacterium ulcerans and Corynebacterium pseudotuberculosis). C. diphtheriae is the

classical etiological agent of diphtheria and the type species of the genus Corynebacterium

(Lehmann & Neumann 1896; Barksdale, 1970). Pathogenic corynebacteria are mostly not

highly virulent, and only immune-compromised patients get infected. However, virulence can

be   dramatically   increased   when   bacteria   are   lysogenized   by   β-corynebacteriophages, which

encode the diphtheria toxin and are integrated into the genome of toxigenic strains. C.

ulcerans and C. pseudotuberculosis are also able to produce the diphtheria toxin due to

infection with tox gene-carrying corynebacteriophages (Groman et al, 1984; Cianciotto &

Groman, 1996).

C. ulcerans is a commensal in domestic and wild animals (Schuhegger et al, 2009;

Dixon, 2010; Sykes et al, 2010) that may serve as reservoir for zoonotic infections with a

diphtheria-like illness. Transmission occurs from animals to humans and usually adults get

infected. High risk of infection is given by consuming raw milk (Bostock et al, 1984; Hart, 1984)

or close contact with domestic animals (Wagner et al, 2010); no person to person transmission

was reported up to now.

5
Introduction

C. pseudotuberculosis is the etiological agent of caseous lymphadenitis in sheep and

goats throughout the world (Dorella et al, 2006; Baird & Fontaine, 2007). Infections due to C.

pseudotuberculosis are rare in humans, but there are some cases of suppurative

lymphadenitis in patients with close contact with sheep. Despite the putative presence of a

diphtheria toxin, classical diphtheria of respiratory tract or skin is not observed. It is important

to know that C. pseudotuberculosis is able to proliferate and persist in macrophages, thus

escaping the immune response of the host since it is a facultative intracellular pathogen

(McKean et al, 2005; Dorella et al, 2006) (Fig. 3).

Fig. 3. Transmission of toxigenic corynebacteria. Transmission of C. diphtheriae from


person to person occurs by close physical contact. Respiratory diphtheria is mainly
disseminated by respiratory droplets but also smear infections are possible, mostly in case of
cutaneous diphtheria. C. ulcerans infections occur via close physical contact with pets and
farm animals or animal bites, while man-to-man transmission was not reported. C.
pseudotuberculosis is transferred to humans from infected cattle, especially sheep and goats,
by close physical contact (Burkovski, 2012).

Based on its medical importance as etiological agent of diphtheria, C. diphtheriae was one of

the first Corynebacterium species for which genome sequences became available (Cerdeno-

Tarraga et al, 2003). With the development of high throughput sequencing techniques, to date

numerous genome sequences of pathogenic corynebacteria are available, including those of

Corynebacterium aurimucosum (Trost et al, 2010a), Corynebacterium bovis (Schröder et al,

6
Introduction

2012a), various C. diphtheriae strains (Sangal et al, 2012a; Sangal et al, 2012b; Trost et al,

2012), C. jeikeium (Tauch et al, 2005), Corynebacterium kroppenstettii (Tauch et al, 2008a),

several C. pseudotuberculosis strains (Trost et al, 2010b; Cerdeira et al, 2011a; Cerdeira et al,

2011b; Lopes et al, 2012; Pethick et al, 2012a; Pethick et al, 2012b; Silva et al, 2011),

Corynebacterium resistens (Schröder et al, 2012b), two C. ulcerans strains (Trost et al, 2011)

and Corynebacterium urealyticum (Tauch et al, 2008b). This wealth of genome information

now provides a basis for the characterization of corynebacterial virulence factors and

corresponding genes at a molecular level.

1.2 Pathogenicity of Corynebacterium diphtheriae

C. diphtheriae causes diphtheria of the upper respiratory tract, which is characterized by sore

throat, low fever and malaise. Symptoms range from mild pharyngitis to severe hypoxia with

pseudomembrane formation due to the toxin. In advanced cases swelling of lymph nodes

results in strong swelling of the neck tissue (bull neck diphtheria) (Hadfield et al, 2000).

Diphtheria also occurs on skin (cutaneous diphtheria), which outnumbers the classical

respiratory diphtheria in tropical areas. Besides these infections, toxigenic and non-toxigenic

C. diphtheriae strains are also associated with systemic infections, e.g. endocarditis,

osteomyelitis, splenic abscesses, meningitis and septic arthritis (Hirata et al, 2008; Puliti et al,

2006), indicating that C. diphtheriae is able to colonize not only epithelia but also deeper parts

of the body.

1.2.1 Adhesion properties of C. diphtheriae

To date, experimental data concerning host pathogen interaction of eukaryotic cell with C.

diphtheriae are limited. Best investigated adhesion factors might be pili structures on the

surface of C. diphtheriae, which are covalently attached to the cell wall peptidoglycan (Navarre

& Schneewind, 1999). Pili formation requires enzymes with transpeptidase activity and sorting

signals of surface proteins (Fischetti et al, 1990; Schneewind et al, 1995; Schneewind et al,

1992). The genome of C. diphtheriae NCTC 13129 encodes three distinct pili clusters

7
Introduction

(spaABC, spaDEF, spaGHI), which are organized together with their sortase encoding genes

(srt, srtB, srtC, srtD, srtE and srtF) (Ton-That & Schneewind, 2003). The corynebacterial pili

consist of a major pilus subunit SpaA (SpaD and SpaH, respectively), a minor subunit SpaB

(SpaE and SpaG, respectively) and the tip protein SpaC (SpaF and SpaI, respectively) (Ton-

That & Schneewind, 2003). SrtA specifically catalyzes the covalent crosslinking of single pilin

monomers, whereas the only housekeeping sortase SrtF is necessary for efficient anchoring of

pili to the cell wall.

Mandlik and co-workers showed (2007) that only the SpaA-type pili contribute to

corynebacterial adherence to human pharyngeal cells (Detroit 562), whereas the SpaD and

SpaH-type pili support adhesion to laryngeal (HEp-2) and lung cells (A549). Mutant strains that

lacked all sortases showed decreased adhesion rates to different epithelial cell lines. In

contrast, the deletion of srtA alone impaired binding to pharyngeal cells (Mandlik et al, 2007).

In addition, strains that express only the SpaA-type pili were able to adhere to pharyngeal cells

properly, but showed deceased adhesion to any other cell line compared to the wild type.

Furthermore, it was shown that the minor pilin subunits do not only decorate the pilus shaft but

are also distributed in clusters all over the bacterial surface. The minor pilins that are directly

anchored to the cell wall are sufficient to mediate specific host cell adhesion, which is in

accordance to data reported on streptococci (Dramsi et al, 2006), indicating that this

observation is a conserved feature in Gram-positive pathogens. The deletion of either spaB or

spaC resulted in significantly lower adhesion rates, and the spaBC double mutant revealed

marginal binding to Detroit 562 cells.

When Ott and co-workers investigated the surface structures of the different wild type

strains by atomic force microscopy, this approach revealed significant differences between

various isolates. Interestingly, not all strains studied by Ott and co-workers showed pili-like

structures on the surface and, on top of that, the pili-like structures observed by atomic force

microscopy differed in length and number (Ott et al, 2010b). Some strains lacked pili

completely, while others revealed short, spike-like pili or long, hair-like protrusions, which had

different biophysical properties (Rheinlaender et al, 2012). Furthermore, adhesion and pili

8
Introduction

formation were not strictly coupled, as strain ISS4060, which lacked pili-like structures showed

adhesion rates comparable to those determined for strain ISS3319, which revealed spike-like

pili (Ott et al, 2010b). In accordance with the pili repertoire observed, RNA hybridization

experiments revealed distinct differences in the expression pattern of pili subunits for the

strains investigated. All detected pili subunits belonged to cluster spaABC or spaGHI. The lack

of PCR products of cluster spaDEF and the absence of SpaD signal in Western blotting

experiments suggested that the spaDEF cluster is absent in these strains and, furthermore,

that the genetic repertoire of C. diphtheriae is rather variable. This idea was further supported

by a recent genome study, in which 12 different C. diphtheriae genome sequences were

analysed. At least two pilus gene clusters in each of the sequenced genomes, with C.

diphtheriae HC04 harboring four pilus gene clusters, were found. Three strains lacked cluster

spaGHI and in one strain cluster spaABC was absent (Trost et al, 2012).

Besides different pili formation, Ott and co-workers observed that investigated C.

diphtheriae isolates differed significantly in their interaction with eukaryotic cell lines. While

around 0.3 to 7% of the inoculated bacteria adhere to the human pharyngeal carcinoma cell

line Detroit 562, up to 10 fold higher adhesion rates were observed for C. diphtheriae to HeLa

cells (Ott et al, 2012b), indicating that adhesion efficiency is not only strain-specific but also

cell line-specific. Interestingly, the toxigenic strain studied, DSM43989, showed always the

lowest adhesion rates irrespective of the host cell line. Furthermore, scanning electron

analysis of HeLa cells infected with dead toxigenic bacteria showed significantly more bacteria

attached to HeLa cells than after infection of HeLa cells with vital toxigenic bacteria. This leads

to the assumption that the toxin might have a detrimental effect on the cells, which itself

impairs pathogen-host interaction. In summary, the reasons for strain-specific differences in

adhesion rates besides pili remain unclear and the characterization of bacterial surface

molecules is necessary for better understanding of the adhesion and infection process.

Interestingly, C. diphtheriae strains display two distinct adherence patterns on HEp-2

cell monolayers, termed localized (LA) and diffuse (DA). Localized adherence is characterized

by small clusters of bacteria, so-called microcolonies, while diffuse adherence is expressed in

9
Introduction

random distribution over the surface of the cells. This is further indicating that more than one

adhesion mechanism is existing.

One important adhesin of C. diphtheriae, which was already described, is the non-

fimbrial surface protein 67-72p (Colombo et al, 2001) that is encoded by a single gene

(dip0733) (Sabbadini et al, 2012). The 67-72p protein (DIP0733) enables C. diphtheriae to

recognize and bind specifically to human erythrocyte receptors, a process named

hemagglutination, which capacitates C. diphtheriae to spread throughout the body via the

blood. However, 67-72p protein is not only able to bind human erythrocytes but also HEp-2

cells (Moreira Lde et al, 2003), and binding to this epithelial cell line was effectively blocked by

anti-67-72p IgG antibodies (Hirata et al, 2004). Interestingly, a negative correlation between

bacterial adhesion properties to HEp-2 cells and erythrocytes of the investigated strains was

found: strains with low hemagglutinating activity showed high adhesion rates to HEp-2 cells,

and vice versa.

Besides proteins as adhesion factors, C. diphtheriae exhibits a 10 kDa lipoglycan on its

surface, termed CdiLAM. The basic structure of CdiLAM shows similarity to mycobacterial

LAM. Unlike the non-fimbrial adhesin 67-72p, CdiLAM does not bind to human erythrocytes,

but it binds HEp-2 cells, and pre-incubation of bacterial cells with anti-CdiLAM IgG antibodies

blocked the adherence of C. diphtheriae strain 241 to HEp-2 cells effectively (Moreira et al,

2008). Additionally, purified CdiLAM was able to inhibit the adherence of C. diphtheriae to

HEp-2 cells. Thus, the CdiLAM functioning as an adhesin to human respiratory cells can be

regarded as a potential virulence factor of C. diphtheriae.

While in the upper examples the pathogen factor is clear, this is not the case for some

other described interactions. One example is fibrinogen: fibrinogen is a major protein in human

plasma and it is predominantly involved in the coagulation cascade through its conversion into

insoluble fibrin. Inflammatory or systemic infections lead to upregulation of fibrinogen

synthesis, indicating its function in pathogen control (Sabbadini et al, 2010). The main role of

fibrinogen is in blood clot formation through its conversion into insoluble fibrin which forms the

matrix of the clot. The conversion of fibrinogen to fibrin is catalyzed by a bacterial enzyme

10
Introduction

termed coagulase that reacts with a cofactor found in blood plasma, and it is produced in the

most virulent Staphylococcus strains (Duthie, 1954) and Yersinia pestis (Beesley et al, 1967;

Rivera et al, 2007). C. diphtheriae is also able to bind to fibrinogen and convert fibrinogen to

fibrin (Sabbadini et al, 2012). Non-toxigenic and toxigenic strains formed bacterial aggregates

in the presence of plasma and the fibrinogen binding occurred in varying intensities. The

fibrinogen polymerisation process seems also to build a fibrin layer on the bacterial cell wall,

and the binding of fibrin to the surface of C. diphtheriae may be an efficient method to avoid

phagocytosis.

Bacterial pathogens do not only decorate their surface with fibrin to hide from host

immune system, but they also use sugar moieties as camouflage and sialidases or

neuramidases play a role in virulence of many pathogenic bacteria. These enzymes belong to

a class of glycosyl hydrolases that catalyze the removal of terminal sialic acid residues from a

variety of glycoconjugates of the host surface (Vimr, 1992). C. diphtheriae exposes sialic acids

on its surface but, to date, no genes involved in sialic acid decoration or metabolism have

been detected. By a search of the GenBank sequence database a putative exo-sialidase

NanH (DIP0543) from C. diphtheriae NCTC 13129 was identified. NanH is able to hydrolyze

2´-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid (MU-Neu5Ac) as a substrate (Kim et al,

2010). Furthermore, C. diphtheriae NanH showed trans-sialylation activity using sialyl-α2,3-

lactose and sialyl-α2,6-lactose as donors. Additionally, it could catalyze the transfer of sialic

acis from other sialoconjugates to MU-Gal and the sialidase-mediated transglycosilation

reaction using sialic acid-conjugated free oligosaccharides and glycoproteins as donors.

Accordingly, C. diphtheriae may transport sialic acids hydrolysed by extracellular sialidases for

synthesis of sialic acid derivates. But it is still to be clarified wether NanH is involved in sialic

acid decoration or not (Kim et al, 2010). Moreover, no host cell molecules involved in

corynebacterial adherence were identified so far.

11
Introduction

1.2.2 Factors involved in the invasion process of C. diphtheriae

C. diphtheriae was generally considered as an extracellular colonizer, with the diphtheria toxin

as the main virulence factor. The upper respiratory tract is the primary site of colonization by

C. diphtheriae, and penicillin treatment failure of severe pharyngitis/tonsillitis could only

partially be explained by penicillin tolerance (von Hunolstein et al, 2002). The occurrence of

diphtheria among vaccinated adults in Rio de Janeiro, as well as an increasing number of

cases of endocarditis caused by non-toxigenic strains, indicates the possibility of other

virulence strategies besides the toxin (Mattos-Guaraldi et al, 2000; Mattos-Guaraldi et al,

2001) and gives rise to the idea that the bacteria are not only able to adhere to host cells of

the respiratory tract, but are also able to gain access into deeper tissues.

Hirata and co-workers demonstrated invasion of HEp-2 cells at varying levels with a

range from ~0.2% to 2.6% for different isolates. Interestingly, there was no correlation

between adhesion and invasion rates of the strains, and the internalization process of the

different strains investigated was completely blocked by cytochalasin E, supporting the idea

that cytoskeletal rearrangements are required for bacterial entry. Furthermore, Hirata and co-

workers showed that actin polymerization, along with a phosphotyrosine signaling event,

seems to be a relevant mechanism during the invasion process in cultured respiratory cells

(Hirata et al, 2008).

Additionally, C. diphtheriae was examined as to its ability to adhere to and enter into

the pharyngeal carcinoma cell line Detroit 562 for better understanding of the kinetics of entry

of non-toxigenic isolates into epithelial cells (Bertuccini et al, 2004). The invasion efficiency of

C. diphtheriae in HEp-2 cells was compared with that observed in Detroit 562 cells by using

gentamicin protection assays. Interestingly, at start of infection, there were significantly more

viable bacteria associated with HEp-2 cells than with Detroit 562 cells. After 48 h, there were

still viable bacteria in Detroit 562 cells but not in HEp-2 cells. In conclusion, C. diphtheriae

shows lower adhesion rates to and invasion rates into Detroit 562 cells compared to HEp-2

cells but it is able to survive significantly longer within Detroit 562 cells. This might indicate

better adaption of C. diphtheriae to its target cell. Cytochalasin D showed a dose-dependent

12
Introduction

inhibitory effect on the internalization process, pointing to the involvement of F-actin in the

bacterial entry. The bacterial binding seems to induce intracellular signal transduction

responsible for actin microfilament rearrangements (Rosenshine et al, 1992). Staurosporine,

an inhibitor of different classes of protein kinases, seemed to contribute to C. diphtheriae

internalization, indicating that the activation of protein kinases negatively controlled bacterial

uptake. However, the possibility to enter the host cell, to hide in epithelial cells, even at low

numbers, and to persist at least 48 h, gives the opportunity to escape from the hosts immune

response and antibiotic treatment.

Investigations of the invasiveness of six non-toxigenic strains as well as the tox+ strain

DSM43989 by Ott and co-workers were carried out by gentamicin protection assays. By this

method, Ott and co-workers demonstrated that the toxigenic strain DSM43989 has the lowest

invasion rate at 2 h post infection. Interestingly, the toxigenic strain showed, in general, lower

adhesion and invasion rates compared to non-toxigenic strains. As in adhesion assays, these

strains differed significantly in invasion rates, ranging from ~0.02% to 0.06% (Ott et al, 2010b).

As in the case of Hirata and co-workers (Hirata et al, 2002), the comparison of these strains in

respect to adhesion and internalization rates revealed that these processes were not strictly

coupled. Additionally, Ott and co-workers observed V-shaped C. diphtheriae dimers within the

host cell by immune-fluorescence microscopy indicating that C. diphtheriae is able to replicate

within the host cells and growth and elimination are parallel processes (Ott et al, 2010b). While

host cell receptors and invasion-associated proteins of the pathogen are still unknown,

bacterial virulence factors beside the toxin have been at least partially characterized on the

molecular level, as described before. In contrast, the molecular mechanism of invasion is more

or less unclear.

Consequently, Ott and co-workers focused their work on the surface-associated protein

DIP1281, a member of the NlpC/P60 family (Anantharaman & Aravind, 2003), which was

annotated as hypothetical invasion-associated protein, and was found as part of the C.

diphtheriae secretome (Hansmeier et al, 2006a). Two non-toxigenic wild type strains as well

as the corresponding DIP1281 mutant strains were tested regarding adhesion to and

13
Introduction

internalization into Detroit 562 cells (Ott et al, 2010a). In these experiments, strain ISS3319

showed higher adhesion rates to Detroit 562 cells, compared to strain ISS4060, while no

significant differences of the number of intracellular bacteria were observed. DIP1281 mutant

strains lacked the ability to adhere to host cells almost completely and, in contrast to the

respective parental strains, no intracellular bacteria were detectable by using gentamicin

protection assays. Further characterization of the mutant strains revealed an increased size of

the single bacteria, an altered less club-like shape and formation of chains of bacteria rather

than the typical V-like cell division forms or palisades of clustered C. diphtheriae. Additionally,

the mutants showed an uneven speckled immuno-fluorescence staining, indicating an altered

surface structure compared to the wild type strains, which was confirmed by SDS-PAGE,

Western blot and 2D-PAGE of the surface proteomes. These clear differences between wild

type and mutant surface protein patterns were also found by using atomic force microscopy.

The wild type strains showed round elevations with defined structures, while the DIP1281

mutant strains lost this fine structure. The results of Ott and co-workers indicate that DIP1281

is predominantly involved in the organization of the outer protein layer of C. diphtheriae rather

than in the separation of the peptidoglycan of dividing bacteria. These rearrangements of the

outer membrane lead to the adhesion and invasion-negative phenotype of the corresponding

mutant strains (Ott et al, 2010a). Tsuge and co-workers reported similar observations in C.

glutamicum R, when the DIP1281 homolog cgR_1596 and other members of the NlpC/P60

protein familiy cgR_2070 were mutated (Tsuge et al, 2008).

Recent studies of Sabbadini and co-workers demonstrated the involvement of the

surface-exposed non-fimbrial 67-72p protein (DIP0733), previously described as

adhesin/hemagglutinin (Colombo et al, 2001; Hirata et al, 2004), in the internalization of C.

diphtheriae into HEp-2 cells (Sabbadini et al, 2012). Furthermore, the intracellular viability of

two different C. diphtheriae strains was significantly reduced after pretreatment of bacterial

cells with 67-72p antibodies, while incubation with 67-72p of HEp-2 cells or bacterial

suspensions also revealed decreased internalization rates. However, since the internalization

process was not completely blocked due to treatments with 67-72p antibodies, the

14
Introduction

involvement of other bacterial factors in this process cannot be excluded (Sabbadini et al,

2012).

The identification of new, up to now unknown virulence factors might be fostered in

future, not only by the availability of various C. diphtheriae gene sequences (Cerdeno-Tarraga

et al, 2003; Sangal et al, 2012a; Sangal et al, 2012b; Trost et al, 2012), but also by the

establishment of easy to handle invertebrate infection model systems. The nematode

Caenorhabditis elegans and larvae of the greater wax moth Galleria mellonella were found to

be suitable in this respect (Ott et al, 2012a).

1.2.3 C. diphtheriae and host immune response

The innate immune system provides the first line of defense against infectious diseases;

anatomic barriers such as skin and optimized phagocytic cells (macrophages and neutrophils)

play a main role in innate immunity, together with a large battery of antimicrobial compounds

synthesized by the host cell. Thereby, most of the microorganisms infecting healthy

vertebrates are readily cleared within a few days after infection, before activating the adaptive

immune response. Invasion of microorganisms into vertebrate cells is initially recognized by

the innate immune system through a limited number of germline-encoded pattern-recognition

receptors (PRRs) (Akira et al, 2006), including the Toll-like receptors (TLRs) with the first

described member Toll. Members of the TLR family are able to recognize Gram-negative cell

wall components, such as lipopolysaccharide (LPS). Gram-positive bacterial cell walls do not

contain LPS but LTA (lipoteichoic acids), or in case of C. diphtheriae mycolic acids that might

also stimulate the innate immune system. The activation of intracellular signaling pathways

leads to the transcription of genes involved in antimicrobial host defense mechanisms. This

was first investigated for C. diphtheriae in respect to septic arthritis. The clinical pattern of

human diphtheroid arthritis can occur as an acute, purulent process or as a chronic arthritis

with plasmacytic or histiocytic inflammation. Furthermore, patients are in general bad condition

or have a preexisting disease that impairs the immune system (Tiley et al, 1993; Patey et al,

1997; Barakett et al, 1993; Guran et al, 1979). Interestingly, non-toxigenic C. diphtheriae

15
Introduction

strains are able to induce lethal infections in mice by intravenous inoculation of CD-1 mice in a

dose-dependent and strain-specific manner (Puliti et al, 2006). Clinical signs of arthritis

occurred 3 days post infection, most frequently in ankles and wrists. By quantitative monitoring

of C. diphtheriae, Puliti and co-workers also demonstrated that C. diphtheriae bacteria were

cleared from the bloodstream within 5 days after infection, but the bacteria are able to persist

in kidneys and spleens for more than two weeks after infection. Additionally, Puliti and co-

workers brought the evidence that the investigated non-toxigenic strains induce systemic and

local IL-6, IL-1 and TNF secretion, which might contribute to articular damage in cases of

septic arthritis. Interestingly, induction of high and early IFN- production was only observed in

mice infected with ISS3319, indicating that IFN- production counterbalances the

proinflammatory response.

Induction of host response was investigated in vitro on a more molecular level by Ott

and co-workers, using luciferase expressing reporter cell lines, monitoring the induction of

various host signaling pathways (Table 1). Furthermore, Ott and co-workers demonstrated that

different non-toxigenic C. diphtheriae wild   type   strains   are   able   to   induce   the   NFĸB  

transduction pathway in epithelial cells in a strain-specific manner; induction only occurs

during incubation with living bacteria and requires invasion of the bacteria into the epithelial

cells (Ott et al, 2012b).

16
Introduction

Table 1: Induction of eukaryotic inflammatory pathways. Infection of epithelial cells


lentiviral transfected with luciferase-coupled reporter systems specific for inflammatory
pathways with different C. diphtheriae wild type strains indicated by - (no signal) and plus
(signal), with number of +s indicating signal strength; n.t. not tested.
HeLa Detroit 562

NFB STAT5RE AP-1 NFAT SIE NFB AP-1

ISS3319 ++ ++ ++ ++ - ++ -

ISS4060 +++ ++ ++ ++ - - -

ISS4746 + - - - n.t. ++ +

ISS4749 + - - - n.t. - -

DSM33988 ++ + - +++ n.t. - +

DSM43989 - - n.t. +++ n.t. - -

DSM44123 + (+) n.t. + n.t. - -

(Ott et al, 2012b and unpublished observation)

1.2.4 Phagocytosis and induction of apoptosis

Non-opsonic phagocytosis is mediated by phagocyte receptors that recognize structures on

microbial surfaces. There are three mechanisms of induction of non-opsonic phagocytosis

discussed: carbohydrate-protein interactions, protein-protein interactions and hydrophobic

interactions (Ofek et al, 1995). It is known that the diphtheria toxin can fully activate human

macrophages and phagocytosis was significantly impaired after pro-longed incubation with

diphtheria toxin (Saelinger et al, 1975; D'Onofrio & Paradisi, 1983). Dos Santos and co-

workers investigated the ability of homologous toxigenic and non-toxigenic C. diphtheriae

strains to be phagocytosed by the human macrophage cell line U937 in the absence of

immune serum (opsonins) and found that human macrophages can act differently in the way

of killing toxigenic and non-toxigenic C. diphtheriae strain. The presence of diphtheria toxin

seems to enhance adhesion to as well as internalization into U937 cells and, therefore, favors

non-opsonic phagocytosis (dos Santos et al, 2010). Interestingly, the non-toxigenic strain

adhered weakly to U937 cells, and only a very low number of bacteria were internalized,

indicating that C. diphtheriae might be able to escape non-opsonic phagocytosis by human

17
Introduction

macrophages. In conclusion, there must be other receptors responsible for binding and

internalization of the tox+ strain than those responsible for binding of diphtheria toxin. Non-

opsonic phagocytosis of both strains toxigenic and non-toxigenic induced cytopathogenicity in

U937 cells; more than 42% of U937 cells were killed during infection with the tox+ strain (3h

post infection), while about 18% were killed during infection with the non-toxigenic strain.

Furthermore, C. diphtheriae seems to induce apoptosis and necrosis in human macrophages,

irrespective of the presence of diphtheria toxin. The toxigenic strain caused higher levels of

DNA fragmentation in U937 macrophages, but also the tox- strain was also able to induce

apoptosis and necrosis.

The 67-72p protein is not only able to interact with macrophages but also with epithelial

cells. This protein seems to be involved in the induction of apoptosis and necrosis in non-

phagocytic epithelial cells. The incubation of HEp-2 cells with 67-72p adsorbed microspheres

and homologous C. diphtheriae strain CDC-E8392 obtained cytoskeletal changes with

accumulations of polymerized actin in HEp-2 cells (Sabbadini et al, 2012). These results

indicate that the 67-72p protein (DIP0733) is as a bacterial factor involved in the mechanism of

recruitment of host actin filaments. Vacuolization and DNA fragmentation was observed after

12 h, and a high occurrence of apoptotic bodies appeared after 24h post treatment with 67-

72p. DNA condensation and aggregation were observed by DAPI staining of 67-72p treated

cells. Furthermore, the incubation of HEp-2 cells with 67-72p caused a reduction in cell

volume, which is a ubiquitous aspect of apoptosis, termed apoptotic volume decrease (Bortner

& Cidlowski, 2002). The ability of C. diphtheriae to kill non-phagocytic cells might be beneficial

for its survival and invasion of deeper tissue and thus to spread over the whole body. In

addition, the induction of apoptosis and necrosis during C. diphtheriae infection is not directly

coupled to diphtheria toxin production. The fact that human macrophages are able to

internalize C. diphtheriae bacilli in the absence of immune serum indicates that there is an

antibody-independent mechanism as resistance to infection. On the other hand, the

observation that the non-toxigenic strain was able to survive longer periods within the

18
Introduction

macrophages might indicate that human macrophages may be not effective in killing C.

diphtheriae in the absence of opsonins (dos Santos et al, 2010).

Potassium transporters seem to be involved in one of these resistance mechanisms.

When Ochrombel and co-workers investigated potassium uptake systems in corynebacteria

(Ochrombel et al, 2011), they found that pathogenic corynebacteria such as C. diphtheriae and

C. jeikeium exhibit more potassium transporters than non-pathogens, such as C. glutamicum.

Furthermore, C. glutamicum K+ transporter mutants were eliminated faster by macrophages

and less stress resistant. This is also true for C. diphtheriae K+ uptake mutants (Ott,

unpublished), which turn K+ uptake systems of pathogenic corynebacteria into potential

virulence factors with regard to survival in an acid environment, like the inside of

macrophages. Figure 4 gives an overview about all known corynebacterial K+ transporter

systems.

19
Introduction

Fig. 4: Different types of potassium transporters in Actinobacteria. Proteins similar to


known potassium transporter subunits in Escherichia coli (Ecoli, Kch, KefB, TrkH, KdpA) or
Streptomyces coelicolor (Scoel, KcsA) were identified by genomicBlast searches against the
genomes of Corynebacterium aurimucosum (Cauri), Corynebacterium diphtheriae biotype
gravis NCTC13129 (Cdiph), Corynebacterium efficiens YS-314 (Ceffi), Corynebacterium
glutamicum ATCC 13032 (Cglut), Corynebacterium glutamicum R (CglutR), Corynebacterium
jeikeium K411 (Cjeik), Corynebacterium urealyticum DSM7109 (Curea), Corynebacterium
kroppenstedtii DSM44385 (Ckrop), Mycobacterium tuberculosis H37RV (Mtube),
Mycobacterium abscessus ATCC 19977 (Mabsc), Nocardia farcinica IFM 10152 (Nfarc),
Propionibacterium acnes KPA171202 (Pacne), Streptomyces avermitilis MA-4680 (Saver), S.
coelicolor A3(2) M145 (Scoel), Bifidobacterium longum NCC2705 (Blong) and Clavibacter
michiganensis subsp. michiganensis NCPPB 382 (Cmich) (Ochrombel et al, 2011).

20
Introduction

In summary, the interaction of the Gram-positive pathogen C. diphtheriae with its host is much

more complex than initially expected. Up to now, several different interaction mechanisms

were elucidated (summarized in Figure 5), but there is still room for research, especially

regarding interactions at the molecular level. Based on the already described results and

ongoing research, C. diphtheriae might work as a model for other pathogenic, but molecular

biologically less addressable corynebacteria such as C. jeikeium.

macrophage

erythrocytes
phagocytosis

non-opsonic
phagocytosis
agglutination
hemagglutination

Fbn
fibrin fibrin
receptor
neuraminidase

67-72p adhesin / invasin


Corynebacterium diphtheriae

trans- AB toxin
SpaB
cdiLAM sialidase production
adhesin
SpaA
∆  DIP1281 sialidase
receptor
SpaC
adhesion toxin
SpaB receptor
receptor
SpaC
receptor signaling cascade

endocytosis

translocation
activation of inflammatory pathways
NFĸB,  NFAT,  AP-1, STAT5RE
protein synthesis

epithelial cell

21
Introduction

Fig. 5. Infection properties of C. diphtheriae. The most prominent virulence factor of C.


diphtheriae is   the   diphtheria   toxin,   which   is   encoded   by   a   β-corynephage integrated in the
bacterial chromosome. This AB toxin is released by the bacterial cell and binds the toxin
receptor (HB-EGF) on the host´s surface. The toxin gets internalized by endocytosis, which
leads to translocation of the A-fragment and inhibition of the protein biosynthesis by binding of
the elongation factor 2. Best investigated adhesion factors of C. diphtheriae might be the pili
structures. Strain NCTC 13129 exhibits three different pilus types, the SpaABC type pilus is
depicted here exemplarily. SpaA forms the pilus shaft, SpaB is linked to SpaA via
transpeptidation and SpaC forms the tip of the pilus. SpaB and SpaC are able to bind
receptors on the surface of the host cell, which enables colonization of the cell. Besides pili
formation the non-fimbrial protein 67-72p (DIP0733) might be an important adhesin. Via this
protein C. diphtheriae is able to bind erythrocytes, a process called hemagglutination, which
facilitates C. diphtheriae to disseminate throughout the body via the blood. Furthermore, the
67-72p protein seems to be involved in the internalization process and might contribute to
apoptosis and necrosis of the host cell. Neuraminidase-secreting strains were also found to be
capable of agglutinating human erythrocytes with high titers. Neuraminidases or sialidases
belong to a class of glycosyl hydrolases that catalyze the removal of terminal sialic acid
residues from a variety of glycoconjugates of the host surface. The sialidase NanH is able to
hydrolyze 2´-(4-methylumbelliferyl)-α-D-N-acetylneuraminic acid as a substrate and showed
trans-sialylation activity using sialyl-α2,3-lactose and sialyl-α2,6-lactose as donors. But it is still
to be clarified wether NanH is involved in sialic acids decoration or not. Besides proteins as
adhesion factors C. diphtheriae exhibits a 10 kDa lipoglycan on its surface, the CdiLAM, which
binds HEp-2 cells and can be regarded as a potential virulence factor of C. diphtheriae.
Additionally, C. diphtheriae is able to bind to fibrinogen and convert fibrinogen to fibrin,
resulting in a fibrin layer on the bacterial surface that may be an efficient method to avoid
phagocytosis. Since C. diphtheriae was expected to be an extracellular colonizer it is very
important to know that C. diphtheriae is able to internalize into non-phagocytic cells in a strain-
specific manner. The invasion process seems to involve actin polymerization along with a
phosphotyrosine signaling event in cultured respiratory cells. Investigations of DIP1281
(annotated as invasion-associated protein) revealed the involvement in the organization of the
outer protein layer, which resulted in an adhesion- and invasion-negative phenotype of the
corresponding mutant strains. Furthermore, infection of epithelial cells leads to inflammatory
host  response,  especially  the  activation  of  the  NFĸB  transduction pathway. NFĸB  induction  is  
strictly dependent on intracellular bacteria, since bacterial adhesion was not sufficient to
activate inflammatory pathways. In contrast to the bacterial property to enter epithelial cells,
U937 human macrophages are able to ingest C. diphtheriae by non-opsonic phagocytosis. C.
diphtheriae is able to persist inside macrophages and to induce apoptosis and necrosis in the
cell.

22
Aim of the work

2 Aim of the work


C. diphtheriae was generally considered as an extracellular pathogen colonizing the mucosal

surfaces on tonsils, larynx and pharynx, with the diphtheria toxin as the main virulence factor.

The fact that non-toxigenic C. diphtheriae strains can cause invasive and systemic infections

such as endocarditis, splenic abcesses, mycotic aneurysm, osteomyelitis, pneumonia and

septic arthritis (Isaac-Renton et al, 1981; Hirata et al, 2008; Puliti et al, 2006; Honma et al,

2009; Muttaiyah et al, 2011), indicates that there must be other virulence factors besides the

toxin. With the development of high throughput sequencing techniques, to date, numerous

genome sequences of pathogenic corynebacteria are available including various C.

diphtheriae strains (Sangal et al, 2012a; Sangal et al, 2012b; Trost et al, 2012). This gives new

opportunities of global analysis techniques in respect to genomics and proteomics and

background for experiments at the molecular level.

The aim of this study was to elucidate the interaction of C. diphtheriae with host cells with

regard to adhesion, invasion, pili formation and host cell response. To address this, infection

assays with eukaryotic cell lines were carried out, as well as insertions mutagenesis of

putative virulence factors, proteomics, immuno-fluorescence microscopy, scanning electron

microscopy and atomic force microscopy.

23
Publications

3 Publications
3.1 Corynebacterium diphtheriae invasion-associated protein
(DIP1281) is involved in cell surface organization, adhesion and
internalization in epithelial cells (Ott et al, 2010a)

24
Publications
Ott et al. BMC Microbiology 2010, 10:2
http://www.biomedcentral.com/1471-2180/10/2

RESEARCH ARTICLE Open Access

Corynebacterium diphtheriae invasion-associated


protein (DIP1281) is involved in cell surface
organization, adhesion and internalization
in epithelial cells
Lisa Ott1, Martina Höller1, Roman G Gerlach2,4, Michael Hensel2,5, Johannes Rheinlaender3, Tilman E Schäffer3,
Andreas Burkovski1*

Abstract
Background: Corynebacterium diphtheriae, the causative agent of diphtheria, is well-investigated in respect to toxin
production, while little is known about C. diphtheriae factors crucial for colonization of the host. In this study, we
investigated the function of surface-associated protein DIP1281, previously annotated as hypothetical invasion-
associated protein.
Results: Microscopic inspection of DIP1281 mutant strains revealed an increased size of the single cells in
combination with an altered less club-like shape and formation of chains of cells rather than the typical V-like
division forms or palisades of growing C. diphtheriae cells. Cell viability was not impaired. Immuno-fluorescence
microscopy, SDS-PAGE and 2-D PAGE of surface proteins revealed clear differences of wild-type and mutant protein
patterns, which were verified by atomic force microscopy. DIP1281 mutant cells were not only altered in shape
and surface structure but completely lack the ability to adhere to host cells and consequently invade these.
Conclusions: Our data indicate that DIP1281 is predominantly involved in the organization of the outer surface
protein layer rather than in the separation of the peptidoglycan cell wall of dividing bacteria. The adhesion- and
invasion-negative phenotype of corresponding mutant strains is an effect of rearrangements of the outer surface.

Background diphtheria, but can cause other infections. Non-toxi-


Corynebacterium diphtheriae is the causative agent of genic strains have been increasingly documented [4-6]
diphtheria, a toxaemic localized infection of the respira- and found to be the cause of invasive diseases such as
tory tract. By vaccination diphtheria is well-controlled in endocarditis, bacteraemia, pneumonia, osteomyelitis,
e. g. Western Europe [1-3]; however, this disease is still spleen abscesses, and septic arthritis ([7] and references
a cause of morbidity and mortality in less developed therein). These systemic infections caused by C.
countries. While the production of diphtheria toxin has diphtheriae suggest that this pathogen is not only able
been well-established as a major virulence factor, little is to attach to host epithelial cells, but must be able to
known about C. diphtheriae factors crucial for coloniza- gain access to deeper tissues by unknown portals of
tion of the host and corresponding host receptors recog- entry and to persist in these tissues.
nized by these factors, although colonization is an A possible clue for the background of persistence of
essential step of pathogenicity. C. diphtheriae came from investigations of adherence
In the last decades it has become evident that and invasion of toxigenic and non-toxigenic strains.
C. diphtheriae is not only the aetiological agent of Using a combination of gentamicin protection assays
and thin-section electron microscopy, Hirata and co-
workers [8] showed that toxigenic C. diphtheriae were
* Correspondence: aburkov@biologie.uni-erlangen.de
1
Friedrich-Alexander-Universität Erlangen-Nürnberg, Lehrstuhl für not only able to adhere to laryngeal HEp-2 cells, but
Mikrobiologie, Staudtstr. 5, 91058 Erlangen, Germany also enter these cells and survive after internalization.
© 2010 Ott et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons
Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in
any medium, provided the original work is properly cited.

25
Publications
Ott et al. BMC Microbiology 2010, 10:2 Page 2 of 9
http://www.biomedcentral.com/1471-2180/10/2

Similar observations were made for non-toxigenic of the NlpC/P60 family [17]. NlpC/P60 proteins define a
strains [9] showing that also pharyngeal Detroit large superfamily of several diverse groups of proteins
562 cells can be invaded by C. diphtheriae. In this study, including putative proteases and probably invasion-asso-
living intracellular bacteria were detected up to 48 h ciated proteins. They are found in bacteria, bacterio-
after infection. phages, RNA viruses, and eukaryotes and various
While host cell receptors and invasion-associated pro- members are highly conserved among non-pathogenic
teins of the pathogen are still unknown, bacterial adhe- and pathogenic corynebacteria [18]. C. diphtheriae pro-
sion factors have been recently at least partially tein DIP1281 was, as its homologs Ce1659, Cg1735, and
characterized on the molecular level. C. diphtheriae is JK0967 in Corynebacterium efficiens, Corynebacterium
able to assemble three distinct pili on its surface. glutamicum, and Corynebacterium jeikeium, previously
Mutant analyses showed that the SpaA-type pilus is suf- annotated as hypothetical invasion-associated protein
ficient for adhesion to pharynx cells, shaft proteins are and was therefore in the focus of this study.
not crucial for pathogen-host interaction, while adher-
ence to pharyngeal cells is greatly diminished when Results
minor pili proteins SpaB and SpaC are lacking [10]. The Adhesion and invasion of C. diphtheriae wild type
results obtained in this study also indicated the exis- and mutant strains
tence of other proteins besides pili subunits involved in As a basis for further analyses of DIP1281 mutants,
adhesion to larynx, pharynx, and lung epithelial cells, strains ISS3319 and ISS4060, which were already shown
since a total loss of attachment to pharyngeal cells due to be adhesion- and invasion-competent [9], were tested
to mutagenesis of pili- and sortase-encoding genes for adhesion to and internalization in Detroit562 (D562)
could not be observed and attachment to lung or larynx cells. Using a slightly modified protocol (compared to
cells was less affected by the mutations. This is in line [9]) with increased number of washing steps, we were
with a number of studies suggesting the multi-factorial able to generate highly reproducible infection conditions
mechanism of adhesion (reviewed in [11]). Furthermore, (Table 1). In these experiments, strain ISS3319 showed
Hirata and co-workers [12] described two distinct pat- a higher number of adherent bacteria compared to
terns of adherence to HEp-2 cells, a localized and a dif- strain ISS4060 (corresponding to adhesion rates of
fuse form, an observation that hint also to the existence 2.66 ± 0.12% for ISS3319 and 2.16 ± 0.29% for ISS4060),
of several adhesion factors. This idea is in accordance while statistically relevant differences of the number of
with the situation in other bacteria such as Salmonella invaded epithelial cells were not observed (Table 1).
enterica where a high number of different factors are After establishing infection conditions for the wild-
crucial for pathogenesis [13]. The involvement of differ- type strains, dip1281 gene disruption mutants Lilo1
ent C. diphtheriae proteins to adherence to distinct cell (ISS3319::pK18 mob’dip1281’’) and Lilo2 (ISS4060::pK18
types is further supported by work on adhesion to mob’dip1281’’) were analyzed. DIP1281 mutant strains
human erythrocytes, showing that non-fimbrial surface lacked the ability to adhere to host cells almost comple-
proteins 67p and 72p, which were up to now only char- tely (with adhesion rates of 0.03 ± 0.01% for Lilo1 and
acterized by their mass, are involved in this process [14]. 0.04 ± 0.01% for Lilo2) and in contrast to the wild-type
Interestingly, besides strain-specific differences in adher- no internalized bacteria were detectable for strain Lilo1
ences (see references cited above), also growth-depen- and Lilo2 (Table 1).
dent effects were observed. In a study using two
toxigenic C. diphtheriae strains and erythrocytes as well Table 1 Adhesion of C. diphtheriae to epithelial cells and
as HEp-2 cells, de Oliveira Moreira and co-workers [15] internalization. D562 cells (2 × 105 cells per well) were
showed an effect of iron supply on hemagglutination infected with C. diphtheriae (4 × 107 cfu/ml) leading to a
and lectin binding properties of the microorganisms. multiplicity of infection (MOI) of 200.
Also in this study, strain-specific differences in adher- Strain Viable bacteria (CFU/ml)a
ence were detected. adherentb internalizedc
While pathogen factors responsible for adhesion are at ISS3319 5
10.1 × 10 ± 1.4 × 10 5
1.6 × 103 ± 1.0 × 102
least partially known, the molecular background of inva- ISS4060 5
3.5 × 10 ± 1.0 × 10 5
3.0 × 103 ± 1.4 × 103
sion is more or less unclear. Since we were interested in Lilo1 2
1.6 × 10 ± 2.1 × 10 2
n. d.
this process, we started a functional genetics approach Lilo2 9.3 ± 10.6 n. d.
to identify proteins involved in invasion, based on a a
values represent the means and standard deviations of three separate
recently published work presenting a comprehensive experiments
analysis of proteins secreted by C. diphtheriae [16]. In b
c
average number of bacteria recovered on agar plates after 1.5 h of infection
average number of bacteria recovered on agar plates after 1.5 h of infection
this study, we focused on one of these identified pro- and further 2 h of treatment with gentamicin
teins, the surface-associated protein DIP1281, a member n. d.: not detectable

26
Publications
Ott et al. BMC Microbiology 2010, 10:2 Page 3 of 9
http://www.biomedcentral.com/1471-2180/10/2

Basic characterization of DIP1281 mutant strains staining, we could show that disruption of chains by vig-
Microscopic inspection of the mutant strains revealed orous vortexing (5 min) was not detrimental to the bac-
unexpectedly an increased size of the single bacteria in teria (Figure 2C and 2F), indicating that mutant strains
combination with an altered less club-like shape and have a fully functional and rigid peptidoglycan layer.
formation of chains of bacteria rather than the typical For all strains, ISS3319, ISS4060, Lilo1 and Lilo2,
V-like division forms or palisades of clustered identical doubling times of about 70 min were observed.
C. diphtheriae. Immuno-fluorescence microscopy carried Interestingly, with a final optical density (OD 600 ) of
out for control verified that observation (Figure 1). approx. 13, the mutants reached a more than fourfold
Additionally, this approach showed an uneven, speckled higher OD600 compared to the corresponding wild-type
staining of the mutants, indication an altered surface strains, which reached final optical densities between 2.5
structure compared to the wild-type strains. and 3. This observation corresponds nicely with the
To analyse, if all bacteria within the observed chains increased colony size of the mutants (data not shown)
of mutants were still viable or if changes were correlated and suggests that the altered bacterial size and form has
with detrimental effects on survival of bacteria, we car- no severe impact on light scattering and consequently
ried out LIVE/DEAD staining. No significant differences OD measurement.
were observed between wild-type and mutants in respect Analysis of surface proteins
to viability, in all cases the majority of bacteria were Since we assumed that the altered shape of the mutants
fully viable and exclusively stained by SYTO9 green and might be correlated with an altered cell surface, espe-
not by propidium iodide (Figure 2). During manipula- cially in the light of the immuno-fluorescence micro-
tion of bacteria (washing steps, resuspension of pellets), scopy approach (Figure 1), which showed a different
we observed that chains of mutants were occasionally antibody binding compared to the wild-type, we isolated
broken down to smaller units. Using LIVE/DEAD the surface proteins of wild-type and mutant strains.

Figure 1 Immuno-fluorescence microscopy of C. diphtheriae wild-type and mutant strains. An antiserum directed against the surface
proteome of C. diphtheriae was used as primary antibody; Alexa Fluor 488 goat anti-rabbit was used as secondary antibody. A: ISS3319, B: Lilo1,
C: ISS4060, D: Lilo2.

27
Publications
Ott et al. BMC Microbiology 2010, 10:2 Page 4 of 9
http://www.biomedcentral.com/1471-2180/10/2

Figure 2 LIVE/DEAD staining of C. diphtheriae wild-type and mutant strains. Green fluorescent bacteria have a functional cytoplasmic
membrane and are stained green, red propidium iodide staining indicates non-viable cells. A: ISS3319, B-C: Lilo1, D: ISS4060, E-F: Lilo2, C and F:
cells subjected to 5 min of vigorous vortexing.

When these were subjected to SDS-PAGE and silver striking differences were observed when the cell surface
staining, significant differences in protein patterns were of different C. diphtheriae strains was examined. In the
observed (Figure 3A). Compared to the protein bands of wild-type strain ISS3319 (Figure 4A) round elevations
the wild-type strains, DIP1281 mutants showed a with a lateral diameter of 10-40 nm and a height of 1-4
decrease of bands in the upper molecular weight range nm can be seen (Figure 4A, upper row). The complemen-
accompanied by an increased number of bands in the tary phase images, which reflect adhesive and elastic tip-
lower molecular weight range. These changes were con- sample interactions, show a similar, highly structured
firmed, when Western blot experiments were carried surface structure (Figure 4A, lower row). In the mutant
out (Figure 3B), which also showed a dramatic change strain Lilo1 (Figure 4B), a loss of this fine structure was
and decrease of immuno-reactive bands. As a third observed: Elongated elevations can be seen with a width
experimental approach to analyse surface proteins, 2-D of 50-100 nm (Figure 4B, upper row). Their height is
PAGE was carried out (gels for strains ISS3319 and similar as in the case of the wild-type strain. Differences
Lilo1 are shown in Figure 3C; ISS4060 and Lilo2 gave in surface structure are especially obvious when compar-
comparable results, data not shown). As in the SDS- ing the complementary phase images (lower rows in
PAGE experiments, the mutant showed a decrease of Figure 4A and 4B). Analyses of strains ISS4060 and Lilo2
proteins in the upper molecular weight range and an gave similar results (data not shown).
increased number of spots in the lower molecular
weight range. Furthermore, in comparison to the wild- Discussion
type, the mutant showed a dramatic increased number In this study, the function of the surface-associated pro-
of multiple spots. The molecular background of these tein DIP1281, a member of the NlpC/P60 family was
multiple protein forms is unclear. investigated, which was annotated as hypothetical inva-
Surface structure of wild-type and mutant strains sion-associated protein. By fluorescence staining and
The altered immuno-staining of the mutant strain sur- atomic force microscopy, we could show that DIP1281
faces and the clear differences of wild-type and mutant mutations cause formation of chains of bacteria, rear-
protein patterns revealed by SDS-PAGE and 2-D PAGE rangements of cell surface structures, and dramatic
prompted us to perform a more detailed investigation of changes in protein patterns. Our data indicate that
the cell surface of C. diphtheriae by atomic force micro- DIP1281 is not crucial for the separation of the peptido-
scopy. Compared to the surface structure of C. glutami- glycan layer of dividing bacteria, since disruption of
cum, which was investigated for several strains in great chains did not decrease the viability of bacteria. Conse-
detail by atomic force microscopy [19-21], C. diphtheriae quently, DIP1281 function seems to be limited to the
shows a more structured surface (Figure 4). Furthermore, outer protein layer of C. diphtheriae, which is not

28
Publications
Ott et al. BMC Microbiology 2010, 10:2 Page 5 of 9
http://www.biomedcentral.com/1471-2180/10/2

Figure 3 Analysis of surface proteins. Surface proteins were isolated from C. diphtheriae wild-type and mutant strains and subjected to SDS-
PAGE (A), Western blotting (B), and 2-D PAGE (C). For SDS-PAGE 25 μg of protein prepared from strains ISS3319 (lane 2), Lilo1 (lane 3), ISS4060
(lane 4), and Lilo2 (lane 5) were applied per lane on a 10% polyacrylamide gel and silver-stained after electrophoresis. Molecular weight of
marker proteins (lane 1, from top to bottom): 250, 130, 95, 72, 55, 36, 28, 17, 11 kDa. Western blotting was carried out after SDS-PAGE using a
polyclonal antiserum directed against C. diphtheriae DSM44123 surface proteins. For 2-D PAGE surface protein preparations were separated
according to their isoelectric point and molecular mass using a pH range of 3-10 for isoelectric focussing and 12.5% polyacrylamide gels for SDS-
PAGE. Gels were stained with Coomassie Brilliant Blue. Molecular weight of marker proteins (from top to bottom): 150, 120, 100, 85, 70, 60, 50,
40, 30, 25, 20, 15 kDa.

uniformly organized in a surface layer lattice, but com- this study, cell separation was not impaired in respect to
prises more than 50 different proteins [16]. If the other separation of peptidoglycan and mycolic acid layers of
NlpC/P60 family members in C. diphtheriae besides daughter cells, but mainly restricted to the surface pro-
DIP1281, namely DIP0640, DIP1621, and DIP1622 [18] tein layer of the bacteria. However, using transmission
have similar functions in cell surface layer organization electron microscopy of thin sections of cells, in this
is unknown and has to be investigated in future projects. study also formation of multiple septa within single bac-
Tsuge and co-workers reported cell separation defects teria was observed in response to cgR_1596 mutations.
in Corynebacterium glutamicum R, when the DIP1281 Furthermore, growth of mutant strains was examined.
homolog cgR_1596 and another member of the NlpC/ In contrast to the situation in C. diphtheriae, where we
P60 protein family cgR_2070 were mutated [22]. Also in found an unaltered growth rate and a strongly increased

29
Publications
Ott et al. BMC Microbiology 2010, 10:2 Page 6 of 9
http://www.biomedcentral.com/1471-2180/10/2

Figure 4 Ultrastructural analysis of the cell surface of C. diphtheriae strains. (A) ISS3319, (B) Lilo1; red boxes in the low magnification
images on the left hand side mark three areas shown with a higher magnification on the right hand side (upper row: topography/height, lower
row: phase). Colour scale bars at the right hand side give height and phase magnitudes.

biomass formation caused by lack of DIP1281, in of the outer surface of bacteria. Specific interaction part-
C. glutamicum R mutation of cgR_1596 led to a slightly ners for DIP1281 and its homologs in other corynebac-
decreased growth rate and unaltered final optical density teria are unknown and might be the focus of further
of the culture. The exact function of the NlpC/P60 pro- studies to unravel the specific functions and targets of
tein family members in C. glutamicum was also not these proteins on a molecular level.
unravelled until now.
In respect to adhesion and internalization of Methods
C. diphtheriae to epithelial cells, the results obtained in Bacterial strains and growth
this study suggest that DIP1281 is crucial for localiza- Strains used in this study are listed in Table 2. Escheri-
tion and function of adhesion and invasion factors and chia coli DH5aMCR was grown in Luria Bertani (LB)
consequently, structural alterations caused by lack medium at 37°C, C. diphtheriae in Heart Infusion (HI)
DIP1281 prevent adhesion of corresponding mutants to broth at 37°C. If appropriate, kanamycin was added
host cells and invasion into these cells. (30 μg/ml for E. coli; 50 μg/ml for C. diphtheriae).
Preparation of C. diphtheriae protein extracts
Conclusions To prepare surface proteins, bacteria were grown in
C. diphtheriae protein DIP1281 was, as its homologs 20 ml HI broth (with kanamycin added for the mutant
Ce1659, Cg1735, and JK0967 in Corynebacterium effi- strains) for approximately six hours and used to inocu-
ciens, Corynebacterium glutamicum, and Corynebacter- late 250 ml HI broth for overnight growth. Bacteria
ium jeikeium, previously annotated as hypothetical were harvested by centrifugation at 5,000 × g for 20
invasion-associated protein. Generation and analyses of min, washed twice with pre-cooled (4°C) 50 mM Tris-
mutant strains indicate that DIP1281 is predominantly HCl buffer (pH 7.2), resuspended in 50 mM Tris-HCl
involved in the organization of the outer surface protein (pH 7.2) containing 2% 3-[(3-choamidopropyl)-dimethy-
layer of C. diphtheriae rather than in the separation of lammonio] propanesulfonate (CHAPS) and incubated on
the peptidoglycan cell wall of dividing bacteria. The ice overnight, followed by centrifugation at 3,500 × g
adhesion- and invasion-negative phenotype of corre- and 4°C for 30 min to separate the cell surface proteins.
sponding mutant strains is an effect of rearrangements After filtration of the protein solution using 0.45 μm

30
Publications
Ott et al. BMC Microbiology 2010, 10:2 Page 7 of 9
http://www.biomedcentral.com/1471-2180/10/2

Table 2 Bacterial strains and eukaryotic cells used in this 20 mM DTT, 2% CHAPS, a trace of bromophenol blue,
study. and 0.5% Pharmalyte 3-10). IEF was performed with
Strains Description Reference commercially available IPG strips (18 cm, pH 3-10)
C. diphtheriae and the Ettan IPGphor II (GE Healthcare, Munich,
DSM44123 non-toxigenic isolate, type DSMZ Germany). The following voltage profile was used for
strain (Braunschweig) IEF: 1 h, 0 V; 12 h, 30 V; 2 h, 60 V; 1 h, 500 V; 1 h,
ISS3319 C. diphtheriae var. mitis, non- [9] 1000 V followed by a linear increase to 8000 V. The
toxigenic isolate final phase of 8000 V was terminated after 90,000 Vh.
ISS4060 C. diphtheriae var. gravis, non- [9] The IPG strips were equilibrated for 30 min each in 5
toxigenic isolate
ml of solution C (6 M urea, 50 mM Tris-HCl (pH 6.8),
Lilo1 ISS3319 DIP1281:: This study
pK18mob'DIP1281''
30% glycerol, 2% SDS, 1% DTT) and in 5 ml of solution
Lilo2 ISS4060 DIP1281:: This study
D (6 M urea, 50 mM Tris-HCl (pH 6.8), 30% glycerol,
pK18mob'DIP1281'' 2% SDS, 4% iodacetamide). The isolated proteins were
E. coli separated in 12.5% acrylamide/bis-acrylamide gels
DH5aMCR endA1 supE44 thi-1 l- recA1 [28] (37.5:1) with an Ettan Dalt II system (GE Healthcare,
gyrA96 relA1 deoR ∆(lacZYA- Munich, Germany) applying approximately 15 mA per
argF) U196 !80∆lacZ ∆M15 gel. To visualize the separated proteins, gels were
mcrA ∆(mmr hsdRMSmcrBC)
stained in Coomassie staining solution (5% methanol,
Cell lines
42.5% ethanol, 10% acetic acid, 0.25% Serva-G250), and
Detroit562 human hypopharyngeal [29]
carcinoma cells
destained with 10% acetic acid.
Immuno-fluorescence
For immuno-fluorescence staining a rabbit antiserum
pore-size filters (SARSTEDT, Nümbrecht, Germany), directed against the C. diphtheriae surface proteome
further preparation of the surface proteins by phenolic was used as primary antibody. As secondary antibody
acid extraction and methanol precipitation followed a Alexa-Fluor 488 (green) goat anti-rabbit IgGs were
protocol described by Watt and co-workers [23]. The applied. All antibodies were diluted in blocking solution
precipitated proteins were harvested by centrifugation at (2% goat serum, 2% BSA). Bacterial cells were dried on
3,500 × g and 4°C for 30 min. The pellet was washed coverslips (37°C), fixed with 3% PFA (10 min at room
twice with 3 ml of 70% ethanol (-20°C) and once with temperature) and finally washed thrice with 1 × PBS.
3 ml of acetone (-20°C). Finally, the protein pellet was Bacterial cells were incubated in staining solution for at
dried on ice and solubilised in 450 μl of dehydration least 1 h at room temperature and washed thrice with
buffer (8 M urea, 20 mM DTT, 2% CHAPS). Protein PBS between staining steps. Coverslips were mounted
concentrations were determined spectrophotometrically on glass slides using Fluoroprep (Biomerieux, Craponne,
using a NanoDrop spectrophotometer (peqLab, Erlan- France). Imaging was done on an AxioVert 200 M
gen, Germany). Surface proteins prepared from strain inverted optical microscope (Carl Zeiss Micromaging
DSM44123 were used for the immunization of rabbits GmbH, Jena, Germany). For additional image processing
to generate C. diphtheriae surface protein-specific anti- Photoshop CS2 (Adobe, Munich, Germany) was used.
sera (Eurogentec, Liege, Belgium). LIVE/DEAD staining
SDS-PAGE, silver staining, and Western blot analysis Overnight cultures grown in 20 ml HI broth plus kana-
Proteins of the cell surface fraction of wild-type and mycin added for the mutant strains were washed once
mutant strains were separated using Tricine-buffered in 20 ml 1 × PBS and resuspended in 10 ml 1 × PBS.
10% SDS gels as described [24]. After SDS-PAGE pro- To distinguish live and dead bacteria the LIVE/DEAD
tein bands were visualized by silver staining [25]. For Baclight Bacterial Viability Kit for microscopy (Invitro-
Western blotting, the SDS gel-separated proteins were gen, Eugene, OR) was used according to the supplier’s
transferred onto a polyvinylidene difluoride membrane protocol. Imaging was done on an AxioVert 200 M
by electroblotting (PVDF, Roth, Karlsruhe, Germany) inverse microscope (Carl Zeiss Micromaging GmbH,
and incubated with C. diphtheriae surface protein-speci- Jena, Germany).
fic antisera generated in rabbits. Antibody binding was Atomic force microscopy (AFM)
visualized by using goat anti-rabbit IgG coupled to alka- Overnight cultures grown in 20 ml HI broth plus kana-
line phosphatase and the BCIP/NBT alkaline phospha- mycin added for the mutant strains were washed five
tase substrate (Sigma-Aldrich, Darmstadt, Germany). times in 20 ml ice cold distilled water and finally resus-
2-D-PAGE of C. diphtheriae surface proteins pended in 10 ml ice cold distilled water. 5 μl of each
2-D polyacryalmide gels were loaded with 300 μg of sample were fixed on a glass slide by drying using com-
proteins dissolved in 450 μl of solution B (8 M urea, pressed air. An AFM instrument (MFP-3D, Asylum

31
Publications
Ott et al. BMC Microbiology 2010, 10:2 Page 8 of 9
http://www.biomedcentral.com/1471-2180/10/2

Research, Santa Barbara, CA) with standard silicon can- the following primers: 5’- cgc gcg ctc gcg ggc acg tca
tilever probes (NCH-W, Nanosensors, Neuchatel, Swit- gga agc tg - 3’; 5’- cgc gcg ccc ggg cga atc caa ttt tat taa
zerland) was used under ambient laboratory conditions aa - 3’. Using the AvaI and XmaI sites introduced in via
and operated in tapping mode. AFM topography and the PCR primers (shown in bold) the DNA fragment
phase images were recorded simultaneously. was ligated to AvaI/XmaI-restricted and dephosphory-
Adhesion assays lated pK18 mob DNA [27]. The resulting plasmid pK18
D562 cells were seeded in 24 well plates (Greiner bio- mobDIP1281’ was amplified in E. coli DH5aMCR. One
one Cellstar, Frickenhausen, Germany) at a density of microgram of unmethylated plasmid isolated from this
2 × 105 cells per well 48 h prior to infection. Bacteria E. coli strain was used to transform C. diphtheriae using
were inoculated to an OD600 of 0.1 from overnight cul- a GenePulser II (Bio-Rad, Munich Germany). Electropo-
tures and grown in HI broth for 3.5 h. Subsequently, rated cells were added to 1 ml of HI broth containing
the bacteria were harvested by centrifugation and 1% glucose and incubated for 2 h at 37°C. An appropri-
adjusted to an OD600 of 0.2. A master mix of the inocu- ate volume of culture was plated on medium containing
lum was prepared in DMEM (Dulbecco’s modified kanamycin. Since pK18 mob cannot be replicated in
Eagle’s medium, PAA; high glucose, 10% FCS, 2 mM C. diphtheriae, kanamycin-resistant C. diphtheriae car-
glutamine) without penicillin/streptomycin and cells ried the vector integrated via recombination in the
were infected for 90 min at a MOI of 200 (viable counts chromosomal DIP1281 gene and were designated Lilo1
experiments). The cells were washed with PBS nine (resulting from the strain ISS3319) and Lilo2 (resulting
times, detached with 500 μl trypsin solution (0.12% tryp- from the strain ISS4060).
sin, 0.01% EDTA in PBS) per well (5 min, 37°C, 5%
CO2 , 90% humidity) and lysed with 0.025% Tween 20
Acknowledgements
for 5 min at 37°C. Serial dilutions were made in pre- The authors wish to thank C. v. Hunolstein (Istituto Superiore di Sanita’,
chilled 1 × PBS and plated on HI plates to determine Rome) for providing strain ISS3319 and ISS4060, A. Völzke (Erlangen) for
the number of cfu. The assay is modification of a pre- preparation of surface proteins for antibody generation and the Deutsche
Forschungsgemeinschaft for financial support in frame of SFB 796 (projects
viously described one [9]. B5 and Z).
Epithelial cell invasion model
D562 cells were seeded in 24 well plates (Greiner bio- Author details
1
Friedrich-Alexander-Universität Erlangen-Nürnberg, Lehrstuhl für
one Cellstar, Frickenhausen, Germany) at a density of Mikrobiologie, Staudtstr. 5, 91058 Erlangen, Germany. 2Mikrobiologisches
2 × 105 cells per well 48 h prior to infection. Overnight Institut des Universitätsklinikums Erlangen, Wasserturmstr. 3-5, 91054,
cultures grown in HI were re-inoculated to an OD600 of Erlangen, Germany. 3Friedrich-Alexander-Universität Erlangen-Nürnberg,
Lehrstuhl für Angewandte Physik, Staudtstr. 5, 91058 Erlangen, Germany.
0.1 in fresh medium and grown aerobically for another 4
Robert-Koch-Institut, Burgstr. 37, 38855 Wernigerode, Germany.
3.5 h. An inoculum of approximately 8 × 107 bacteria 5
Arbeitsgruppe Mikrobiologie, Universität Osnabrück, Barbarastr. 11, 49076
ml -1 (MOI = 200) was prepared in DMEM without Osnabrück, Germany.

penicillin/streptomycin and 500 μl per well were used to Authors’ contributions


infect the D562 cells. The plates were centrifuged for LO carried out growth mutagenesis experiments, invasion assays,
5 min at 500 × g to synchronize infection and subse- fluorescence microscopy, protein preparation and analysis, MHö carried out
adhesion experiments, RGG and MHe supported LO and MHö in respect to
quently incubated for 90 min (37°C, 5% CO 2 , 90% cell culture, adhesion and invasion analysis and fluorescence microscopy,
humidity). The cells were washed thrice with PBS and AFM experiments were carried out in cooperation with JR and TES, AB
500 μl of DMEM containing 100 μg ml -1 gentamicin supervised the experiments of LO and MHö and was responsible for the
draft and final version of the manuscript. All authors read and approved the
was applied to each well to kill remaining extracellular final manuscript.
bacteria. After 2 h of incubation the cell layers were
washed thrice with PBS, detached by adding 500 μl tryp- Received: 23 July 2009
Accepted: 5 January 2010 Published: 5 January 2010
sin solution (0.12% trypsin, 0.01% EDTA in PBS) per
well (5 min, 37°C, 5% CO2, 90% humidity), and lysed for References
5 min at 37°C with 0.025% Tween 20 to liberate the 1. Galazka A: The changing epidemiology of diphtheria in the vaccine era.
intracellular bacteria. Serial dilutions of the inoculum J Infec Dis 2000, 181(suppl 1):S2-S9.
2. Hadfield TL, McEvoy P, Polotsky Y, Tzinserling A, Yakovlev AA: The
and the lysates were plated on HI plates to determine pathology of diphtheria. J Infect Dis 2000, 181(suppl 1):S116-S120.
the number of colony forming units (cfu). 3. von Hunolstein C, Alfarone G, Scopetti F, Pataracchia M, La Valle R, Franchi
Construction of mutant strains F, Pacciani L, Manera A, Giammanco A, Farinelli S, Engler K, De Zoysa A,
Efstratiou A: Molecular epidemiology and characteristics of
For plasmid isolation, transformation and cloning, stan- Corynebacterium diphtheriae and Corynebacterium ulcerans strains
dard techniques were used [26]. For chromosomal dis- isolated in Italy during the 1990s. J Med Microbiol 2003, 52:181-188.
ruption of the C. diphtheriae DIP1281 gene an 582 bp 4. Funke G, Altwegg M, Frommel L, von Graevenitz AA: Emergence of related
nontoxigenic Corynebacterium diphtheriae biotype mitis strains in
internal DNA fragment was amplified via PCR using Western Europe. Emerg Infect Dis 1999, 5:477-480.
chromosomal DNA of strain ISS3319 as template and

32
Publications
Ott et al. BMC Microbiology 2010, 10:2 Page 9 of 9
http://www.biomedcentral.com/1471-2180/10/2

5. Hamour AA, Efstratiou A, Neill R, Dunbar EM: Epidemiology and molecular 26. Sambrook J, Fritsch EF, Maniatis T: Molecular Cloning: A Laboratory
characterisation of toxigenic Corynebacterium diphtheriae var mitis from Manual. Cold Spring Habor Laboratory Press Cold Spring Habor, NY, 2 1989.
a case of cutaneous diphtheria in Manchester. J Infect 1995, 31:153-157. 27. Schäfer A, Tauch A, Jäger W, Kalinowski J, Thierbach G, Pühler A: Small
6. Romney MG, Roscoe DL, Bernard K, Lai S, Efstratiou A, Clarke AM: mobilizable multi-purpose cloning vectors derived from the Escherichia
Emergence of an invasive clone of nontoxigenic Corynebacterium coli plasmids pK18 and pK19: Selection of defined deletions in the
diphtheriae in the urban poor population of Vancouver, Canada. J Clin chromosome of Corynebacterium glutamicum. Gene 1994, 145:69-73.
Microbiol 2006, 44:1625-1629. 28. Grant SGN, Jessee J, Bloom FR, Hanahan D: Differential plasmid rescue
7. Puliti M, von Hunolstein C, Marangi M, Bistoni F, Tissi L: Experimental from transgenic mouse DNAs into Escherichia coli methylation-restriction
model of infection with non-toxigenic strains of Corynebacterium mutants. Proc Natl Acad Sci USA 1990, 87:4645-4649.
diphtheriae and development of septic arthritis. J Med Microbiol 2006, 55: 29. Peterson WDJr, Stulberg CS, Swanborg NK, Robinson AR: Glucose-6-
229-235. phosphate dehydrogenase isoenzymes in human cell cultures
8. Hirata RJr, Napoleao F, Monteiro-Leal LH, Andrade AFB, Nagao PE, Formiga determined by sucrose-agar gel and cellulose acetate zymograms. Proc
LCD, Fonseca LS, Mattos-Guaraldi AL: Intracellular viability of toxigenic Soc Exp Biol Med 1968, 128:772-776.
Corynebacterium diphtheriae strains in HEp-2 cells. FEMS Microbiol Lett
2002, 215:115-119. doi:10.1186/1471-2180-10-2
9. Bertuccini L, Baldassarri L, von Hunolstein C: Internalization of non- Cite this article as: Ott et al.: Corynebacterium diphtheriae invasion-
toxigenic Corynebacterium diphtheriae by cultured human respiratory associated protein (DIP1281) is involved in cell surface organization,
epithelial cells. Microbial Path 2004, 37:111-118. adhesion and internalization
10. Mandlik A, Swierczynski A, Das A, Ton-That H: Corynebacterium diphtheriae in epithelial cells. BMC Microbiology 2010 10:2.
employs specific minor pilins to target human pharyngeal epithelial
cells. Mol Microbiol 2007, 64:111-124.
11. Mattos-Guaraldi AL, Formiga LCD, Pereira GA: Cell surface components and
adhesion in Corynebacterium diphtheriae. Micr Infect 2000, 2:1507-1512.
12. Hirata RJr, Souza SMS, Rocha de Souza CM, Andrade AFB, Monteiro-Leal LH,
Formiga LCD, Mattos-Guaraldi AL: Patterns of adherence to HEp-2 cells
and actin polymerization by toxigenic Corynebacterium diphtheriae
strains. Microbial Path 2004, 36:125-130.
13. Gerlach RG, Hensel M: Salmonella pathogenicity islands in host specificity,
host pathogen-interactions and antibiotics resistance of Salmonella
enterica. Berl Munch Tierärztl Wochenschr 2007, 120:317-327.
14. Colombo AV, Hirata RJr, Rocha de Souza CM, Monteiro-Leal LH, Previato JO,
Formiga LCD, Andrade AFB, Mattos-Guaraldi AL: Corynebacterium
diphtheriae surface proteins as adhesins to human erythrocytes. FEMS
Microbiol Lett 2001, 197:235-239.
15. de Oliveira Moreira L, Andrade AFB, Vale MD, Souza SMS, Hirata RJr, Asad
LOB, Asad NR, Monteiro-Leal LH, Previato JO, Mattos-Guaraldi AL: Effects of
iron limitation on adherence and cell surface carbohydrates of
Corynebacterium diphtheriae strains. Applied Environ Microbiol 2003,
69:5907-5913.
16. Hansmeier N, Chao T-C, Kalinowski J, Pühler A, Tauch A: Mapping and
comprehensive analysis of the extracellular and cell surface proteome of
the human pathogen Corynebacterium diphtheriae. Proteomics 2006,
6:2465-2476.
17. Anantharaman V, Aravind L: Evolutionary history, structural features and
biochemical diversity of the NlpC/P60 superfamily of enzymes. Genome
Biol 2003, 4:R11.
18. Amon J, Lüdke A, Titgemeyer F, Burkovski A: General and regulatory
proteolysis in corynebacteria. Corynebacteria: genomics and molecular
biology Caister Academic Press, Norfolk, UKBurkovski A 2008, 295-311.
19. Anselmetti D, Hansmeier N, Kalinowski J, Martini J, Merkle T, Palmisano R,
Ros R, Schmied K, Sischka A, Toensing K: Analysis of subcellular surface
structure, function and dynamics. Anal Bioanal Chem 2007, 387: 83-89.
20. Hansmeier N, Albersmeier A, Tauch A, Damberg T, Ros R, Anselmetti D,
Pühler A, Kalinowski J: The surface (S)-layer gene cspB of Corynebacterium
glutamicum is transcriptionally activated by a LuxR-type regulator and
located on a 6 kb genomic island absent from the type strain ATCC
13032. Microbiology 2006, 152:923-935.
21. Hansmeier N, Bartels FW, Ros R, Anselmetti D, Tauch A, Pühler A, Kalinowski
J: Classification of hyper-variable Corynebacterium glutamicum surface Submit your next manuscript to BioMed Central
layer proteins by sequence analyses and atomic force microscopy. J
Biotechnol 2004, 112:117-193.
and take full advantage of:
22. Tsuge Y, Ogino H, Teramoto H, Inui M, Yukawa H: Deletion of cg_1596
and cg_ encoding NlpC/P60 proteins, causes a defect in cell separation • Convenient online submission
in Corynebacterium glutamicum R. J Bacteriol 2070, 190:8204-8214. • Thorough peer review
23. Watt SA, Wilke A, Patschkowski T, Niehaus K: Comprehensive analysis of
the extracellular proteins from Xanthomonas campestris pv. campestris • No space constraints or color figure charges
B100. Proteomics 2005, 5:153-167. • Immediate publication on acceptance
24. Schägger H, von Jagow G: Tricine-sodium dodecyl sulfate-polyacrylamide
• Inclusion in PubMed, CAS, Scopus and Google Scholar
gel eletrophoresis for the separation of proteins in the range from 1 to
100 kDa. Anal Biochem 1987, 166:368-379. • Research which is freely available for redistribution
25. Blum M, Beier H, Gross HJ: Improved silverstaining of plant proteins, RNA
and DNA in polyacrylamid gels. Electrophoresis 1987, 8:93-99. Submit your manuscript at
www.biomedcentral.com/submit

33
Publications

3.2 Strain-specific differences in pili formation and the interaction of


Corynebacterium diphtheriae with host cells (Ott et al, 2010b)

34
Publications
Ott et al. BMC Microbiology 2010, 10:257
http://www.biomedcentral.com/1471-2180/10/257

RESEARCH ARTICLE Open Access

Strain-specific differences in pili formation and


the interaction of Corynebacterium diphtheriae
with host cells
Lisa Ott1, Martina Höller1, Johannes Rheinlaender2, Tilman E Schäffer2, Michael Hensel3,4, Andreas Burkovski1*

Abstract
Background: Corynebacterium diphtheriae, the causative agent of diphtheria, is well-investigated in respect to toxin
production, while little is known about C. diphtheriae factors crucial for colonization of the host. In this study, we
investigated strain-specific differences in adhesion, invasion and intracellular survival and analyzed formation of pili
in different isolates.
Results: Adhesion of different C. diphtheriae strains to epithelial cells and invasion of these cells are not strictly
coupled processes. Using ultrastructure analyses by atomic force microscopy, significant differences in
macromolecular surface structures were found between the investigated C. diphtheriae strains in respect to number
and length of pili. Interestingly, adhesion and pili formation are not coupled processes and also no correlation
between invasion and pili formation was found. Using RNA hybridization and Western blotting experiments, strain-
specific pili expression patterns were observed. None of the studied C. diphtheriae strains had a dramatic
detrimental effect on host cell viability as indicated by measurements of transepithelial resistance of Detroit 562
cell monolayers and fluorescence microscopy, leading to the assumption that C. diphtheriae strains might use
epithelial cells as an environmental niche supplying protection against antibodies and macrophages.
Conclusions: The results obtained suggest that it is necessary to investigate various isolates on a molecular level
to understand and to predict the colonization process of different C. diphtheriae strains.

Background able to gain access to deeper tissues and to persist inside


Corynebacterium diphtheriae is the causative agent of tissues or cells.
diphtheria, a toxaemic localized infection of the respira- A possible clue for the background of persistence of
tory tract. While this disease is well-controlled by vacci- C. diphtheriae came from investigations of adherence
nation against the diphtheria toxin in e. g. Western and invasion of toxigenic and non-toxigenic strains by
Europe [1-3], it is still a severe health problem in less different groups. Using a combination of gentamicin
developed countries. Furthermore, C. diphtheriae is not protection assays and thin-section electron microscopy,
only the aetiological agent of diphtheria, but can cause Hirata and co-workers [9] showed that toxigenic
other infections as well. Non-toxigenic strains have been C. diphtheriae are not only able to adhere to laryngeal
increasingly documented [4-6] and found to be the HEp-2 cells, but also enter these cells and survive after
cause of invasive diseases such as endocarditis, bacterae- internalization. Similar observations were made for non-
mia, pneumonia, osteomyelitis, spleen abscesses, and toxigenic strains [10] showing that also pharyngeal
septic arthritis [7,8]. As indicated by these systemic Detroit 562 cells can be invaded by C. diphtheriae and
infections, C. diphtheriae is not only able to attach to that viable intracellular bacteria can be detected up to
host epithelial cells of larynx and pharynx, but must be 48 h after infection.
While host cell receptors and invasion-associated pro-
* Correspondence: aburkov@biologie.uni-erlangen.de teins of the pathogen are still unknown, bacterial adhe-
1
Lehrstuhl für Mikrobiologie, Friedrich-Alexander-Universität Erlangen- sion factors have been recently at least partially
Nürnberg, Nürnberg, Germany
Full list of author information is available at the end of the article
characterized on the molecular level. C. diphtheriae

© 2010 Ott et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons
Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in
any medium, provided the original work is properly cited.

35
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 2 of 12
http://www.biomedcentral.com/1471-2180/10/257

strain NCTC13129 is able to assemble three distinct Once attached to the surface of an epithelial cell,
types of pili on its surface [11,12]. Mutant analyses C. diphtheriae might invade the host cell and persist
showed that the SpaA-type pilus is sufficient for adhe- within the cell. In order to investigate this process for
sion of this strain to pharynx cells, shaft proteins are the different strains studied here, gentamicin protection
not crucial for pathogen-host interaction, and adherence assays were carried out. For this purpose, cells were
to pharyngeal cells is greatly diminished when minor incubated for 1.5 h with bacteria, gentamicin was added
pili proteins SpaB and SpaC are lacking [13]. The results to kill remaining extracellular C. diphtheriae and survi-
obtained in other studies indicated the existence of addi- val of intracellular bacteria was analyzed after different
tional proteins besides pili subunits involved in adhesion times of incubation (Fig. 2). When invasion into D562
to larynx, pharynx, and lung epithelial cells, since a total cells was analyzed for the six non-toxigenic strains and
loss of attachment to pharyngeal cells due to mutagen- the toxigenic C. diphtheriae strain after 2 h, tox+ strain
esis of pili- and sortase-encoding genes could not be DSM43989 showed the lowest internalization rate with
observed and attachment to lung or larynx cells was less 0.014 ± 0.007%. As in the adhesion assay, the non-toxi-
affected by the mutations. This is in line with a number genic strains showed in general a higher rate compared
of studies suggesting the multifactorial mechanism of to the toxin-producer strain and again rates differed sig-
adhesion (reviewed in [14]). Furthermore, Hirata and nificantly between the non-toxigenic strains, varying
co-workers [7,15] described three distinct patterns of between 0.018 ± 0.006% for strain ISS4749 and 0.060 ±
adherence to HEp-2 cells, an aggregative, a localized, 0.027 for strain ISS4060 (Fig. 2A). The comparison of
and a diffuse form, an observation that hints also to the strains in respect to adhesion and internalization rates
existence of several adhesion factors and different recep- suggested that although a high adhesion seems to favour
tors on the host cell surface. The involvement of differ- internalization, adhesion and invasion are not strictly
ent C. diphtheriae proteins to adherence to distinct cell coupled processes. Plating and counting of internalized
types is further supported by work on adhesion to cells after 8.5 and 18.5 h revealed decreasing numbers
human erythrocytes, showing that non-fimbrial surface of colony forming units (Fig. 2B-C). Even after 18.5 h,
proteins 67p and 72p, which were up to now only char- no strain was completely eliminated from the cells and
acterized by their apparent mass, are involved in this survival of bacteria ranged from 0.002 ± 0.001% of the
process [16]. Interestingly, besides strain-specific differ- inoculums for DSM43989 to 0.005 ± 0.001% for
ences in adherences (see references cited above), also ISS4060.
growth-dependent effects were observed. In a study In addition to the gentamicin protection assay, which
using two toxigenic C. diphtheriae strains and erythro- gives quantitative data, immune-fluorescence micro-
cytes as well as HEp-2 cells, de Oliveira Moreira and scopy was applied as an independent method to investi-
co-workers [17] showed an effect of iron supply on gate host cell interaction of C. diphtheriae strains. This
hemagglutination and lectin binding properties of the method has the advantage of allowing direct visualiza-
microorganisms. tion, although only on a qualitative level. Using an anti-
In this study, we present a characterization of different serum directed against C. diphtheriae surface proteins
non-toxigenic C. diphtheriae and a toxin-producing and antibody staining before and after permeabilization
strain with respect to adhesion to and internalization of the host cell, internalized C. diphtheriae were
into epithelial cells. Analyses reveal significant strain- detected (Fig. 3). Interestingly, V-shaped C. diphtheriae
specific differences in host colonization and macromole- dimers within the cells were observed. These V-forms
cular surface structures of the studied strains, while are the result of the Corynebacterium-specific snapping
neither of the strains evoked rapid cell damage under division and indicate growing bacteria. Together with a
the conditions tested. tendency towards formation of clusters of cells (Fig. 3C
and 3F), this observation suggests that bacteria replicate
Results within the host cells and growth and elimination
Adhesion of C. diphtheriae to epithelial cells, invasion of described above (Fig. 2A-C) are parallel processes.
host cells and intracellular survival
In this study, adhesion of six non-toxigenic strains and Influence of C. diphtheriae on the transepithelial
one toxin-producing C. diphtheriae to Detroit562 cells resistance of cell monolayers
was analyzed (Fig. 1). In these experiments tox+ strain Some pathogens, such as Salmonella enteric serovar
DSM43989 showed the lowest adhesion rate with 0.34 ± Typhimurium (S. Typhimurium), can cause severe
0.05%. While in general higher than the tox+ strain, the damage on cell membranes and due to the resulting loss
non-toxigenic strains differed significantly in their adhe- of cell integrity, the transepithelial resistance of mono-
sion rate, varying between 0.69 ± 0.12% for strain layers is dramatically reduced (for example see [18]). In
DSM43988 and 7.34 ± 2.33% for strain ISS4749 (Fig. 1). this study, we used S. Typhimurium NCTC12023 as a

36
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 3 of 12
http://www.biomedcentral.com/1471-2180/10/257

Figure 1 Adhesion of C. diphtheriae strains to D562 cell layers. D562 cells were infected with different C. diphtheriae strains. Besides
DSM43989, which is tox+, the isolates are non-toxigenic. The cells were washed with PBS, detached with trypsin solution, lysed with Tween 20,
and the number of colony forming units (cfus) was determined. Adhesion is expressed as percentage of the inoculum, showing means and
standard deviations of ten independent measurements (biological replicates) with 3 samples each (technical replicates). All strains, except
ISS4746 and ISS4749, show statistically significant differences in adhesion rates (students TTEST values below 0.04).

positive control (Fig. 4A) and tested the influence of dif- Ultrastructural analysis of C. diphtheriae strains
ferent C. diphtheriae strains on transepithelial resistance Since we suspected that the differences in adhesion
(Fig. 4B). Infection of Detroit562 monolayers with might be the result of different surface structures, we
S. Typhimurium caused a dramatic break-down of trans- started an ultrastructure analysis of selected C. diphther-
epithelial resistance within 1.5 h while all tested iae. For this purpose, non-toxigenic strains as well as
C. diphtheriae strains including tox+ strain DSM43989 tox+ strain DSM43989 were analyzed by atomic force
had no effect on transepithelial resistance within a time microscopy (Fig. 5A). With this technique, which allows
span of three hours. Since the incubation conditions imaging surfaces topography at high resolution, signifi-
used in this assay are similar to that applied for the cant different macromolecular surface structures were
adhesion and invasion assays, this observation indicates found between the different investigated C. diphtheriae
that a detrimental effect of toxin production can be strains. While for ISS4060 and DSM43988 pili were not
excluded as a reason for the low adhesion and internali- detectable at all, ISS3319 and DSM44123 revealed short,
zation rates of strain DSM43989. spike-like pili, ISS4746, ISS4749 and DSM43989 showed
Overnight incubation of D562 cells with C. diphther- long, hair-like protrusions (Fig. 5A). Also the number of
iae was tested as well. In this case, the Dulbecco’s modi- pili (counted from at least six specimens of each strain)
fied Eagle’s medium had to be exchanged after 3 h with differed significantly (5B). Interestingly, adhesion and
fresh medium to remove not adhered bacteria in order pili formation were not coupled, since ISS3319, which
to avoid that the pH of the medium dropped due to the revealed spike-like pile and ISS4060, lacking these,
bacterial metabolism leading to secondary detrimental showed comparable adhesion rates, while ISS4746 and
effects. In contrast to short term incubation and to the ISS4749 had different numbers of long hair-like pili but
non-toxigenic strains, long term measurement (Fig. 4B, showed identical adhesion rates. Also no correlation
overnight time point) of transepithelial resistance of cell between invasion and pili formation was found. Since
monolayers infected with DSM43989 showed a signifi- strain-specific differences in pili formation have not
cant effect, which might be caused by toxin production. been observed before, the background for this

37
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 4 of 12
http://www.biomedcentral.com/1471-2180/10/257

phenomenon was investigated in more detail in subse-


quent experiments.

Strain-specific expression of pili subunits


To analyze the molecular basis of strain-specific differ-
ences in pili formation, RNA hybridization experiments
were carried out to study the mRNA levels of the C.
diphtheriae spa genes. These genes are organized in
three different clusters together with the corresponding
sortase-encoding genes in the sequenced strain
NCTC13129 [13,19]. The first cluster comprises the
genes spaA, spaB, and spaC, which are most likely orga-
nized as an operon; the second cluster is formed by
spaD and a putative spaE-spaF operon, and a third clus-
ter comprises the spaG, spaH, and spaI gene, which are
most likely independently transcribed. Strain-specific
differences were detected, when probes for the detection
of all genes of cluster I and III were applied in RNA
hybridization experiments (Fig. 6A).
Strongest hybridization signals with spaA, spaB, and
spaC probes were detected with RNA isolated from
strains ISS4746 and ISS4749, slightly lower signal inten-
sities were observed with strain DSM43989, while only
faint signals were obtained for cluster I mRNA for the
other investigated strains. Strong transcription of spaG,
spaH, and spaI were again detected in strains ISS4746
and ISS4749, while other strains did not express cluster
III genes deduced from RNA hybridization experiments.
The data are in accordance with the AFM experiments
presented in Fig. 5, which show formation of a high
number of extended pili for strains ISS4746 and
ISS4749, followed by DSM43989; however, hybridization
signals may differ not only due to mRNA abundance,
but also due to sequence alteration.
To elucidate whether the missing transcripts in var-
ious strains are the result of regulatory processes or
have genetic reasons, PCR experiments were carried
out, which showed that missing transcripts are corre-
lated to lacking PCR products making regulatory
effects unlikely (Fig. 6B). Furthermore, reproducible
strain-specific differences in sizes of the PCR products
were observed for spaA and in band intensities for
Figure 2 Invasion of epithelial cells by C. diphtheriae strains. D562 spaB fragments, suggesting that also sequence devia-
cells were infected with different C. diphtheriae strains (DSM43989 tox+,
all others are non-toxigenic), washed, and incubated 2.0 (A), 8.5 (B) and
tions exist besides strain-specific differences in the spa
18.5 (C) hours with 100 μg ml-1 gentamicin. Subsequently, cells were gene repertoire. Interestingly, we were unable to gener-
washed, detached with trypsin solution, lysed with Tween 20, and the ate any probes for genes of cluster II, comprising spaD,
number of intracellular cfus was determined. Invasion is shown as spaE, and spaF, when chromosomal DNA of the used
percentage of the inoculum internalized (means and standard ISS and DSM strains were used as template, while all
deviations of three independent biological replicates with three
samples each (technical replicates). Statistically relevant differences
genes could be amplified from NCTC13129 DNA (Fig.
between the strains (based on students TTEST values below 0.05) are 7). Deduced from these PCR experiments, these genes
indicated by letters above columns. seem to be absent in the investigated C. diphtheriae
strains. As an additional approach, we tested

38
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 5 of 12
http://www.biomedcentral.com/1471-2180/10/257

Figure 3 Detection of intracellular C. diphtheriae in Detroit562 cells by immune-fluorescence microscopy. D562 cells were seeded on
coverslips 48 h prior to infection and infected with C. diphtheriae (DSM43989 tox+, all others are non-toxigenic) for 4 h with at a MOI of 200 as
described earlier [26]. Antibodies directed against the surface proteome of C. diphtheriae were used as primary, Alexa Fluor 488 goat anti-rabbit
IgGs and Alexa-Fluor 568 goat anti-rabbit IgGs as secondary antibodies (A, D: intact D562, B, E: permeabilized D562, C, F: overlay with blue
F-actin stain Phalloidin-Alexa-Fluor 647, A-C: ISS3319, D-F: ISS4060. Green stain in panels A and D indicate extracellular bacteria. Dark red stain in
panels B and E indicates internalized C. diphtheriae, while adherent bacteria appear in light red. In the overlay (C, F) extracellular C. diphtheriae
appear orange, while internalized bacteria are stained dark red. Scale bars: 20 μm.

Figure 4 Transepithelial resistance of polarized D562 monolayers grown on transwells. (A) Control experiments of cells, which were
incubated without bacteria (open circles) and S. enterica serovar Typhimurium (open squares). (B) Incubation with C. diphtheriae strains
DSM43989 (tox+, open stars), ISS4749 (inverted closed triangles), ISS4746 (closed triangles), ISS4060 closed circles, ISS3319 (closed square),
DSM43988 (closed hexagons), and DSM44123 (closed diamonds). Experiments were carried out independently at least thrice and typical results
are shown.

39
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 6 of 12
http://www.biomedcentral.com/1471-2180/10/257

Figure 5 Ultrastructural analysis of the cell surface of C. diphtheriae strains. (A) Bacteria were fixed on glass slides by drying using
compressed air. Atomic force microscopy was carried out under ambient laboratory conditions and operated in tapping mode. Scale bars: 500
nm. (B) AFM images were analyzed in respect to pili number per bacterium. For each strain pili of at least six bacteria were counted; error bars
indicate deviations from mean values.

expression of SpaD in the different strains by Western signal was detectable in the investigated cell extracts
blot experiments. Cell extracts of strains ISS3319, (data not shown).
ISS4040, ISS4746, ISS4749, DSM43988, DSM43989, To address the hypothesis that pili expression patterns
and DSM44123 as well as purified SpaD protein as a might change, when bacteria were in exposed to host
positive control were separated by SDS-PAGE and sub- cells, Green fluorescent protein (GFP) fluorescence of C.
jected to Western blotting. SpaD-specific antiserum diphtheriae transformed with plasmids carrying spa
reacted exclusively with the SpaD control, while no gene upstream DNA and a promoter-less gfpuv gene

40
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 7 of 12
http://www.biomedcentral.com/1471-2180/10/257

was determined without and after 1.5 h of host cell con- Discussion
tact. However, analysis of 80 to 140 bacteria for GFP In this study, different non-toxigenic C. diphtheriae and
fluorescence before and after host cell contact revealed a toxin-producing strain were characterized in respect
no significant differences (data not shown). to adhesion to and invasion of epithelial cells. All strains
were able to attach to host cells and immuno-fluores-
cence microscopy revealed internalization and growth of
C. diphtheriae within epithelial cells. We could show
that adhesion and invasion are not strictly coupled, indi-
cating that different proteins and mechanisms play a
role in these processes. Despite the fact that the number
of internalized bacteria decreased over time for all inves-
tigated strains, a considerable number of bacteria sur-
vived prolonged internalization for more than 18 h.
Furthermore, V-shaped division forms as well as forma-
tion of microcolonies were observed by fluorescence
microscopy, suggesting that the epithelial cells might
support growth of C. diphtheriae.
While proteins responsible for invasion and intracellu-
lar persistence are completely unknown for C. diphther-
iae, for the sequenced strain NCTC13129 the influence
of pili subunits on adhesion was characterized recently.
It was shown that the minor pili subunits SpaB and
SpaC are crucial for adhesion of strain NCTC13129 to
epithelial cells [13], while pili length is influenced by the
major pili subunits SpaA, SpaD, and SpaH, which form
the shaft of the structure [11,12,19]. The strains investi-
gated here showed significant variations in adhesion
rates and compared to the tox + strain, for all studied
non-toxigenic strains higher adhesion rates were
observed; however, adhesion and pili formation were
not strictly coupled processes. Strain-specific differences
of appearance and numbers of pili-like structures on the
surface of C. diphtheriae strains were shown by ultra-
structural analyses via atomic force microscopy. Addi-
tionally, RNA hybridization and Western blotting
experiments revealed distinct differences in the expres-
sion patterns of pili subunits for the investigated strains.
To our knowledge, this is the first time that isolate-spe-
cific differences in pili formation were characterized.
Mandlik and co-workers [13] showed that type III pili

Figure 6 Strain-specific distribution and expression of pili-


encoding genes. (A) Levels of spa gene transcripts in different
C. diphtheriae strains. Total RNA was isolated from the indicated
C. diphtheriae strains and hybridized with probes monitoring 16SrRNA Figure 7 PCR detection of spa genes in C. diphtheriae strain
for control as well as spa gene transcription. (B) PCR detection of spa NCTC 13129. Chromosomal DNA of C. diphtheriae strain NCTC
genes. Chromosomal DNA of the indicated C. diphtheriae strains was 13129 was used as template for PCR using specific oligonucleotide
used as template for PCR using specific oligonucleotide pairs for the pairs for the indicated spa genes. In all cases, DNA fragments of the
spa genes indicated at the right side of the figure. expected size were amplified.

41
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 8 of 12
http://www.biomedcentral.com/1471-2180/10/257

length of strain NCTC13129 depends on spaH expres- comparisons [23] it is necessary to investigate various
sion and can be manipulated by deletion or overexpres- isolates on a molecular level to understand and to pre-
sion of spaH. These results are supplemented here by dict the colonization process of different C. diphtheriae
showing that this is a phenomenon which occurs also as strains.
natural variation in different C. diphtheriae wild type
isolates. Strains ISS4746 and ISS4749 showed the most Methods
extended pili structures, an observation which is corre- Bacterial strains and growth
lated with high expression of spaA and spaH in these Strains used in this study are listed in Table 1.
strains, while medium-length pili of DSM43989 are cor- C. diphtheriae strains were grown in Heart Infusion
related with lack of spaH expression. As mentioned (HI) broth or on Columbia agar with sheep blood
above, it was shown by mutant analyses of strain (Oxoid, Wesel, Germany) at 37°C. S. Typhimurium and
NCTC13129 that expression of spaB and spaC is crucial Escherichia coli DH5aMCR were grown in Luria Broth
for adhesion to D562 cells [13]. Natural variations of the (LB) [25] at 37°C. If appropriate, kanamycin was added
spaB and spaC expression patterns observed here indi- (30 μg ml-1 for E. coli; 50 μg ml-1 for C. diphtheriae).
cate that this correlation is not as strict as suggested,
since strain ISS4060 shows only low spaB and no spaC Transformation of competent C. diphtheriae
expression but a high adhesion rate, indicating that For preparation of electrocompetent cells, 10 ml of an
other factors are important for adhesion as well and overnight culture of C. diphtheriae were inoculated in
expression of these might differ in various isolates. 200 ml of Brain Heart Infusion (BHI) containing 2% gly-
The lack of any PCR product for spaD, spaE, and spaF cine and 15% sucrose, at 37°C in an orbital shaker until
and the absence of a SpaD signal in Western blotting an OD600 nm of 0.5 was reached. After storing the cells
experiments suggest that these genes are absent in the on ice for 15 min, bacteria were harvested by centrifuga-
investigated strains. All pili-encoding genes of tion (4,000 × g, 4°C), washed thrice with 15% glycerol,
C. diphtheriae are located on pathogenicity islands and resuspended in 1 ml of 15% glycerol. 100 μl aliquots
[20,21]. Based on the genome sequence of strain of the competent cells were frozen in liquid nitrogen
NCTC13129, C. diphtheriae possesses 13 of these geno- and stored at -80°C.
mic islands [20,22] and pili cluster II is located on geno- For transformation the aliquots were thawed on ice.
mic island CDGI-2, which has a size of 17.5 kb and is Plasmid DNA used for transformation was extracted
located directly adjacent to 36.5 kb pathogenicity island from E. coli strain DH5aMCR, which is unable to
CDGI-1, the tox+ corynephage [20]. Data of PCR experi- methylate DNA. One microgram of plasmid DNA was
ments (not shown) indicate that the pili-encoding genes used to transform C. diphtheriae cells using a GenePul-
located on CDGI-2 are missing in all investigated ISS ser II apparatus (Bio-Rad, Munich, Germany) and 200
and DSM strains and consequently the genetic reper- Ω, 2.5 kV, 25 μF. Electroporated cells were added to 1
toire of C. diphtheriae isolates is rather variable. This ml of HI broth containing 1% glucose, incubated for 2 h
observation is in agreement with a recent genome sur- at 37°C, and plated on medium containing 10 μg ml-1
vey of C. diphtheriae C7(-) and PW8 strains [23] indi- kanamycin. Subsequently, transformants were cultivated
cating that 11 of the 13 putative pathogenicity islands of in the presence of 50 μg ml-1 kanamycin.
the sequenced reference strain NCTC13129 are absent
in the C7(-) strain. Construction of GFP reporter plasmids and reporter gene
The importance of bacterial appendices and surface assay
proteins for host cell contact were also shown recently For construction of reporter plasmids, promoter regions
for a non-fimbrial protein, DIP1281, previously anno- of the pili- encoding spaA, spaG, spaH, and spaI genes
tated as invasion-associated protein. This protein is a were amplified from chromosomal DNA by PCR (for
virulence factor involved in cell surface organization, oligonucleotides used see Table 2). The PCR products
adhesion and internalization in epithelial cells. Corre- were inserted into plasmid pEPR1, using the NsiI and
sponding mutant strains lack the ability to adhere to BamHI restriction site upstream of the promoterless
host cells or invade these [24]. gfpuv gene. Plasmids constructed (Table 3) were used to
transform electrocompetent C. diphtheriae and resulting
Conclusions plasmid-carrying strains were applied to analyze spa
The results obtained in this study show that adhesion promoter activity before and in response to host cell
and invasion are not necessarily coupled processes. contact. For this purpose, Detroit562 cells were seeded
Adhesion rates are not strictly correlated with pili for- on coverslips 48 h prior to infection. Adhesion assays
mation and in summary the pili repertoire of the inves- with Detroit562 cells were carried out as described
tigated strains is highly variable. As shown by genome below (4 μl of the inoculi dried on coverslips were used

42
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 9 of 12
http://www.biomedcentral.com/1471-2180/10/257

as no contact control). Subsequently, the infected cells Germany) Minifold I Dot Blotter. Hybridization of
were fixed with 3% para-formaldehyde (PFA) in PBS (10 digoxigenin-labelled RNA probes was detected with X-
min, room temperature), coverslips were mounted on ray films (Hyperfilm MP; GE Healthcare, Munich, Ger-
glass slides using Fluoroprep (Biomerieux, Craponne, many) using alkaline phosphatase-conjugated anti-digox-
France), and imaging was done on a LSM700 confocal igenin Fab fragments and CSPD as light-emitting
laser microscope (Carl Zeiss Micromaging GmbH, Jena, substrate as recommended by the supplier (Roche, Man-
Germany). GFP fluorescence emission of C. diphtheriae nheim, Germany).
without and with 1.5 h of host cell contact was deter-
mined for ISS4746 and the different reporter plasmids SDS-PAGE and Western blotting
(Table 3) by analyzing between 80 and 140 bacteria for Cell extracts of C. diphtheriae strains were separated by
each condition using the open source ImageJ program Tricine-buffered 9.5% SDS gels as described [26], pro-
package (http://rsbweb.nih.gov/ij/index.html). teins were transferred onto a polyvinylidene difluoride
membrane (PVDF, Immobilon-P, pore size 0.45 μm,
RNA preparation, hybridization analyses Millipore, Bedford, MA, USA) by semi-dry electroblot-
Total C. diphtheriae RNA was prepared from 20 ml cul- ting, and immunodetection of SpaD was performed with
tures using the NucleoSpin RNA II Kit (Macherey antibodies directed against this pili subunit produced in
Nagel, Düren, Germany). For the generation of antisense rabbits [13]. Antibody binding was visualised by using
probes, internal DNA fragments of the corresponding appropriate anti-antibodies coupled to alkaline phospha-
genes were amplified by PCR (oligonucleotide primers tase (Sigma-Aldrich, Taufkirchen, Germany) and the
for the different probes are listed in Table 2). The BCIP/NBT alkaline phosphatase substrate (Sigma-
reverse primers encoded the promoter region for T7 Aldrich, Taufkirchen, Germany).
polymerase, which allowed in vitro transcription of
probes using T7 polymerase and subsequent labelling Atomic force microscopy (AFM)
with DIG RNA-labeling mix (Roche, Mannheim, Ger- Overnight cultures grown in 20 ml HI broth were
many). RNA (2.5 μg per time point) was spotted onto washed five times in 20 ml ice cold distilled water and
nylon membranes using a Schleicher & Schuell (Dassel, finally resuspended in 10 ml ice cold distilled water

Table 1 Bacterial strains and eukaryotic cell lines used in this study
Strains Description Reference
C. diphtheriae
DSM43988 non-toxigenic, isolated from throat culture DSMZ, Braunschweig,
Germany
DSM43989 tox+, unknown source DSMZ, Braunschweig,
Germany
DSM44123 non-toxigenic isolate, type-strain, unknown source DSMZ, Braunschweig,
Germany
ISS3319 C. diphtheriae var. mitis, non-toxigenic, isolated from patients affected by [1]
pharyngitis/tonsilitis
ISS4060 C. diphtheriae var. gravis, non-toxigenic, isolated from patients affected by [1]
pharyngitis/tonsilitis
ISS4746 C. diphtheriae var. gravis, non-toxigenic, isolated from patients affected by [21]
pharyngitis/tonsilitis
ISS4749 C. diphtheriae var. gravis, non-toxigenic, isolated from patients affected by [21]
pharyngitis/tonsilitis
NCTC13129 C. diphtheriae var. gravis, non-toxigenic, isolated from pharyngeal membrane, [2]
patient with clinical diphtheria
E. coli
DH5aMCR endA1 supE44 thi-1 l- recA1 gyrA96 relA1 deoR ∆(lacZYA-argF) U196 j80∆lacZ [9]
∆M15mcrA ∆(mmr hsdRMSmcrBC)
Salmonella enterica serovar Typhimurium
(S. Typhimurium)
NCTC12023 wild type identical to ATCC14028 NCTC, Colindale, UK
Cell lines
Detroit562 human hypopharyngeal carcinoma cells [20]

43
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 10 of 12
http://www.biomedcentral.com/1471-2180/10/257

Table 2 Oligonucleotides used in this study


Designation Sequence (5’®3’ direction) Application
DIP0235-as ctt ggt tgc cgg agc agc ctc ctt PCR spaD
DIP0235-s ccg aaa cca aga ccg aga aga ccg tca ag PCR spaD
DIP0237-as gca cac cag tca gcg cca agt cgc PCR spaE
DIP0237-s cgc gac tac gga acc gac acg ctg a PCR spaE
DIP0238-as gaa gtt gaa agg tcg gcc act aca gca a PCR spaF
DIP0238-s aaa ggg cta cta cat caa cat tcc aga cac PCR spaF
DIP2010asT7 cgc gta ata cga ctc act ata ggg tcc atc acg agg aac gac aac ggt ttt aga spaC probe
DIP2010-s cgc tac tcc tat ggg caa gca cct act gat att c spaC probe
DIP2011asT7 cgc gta ata cga ctc act ata ggg tgg tgg cga tgg cca gca gtc cga spaB probe
DIP2011-s tgc agc att cgc cga cga cca acc spaB probe
SondeDIP2013-asT7 ggg ccc taa tac gac tca cta tag gga gga ctg gag tgt tgc gcc g spaA probe
SondeDIP2013-s ggc gtc gaa aat caa gct gg spaA probe
DIP2223asT7 cgc gta ata cga ctc act ata ggg atc ggt aac ttc ctt acg gaa ctt ctc tgg cag spaI probe
DIP2223-s ttg ccc gcg gga act atc gac gga spaI probe
SondeDIP2226-asT7 ggg ccc taa tac gac tca cta tag ggc cca gcc cct gcg acg tc spaH probe
SondeDIP2226-s gga ggg ctg gga ggc agt ca spaH probe
DIP2227asT7 cgc gta ata cga ctc act ata ggg tcg acc ttg gac cag tgg acc tta gcg spaG probe
DIP2227-s ccg gac aga aga ttg ctg ccg agg ca spaG probe
PromDIP2223-as cgc ggg atc cag tag ggc gtc ctt tca gga construction reporter plasmid for spaI
PromDIP2223-s cgc gat gca tgt gac gcc attt tat gta cgc construction reporter plasmid for spaI
PromDIP2226-as cgc ggg atc cag ggt gtt ttc ctt tca gga construction reporter plasmid for spaH
PromDIP2226-s cgc gat gca tcg tca aag tta cgg ccg acc construction reporter plasmid for spaH
PromDIP2227-as cgc ggg atc cag tga aaa cac ctt cta ggg construction reporter plasmid for spaG
PromDIP2227-s cgc gat gca ttg aac cgg aat cat ttc tta construction reporter plasmid for spaG
PromDIP2013-neu-as cgc ggg atc ccc ctc aac tta ttt att tgg caa aaa g construction reporter plasmid for spaA
PromDIP2013-neu-s cgc gat gca tat tgg tga gac tac ttc ctt aaa gct ggt construction reporter plasmid for spaA

(centrifugation steps: 10 min 4,500 × g; resuspension by density of 5 × 10 4 cells per well and cultivated in
vortexing). 5 μl of each sample were fixed on a glass DMEM (Dulbecco’s modified Eagle’s medium, PAA;
slide by drying using compressed air. An AFM instru- high glucose, 10% FCS, 2 mM glutamine) for 14 days
ment (MFP-3D, Asylum Research, Santa Barbara, CA) until they build a transepithelial resistance of at least
with standard silicon cantilever probes (NCH-W, Nano- 1600 Ω·cm-2. Bacteria were subcultured (OD600 of 0.1
sensors, Neuchatel, Switzerland) was used under ambi- from overnight cultures) in 20 ml HI broth for 3.5 h.
ent laboratory conditions and operated in tapping mode The pellet was resuspended in 500 μl 1 × PBS. 50 μl of
[26]. the suspension were used for infection. Measurements
of transepithelial resistance of D562 cells during the
Measurement of transepithelial resistance infection with C. diphtheriae were carried out with a
D562 cells were seeded in transwells (6.5 mm, 0.4 μm, volt-ohm-meter (EVOM2, World Precision Instruments,
polyester membrane, 24 well plate, Corning Costar) at a Berlin, Germany) every 30 min. After 3 h the

Table 3 Plasmids used in this study


Plasmid Description Reference/Source
pEPR1_spaApromoter promoter sequence (500 bp) of C. diphtheriae spaA gene This study
pEPR1_spaGpromoter promoter sequence (500 bp) of C. diphtheriae spaG gene This study
pEPR1_spaHpromoter promoter sequence (500 bp) of C. diphtheriae spaH gene This study
pEPR1_spaIpromoter promoter sequence (500 bp) of C. diphtheriae spaI gene This study
pEPR1 gfpuv, KmR, rep, per, T1, T2 [27]

44
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 11 of 12
http://www.biomedcentral.com/1471-2180/10/257

supernatant of infected D562 cells was removed and the antibodies Alexa-Fluor 488 (green) goat anti-rabbit IgGs
cells were incubated in fresh DMEM overnight to avoid and Alexa-Fluor 568 (red) goat anti-rabbit IgGs were
detrimental effects of excessive bacterial growth. used. Phalloidin Alexa-Fluor 647 was used for staining
the cytoskeleton of D562 cells. All antibodies were
Adhesion assays diluted in blocking solution (2% goat serum, 2% BSA in
D562 cells were seeded in 24 well plates (bio-one Cell- PBS). Cell lines were seeded on round coverslips in 24
star, Greiner, Frickenhausen, Germany) at a density of 2 well plates 48 h prior to infection and fixed after the
× 10 5 cells per well 48 h prior to infection. Bacteria respective assay with 3% PFA in PBS (10 min at room
were subcultured (OD600 of 0.1 from overnight cultures) temperature). For immuno-fluorescence staining the
in HI broth for 3.5 h and adjusted to an OD600 of 0.2. A preparations were washed thrice with 1 × PBS and incu-
master mix of the inoculum was prepared in DMEM bated with primary antibodies for at least 1 h at room
without penicillin/streptomycin at a MOI of 200 (viable temperature, washed thrice with PBS again, and subse-
counts experiments). The plates were centrifuged for 5 quently incubated with Alexa-Fluor 488 (green) goat
min at 500 × g to synchronize infection and subse- anti-rabbit for 45 min. After permeabilization with 0.1%
quently incubated for 1.5 h. The cells were washed with Triton X-100 (5 min room temperature) and three
PBS nine times, detached with 500 μl trypsin solution washing steps with PBS, staining with Alexa-Fluor 568
(0.12% trypsin, 0.01% EDTA in PBS) per well (5 min, (red) goat anti-rabbit was carried out as described
37°C, 5% CO 2 , 90% humidity) and lysed with 0.025% above. F-actin was stained in parallel with Phalloidin-
Tween 20 for 5 min at 37°C. Serial dilutions were made Alexa-Fluor 647 (blue). Coverslips were mounted on
in pre-chilled 1 × PBS and plated on blood agar plates glass slides using Fluoroprep (Biomerieux, Craponne,
to determine the number of colony forming units (cfu). France). Imaging was done on an AxioVert 200 M
From this, the percentage of invasive bacteria was calcu- inverted optical microscope (Carl Zeiss Micromaging
lated [24]. GmbH, Jena, Germany).

Epithelial cell invasion model


Acknowledgements
D562 cells were seeded in 24 well plates (bio-one Cell- The authors wish to thank C. von Hunolstein (Istituto Superiore di Sanità,
star, Greiner, Frickenhausen, Germany) at a density of 2 Rome) for providing strain ISS3319, ISS4060, ISS4746, and ISS4749, as well as
× 105 cells per well 48 h prior to infection. Overnight H. Ton-That (University of Texas Health Science Center, Houston, TX) for
SpaD antibodies, SpaD protein, and chromosomal DNA of strain NCTC13129.
cultures of C. diphtheriae grown in HI were re-inocu- The help of R. G. Gerlach (Mikrobiologisches Institut des Universitätsklinikums
lated to an OD 600 of 0.1 in fresh medium and grown Erlangen) to establish the epithelial resistance assay is gratefully
aerobically for another 3.5 h. An inoculum of approxi- acknowledged. This study was financially supported by the Deutsche
Forschungsgemeinschaft for in frame of SFB 796 (projects B5, C1, and Z).
mately 1.6 × 10 8 bacteria ml -1 (MOI = 200) was pre-
pared in DMEM without penicillin/streptomycin and Author details
1
500 μl per well were used to infect the D562 cells. The Lehrstuhl für Mikrobiologie, Friedrich-Alexander-Universität Erlangen-
Nürnberg, Nürnberg, Germany. 2Lehrstuhl für Angewandte Physik, Friedrich-
plates were centrifuged for 5 min at 500 × g to synchro- Alexander-Universität Erlangen-Nürnberg, Nürnberg, Germany.
nize infection and subsequently incubated for 1.5 h (37° 3
Mikrobiologisches Institut des Universitätsklinikums Erlangen, Erlangen,
C, 5% CO2, 90% humidity). The cells were washed thrice Germany. 4Abteilung Mikrobiologie, Universität Osnabrück, Osnabrück,
Germany.
with PBS and subsequently 500 μl of DMEM containing
100 μg ml-1 gentamicin were applied to each well to kill Authors’ contributions
remaining extracellular bacteria. After 2, 8.5 and 18.5 h LO carried out invasion assays, fluorescence microscopy, TER measurements,
and RNA and protein analyses, MHö carried out adhesion and invasion
of incubation the cell layers were washed thrice with analysis, AFM experiments were carried out in cooperation with JR and TES,
PBS, detached by adding 500 μl trypsin solution (0.12% MHe supported LO and MHö in respect to cell culture, AB supervised the
trypsin, 0.01% EDTA in PBS) per well (5 min, 37°C, 5% experiments of LO and MHö and was responsible for the draft and final
version of the manuscript. All authors read and approved the final
CO2, 90% humidity) and lysed for 5 min at 37°C with manuscript.
0.025% Tween 20 to liberate the intracellular bacteria.
Serial dilutions of the inoculum and the lysates were Received: 11 August 2010 Accepted: 13 October 2010
Published: 13 October 2010
plated on blood agar plates to determine the number of
colony forming units (cfu). References
1. Galazka A: The changing epidemiology of diphtheria in the vaccine era. J
Immuno-fluorescence Infec Dis 2000, 181(suppl 1):S2-S9.
2. Hadfield TL, McEvoy P, Polotsky Y, Tzinserling A, Yakovlev AA: The
For immuno-fluorescence staining an antibody against pathology of diphtheria. J Infect Dis 2000, 181(suppl 1):S116-S120.
the C. diphtheriae surface proteome was used, which 3. von Hunolstein C, Alfarone G, Scopetti F, Pataracchia M, La Valle R,
was raised in rabbits. For antibody generation, surface Franchi F, Pacciani L, Manera A, Giammanco A, Farinelli S, Engler K, De
Zoysa A, Efstratiou A: Molecular epidemiology and characteristics of
proteins were prepared as described [24]. As secondary

45
Publications
Ott et al. BMC Microbiology 2010, 10:257 Page 12 of 12
http://www.biomedcentral.com/1471-2180/10/257

Corynebacterium diphtheriae and Corynebacterium ulcerans strains 24. Ott L, Höller M, Gerlach RG, Hensel M, Rheinlaender J, Schäffer TE,
isolated in Italy during the 1990s. J Med Microbiol 2003, 52:181-188. Burkovski A: Corynebacterium diphtheriae invasion-associated protein
4. Funke G, Altwegg M, Frommel L, von Graevenitz AA: Emergence of related (DIP1281) is involved in cell surface organization, adhesion and
nontoxigenic Corynebacterium diphtheriae biotype mitis strains in internalization in epithelial cells. BMC Microbiol 2010, 10:2.
Western Europe. Emerg Infect Dis 1999, 5:477-480. 25. Sambrook J, Fritsch EF, Maniatis T: Molecular Cloning: A Laboratory
5. Hamour AA, Efstratiou A, Neill R, Dunbar EM: Epidemiology and molecular Manual. Cold Spring Habor Laboratory Press, Cold Spring Habor, NY, 2
characterisation of toxigenic Corynebacterium diphtheriae var mitis from 1989.
a case of cutaneous diphtheria in Manchester. J Infect 1995, 31:153-157. 26. Schägger H, von Jagow G: Tricine-sodium dodecyl sulfate-polyacrylamide
6. Romney MG, Roscoe DL, Bernard K, Lai S, Efstratiou A, Clarke AM: gel eletrophoresis for the separation of proteins in the range from 1 to
Emergence of an invasive clone of nontoxigenic Corynebacterium 100 kDa. Anal Biochem 1987, 166:368-379.
diphtheriae in the urban poor population of Vancouver, Canada. J Clin 27. Knoppová M, Phensaijai M, Veselý M, Zemanova M, Nešvera J, Pátek M:
Microbiol 2006, 44:1625-1629. Plasmid vectors for testing in vivo promoter activities in Corynebacterium
7. Hirata R Jr, Pereira GA, Filardy AA, Gomes DLR, Damasco PV, Rosa ACP, glutamicum and Rhodococcus erythropolis. Curr Microbiol 2007, 55:234-239.
Nagao PE, Pimenta FP, Mattos-Guaraldi AL: Potential pathogenic role of
aggregative-adhering Corynebacterium diphtheriae of different clonal doi:10.1186/1471-2180-10-257
groups in endocarditis. Braz J Med Biol Res 2008, 41:986-991. Cite this article as: Ott et al.: Strain-specific differences in pili formation
8. Puliti M, von Hunolstein C, Marangi M, Bistoni F, Tissi L: Experimental and the interaction of Corynebacterium diphtheriae with host cells. BMC
model of infection with non-toxigenic strains of Corynebacterium Microbiology 2010 10:257.
diphtheriae and development of septic arthritis. J Med Microbiol 2006,
55:229-235.
9. Hirata R Jr, Napoleao F, Monteiro-Leal LH, Andrade AFB, Nagao PE,
Formiga LCD, Fonseca LS, Mattos-Guaraldi AL: Intracellular viability of
toxigenic Corynebacterium diphtheriae strains in HEp-2 cells. FEMS
Microbiol Lett 2002, 215:115-119.
10. Bertuccini L, Baldassarri L, von Hunolstein C: Internalization of non-
toxigenic Corynebacterium diphtheriae by cultured human respiratory
epithelial cells. Microbial Path 2004, 37:111-118.
11. Gaspar AH, Ton-That H: Assembly of distinct pilus structures on the
surface of Corynebacterium diphtheriae. J Bacteriol 2006, 188:1526-1533.
12. Swierczynski A, Ton-That H: Type III pilus of corynebacteria: pilus length is
determined by the level of its major pilin subunit. J Bacteriol 2006,
188:6318-6325.
13. Mandlik A, Swierczynski A, Das A, Ton-That H: Corynebacterium diphtheriae
employs specific minor pilins to target human pharyngeal epithelial
cells. Mol Microbiol 2007, 64:111-124.
14. Mattos-Guaraldi AL, Formiga LCD, Pereira GA: Cell surface components
and adhesion in Corynebacterium diphtheriae. Micr Infect 2000,
2:1507-1512.
15. Hirata R Jr, Souza SMS, Rocha de Souza CM, Andrade AFB, Monteiro-
Leal LH, Formiga LCD, Mattos-Guaraldi AL: Patterns of adherence to HEp-2
cells and actin polymerization by toxigenic Corynebacterium diphtheriae
strains. Microbial Path 2004, 36:125-130.
16. Colombo AV, Hirata R Jr, Rocha de Souza CM, Monteiro-Leal LH,
Previato JO, Formiga LCD, Andrade AFB, Mattos-Guaraldi AL:
Corynebacterium diphtheriae surface proteins as adhesins to human
erythrocytes. FEMS Microbiol Lett 2001, 197:235-239.
17. de Oliveira Moreira L, Andrade AFB, Vale MD, Souza SMS, Hirata R Jr,
Asad LOB, Asad NR, Monteiro-Leal LH, Previato JO, Mattos-Guaraldi AL:
Effects of iron limitation on adherence and cell surface carbohydrates of
Corynebacterium diphtheriae strains. Applied Environ Microbiol 2003,
69:5907-5913.
18. Gerlach RG, Claudio N, Rohde M, Jäckel D, Wagner C, Hensel M:
Cooperation of Salmonella pathogenicity islands 1 and 4 is required to
breach epithelial barriers. Cell Microbiol 2008, 10:2364-2376.
19. Scott JR, Zähner D: Pili with strong attachment: Gram-positive bacteria
do it differently. Mol Microbiol 2006, 62:320-330.
20. Tauch A: Genomics of industrially and medically relevant corynebacteria.
In Corynebacteria: Genomics and Molecular Biology. Edited by: Burkovski A.
Norfolk, UK, Caister Academic Press; 2008:7-32. Submit your next manuscript to BioMed Central
21. Telford JL, Barocchi MA, Margarit I, Rappuoli R, Grandi G: Pili in Gram- and take full advantage of:
positive pathogens. Nature Rev Microbiol 2006, 4:509-519.
22. Cerdeno-Tarraga AM, Efstratiou A, Dover LG, Holden MTG, Pallen M,
• Convenient online submission
Bentley SD, Besra GS, Churcher C, James KD, De Zoysa A, Chillingworth T,
Cronin A, Dowd L, Feltwell T, Hamlin N, Holroyd S, Jagels K, Moule S, • Thorough peer review
Quail MA, Rabbinowitch E, Rutherford KM, Thomson NR, Unwin L, • No space constraints or color figure charges
Whitehead S, Barrell BG, Parkhill J: The complete genome sequence and
analysis of Corynebacterium diphtheriae NCTC13129. Nucleic Acids Res • Immediate publication on acceptance
2003, 31:6516-6523. • Inclusion in PubMed, CAS, Scopus and Google Scholar
23. Iwaki M, Komiya T, Yamamoto A, Ishiwa A, Nagata N, Arakawa Y,
• Research which is freely available for redistribution
Takahashi M: Corynebacterium diphtheriae C7(-) and PW8 strains: Genome
organization and pathogenicity. Infect Immun 2010, 78(9):3791-800.
Submit your manuscript at
www.biomedcentral.com/submit

46
Publications

3.3 The complete genome sequence of Corynebacterium


pseudotuberculosis FRC41 isolated from a 12-year-old girl with
necrotizing lymphadenitis reveals insights into gene-regulatory
networks contributing to virulence (Trost et al, 2010b)

47
Publications
Trost et al. BMC Genomics 2010, 11:728
http://www.biomedcentral.com/1471-2164/11/728

RESEARCH ARTICLE Open Access

The complete genome sequence of


Corynebacterium pseudotuberculosis FRC41
isolated from a 12-year-old girl with necrotizing
lymphadenitis reveals insights into gene-
regulatory networks contributing to virulence
Eva Trost1,2, Lisa Ott3, Jessica Schneider1,2,4, Jasmin Schröder1, Sebastian Jaenicke4, Alexander Goesmann4,
Peter Husemann5, Jens Stoye5, Fernanda Alves Dorella6, Flavia Souza Rocha6, Siomar de Castro Soares6,
Vívian D’Afonseca6, Anderson Miyoshi6, Jeronimo Ruiz7, Artur Silva8, Vasco Azevedo6, Andreas Burkovski3,
Nicole Guiso9, Olivier F Join-Lambert10, Samer Kayal11, Andreas Tauch1*

Abstract
Background: Corynebacterium pseudotuberculosis is generally regarded as an important animal pathogen that rarely
infects humans. Clinical strains are occasionally recovered from human cases of lymphadenitis, such as
C. pseudotuberculosis FRC41 that was isolated from the inguinal lymph node of a 12-year-old girl with necrotizing
lymphadenitis. To detect potential virulence factors and corresponding gene-regulatory networks in this human
isolate, the genome sequence of C. pseudotuberculosis FCR41 was determined by pyrosequencing and functionally
annotated.
Results: Sequencing and assembly of the C. pseudotuberculosis FRC41 genome yielded a circular chromosome with
a size of 2,337,913 bp and a mean G+C content of 52.2%. Specific gene sets associated with iron and zinc
homeostasis were detected among the 2,110 predicted protein-coding regions and integrated into a gene-
regulatory network that is linked with both the central metabolism and the oxidative stress response of FRC41.
Two gene clusters encode proteins involved in the sortase-mediated polymerization of adhesive pili that can
probably mediate the adherence to host tissue to facilitate additional ligand-receptor interactions and the delivery
of virulence factors. The prominent virulence factors phospholipase D (Pld) and corynebacterial protease CP40 are
encoded in the genome of this human isolate. The genome annotation revealed additional serine proteases,
neuraminidase H, nitric oxide reductase, an invasion-associated protein, and acyl-CoA carboxylase subunits involved
in mycolic acid biosynthesis as potential virulence factors. The cAMP-sensing transcription regulator GlxR plays a
key role in controlling the expression of several genes contributing to virulence.
Conclusion: The functional data deduced from the genome sequencing and the extended knowledge of virulence
factors indicate that the human isolate C. pseudotuberculosis FRC41 is equipped with a distinct gene set promoting
its survival under unfavorable environmental conditions encountered in the mammalian host.

* Correspondence: tauch@cebitec.uni-bielefeld.de
1
Institut für Genomforschung und Systembiologie, Centrum für
Biotechnologie, Universität Bielefeld, Universitätsstraße 27, D-33615 Bielefeld,
Germany
Full list of author information is available at the end of the article

© 2010 Trost et al; licensee BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons
Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in
any medium, provided the original work is properly cited.

48
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 2 of 17
http://www.biomedcentral.com/1471-2164/11/728

Background necrotizing lymphadenitis [12]. This patient had no


Corynebacterium pseudotuberculosis is generally regarded underlying disease or predisposing conditions. The diag-
as an important animal pathogen and the etiological agent nosis of necrotizing lymphadenitis was supported by 16
of a disease that is commonly called caseous S rDNA gene sequencing, a taxonomic classification of
lymphadenitis [1,2]. This bacterium is predominantly iso- the bacterial isolate and Western blot assays revealing
lated from sheep and goats (biovar ovis), but has been the presence of phospholipase D in the patient’s serum.
recognized also in other animals, including horses and cat- The patient relapsed many times despite a surgical drai-
tle (biovar equi) [1]. The importance of caseous lymphade- nage and an adapted antimicrobial treatment, although
nitis varies greatly around the world, but this disease is C. pseudotuberculosis FRC41 turned out to be highly
found in all major sheep and goat production areas [2]. C. susceptible to a wide range of antibiotics in vitro. How-
pseudotuberculosis is a significant cause of morbidity in ever, the patient recovered after a broad spectrum intra-
sheep and goats, and caseous lymphadenitis in these ani- venous antimicrobial therapy with imipenem-cilastatin,
mals resulted in economic losses, for instance in wool, rifampin and ofloxacin for four months, followed by an
milk and meat production [2,3]. C. pseudotuberculosis is a oral therapy with rifampin and ofloxacin for six months
facultative intracellular pathogen that is able to survive [12]. Here, we report the functional annotation of the
and grow in macrophages, thus escaping the immune complete genome sequence of C. pseudotuberculosis
response of the host [1,4]. A close phylogenetic relation- FRC41, the detection of potential virulence factors and
ship between C. pseudotuberculosis and Corynebacterium the deduced gene-regulatory networks controlling their
ulcerans was proposed as both species are unique among expression.
the corynebacteria in producing phospholipase D. The
sphingomyelin-degrading enzyme is regarded as the major Results and discussion
virulence factor for C. pseudotuberculosis [5,6]. This exo- Pyrosequencing and annotation of the C.
toxin promotes the hydrolysis of ester bonds in sphingo- pseudotuberculosis FRC41 genome
myelin in mammalian cell membranes and contributes to The DNA sequence of the C. pseudotuberculosis FRC41
the spread of the bacterium from the initial site of infec- chromosome was determined by a whole-genome shotgun
tion to secondary sites within the host. approach using pyrosequencing. A quarter of a sequencing
Although infections due to C. pseudotuberculosis are run with the Genome Sequencer FLX Instrument yielded
predominantly observed in sheep and goats, infections 286,938 reads and 94,447,635 bases that were assembled
due to this pathogen also occur in humans [7,8]. The into ten large contigs (≥ 500 bases) and one small contig
number of human infections is rare, but it might be (313 bases), indicating a very low number of repetitive
underestimated as corynebacteria are often considered sequences in the C. pseudotuberculosis FRC41 genome. A
as skin contaminants in clinical specimens [9]. Published search for repetitive DNA elements in the complete gen-
cases of human infections by C. pseudotuberculosis ome sequence revealed the absence of insertion sequences
usually presented as suppurative lymphadenitis [7,8], in C. pseudotuberculosis FRC41, whereas the small contig
with the exception of one case of eosinophilic pneumo- was present in three tandem copies in the assembled chro-
nia [10]. Most patients revealed a classical risk exposure mosome (data not shown). The remaining gaps were
of close contact with animals, in particular with sheep. closed by a PCR strategy that was supported by the related
C. pseudotuberculosis infects humans via superficial reference contig arrangement tool r2cat [13], using the
wounds, forming abscesses in the regional draining Corynebacterium diphtheriae NCTC 13129 genome
lymph nodes after an incubation period ranging from sequence as a reference [14]. The final assembly of the
weeks to months. In most of the published cases, anti- DNA sequences yielded a circular chromosome with a size
biotic treatment alone was unsuccessful and the antimi- of 2,337,913 bp and a mean G+C content of 52.2% (Figure
crobial therapy was therefore supplemented by surgical 1A). Considering the final size of the C. pseudotuberculosis
interventions [7,8]. The general problem in achieving an FRC41 chromosome, a 40-fold coverage was initially
effective treatment of C. pseudotuberculosis infections in obtained by pyrosequencing.
humans and animals is probably related to the faculta- The annotation of the C. pseudotuberculosis FRC41
tive intracellular lifestyle of this bacterium, as it can sur- genome sequence was performed with the GenDB soft-
vive and multiply in macrophages [4]. The cell death ware system [15] and resulted in the detection of 2,110
and subsequent release of the pathogen lead to necrotic protein-coding regions. Furthermore, 49 tRNA genes
lesions and the formation of thick collagen capsules that were predicted by the tRNAscan-SE program [16] and
cannot be penetrated by antibiotics [11]. four rrn operons were detected on the leading strands
In the present study, we characterize the genome of of the chromosome (Figure 1B). A plot of the calculated
C. pseudotuberculosis FRC41 that was isolated from the G/C skew [(G-C)/(G+C)] indicated a bi-directional repli-
inguinal lymph node of a 12-year-old French girl with cation mechanism of the C. pseudotuberculosis

49
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 3 of 17
http://www.biomedcentral.com/1471-2164/11/728

Figure 1 Annotation and analysis of the C. pseudotuberculosis FRC41 genome sequence. (A), Plot of the annotated C. pseudotuberculosis
FRC41 chromosome. The circles represent from the outside: circle 1, DNA base position [kb]; circle 2, protein-coding regions transcribed
clockwise; circle 3, protein-coding regions transcribed anticlockwise; circle 4, G/C skew plotted using a 10-kb window; circle 5, G+C content
plotted using a 10-kb window. The protein-coding regions are coloured according to their functional classification into the Clusters of
Orthologous Groups of proteins [111]. (B), Architecture imparting sequences in the C. pseudotuberculosis FRC41 chromosome. The distribution of
the octamers G(A/T/C)GGGGGA and (T/C)GGGGGAG on the leading and lagging strands is shown. The origin of replication (oriC) is marked. The
deduced dif locus is located at around 1.1 Mbp of the chromosomal map. The 28-bp sequence of the predicted dif region is shown. The
location of the four rRNA operons (rrnA-rrnD) on the leading strands is indicated. (C), Synteny between the chromosomes of C.
pseudotuberculosis FRC41 and C. diphtheriae NCTC 13129. The X-Y plot is composed of dots forming syntenic regions between both
chromosomes. The dots represent predicted C. pseudotuberculosis FRC41 proteins having an orthologue in the genome of C. diphtheriae NCTC
13129 with co-ordinates corresponding to the position of the respective coding region in each genome sequence and indicated in kb.
Orthologous proteins were detected by reciprocal best BLASTP matches.

chromosome (Figure 1A). According to the presence proteome of C. pseudotuberculosis FRC41 with the com-
and distribution of six conserved DnaA boxes, the oriC plete set of proteins encoded in the genome of
is located downstream of the dnaA coding region [17]. C. diphtheriae NCTC 13129 [14]. This comparative con-
The G/C skew and the biased distribution of architec- tent analysis at the proteome level revealed that 1610
ture imparting sequences (AIMS) on the leading and proteins (76.3%) of C. pseudotuberculosis FRC41 share a
lagging strands indicated the presence of a dif region homologue in the genome of C. diphtheriae NCTC
[18] at the expected position of 180° from oriC, dividing 13129 (data not shown). The characteristic features of
the chromosome of C. pseudotuberculosis FRC41 into C. pseudotuberculosis FRC41 are apparently based on a
two replichores of similar size (Figure 1B). Synteny ana- distinct gene set, defining its lifestyle and pathogenicity,
lysis by reciprocal best matches with BLASTP [19] such as the pld gene encoding phospholipase D [22]. In
revealed a highly conserved order of orthologous genes the following sections, we describe a collection of rele-
between the chromosomes of C. pseudotuberculosis vant genes contributing to the lifestyle and pathogenicity
FRC41 and C. diphtheriae NCTC 13129 (Figure 1C), of C. pseudotuberculosis FRC41 and deduce their inte-
which is consistent with the close phylogenetic relation- gration into a transcriptional gene-regulatory network.
ship of both species [1] and the observation that genetic
rearrangements are rare in the genomes of species The transcriptional regulatory repertoire of C.
belonging to the main lineage of the genus Corynebac- pseudotuberculosis FRC41
terium [20,21]. The calculated reciprocal best BLASTP The repertoire of candidate transcription regulators
hits [19] were used also to compare the predicted encoded in the C. pseudotuberculosis FRC41 genome

50
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 4 of 17
http://www.biomedcentral.com/1471-2164/11/728

was deduced from the functional genome annotation representatives (Figure 2). The largest family of DNA-
taking into account the comprehensive knowledge of binding transcription regulators is TetR with 11 mem-
the reconstructed gene-regulatory network of Coryne- bers, followed by GntR with 5 proteins. The TetR family
bacterium glutamicum ATCC 13032 [23]. A collection of transcription regulators is widely distributed among
of 83 genes encoding DNA-binding transcription regula- bacterial species [27] and is also the most prevalent
tors, sigma factors and response regulators of two- group of regulatory proteins in other corynebacteria
component systems can be regarded as the minimal reg- [25]. It is noteworthy that the detected collection of
ulatory repertoire of C. pseudotuberculosis FRC41 transcription regulators includes only 23 out of the 24
(Figure 2). This set of candidate transcription regulators proteins that hitherto constituted the core of DNA-
represents 3.9% of the predicted protein-coding genes of binding transcription regulators in corynebacteria, as C.
the C. pseudotuberculosis FRC41 genome. This value is pseudotuberculosis FRC41 lacks a gene for an orthologue
in agreement with previous observations that less than of the conserved TetR regulator MfsR (Cg0454) [23,25].
10% of the total number of predicted proteins are asso- The knowledge of the transcriptional regulatory reper-
ciated with transcriptional regulatory processes in toire of C. pseudotuberculosis FRC41 was combined
bacteria [24] and is in the range known from other with regulons contributing to virulence by bioinformatic
pathogenic corynebacteria, such as C. diphtheriae and motif searches for DNA-binding sites of prominent reg-
Corynebacterium jeikeium [25]. The collection of poten- ulatory proteins.
tial transcription regulators was grouped into regulatory
protein families according to their amino acid sequence Iron regulation in physiology and virulence of C.
similarities and domain organizations [26]. This bioin- pseudotuberculosis
formatic classification assigned the candidate transcrip- For most bacteria, iron is essential as a cofactor for
tion regulators to 31 regulatory protein families, with proteins involved in important cellular functions, such
one regulator (cpfrc_01413) remaining unclassified. The as DNA biosynthesis and respiration [28]. Thus, iron
regulatory protein families detected in C. pseudotubercu- acquisition is a vital function for bacterial survival. As
losis FRC41 vary significantly in their number of iron limitation is a common strategy by which a

Figure 2 Classification of the predicted transcriptional regulatory repertoire of C. pseudotuberculosis FRC41 into regulatory protein
families. The number of candidate transcription regulators assigned to a regulatory protein family is shown.

51
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 5 of 17
http://www.biomedcentral.com/1471-2164/11/728

mammalian host suppresses bacterial growth, iron has C. pseudotuberculosis, for instance, encode an iron
a decisive role in infectious diseases. On the one hand, uptake system that is regulated by iron in vitro [29]. A
pathogenic bacteria have to compete for iron in the fag mutant showed a reduced virulence in a goat
host so that they can multiply and establish a success- model of caseous lymphadenitis when compared with a
ful infection. On the other hand, they must regulate wild-type control, although no defect in iron utilization
iron metabolism to prevent excess iron that can initi- by the mutant strain was determined [29]. As the
ate the generation of toxic oxygen radicals from nor- expression of the fagABCD genes contributed to the
mal products of metabolism by Fenton chemistry. virulence of C. pseudotuberculosis from animal sources,
Bacteria have solved the problem of iron acquisition the complete genome sequence of FRC41 was screened
and homeostasis by encoding a variety of high-affinity for the presence of these genes, additional iron uptake
uptake systems that are tightly regulated at the tran- systems and the responsible transcription regulator
scriptional level [28]. The fagCBA-fagD genes of (Figure 3).

Figure 3 Regulons involved in iron regulation of C. pseudotuberculosis FRC41. The DtxR and RipA regulons controlling iron homeostasis
and the respective gene-regulatory interactions were deduced from a genome-scale network transfer approach [114] and the combined use of
hidden Markov models and position weight matrices [74]. The assignment of the transcription regulators into the regulatory protein families of
C. pseudotuberculosis FRC41 is indicated. Predicted DNA-binding sites are listed by sequence and are shown as black boxes, regulated target
genes are shown as arrows and coloured as follows: grey, regulatory gene; yellow, gene involved in iron uptake and iron storage; orange, gene
involved in central metabolism; red, gene contributing to the oxidative stress response. The highly conserved coregulation of the hmuO gene by
DtxR and the response regulators HrrA and ChrA in corynebacteria is indicated [23]. The 19-bp consensus sequence of the DtxR-binding site of
C. pseudotuberculosis FRC41 is shown as DNA sequence logo. The 16 predicted DNA-binding sites of DtxR were used as input data for the
WebLogo tool [115].

52
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 6 of 17
http://www.biomedcentral.com/1471-2164/11/728

The dtxR gene of C. pseudotuberculosis FRC41 product of the ciuE gene is similar to aerobactin bio-
encodes a homologue of the diphtheria toxin repressor synthesis enzymes [39]. The expression of the ciuA gene
DtxR that is activated by iron and controls a complex encoding the lipoprotein receptor of the ABC transport
gene-regulatory network involved in iron homeostasis in system has been detected in vivo by a reporter transpo-
corynebacteria [30,31]. The detection of a putative son system [40]. The ciu gene region detected in the
DtxR-binding site in the fagA-fagD intergenic region genome of C. pseudotuberculosis FRC41 is similar to the
supports the previously observed differential expression ciu gene locus from C. diphtheriae NCTC 13129, with
pattern of the fagABC operon in vitro [29]. We com- the exception that it lacks the ciuG gene, encoding a
bined the functional annotation of the C. pseudotubercu- protein of unknown function [41].
losis FRC41 genome sequence with a bioinformatic The second DtxR-regulated gene cluster related to
motif search for DtxR-binding sites using a hidden Mar- siderophore biosynthesis and excretion includes four
kov model and a position weight matrix with input data genes probably constituting the biosynthesis pathway
from the DtxR regulon of C. glutamicum [31]. In this (ogs, ocd, odc and tsb), a gene encoding an efflux pro-
way, additional genes were assigned to the DtxR regulon tein (mdtK) and four genes encoding an ABC-type
of C. pseudotuberculosis FRC41, including several gene transporter (stsABCD) (Figure 3). Ornithine cyclodea-
clusters involved in the utilization of various host com- minase (ocd) and ornithine decarboxylase (odc) as well
pounds as iron sources (Figure 3). One gene region as monooxygenase (ogs) and synthetase (tsb) functions
revealed similarity to the hemin utilization system Hmu- encoded in this gene cluster are components of widely
TUV from C. diphtheriae [32]. The corresponding distributed routes for siderophore biosynthesis [39].
hemin binding protein HtaA is probably associated with Additional DtxR-binding sites were detected in front of
the cell envelope and involved in the utilization of heme ftn (ferritin) and fas (fatty acid synthase) and the
iron [33]. Two gene clusters assigned to the DtxR regu- sdhCAB (succinate dehydrogenase) operon that are
lon of C. pseudotuberculosis FRC41 include htaA-like also part of the DtxR regulon in C. glutamicum
genes (htaD and htaF) that are associated with genes [31,42]. Ferritins act primarily in iron storage and are
encoding membrane proteins (htaE and htaG), suggest- thus central to the natural regulation of iron in the
ing a role of these clusters in the acquisition of iron bacterial cell [28].
from the host (Figure 3). The hmuO gene of the DtxR Furthermore, the ripA gene encoding a DNA-binding
regulon encodes heme oxygenase that releases iron from transcription regulator of the AraC protein family was
the protoporphyrin ring of heme and facilitates the assigned to the DtxR regulon of C. pseudotuberculosis
acquisition of iron from heme and hemoglobin [34]. FRC41 (Figure 3). The expression of the orthologous
Because of the potential toxicity of both, iron and heme, ripA gene from C. glutamicum is also controlled directly
the expression of hmuO in C. diphtheriae is under com- by DtxR [42]. Under iron limitation, the RipA protein
plex control, comprising the iron-responsive repressor acts as a repressor of genes coding for iron proteins in
DtxR and the heme-dependent activators ChrA and C. glutamicum [42]. Candidate RipA-binding sites were
HrrA that are part of the two-component signal trans- detected by bioinformatic pattern searches in the gen-
duction systems ChrA-ChrS and HrrA-HrrS [35,36]. A ome sequence of C. pseudotuberculosis FRC41 in front
BLAST search across the C. pseudotuberculosis FRC41 of the acn (aconitase) gene and upstream of the sdhCAB
genome revealed two response regulators that share (succinate dehydrogenase) and cydABDC operons (cyto-
similarity with ChrA and HrrA. As the hrrA gene of C. chrome bd-type menaquinol oxidase and ABC-type
pseudotuberculosis FRC41 is part of the DtxR regulon, a transporter), thereby linking the availability of iron with
complex hierarchical control of hmuO gene expression the expression of genes in the citrate cycle and the
might be established in this bacterium (Figure 3). respiratory energy metabolism of C. pseudotuberculosis.
Iron acquisition can moreover involve the synthesis Due to its high oxygen affinity, the cytochrome bd oxi-
and secretion of high-affinity iron chelators, termed dase is used in many bacteria under microaerobic
siderophores, which are synthesized by nonribosomal growth conditions [43]. Additional RipA-binding sites
peptide synthetases or by biosynthesis pathways inde- were detected in front of the katA (catalase) gene and in
pendent of these multimodular enzymes [37,38]. The the dps-fpg1 (DNA protection during starvation protein,
genome of C. pseudotuberculosis FRC41 contains two formamidopyrimidine-DNA glycosylase) intergenic
DtxR-regulated gene clusters that are probably asso- region, integrating protective functions into the RipA
ciated with pathways for siderophore biosynthesis inde- regulon (Figure 3). Dps-like proteins effectively protect
pendent of nonribosomal peptide synthetases (Figure 3). DNA against oxidizing agents by nonspecific DNA-bind-
The ciu locus comprises the ciuABCD (ABC-type trans- ing and physical sequestration that limits DNA accessi-
porter), ciuE (siderophore biosynthesis-related protein) bility to detrimental factors [44]. They also act as
and ciuF (putative efflux protein) genes. The predicted iron-binding and storage proteins and catalyze the

53
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 7 of 17
http://www.biomedcentral.com/1471-2164/11/728

oxidation of ferrous iron to ferric iron by H2O2, which the enzymatic activity of catalase [56]. Superoxide dis-
prevents the formation of hydroxyl radicals by the Fen- mutases were classified into three evolutionarily distinct
ton reaction [45]. The lsr2 gene of C. pseudotuberculosis families according to the type of metal cofactors. Most
FRC41 encodes the multifunctional histone-like protein bacteria possess either a manganese-dependent (Mn-
and transcription regulator Lsr2 (Figure 2) that shares a SOD) or an iron-dependent (Fe-SOD) superoxide
number of physical properties with Dps-like proteins dismutase in their cytoplasm, while secreted copper,
and the ability to bind DNA sequences with little speci- zinc-dependent superoxide dismutases (Cu,Zn-SODs)
ficity [46]. Like Dps, the Lsr2 protein may physically have been detected in pathogenic or endosymbiontic
protect corynebacterial DNA against reactive oxygen bacteria [57]. The genome of C. pseudotuberculosis
intermediates [47]. Formamidopyrimidine-DNA glycosy- FRC41 encodes two types of superoxide dismutases, a
lase is a primary participant in the repair of DNA cytoplasmic Mn-SOD (SodA) and a secreted Cu,Zn-
lesions caused by oxidative damage [48]. A second gene SOD (SodC) that is characterized by a lipobox motif
(fpg2) encoding formamidopyrimidine-DNA glycosylase and may be anchored in the cell membrane [58]. The
was detected in the genome of C. pseudotuberculosis extracellular location of SodC suggests that it may pro-
FRC41, but the upstream region of fpg2 apparently lacks tect the surface of C. pseudotuberculosis cells against
a RipA-binding site. Catalase is another important pro- superoxide generated externally by the mammalian host
tective component in the bacterial oxidative stress cells. Likewise, the mycobacterial SodC protein contri-
response which is involved in the detoxification of H2O2 butes to the resistance of Mycobacterium tuberculosis
[49]. Hence, the gene composition of the DtxR-RipA against oxidative burst products generated by activated
gene-regulatory network revealed the interdependence macrophages [59,60]. The protective activity of Cu,Zn-
of iron metabolism and oxidative stress response and SODs has been associated with virulence in many bac-
the regulatory connection between distinct physiological teria, such as Neisseria meningitidis and Haemophilus
functionalities of the corynebacterial cell, including an ducreyi [61,62]. However, further experimental work is
important role in virulence of C. pseudotuberculosis. necessary to elucidate which protective enzyme contri-
butes to the virulence of C. pseudotuberculosis.
Manganese and zinc regulation in physiology and As manganese and zinc ions are apparently involved as
virulence of C. pseudotuberculosis cofactors in the oxidative stress response of pathogens, the
Peroxynitrite and other reactive nitrogen and oxygen corresponding regulons involved in metal ion uptake were
intermediates are produced by macrophages as part of identified in the genome sequence of C. pseudotuberculosis
their antimicrobial response [50]. Consequently, many FRC41 (Figure 4B). The uptake of manganese in C. pseu-
pathogenic bacteria have evolved protection mechanisms dotuberculosis FRC41 is mediated by a typical manganese
against these reactive nitrogen and oxygen intermediates ABC-type transport system (mntABCD) that is negatively
that have potent antimicrobial activity [51]. Four genes controlled at the transcriptional level by the metalloregula-
encoding protective enzymes probably involved in corre- tor MntR (Figure 4B). The manganese ABC transporter is
sponding detoxification reactions were identified in the composed of: a lipoprotein receptor (MntA), anchored to
genome sequence of C. pseudotuberculosis FRC41, the cell membrane and functioning as an extracellular
including alkyl hydroperoxide reductase (ahpCD), man- cation-binding protein; a cytoplasmic ATP-binding protein
ganese-dependent superoxide dismutase (sodA) and cop- (MntB); and two integral membrane proteins (MntC and
per, zinc-dependent superoxide dismutase (sodC). A MntD) that mediate the cation flux [63]. A homologous
bioinformatic pattern search with actinobacterial OxyR- gene cluster and its manganese-dependent transcriptional
binding sites as input data revealed that these genes are control by the metalloregulator MntR have been examined
most likely under transcriptional control by OxyR (Fig- in the genome of C. diphtheriae [64]. The uptake of zinc
ure 4A). The oxyR gene of C. pseudotuberculosis FRC41 ions in C. pseudotuberculosis FRC41 is probably mediated
is linked to the ahpCD genes and its gene product may by two ABC transport systems (znuB1C1A1 and znu-
act as a repressor of gene expression [52]. AhpC is a B2A2C2), as both gene regions are specified by the pre-
member of a large family of peroxidases that contribute sence of Zur-binding sites (Figure 4C). The Zur protein is
to the antioxidant defense in bacteria [53]. The AhpC a metalloregulator of the ferric uptake regulator (FUR)
protein directly reduces peroxides and is in turn family of DNA-binding transcription regulators [65,66].
reduced by AhpD [54]. The mycobacterial AhpC protein The znu ABC transporter genes are key components of
also catalyzes the rapid conversion of peroxynitrite to actinobacterial Zur regulons and their expression is gener-
nitrite to avoid the formation of deleterious nitrogen ally repressed by Zur in a zinc-dependent manner [66].
dioxide and hydroxyl radicals [55]. Superoxide dismu- Zinc resistance might be facilitated in C. pseudotuberculo-
tase converts superoxide anions into molecular oxygen sis FRC41 by the ArsR-type transcription regulator Znr
and H 2 O2 , the latter being broken in turn to H2 O by that probably controls the expression of the czcE gene

54
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 8 of 17
http://www.biomedcentral.com/1471-2164/11/728

Figure 4 Regulons involved in the oxidative stress response and in zinc and manganese regulation of C. pseudotuberculosis FRC41. (A),
The OxyR regulon controlling the oxidative stress response. (B), The MntR operon controlling the uptake of manganese. (C), The Zur regulon
controlling zinc metabolism. The assignment of the transcription regulators into the regulatory protein families of C. pseudotuberculosis FRC41 is
indicated. The regulatory interactions were deduced from a genome-scale network transfer approach [114] and the combined use of a hidden
Markov model and a position weight matrix [74]. Predicted DNA-binding sites are shown as black boxes and indicated by DNA sequence. The
regulated target genes are shown as arrows and coloured as follows: grey, regulatory gene; red, gene contributing to the oxidative stress
response; dark blue, gene involved in maganese uptake; light blue, gene involved in zinc metabolism.

encoding a cobalt/zinc/cadmium efflux system [23,67,68]. may fulfill novel functions in sensing the presence of
The yciC gene coding for a putative P-loop GTPase of the zinc in the environment.
COG0523 protein family is also part of the Zur regulon in
C. pseudotuberculosis FRC41 (Figure 4C). The YciC pro- Genes encoding adhesive pili in C. pseudotuberculosis
tein may function as a metallochaperone/insertase to FRC41
enable the in vivo assembly of zinc-containing proteins The complete set of predicted protein-coding regions of
under environmental conditions of zinc deficiency [69]. C. pseudotuberculosis FCR41 was subsequently screened
Moreover, the genome-wide motif search for Zur- for the presence of further proteins containing a typical
binding sites in C. pseudotuberculosis FRC41 detected sorting signal. This approach revealed ten additional
three genes (cpfrc_00168, cpfrc_00313, cpfrc_02059) proteins without any conserved domain organization
encoding proteins with a carboxy-terminal sorting [56] that were annotated as hypothetical proteins with
(LPxTG) signal that is generally used by Gram-positive LPxTG motif and, more interestingly, six proteins show-
bacteria to anchor proteins to the cell wall [70]. The ing similarity to subunits of adhesive pili from
Cpfrc_00168 protein contains two CnaB-like domains C. diphtheriae NCTC 13129 (Figure 5). The correspond-
that may be involved in the positioning of a ligand bind- ing coding regions are organized in two gene clusters
ing domain away from the corynebacterial cell surface that include sortase genes involved in the process of
and is encoded adjacent to a putative sialoprotein-bind- pilus polymerization [71,72]. The housekeeping sortase
ing protein. The secreted proteins Cpfrc_00313 and gene (cpfrc_02014; srtD) of C. pseudotuberculosis
Cpfrc_02059 contain actinobacterial surface-anchored FCR41, necessary for the cell wall anchoring of pilin
protein domains for their covalent attachment to the monomers and pilus polymers [73], is located elsewhere
cell wall [56]. The Cpfrc_00313 protein is encoded next in the chromosome. The adhesive pili of C. pseudotuber-
to the components of a transporter and may act culosis FCR41 consist of major pilin subunits (SpaA,
together with the Cpfrc_00314 protein as a substrate SpaD), minor pilin subunits (SpaB, SpaE) and tip pro-
receptor for this system. Hence, the reconstruction of teins (SpaC, SpaF) that are characterized by conserved
regulons participating in metal ion uptake of C. pseudo- amino acid motifs (Figure 5). The function of the
tuberculosis FRC41 led to the detection of genes that hypothetical proteins SpaX and SpaY encoded in the

55
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 9 of 17
http://www.biomedcentral.com/1471-2164/11/728

Figure 5 Gene regions encoding adhesive pili of C. pseudotuberculosis FRC41. The gene clusters involved in the synthesis of adhesive (Spa-
like) pili of C. pseudotuberculosis FRC41 are shown. The gene clusters encode sortases required for the assembly of the pilus (blue), major pilins
(red), minor pilins (yellow), pilus tip proteins (green), and proteins of unknown function (grey). The detected sorting (LPxTG) signals are indicated.
Specifically marked in the major pilin proteins are the characteristic pilin boxes (blue) and E-boxes (white). The predicted binding of the
transcription regulator GlxR in the spaA-srtB intergenic region is shown.

spa gene regions of C. pseudotuberculosis FCR41 is cur- genome sequence was screened for further candidate
rently unknown. According to alkaline phosphatase genes encoding virulence factors (Table 1). The major
(phoZ) gene fusions generated with a reporter transpo- virulence factor of C. pseudotuberculosis is the sphingo-
son system in C. pseudotuberculosis, at least the SpaABC myelin-degrading phospholipase D that facilitates the
pilus is expressed in this species [1]. The spaABC gene persistence and spread of the bacterium within the host
cluster contains a putative DNA-binding site for the [5,6]. The expression of the pld gene is regulated by
cAMP-sensing transcription regulator GlxR in the spaA- multiple environmental stimuli, including heat, and
srtB intergenic region (Figure 5), thereby connecting the plays a role in the reduction of macrophage viability fol-
expression of pilus genes with a global gene-regulatory lowing infection [78,79]. The secreted corynebacterial
network in C. pseudotuberculosis [74,75]. The adhesive protease CP40 was detected previously as a protective
pili of C. diphtheriae NCTC 13129 are covalently antigen of C. pseudotuberculosis and shown to be of the
anchored to the cell wall and can mediate the initial serine protease type, although BLAST searches revealed
adhesion to host tissues and other bacterial cells [76]. homology to endoglycosidases [80]. The extracellular
The adherence of C. diphtheriae to pharyngeal epithelial CP40 enzyme may contribute to the virulence of
cells is mediated also by the minor pilin of the adhesive C. pseudotuberculosis by its proteolytic activity, but the
pilus, as this pilin subunit is covalently anchored to the enzymatic activity of CP40 was not detectable in culture
cell wall and can provide tight contact between the bac- supernatants [80]. However, vaccination of sheep with
terial cell and the host tissue in the absence of a pilus this antigen resulted in protection against infection with
shaft [77]. Considering a similar functioning of the pre- C. pseudotuberculosis, probably by affecting directly the
dicted SpaABC and SpaDEF pilus proteins from C. pseu- function of the CP40 protein and indirectly the growth
dotuberculosis, either a complete pilus structure or the of the pathogen [11]. The genome sequence of C. pseu-
minor pilins SpaB and SpaE can probably make the dotuberculosis FRC41 revealed three additional genes
initial contact with host cell receptors to enable addi- encoding secreted proteases, including two subtilisin-
tional ligand-receptor interactions and to facilitate the like serine proteases and one trypsin-like serine protease
efficient delivery of virulence factors and intracellular (Table 1). Extracellular proteases may exhibit a wide
invasion. range of pathogenic potentials when interacting with the
defense mechanisms and tissue components of the host.
Candidate virulence factors of C. pseudotuberculosis Redundant enzymatic systems are moreover suitable to
FRC41 and their integration into a gene-regulatory promote the survival of pathogens under unfavorable
network environmental conditions encountered in the infected
The observation that adhesive pili promote the adher- host [81]. The genome sequence of C. pseudotuberculo-
ence of C. diphtheriae to host tissue suggests that the sis FRC41 also encodes a secreted protein of the SGNH-
SpaABC and SpaDEF pili from C. pseudotuberculosis hydrolase subfamily (Table 1). SGNH-hydrolases are a
FRC41 can be regarded as potential virulence factors diverse family of lipases and esterases which are known
[77]. To extend the view on proteins contributing to the to act as virulence factors in other bacteria, such as
pathogenicity of C. pseudotuberculosis FRC41, the Streptomyces scabies, the causal agent of the potato scab

56
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 10 of 17
http://www.biomedcentral.com/1471-2164/11/728

Table 1 Candidate determinants contributing to virulence of C. pseudotuberculosis FRC41


Identifier Gene Predicted protein function
cpfrc_00029 pld phospholipase D (sphingomyelin-degrading enzyme)
cpfrc_01895 cpp corynebacterial protease CP40 (serine protease)
cpfrc_00397 - secreted subtilisin-like serine protease
cpfrc_01634 - secreted subtilisin-like serine protease
cpfrc_00562 - secreted trypsin-like serine protease
cpfrc_00536 - secreted SGNH-hydrolase
cpfrc_00386 nanH neuraminidase H (sialidase)
cpfrc_01079 rpfI resuscitation-promoting factor interacting protein (D,L-endopeptidase)
cpfrc_00594 rpfA resuscitation-promoting factor A (muralytic enzyme)
cpfrc_00679 rpfB resuscitation-promoting factor B (muralytic enzyme)
cpfrc_00128 nor nitric oxide reductase
cpfrc_00565 nrpS1 nonribosomal peptide synthetase 1
cpfrc_01801 nrpS2 nonribosomal peptide synthetase 2
cpfrc_00492 dtsR1 acetyl-CoA carboxylase b-subunit involved in fatty acid synthesis
cpfrc_00491 dtsR2 acyl-CoA carboxylase b-subunit involved in mycolic acid synthesis
cpfrc_01953 accD3 acyl-CoA carboxylase b-subunit involved in mycolic acid synthesis

disease [56]. Further experimental studies are required adhesion of corynebacteria [86]. The homologue of RpfI
to elucidate whether the expression of the secreted in M. tuberculosis (named RipA) revealed endopeptidase
enzymes promotes the virulence of C. pseudotuberculo- activity and interacts with the resuscitation-promoting
sis FRC41. factor RpfB, representing a peptidoglycan glycosidase
Another candidate virulence factor of C. pseudotuber- [86,87]. Two genes (rpfA and rpfB) encoding resuscita-
culosis FRC41 is the extracellular neuraminidase NanH tion-promoting factors are present in the genome of
(Table 1). Neuraminidases, or sialidases, belong to a C. pseudotuberculosis FRC41 (Table 1). Important roles
class of glycosyl hydrolases that catalyze the removal of in pathogenesis for peptidoglycan hydrolytic enzymes
terminal sialic acid residues from a variety of glycocon- have been proposed [88] and an analogous system com-
jugates and can contribute to the recognition of sialic bining the activities of a muramidase and an endopepti-
acids exposed on host cell surfaces [82,83]. The homolo- dase contributed to the virulence of Listeria
gous counterpart of NanH was recently characterized in monocytogenes [89]. As previously demonstrated in
C. diphtheriae KCTC3075 and shown to be a protein C. glutamicum [74,90], the expression of rpfI, rpfA and
containing neuraminidase and trans-sialidase activities rpfB in C. pseudotuberculosis FRC41 is probably under
[84]. Trans-sialidases located on the bacterial cell sur- complex control by three regulatory proteins, GlxR,
face can be used for the decoration of sugar moiety RamB and RamA (Figure 6).
acceptors with sialic acid to enable the invasion of hosts Another potential virulence factor of C. pseudotuber-
under certain conditions. The trans-sialidase activity is culosis FRC41 is represented by the nor gene encoding
of importance for many pathogenic bacteria and the nitric oxide reductase (Table 1). This enzyme is gener-
corresponding proteins are therefore considered poten- ally involved in the detoxification of nitric oxide and
tial virulence factors [82]. Iron limitation reduced the consequently necessary for the long-term persistence of
number of sialic acid residues on the surface of C. pathogens in macrophages [91]. The toxic properties of
diphtheriae cells and their adhesive properties, indicat- nitric oxide are used by the host immune system to kill
ing that the expression of dissimilar virulence determi- or slow down the growth of pathogenic bacteria [51].
nants is coordinately controlled by a gene-regulatory Interestingly, the expression of the nor gene was not
system [85]. By bioinformatic pattern searches, a GlxR- induced upon the infection of macrophages by animal
binding site was detected in the upstream region of the C. pseudotuberculosis [4]. As the expression of nor is
nanH gene of C. pseudotuberculosis FRC41, suggesting typically activated by a transcription regulator in
that the cAMP-sensing transcription regulator GlxR response to the presence of nitric oxide [92], the regula-
might be involved in the control of this virulence factor tory pattern of nor transcription and its contribution to
gene (Figure 6). the protection against nitric oxide remains unclear.
Likewise, a DNA-binding site for GlxR was detected in The previous search for macrophage-induced genes of
front of the rpfI gene encoding an invasion-associated animal C. pseudotuberculosis by means of a cloned pro-
protein that is involved in cell surface organization and moter library provided two gene tags showing significant

57
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 11 of 17
http://www.biomedcentral.com/1471-2164/11/728

Figure 6 Regulatory interactions involved in the control of potential virulence factors of C. pseudotuberculosis FRC41. Transcription
regulators controlling the expression of candidate virulence factors are shown. The regulatory interactions were deduced from a genome-scale
network transfer approach [114] and the combined use of a hidden Markov model and a position weight matrix [74]. The assignment of the
transcription regulators into the regulatory protein families of C. pseudotuberculosis FRC41 is indicated. Predicted DNA-binding sites of RamA and
RamB are shown as white boxes and predicted DNA-binding sites of GlxR as black boxes. Regulated target genes are shown as arrows and
coloured as follows: grey, regulatory gene; yellow, gene involved in iron storage; orange, gene of central metabolism or fatty acid/mycolic acid
biosynthesis; violet, gene involved in the resuscitation process; light green, gene involved in the assembly of adhesive pilus; dark green, other
potential virulence factor gene. The 16-bp consensus sequence of the GlxR-binding site of C. pseudotuberculosis FRC41 is shown as DNA
sequence logo. The 17 predicted DNA-binding sites of GlxR were used as input data for the WebLogo tool [115].

58
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 12 of 17
http://www.biomedcentral.com/1471-2164/11/728

induction rates in macrophages [4]. The nucleotide of C. pseudotuberculosis in suspension, the strong aggre-
sequence of the respective gene tags revealed similarity gation within macrophages, the formation of thick
to nonribosomal peptide synthetases (44-fold induction) capsules after the release of the pathogen from macro-
and to the b-subunit of acyl-CoA carboxylases (24-fold phages [11], and the occurrence of virulent and attenu-
induction), respectively. The genome sequence of C. ated strains [98].
pseudotuberculosis FRC41 encodes two nonribosomal
peptide synthetases, NrpS1 and NrpS2 (Table 1). These Conclusions
modular enzymes are used by microorganisms to partici- The complete genome sequence of C. pseudotuberculosis
pate in the synthesis of many secondary metabolites, FRC41 of human origin provides detailed insights into
including for instance siderophores and antibiotics [93]. the gene repertoire contributing to the virulence of this
As both nrpS genes were not assigned to the iron-respon- bacterium that was isolated from a rare case of necrotiz-
sive DtxR regulon of C. pseudotuberculosis FRC41 and ing lymphadenitis [12]. By combining in silico data
siderophore biosynthesis is carried out by alternative obtained from the genome annotation with previous
pathways independent of nonribosomal peptide synthe- experimental knowledge, occasional observations on
tases, a physiological role in iron metabolism of the two genes that affect the virulence of C. pseudotuberculosis
proteins cannot be deduced from the current data. How- were integrated into a global view on the pathogenicity
ever, the strong upregulation of gene expression in of this species. The reconstruction of the DtxR regulon
macrophages points toward a protective or toxic function for instance provides a comprehensive set of genes
during the infection of at least one nonribosomal peptide involved in the acquisition of iron and extends the
synthetase [4]. A role in virulence of a secondary metabo- initial observation that a fagB mutant was not impaired
lite produced by a nonribosomal peptide synthetase has in iron uptake [29]. A systematic mutational characteri-
been demonstrated in Streptomyces acidiscabies. This zation of the newly detected iron transporters and side-
phytopathogen produces thaxtomin A which is necessary rophore biosynthesis gene clusters may help to dissect
for the infection of potato tubers [94]. the contribution of each system to the virulence of
Three genes coding for b-subunits of acyl-CoA car- C. pseudotuberculosis FRC41. It is very likely that the
boxylases are present in the genome of C. pseudotuber- ciu siderophore biosynthesis and transport system may
culosis FRC41 (Table 1). These genes are located in complement the uptake of iron by the FagABCD trans-
highly conserved regions of corynebacterial genomes porter under certain environmental conditions, as the
and are essential for either fatty acid synthesis (dtsR1) ciuA gene was shown earlier to be expressed in C. pseu-
or mycolic acid synthesis (dtsR2 and accD3) [95]. The dotuberculosis [1]. Likewise, a reporter transposon sys-
acetyl-CoA carboxylase of C. glutamicum consists of the tem indicated that a gene coding for a fimbrial subunit
biotinylated a-subunit AccBC, the b-subunit AccD1 is expressed during growth of C. pseudotuberculosis in
(DtsR1) and the small AccE protein. The acyl-CoA car- standard medium [40]. As the genome sequence of
boxylase involved in mycolic acid synthesis of C. gluta- C. pseudotuberculosis FRC41 revealed two gene clusters
micum consists of the two b-subunits AccD2 and encoding adhesive pili, it is interesting to examine how
AccD3 (DtsR2 and AccD3) in addition to AccBC and the respective pilin monomers or pilus polymers contri-
AccE [95,96]. The expression of the respective genes in bute to the adherence of C. pseudotuberculosis to host
C. pseudotuberculosis FRC41 is probably controlled by tissue and how their expression is controlled. We
the global transcription regulator GlxR (Figure 6), pro- detected a DNA-binding site for the cAMP-sensing
viding hints that this regulatory protein has a key func- transcription regulator GlxR in the spaA-srtB intergenic
tion in connecting both the central metabolism and the region, whereas the iron-responsive regulator DtxR was
expression of candidate virulence factors in this patho- proposed to control the assembly of pilin subunits in
genic bacterium. Mycolic acids, which are long-chain a- C. diphtheriae NCTC 13129 [100]. Another previous
alkyl-b-hydroxy fatty acids, are important components study provided two DNA sequence tags for macro-
of the corynebacterial cell wall [97] and probably asso- phage-induced genes [4] that were linked to the nonri-
ciated with the pathogenicity of C. pseudotuberculosis bosomal peptide synthetase genes nrpS1 and nrpS2 and
[98]. They can provide a thick layer at the outer surface to the acyl-CoA carboxylase subunit genes dtsR1, dtsR2
of the cell that protects the bacterium from antibiotics and accD3 by means of the genome sequence of
and the host’s immune system [99]. Moreover, the non- C. pseudotuberculosis FRC41. The detection of the latter
covalently bound trehalose dimycolate is a well-estab- genes is consistent with the observation that the cell
lished immunostimulatory compound with toxic surface of C. pseudotuberculosis is an important factor
properties [97]. Variations in the amount of mycolic contributing to virulence [98]. Future work should com-
acids and differences in other cell surface properties pare the knowledge deduced from the genome sequence
provide a basis for explaining the aggregation capacity of C. pseudotuberculosis FRC41 with the genetic

59
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 13 of 17
http://www.biomedcentral.com/1471-2164/11/728

information generated from other human isolates or ani- Software (Version 2.3). According to the 454 Newbler
mal pathogens of the different biovars [101], or even Metrics file, 286,938 reads representing 94,447,635 bases
extend the genome comparison to the pangenomic level. were assembled. Using the default cutoff of 500 bases
This global comparative approach with a larger set of for the size classification of the contigs, ten large contigs
sequenced genomes may provide comprehensive insights (≥ 500 bases) and one small contig (313 bases) were
into the distinctive features of each biovar or strain. As obtained to give a total size of 2,319,243 bases. The gap
the currently available commercial vaccines are unable closure process was supported by the related reference
to fully protect susceptible animals [102,103], the contig arrangement tool r2cat, using the C. diphtheriae
extended knowledge of potential virulence factors and NCTC 13129 genome sequence as a reference [13]. The
novel antigens of C. pseudotuberculosis might be helpful remaining gaps in the genome sequence were closed by
for the design of more effective vaccines and molecular PCR with Phusion hot start high-fidelity DNA polymer-
diagnostics to control caseous lymphadenitis in sheep ase (Finnzymes) and genomic template DNA. All pri-
and goats to reduce thereby the occupational risk expo- mers used in this study were synthesized by Metabion.
sure for humans. The PCR assays were carried out with a TProfessional
PCR thermocycler (Biometra) according to standard
Methods protocols (Finnzymes). The amplified DNA fragments
Bacterial strain and growth conditions linking the individual contigs were sequenced by IIT
C. pseudotuberculosis FRC41 was isolated from the ingu- Biotech. All DNA sequences were uploaded into the
inal lymph node of a 12-year-old French girl with necro- Consed program [104] to generate the complete genome
tizing lymphadenitis [12]. The taxonomic identification sequence of C. pseudotuberculosis FRC41.
of this clinical isolate was recently confirmed using mul-
tiplex PCR and the nucleotide sequence of the pld gene Bioinformatic analysis of the complete genome sequence
(N. Guiso, unpublished data). C. pseudotuberculosis The assembled sequence of C. pseudotuberculosis FRC41
FRC41 was routinely grown in brain-heart-infusion (BHI) was uploaded into the bacterial genome annotation sys-
broth or on Columbia agar with sheep blood at 37°C. tem GenDB [15]. The annotation of the complete gen-
ome sequence was performed as described previously
Preparation of chromosomal DNA for genome sequencing [105], followed by manual curation. Analyses of the pre-
The preparation of chromosomal DNA from C. pseudo- dicted gene content and the metabolic properties of
tuberculosis FRC41 was performed as follows: 50-ml ali- C. pseudotuberculosis FRC41 were accomplished by the
quots of cultures grown for 48-72 h were centrifuged at computer programs EDGAR [19] and CARMEN [106].
4°C and 2,000 × g for 20 min. The cell pellets were The synteny between the genomes of C. pseudotubercu-
resuspended in 0.6 ml Tris/NaCl buffer [10 mM Tris losis FRC41 and C. diphtheriae NCTC 13129 was calcu-
(pH 7.0), 10 mM EDTA (pH 8.0), 300 mM NaCl] and lated by the EDGAR software [19]. The origin of
transferred to VK01 Precellys lysing tubes. The cells chromosomal DNA replication was predicted with the
were lysed by means of a Precellys 24-Dual Tissue Ori-Finder tool [17]. The genome sequence of C. pseu-
Homogenizer, using two cycles of 6,500 rpm for 15 sec dotuberculosis FRC41 has been deposited in the Gen-
with an interval of 30 sec. The chromosomal DNA was Bank database with accession number CP002097.
purified by phenol/chloroform/isoamyl alcohol (25:24:1)
extraction and precipitated with ethanol. The DNA con- Bioinformatic analysis of the regulatory repertoire
centration was determined with a Tecan Infinite 200 The detection of the transcriptional regulatory reper-
Microplate Reader. toire of C. pseudotuberculosis FRC41 was performed by
a combined bioinformatic approach using several tools
Sequencing of the C. pseudotuberculosis FRC41 genome and programs [26]. Proteins containing putative DNA-
A total of 5 μg of purified chromosomal DNA was used binding domains were detected by means of the HMM
for constructing a single-stranded template DNA library. library and genome assignments server Superfamily ver-
The DNA concentration of the library was measured by sion 1.75 [107]. To identify among the set of potential
using the Agilent RNA 6000 Nano Kit. The preparation DNA-binding proteins those representing transcription
of the single-stranded template DNA library and DNA regulators, hidden Markov model (HMM) profiles of
sequencing were performed according to manufacturer’s regulatory protein families were downloaded from the
protocols (Roche Applied Science). The sequencing of Pfam database version 24.0 [108] and used for searches
C. pseudotuberculosis DNA was carried out with the against the predicted C. pseudotuberculosis FRC41 pro-
Genome Sequencer FLX Instrument and Titanium teins by applying the HMMsearch module of the profile
chemistry (Roche Applied Science). The sequence data hidden Markov model software HMMER [109]. More-
were assembled with the GS de novo Assembler over, the helix-turn-helix (HTH) recognition tool [110]

60
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 14 of 17
http://www.biomedcentral.com/1471-2164/11/728

integrated in the GenDB platform was applied to scan Genominformatik, Technische Fakultät, Universität Bielefeld, Universitätsstraße
25, D-33615 Bielefeld, Germany. 6Laboratório de Genética Celular e
the putative DNA-binding transcription regulators for Molecular, Departamento de Biologia Geral, Instituto de Ciências Biológicas,
the presence of HTH motifs. The classification of Universidade Federal de Minas Gerais, Av. Antonio Carlos 6627, Pampulha,
C. pseudotuberculosis FRC41 proteins into Clusters of Belo Horizonte, MG, Brazil. 7Cellular and Molecular Parasitology Laboratory,
Rene Rachou Research Center, Oswaldo Cruz Foundation (FIOCRUZ), Belo
Orthologous Groups of proteins [111] during genome Horizonte, MG, Brazil. 8Instituto de Ciências Biológicas, Universidade Federal
annotation provided further indications on the role of do Pará, Rua Augusto Corrêa, 01 - Guamá, Belém, PA, Brazil. 9Institut Pasteur,
the predicted proteins in transcriptional regulation. The Unité de Prévention et Thérapies Moléculaires des Maladies Humaines,
National Centre of Reference of Toxigenic Corynebacteria, 25 rue du Dr.
genome-wide search was extended by using data on Roux, 75724 Paris Cedex 15, France. 10Medical Faculty, University Paris
known transcription regulators from C. glutamicum Descartes, Hospital Necker Enfants Malades, Department of Microbiology,
[23]. A validation step was included by performing 147 rue de Sèvres, 75015 Paris, France. 11Medical Faculty, University Rennes
1, Hospital Pontchaillou, Department of Microbiology, 2 rue Henri le Guilloux,
BLASTP [112] searches against the NCBI protein data- 35000 Rennes, France.
base and evaluating results generated with the Con-
served Domain Search program [56]. During the final Authors’ contributions
ET sequenced and annotated the FRC41 genome and prepared the
step of data analysis, the DNA-binding transcription manuscript. JSchn implemented and maintained the GenDB project for
regulators of C. pseudotuberculosis FRC41 were grouped annotation. JSchr participated in the gene-regulatory network analysis. SJ
into regulatory protein families [23,26]. and AG provided bioinformatic support. PH and JSt participated in the gap
closure process. FAD and FSR purified the genomic DNA. LO, SCS, VD, AM,
The detection of DNA-binding sites in the genome JR and AS participated in data evaluation. NG performed the taxonomic
sequence of C. pseudotuberculosis FRC41 followed a classification of FRC41. OFJL and SK discovered FRC41 and provided the
combined workflow, using both position weight matrices isolate for the sequencing project. VA, AB and AT supervised the project. All
authors read and approved the final version of the manuscript.
and hidden Markov models [74]. The programs PoSSuM-
search [113] and HMMsearch [109] were applied to scan Received: 8 September 2010 Accepted: 30 December 2010
the complete genome sequence of C. pseudotuberculosis Published: 30 December 2010
FRC41. As HMMsearch does not support searching in
References
both directions of a double strand, scanning of the 1. Dorella FA, Pacheco LG, Oliveira SC, Miyoshi A, Azevedo V: Corynebacterium
reverse complementary DNA sequence was implemen- pseudotuberculosis: microbiology, biochemical properties, pathogenesis
ted. PoSSuMsearch was configured for lazy probability and molecular studies of virulence. Vet Res 2006, 37:201-218.
2. Baird GJ, Fontaine MC: Corynebacterium pseudotuberculosis and its role in
evaluation [113]. The comprehensive set of validated ovine caseous lymphadenitis. J Comp Pathol 2007, 137:179-210.
DNA-binding sites sequences from C. glutamicum was 3. Paton MW, Rose IR, Hart RA, Sutherland SS, Mercy AR, Ellis TM, Dhaliwal JA:
downloaded from the reference database CoryneRegNet New infection with Corynebacterium pseudotuberculosis reduces wool
production. Aust Vet J 1994, 71:47-49.
[114] and used as input for PoSSuMsearch and 4. McKean S, Davies J, Moore R: Identification of macrophage induced
HMMsearch. The respective gene-regulatory network genes of Corynebacterium pseudotuberculosis by differential fluorescence
transfer between C. glutamicum and other corynebacteria induction. Microbes Infect 2005, 7:1352-1363.
5. McNamara PJ, Bradley GA, Songer JG: Targeted mutagenesis of the
on the genome-scale was described previously [114]. E- phospholipase D gene results in decreased virulence of Corynebacterium
value cut-offs were used as described for the genome- pseudotuberculosis. Mol Microbiol 1994, 12:921-930.
wide pattern recognition approach in C. glutamicum 6. Songer JG: Bacterial phospholipases and their role in virulence. Trends
Microbiol 1997, 5:156-161.
[75]. Sequence logos of the detected DNA-binding sites 7. Bregenzer T, Frei R, Ohnacker H, Zimmerli W: Corynebacterium
were generated with WebLogo version 2.8.2 [115]. pseudotuberculosis infection in a butcher. Clin Microbiol Infect 1997,
3:696-698.
8. Peel MM, Palmer GG, Stacpoole AM, Kerr TG: Human lymphadenitis due to
Acknowledgements Corynebacterium pseudotuberculosis: report of ten cases from Australia
ET and JSchn acknowledge the receipt of scholarships which are embedded and review. Clin Infect Dis 1997, 24:185-191.
into the Cluster Industrial Biotechnology CLIB2021 and financed by Bielefeld 9. Funke G, von Graevenitz A, Clarridge JE, Bernard KA: Clinical microbiology
University and the Ministry of Innovation, Science, Research and Technology of coryneform bacteria. Clin Microbiol Rev 1997, 10:125-159.
of North Rhine-Westphalia. LO and AB were supported by the Deutsche 10. Keslin MH, McCoy EL, McCusker JJ, Lutch JS: Corynebacterium
Forschungsgemeinschaft in frame of SFB 796 (project B5). The valuable pseudotuberculosis. A new cause of infectious and eosinophilic
contribution of the CeBiTec sequencing team (A. Albersmeier, K. Brinkrolf, S. pneumonia. Am J Med 1979, 67:228-231.
Schatschneider, R. Szczepanowski, P. Viehoever, A. Winkler) to this genome 11. Walker J, Jackson HJ, Eggleton DG, Meeusen EN, Wilson MJ, Brandon MR:
project is gratefullly acknowledged. The authors thank the sequencing team Identification of a novel antigen from Corynebacterium
from IIT Biotech for the help during the gap closure process. pseudotuberculosis that protects sheep against caseous lymphadenitis.
Infect Immun 1994, 62:2562-2567.
Author details 12. Join-Lambert OF, Ouache M, Canioni D, Beretti JL, Blanche S, Berche P,
1
Institut für Genomforschung und Systembiologie, Centrum für Kayal S: Corynebacterium pseudotuberculosis necrotizing lymphadenitis in
Biotechnologie, Universität Bielefeld, Universitätsstraße 27, D-33615 Bielefeld, a twelve-year-old patient. Pediatr Infect Dis J 2006, 25:848-851.
Germany. 2CLIB Graduate Cluster Industrial Biotechnology, Centrum für 13. Husemann P, Stoye J: r2cat: synteny plots and comparative assembly.
Biotechnologie, Universität Bielefeld, Universitätsstraße 27, D-33615 Bielefeld, Bioinformatics 2010, 26:570-571.
Germany. 3Lehrstuhl für Mikrobiologie, Friedrich-Alexander-Universität 14. Cerdeno-Tarraga AM, Efstratiou A, Dover LG, Holden MT, Pallen M,
Erlangen-Nürnberg, Staudtstraße 5, D-91058 Erlangen, Germany. Bentley SD, Besra GS, Churcher C, James KD, De Zoysa A, et al: The
4
Bioinformatics Resource Facility, Centrum für Biotechnologie, Universität complete genome sequence and analysis of Corynebacterium diphtheriae
Bielefeld, Universitätsstraße 27, D-33615 Bielefeld, Germany. 5AG NCTC13129. Nucleic Acids Res 2003, 31:6516-6523.

61
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 15 of 17
http://www.biomedcentral.com/1471-2164/11/728

15. Meyer F, Goesmann A, McHardy AC, Bartels D, Bekel T, Clausen J, 37. Crosa JH, Walsh CT: Genetics and assembly line enzymology of
Kalinowski J, Linke B, Rupp O, Giegerich R, Pühler A: GenDB-an open siderophore biosynthesis in bacteria. Microbiol Mol Biol Rev 2002,
source genome annotation system for prokaryote genomes. Nucleic Acids 66:223-249.
Res 2003, 31:2187-2195. 38. Miethke M, Marahiel MA: Siderophore-based iron acquisition and
16. Lowe TM, Eddy SR: tRNAscan-SE: a program for improved detection of pathogen control. Microbiol Mol Biol Rev 2007, 71:413-451.
transfer RNA genes in genomic sequence. Nucleic Acids Res 1997, 39. Challis GL: A widely distributed bacterial pathway for siderophore
25:955-964. biosynthesis independent of nonribosomal peptide synthetases.
17. Gao F, Zhang CT: Ori-Finder: a web-based system for finding oriCs in Chembiochem 2005, 6:601-611.
unannotated bacterial genomes. BMC Bioinformatics 2008, 9:79. 40. Dorella FA, Estevam EM, Pacheco LG, Guimaraes CT, Lana UG, Gomes EA,
18. Hendrickson H, Lawrence JG: Mutational bias suggests that replication Barsante MM, Oliveira SC, Meyer R, Miyoshi A, Azevedo V: In vivo
termination occurs near the dif site, not at Ter sites. Mol Microbiol 2007, insertional mutagenesis in Corynebacterium pseudotuberculosis: an
64:42-56. efficient means to identify DNA sequences encoding exported proteins.
19. Blom J, Albaum SP, Doppmeier D, Pühler A, Vorhölter FJ, Zakrzewski M, Appl Environ Microbiol 2006, 72:7368-7372.
Goesmann A: EDGAR: a software framework for the comparative analysis 41. Kunkle CA, Schmitt MP: Analysis of a DtxR-regulated iron transport and
of prokaryotic genomes. BMC Bioinformatics 2009, 10:154. siderophore biosynthesis gene cluster in Corynebacterium diphtheriae. J
20. Ventura M, Canchaya C, Tauch A, Chandra G, Fitzgerald GF, Chater KF, van Bacteriol 2005, 187:422-433.
Sinderen D: Genomics of Actinobacteria: tracing the evolutionary history 42. Wennerhold J, Krug A, Bott M: The AraC-type regulator RipA represses
of an ancient phylum. Microbiol Mol Biol Rev 2007, 71:495-548. aconitase and other iron proteins from Corynebacterium under iron
21. Trost E, Götker S, Schneider J, Schneiker-Bekel S, Szczepanowski R, Tilker A, limitation and is itself repressed by DtxR. J Biol Chem 2005,
Viehoever P, Arnold W, Bekel T, Blom J, et al: Complete genome sequence 280:40500-40508.
and lifestyle of black-pigmented Corynebacterium aurimucosum ATCC 43. Kana BD, Weinstein EA, Avarbock D, Dawes SS, Rubin H, Mizrahi V:
700975 (formerly C. nigricans CN-1) isolated from a vaginal swab of a Characterization of the cydAB-encoded cytochrome bd oxidase from
woman with spontaneous abortion. BMC Genomics 2010, 11:91. Mycobacterium smegmatis. J Bacteriol 2001, 183:7076-7086.
22. Yozwiak ML, Songer JG: Effect of Corynebacterium pseudotuberculosis 44. Martinez A, Kolter R: Protection of DNA during oxidative stress by the
phospholipase D on viability and chemotactic responses of ovine nonspecific DNA-binding protein Dps. J Bacteriol 1997, 179:5188-5194.
neutrophils. Am J Vet Res 1993, 54:392-397. 45. Zhao G, Ceci P, Ilari A, Giangiacomo L, Laue TM, Chiancone E, Chasteen ND:
23. Schröder J, Tauch A: Transcriptional regulation of gene expression in Iron and hydrogen peroxide detoxification properties of DNA-binding
Corynebacterium glutamicum: the role of global, master and local protein from starved cells. A ferritin-like DNA-binding protein of
regulators in the modular and hierarchical gene regulatory network. Escherichia coli. J Biol Chem 2002, 277:27689-27696.
FEMS Microbiol Rev 2010, 34:685-737. 46. Gordon BR, Li Y, Wang L, Sintsova A, van Bakel H, Tian S, Navarre WW,
24. Rodionov DA: Comparative genomic reconstruction of transcriptional Xia B, Liu J: Lsr2 is a nucleoid-associated protein that targets AT-rich
regulatory networks in bacteria. Chem Rev 2007, 107:3467-3497. sequences and virulence genes in Mycobacterium tuberculosis. Proc Natl
25. Brinkrolf K, Brune I, Tauch A: The transcriptional regulatory network of the Acad Sci USA 2010, 107:5154-5159.
amino acid producer Corynebacterium glutamicum. J Biotechnol 2007, 47. Colangeli R, Haq A, Arcus VL, Summers E, Magliozzo RS, McBride A,
129:191-211. Mitra AK, Radjainia M, Khajo A, Jacobs WR, et al: The multifunctional
26. Brune I, Brinkrolf K, Kalinowski J, Pühler A, Tauch A: The individual and histone-like protein Lsr2 protects mycobacteria against reactive oxygen
common repertoire of DNA-binding transcriptional regulators of intermediates. Proc Natl Acad Sci USA 2009, 106:4414-4418.
Corynebacterium glutamicum, Corynebacterium efficiens, Corynebacterium 48. Zaika EI, Perlow RA, Matz E, Broyde S, Gilboa R, Grollman AP, Zharkov DO:
diphtheriae and Corynebacterium jeikeium deduced from the complete Substrate discrimination by formamidopyrimidine-DNA glycosylase: a
genome sequences. BMC Genomics 2005, 6:86. mutational analysis. J Biol Chem 2004, 279:4849-4861.
27. Ramos JL, Martinez-Bueno M, Molina-Henares AJ, Teran W, Watanabe K, 49. Hassett DJ, Cohen MS: Bacterial adaptation to oxidative stress:
Zhang X, Gallegos MT, Brennan R, Tobes R: The TetR family of implications for pathogenesis and interaction with phagocytic cells.
transcriptional repressors. Microbiol Mol Biol Rev 2005, 69:326-356. Faseb J 1989, 3:2574-2582.
28. Andrews SC, Robinson AK, Rodriguez-Quinones F: Bacterial iron 50. Nathan C, Shiloh MU: Reactive oxygen and nitrogen intermediates in the
homeostasis. FEMS Microbiol Rev 2003, 27:215-237. relationship between mammalian hosts and microbial pathogens. Proc
29. Billington SJ, Esmay PA, Songer JG, Jost BH: Identification and role in Natl Acad Sci USA 2000, 97:8841-8848.
virulence of putative iron acquisition genes from Corynebacterium 51. Zahrt TC, Deretic V: Reactive nitrogen and oxygen intermediates and
pseudotuberculosis. FEMS Microbiol Lett 2002, 208:41-45. bacterial defenses: unusual adaptations in Mycobacterium tuberculosis.
30. Oram DM, Avdalovic A, Holmes RK: Analysis of genes that encode DtxR- Antioxid Redox Signal 2002, 4:141-159.
like transcriptional regulators in pathogenic and saprophytic 52. Storz G, Tartaglia LA: OxyR: a regulator of antioxidant genes. J Nutr 1992,
corynebacterial species. Infect Immun 2004, 72:1885-1895. 122:627-630.
31. Brune I, Werner H, Hüser AT, Kalinowski J, Pühler A, Tauch A: The DtxR 53. Wood ZA, Schroder E, Robin Harris J, Poole LB: Structure, mechanism and
protein acting as dual transcriptional regulator directs a global regulation of peroxiredoxins. Trends Biochem Sci 2003, 28:32-40.
regulatory network involved in iron metabolism of Corynebacterium 54. Guimaraes BG, Souchon H, Honore N, Saint-Joanis B, Brosch R, Shepard W,
glutamicum. BMC Genomics 2006, 7:21. Cole ST, Alzari PM: Structure and mechanism of the alkyl
32. Drazek ES, Hammack CA, Schmitt MP: Corynebacterium diphtheriae genes hydroperoxidase AhpC, a key element of the Mycobacterium tuberculosis
required for acquisition of iron from haemin and haemoglobin are defense system against oxidative stress. J Biol Chem 2005,
homologous to ABC haemin transporters. Mol Microbiol 2000, 36:68-84. 280:25735-25742.
33. Allen CE, Schmitt MP: HtaA is an iron-regulated hemin binding protein 55. Bryk R, Griffin P, Nathan C: Peroxynitrite reductase activity of bacterial
involved in the utilization of heme iron in Corynebacterium diphtheriae. J peroxiredoxins. Nature 2000, 407:211-215.
Bacteriol 2009, 191:2638-2648. 56. Marchler-Bauer A, Anderson JB, Derbyshire MK, DeWeese-Scott C,
34. Schmitt MP: Utilization of host iron sources by Corynebacterium Gonzales NR, Gwadz M, Hao L, He S, Hurwitz DI, Jackson JD, et al: CDD: a
diphtheriae: identification of a gene whose product is homologous to conserved domain database for interactive domain family analysis.
eukaryotic heme oxygenases and is required for acquisition of iron from Nucleic Acids Res 2007, 35:D237-240.
heme and hemoglobin. J Bacteriol 1997, 179:838-845. 57. Merkamm M, Guyonvarch A: Cloning of the sodA gene from
35. Schmitt MP: Transcription of the Corynebacterium diphtheriae hmuO gene Corynebacterium melassecola and role of superoxide dismutase in
is regulated by iron and heme. Infect Immun 1997, 65:4634-4641. cellular viability. J Bacteriol 2001, 183:1284-1295.
36. Bibb LA, Kunkle CA, Schmitt MP: The ChrA-ChrS and HrrA-HrrS signal 58. Babu MM, Priya ML, Selvan AT, Madera M, Gough J, Aravind L, Sankaran K:
transduction systems are required for activation of the hmuO promoter A database of bacterial lipoproteins (DOLOP) with functional
and repression of the hemA promoter in Corynebacterium diphtheriae. assignments to predicted lipoproteins. J Bacteriol 2006, 188:2761-2773.
Infect Immun 2007, 75:2421-2431.

62
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 16 of 17
http://www.biomedcentral.com/1471-2164/11/728

59. Dussurget O, Stewart G, Neyrolles O, Pescher P, Young D, Marchal G: Role 81. Dubin G: Extracellular proteases of Staphylococcus spp. Biol Chem 2002,
of Mycobacterium tuberculosis copper-zinc superoxide dismutase. Infect 383:1075-1086.
Immun 2001, 69:529-533. 82. Mattos-Guaraldi AL, Duarte Formiga LC, Pereira GA: Cell surface
60. Piddington DL, Fang FC, Laessig T, Cooper AM, Orme IM, Buchmeier NA: components and adhesion in Corynebacterium diphtheriae. Microbes Infect
Cu,Zn superoxide dismutase of Mycobacterium tuberculosis contributes 2000, 2:1507-1512.
to survival in activated macrophages that are generating an oxidative 83. Vimr ER, Kalivoda KA, Deszo EL, Steenbergen SM: Diversity of microbial
burst. Infect Immun 2001, 69:4980-4987. sialic acid metabolism. Microbiol Mol Biol Rev 2004, 68:132-153.
61. Wilks KE, Dunn KL, Farrant JL, Reddin KM, Gorringe AR, Langford PR, Kroll JS: 84. Kim S, Oh DB, Kwon O, Kang HA: Identification and functional
Periplasmic superoxide dismutase in meningococcal pathogenicity. Infect characterization of the NanH extracellular sialidase from
Immun 1998, 66:213-217. Corynebacterium diphtheriae. J Biochem 2010, 147:523-533.
62. San Mateo LR, Toffer KL, Orndorff PE, Kawula TH: Neutropenia restores 85. Moreira Lde O, Andrade AF, Vale MD, Souza SM, Hirata R Jr, Asad LM,
virulence to an attenuated Cu,Zn superoxide dismutase-deficient Asad NR, Monteiro-Leal LH, Previato JO, Mattos-Guaraldi AL: Effects of iron
Haemophilus ducreyi strain in the swine model of chancroid. Infect limitation on adherence and cell surface carbohydrates of
Immun 1999, 67:5345-5351. Corynebacterium diphtheriae strains. Appl Environ Microbiol 2003,
63. Papp-Wallace KM, Maguire ME: Manganese transport and the role of 69:5907-5913.
manganese in virulence. Annu Rev Microbiol 2006, 60:187-209. 86. Ott L, Holler M, Gerlach RG, Hensel M, Rheinlaender J, Schaffer TE,
64. Schmitt MP: Analysis of a DtxR-like metalloregulatory protein, MntR, from Burkovski A: Corynebacterium diphtheriae invasion-associated protein
Corynebacterium diphtheriae that controls expression of an ABC metal (DIP1281) is involved in cell surface organization, adhesion and
transporter by an Mn(2+)-dependent mechanism. J Bacteriol 2002, internalization in epithelial cells. BMC Microbiol 2010, 10:2.
184:6882-6892. 87. Kana BD, Mizrahi V: Resuscitation-promoting factors as lytic enzymes for
65. Lee JW, Helmann JD: Functional specialization within the Fur family of bacterial growth and signaling. FEMS Immunol Med Microbiol 2010,
metalloregulators. Biometals 2007, 20:485-499. 58:39-50.
66. Schröder J, Jochmann N, Rodionov DA, Tauch A: The Zur regulon of 88. Boneca IG: The role of peptidoglycan in pathogenesis. Curr Opin Microbiol
Corynebacterium glutamicum ATCC 13032. BMC Genomics 2010, 11:12. 2005, 8:46-53.
67. Grover A, Sharma R: Identification and characterization of a major Zn(II) 89. Lenz LL, Mohammadi S, Geissler A, Portnoy DA: SecA2-dependent
resistance determinant of Mycobacterium smegmatis. J Bacteriol 2006, secretion of autolytic enzymes promotes Listeria monocytogenes
188:7026-7032. pathogenesis. Proc Natl Acad Sci USA 2003, 100:12432-12437.
68. Riccardi G, Milano A, Pasca MR, Nies DH: Genomic analysis of zinc 90. Jungwirth B, Emer D, Brune I, Hansmeier N, Pühler A, Eikmanns BJ, Tauch A:
homeostasis in Mycobacterium tuberculosis. FEMS Microbiol Lett 2008, Triple transcriptional control of the resuscitation promoting factor 2
287:1-7. (rpf2) gene of Corynebacterium glutamicum by the regulators of acetate
69. Haas CE, Rodionov DA, Kropat J, Malasarn D, Merchant SS, de Crecy- metabolism RamA and RamB and the cAMP-dependent regulator GlxR.
Lagard V: A subset of the diverse COG0523 family of putative metal FEMS Microbiol Lett 2008, 281:190-197.
chaperones is linked to zinc homeostasis in all kingdoms of life. BMC 91. Luthra A, Malik SS, Ramachandran R: Cloning, purification and
Genomics 2009, 10:470. comparative structural analysis of two hypothetical proteins from
70. Navarre WW, Schneewind O: Surface proteins of gram-positive bacteria Mycobacterium tuberculosis found in the human granuloma during
and mechanisms of their targeting to the cell wall envelope. Microbiol persistence and highly up-regulated under carbon-starvation conditions.
Mol Biol Rev 1999, 63:174-229. Protein Expr Purif 2008, 62:64-74.
71. Ton-That H, Schneewind O: Assembly of pili in Gram-positive bacteria. 92. Tucker NP, D’Autreaux B, Studholme DJ, Spiro S, Dixon R: DNA binding
Trends Microbiol 2004, 12:228-234. activity of the Escherichia coli nitric oxide sensor NorR suggests a
72. Ton-That H, Marraffini LA, Schneewind O: Sortases and pilin elements conserved target sequence in diverse proteobacteria. J Bacteriol 2004,
involved in pilus assembly of Corynebacterium diphtheriae. Mol Microbiol 186:6656-6660.
2004, 53:251-261. 93. Donadio S, Monciardini P, Sosio M: Polyketide synthases and
73. Swaminathan A, Mandlik A, Swierczynski A, Gaspar A, Das A, Ton-That H: nonribosomal peptide synthetases: the emerging view from bacterial
Housekeeping sortase facilitates the cell wall anchoring of pilus genomics. Nat Prod Rep 2007, 24:1073-1109.
polymers in Corynebacterium diphtheriae. Mol Microbiol 2007, 66:961-974. 94. Healy FG, Wach M, Krasnoff SB, Gibson DM, Loria R: The txtAB genes of the
74. Kohl TA, Baumbach J, Jungwirth B, Pühler A, Tauch A: The GlxR regulon of plant pathogen Streptomyces acidiscabies encode a peptide synthetase
the amino acid producer Corynebacterium glutamicum in silico and in required for phytotoxin thaxtomin A production and pathogenicity. Mol
vitro detection of DNA binding sites of a global transcription regulator. J Microbiol 2000, 38:794-804.
Biotechnol 2008, 135:340-350. 95. Gande R, Dover LG, Krumbach K, Besra GS, Sahm H, Oikawa T, Eggeling L:
75. Kohl TA, Tauch A: The GlxR regulon of the amino acid producer The two carboxylases of Corynebacterium glutamicum essential for fatty
Corynebacterium glutamicum: Detection of the corynebacterial core acid and mycolic acid synthesis. J Bacteriol 2007, 189:5257-5264.
regulon and integration into the transcriptional regulatory network 96. Gande R, Gibson KJ, Brown AK, Krumbach K, Dover LG, Sahm H,
model. J Biotechnol 2009, 143:239-246. Shioyama S, Oikawa T, Besra GS, Eggeling L: Acyl-CoA carboxylases (accD2
76. Mandlik A, Das A, Ton-That H: The molecular switch that activates the cell and accD3), together with a unique polyketide synthase (Cg-pks), are
wall anchoring step of pilus assembly in gram-positive bacteria. Proc key to mycolic acid biosynthesis in Corynebacterianeae such as
Natl Acad Sci USA 2008, 105:14147-14152. Corynebacterium glutamicum and Mycobacterium tuberculosis. J Biol Chem
77. Mandlik A, Swierczynski A, Das A, Ton-That H: Corynebacterium diphtheriae 2004, 279:44847-44857.
employs specific minor pilins to target human pharyngeal epithelial 97. Chami M, Andreau K, Lemassu A, Petit JF, Houssin C, Puech V, Bayan N,
cells. Mol Microbiol 2007, 64:111-124. Chaby R, Daffe M: Priming and activation of mouse macrophages by
78. McKean SC, Davies JK, Moore RJ: Probing the heat shock response of trehalose 6,6’-dicorynomycolate vesicles from Corynebacterium
Corynebacterium pseudotuberculosis: the major virulence factor, glutamicum. FEMS Immunol Med Microbiol 2002, 32:141-147.
phospholipase D, is downregulated at 43 degrees C. Res Microbiol 2007, 98. Hard GC: Electron microscopic examination of Corynebacterium ovis. J
158:279-286. Bacteriol 1969, 97:1480-1485.
79. McKean SC, Davies JK, Moore RJ: Expression of phospholipase D, the 99. Takayama K, Wang C, Besra GS: Pathway to synthesis and processing of
major virulence factor of Corynebacterium pseudotuberculosis, is mycolic acids in Mycobacterium tuberculosis. Clin Microbiol Rev 2005,
regulated by multiple environmental factors and plays a role in 18:81-101.
macrophage death. Microbiology 2007, 153:2203-2211. 100. Yellaboina S, Ranjan S, Chakhaiyar P, Hasnain SE, Ranjan A: Prediction of
80. Wilson MJ, Brandon MR, Walker J: Molecular and biochemical DtxR regulon: identification of binding sites and operons controlled by
characterization of a protective 40-kilodalton antigen from Diphtheria toxin repressor in Corynebacterium diphtheriae. BMC Microbiol
Corynebacterium pseudotuberculosis. Infect Immun 1995, 63:206-211. 2004, 4:38.

63
Publications
Trost et al. BMC Genomics 2010, 11:728 Page 17 of 17
http://www.biomedcentral.com/1471-2164/11/728

101. D’Afonseca V, Prosdocimi F, Dorella FA, Pacheco LG, Moraes PM, Pena I,
Ortega JM, Teixeira S, Oliveira SC, Coser EM, et al: Survey of genome
organization and gene content of Corynebacterium pseudotuberculosis.
Microbiol Res 2010, 165:312-320.
102. D’Afonseca V, Moraes PM, Dorella FA, Pacheco LG, Meyer R, Portela RW,
Miyoshi A, Azevedo V: A description of genes of Corynebacterium
pseudotuberculosis useful in diagnostics and vaccine applications. Genet
Mol Res 2008, 7:252-260.
103. Dorella FA, Pacheco LG, Seyffert N, Portela RW, Meyer R, Miyoshi A,
Azevedo V: Antigens of Corynebacterium pseudotuberculosis and
prospects for vaccine development. Expert Rev Vaccines 2009, 8:205-213.
104. Gordon D, Abajian C, Green P: Consed: a graphical tool for sequence
finishing. Genome Res 1998, 8:195-202.
105. Tauch A, Kaiser O, Hain T, Goesmann A, Weisshaar B, Albersmeier A, Bekel T,
Bischoff N, Brune I, Chakraborty T, et al: Complete genome sequence and
analysis of the multiresistant nosocomial pathogen Corynebacterium
jeikeium K411, a lipid-requiring bacterium of the human skin flora. J
Bacteriol 2005, 187:4671-4682.
106. Schneider J, Vorhölter FJ, Trost E, Blom J, Musa YR, Neuweger H,
Schatschneider S, Tauch A, Goesmann A: CARMEN: In silico reconstruction
of organism-specific metabolic networks using SBML. Genet Mol Res 2010,
9:1660-1672.
107. Gough J, Karplus K, Hughey R, Chothia C: Assignment of homology to
genome sequences using a library of hidden Markov models that
represent all proteins of known structure. J Mol Biol 2001, 313:903-919.
108. Finn RD, Mistry J, Tate J, Coggill P, Heger A, Pollington JE, Gavin OL,
Gunasekaran P, Ceric G, Forslund K, et al: The Pfam protein families
database. Nucleic Acids Res 2010, 38:D211-222.
109. Eddy SR: Profile hidden Markov models. Bioinformatics 1998, 14:755-763.
110. Dodd IB, Egan JB: Improved detection of helix-turn-helix DNA-binding
motifs in protein sequences. Nucleic Acids Res 1990, 18:5019-5026.
111. Tatusov RL, Galperin MY, Natale DA, Koonin EV: The COG database: a tool
for genome-scale analysis of protein functions and evolution. Nucleic
Acids Res 2000, 28:33-36.
112. Altschul SF, Madden TL, Schaffer AA, Zhang J, Zhang Z, Miller W,
Lipman DJ: Gapped BLAST and PSI-BLAST: a new generation of protein
database search programs. Nucleic Acids Res 1997, 25:3389-3402.
113. Beckstette M, Homann R, Giegerich R, Kurtz S: Fast index based algorithms
and software for matching position specific scoring matrices. BMC
Bioinformatics 2006, 7:389.
114. Baumbach J, Wittkop T, Kleindt CK, Tauch A: Integrated analysis and
reconstruction of microbial transcriptional gene regulatory networks
using CoryneRegNet. Nat Protoc 2009, 4:992-1005.
115. Crooks GE, Hon G, Chandonia JM, Brenner SE: WebLogo: a sequence logo
generator. Genome Res 2004, 14:1188-1190.

doi:10.1186/1471-2164-11-728
Cite this article as: Trost et al.: The complete genome sequence of
Corynebacterium pseudotuberculosis FRC41 isolated from a 12-year-old
girl with necrotizing lymphadenitis reveals insights into gene-regulatory
networks contributing to virulence. BMC Genomics 2010 11:728.

Submit your next manuscript to BioMed Central


and take full advantage of:

• Convenient online submission


• Thorough peer review
• No space constraints or color figure charges
• Immediate publication on acceptance
• Inclusion in PubMed, CAS, Scopus and Google Scholar
• Research which is freely available for redistribution

Submit your manuscript at


www.biomedcentral.com/submit

64
Publications

3.4 Impact of improved potassium accumulation on pH homeostasis,


membrane potential adjustment and survival of Corynebacterium
glutamicum (Ochrombel et al, 2011)

65
Publications
Biochimica et Biophysica Acta 1807 (2011) 444–450

Contents lists available at ScienceDirect

Biochimica et Biophysica Acta


j o u r n a l h o m e p a g e : w w w. e l s e v i e r. c o m / l o c a t e / b b a b i o

Impact of improved potassium accumulation on pH homeostasis, membrane


potential adjustment and survival of Corynebacterium glutamicum
Ines Ochrombel a, Lisa Ott b, Reinhard Krämer a, Andreas Burkovski b, Kay Marin a,⁎
a
Institute of Biochemistry, University of Cologne, 50674 Cologne, Germany
b
Lehrstuhl für Mikrobiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, 91058 Erlangen, Germany

a r t i c l e i n f o a b s t r a c t

Article history: Metal ion uptake is crucial for all living cells and an essential part of cellular bioenergetic homeostasis. In this
Received 22 July 2010 study the uptake and the impact of the most abundant internal cation, potassium, were investigated in
Received in revised form 26 January 2011 Actinobacteria, a group of high G + C Gram-positives with a number of prominent biotechnologically and
Accepted 28 January 2011
medically important members. Genome analyses revealed a variety of different potassium uptake systems in
Available online 2 February 2011
this monophyletic group ranging from potassium channels common in virtually all Actinobacteria to different
Keywords:
active carriers that were present predominantly in pathogenic members able to cope with various stress
Potassium transport conditions. By applying Corynebacterium glutamicum as model system we provide experimental evidence that
Potassium channel under optimal conditions a potassium channel is sufficient in bacteria for the maintenance of internal pH and
Corynebacterium membrane potential ensuring survival of cells under stress conditions. Under potassium limitation, however,
pH homeostasis viability of C. glutamicum was increased under acidic stress or during desiccation when a functional KtrAB
Membrane potential adjustment potassium transporter from the pathogen Corynebacterium jeikeium was heterologously expressed. We
provide experimental evidence that the KtrAB mediated enhanced potassium accumulation improved
maintenance of internal pH and membrane potential. The results indicate that the occurrence of active
potassium transport systems correlates with an improved potassium-dependent bioenergetic homeostasis
and survival of bacterial cells under stress conditions.
© 2011 Elsevier B.V. All rights reserved.

1. Introduction third type of uptake system is the secondary Kup-type transporter


that is present in many Gram-positive and Gram-negative bacteria. All
Ion homeostasis is an essential part of life. Since the cell membrane secondary active carriers are supposed to use the membrane potential
is impermeable for ions, transport systems are mandatory for ion for uptake of potassium [3]. Additionally, several ligand-gated channel
uptake and extrusion and carrier proteins regulate the internal ion proteins are involved in potassium uptake [4,5].
content. The alkali metal ion potassium plays an outstanding role in The distribution of potassium transport systems in bacteria was
this respect [1]. With concentrations between 0.1 and 0.6 M proposed to be correlated with the potassium availability in their
potassium is the most abundant cation in bacteria, while in general natural habitat [6]. Bacteria like Escherichia coli or Bacillus subtilis
only traces are available in the environment (0.1–10 mM). It is harbor different types of potassium carriers and maintain high
involved in membrane potential adjustment and required for the internal potassium levels under both stress and standard cultivation
activity of the respiratory chain [2]. Potassium acts as second conditions [7,8], while Corynebacterium glutamicum, Klebsiella pneu-
messenger for stress signalling and as regulatory element for moniae and Bacillus stearothermophilus do not require potassium
transcription control as well as counter ion for glutamate and other under optimal growth conditions and can be propagated in its virtual
compounds during osmotic stress response [3]. The impact of absence [1,2]. However, under acidic stress C. glutamicum accumulates
potassium for bacterial life is indicated by the diversity of transport high cytoplasmic potassium concentrations as well. A Kup-type
systems present for this particular ion. In many enteric bacteria transporter and a potassium channel encoding gene were found in
primary active P-type ATPases of the Kdp-type are present in addition the C. glutamicum genome, respectively. Interestingly, no activity was
to secondary carriers of the Trk-type. Ktr-type systems are related to observed for the secondary active potassium carrier Kup and the
Trk transporters and are present in many Gram-positive bacteria. A potassium channel CglK was found to represent the main uptake
system under standard conditions [2]. CglK represents the first
example of a bacterial potassium channel for which the activity was
⁎ Corresponding author at: University of Cologne, Institute of Biochemistry,
Zuelpicher Str. 47, 50674 Cologne, Germany. Tel.: +49 221 470 6476; fax: + 49 221
proven in its natural membrane environment. Additionally, the
470 5091. analysis of CglK in C. glutamicum unravelled for the first time the
E-mail address: kay.marin@uni-koeln.de (K. Marin). physiological function of a bacterial potassium channel, namely, the

0005-2728/$ – see front matter © 2011 Elsevier B.V. All rights reserved.
doi:10.1016/j.bbabio.2011.01.008

66
Publications
I. Ochrombel et al. / Biochimica et Biophysica Acta 1807 (2011) 444–450 445

decease of the membrane potential and the maintenance of a neutral gas-permeable membrane in a volume of 200 μl shaken at 1200 rpm.
internal pH under conditions of acidic stress. The CglK channel is Plates were prepared by the addition of 15 g l− 1 agar to the medium. For
sufficient for potassium accumulation in presence of high potassium all experiments precultivation was performed as described [2] and
concentrations or at conditions of low potassium demand. However, if experiments were performed with cultures entered the exponential
the potassium availability is limited or the requirement for potassium growth phase. Whereas BHI contains 10 mM potassium, MMI contains
is increased the sole presence of a potassium channel might restrict 37 mM potassium. The pH was adjusted by appropriate buffer
growth and consequently active carriers might be required [6]. substances (250 mM) as described [11]. If necessary, the medium was
Taxonomically, C. glutamicum belongs to the Actinobacteria that are supplemented with kanamycin (25 μg ml− 1) and for induction of
widespread in nature and live as soil bacteria, saprophytes, members expression with 0.5 mM isopropyl-β-D-thiogalactopyranoside (IPTG).
of the microflora of humans or animal and plant pathogens. Medically Growth was followed by measuring the OD600. Standard molecular
important members are Mycobacterium tuberculosis, Nocardia farci- cloning techniques were applied using E. coli DH5αMCR cells grown in
nica, Propionibacterium acnes, Corynebacterium diphtheriae and Cory- Luria-Bertani (LB) medium at 37 °C.
nebacterium jeikeium, while C. glutamicum, Corynebacterium efficiens, For amplification of the ktr locus of C. diphtheriae ISS3319 or C.
Bifidobacterium longum as well as Streptomyces coelicolor are impor- jeikeium K411, primers listed in Table 1 were applied. The genomic
tant in biotechnological processes [9]. fragments, harboring both ktrB and ktrA as well as 500 bp upstream of
In this study we analyzed the monophyletic group of Actinobac- ktrB, including the putative promoter region and the ribosome
teria regarding the equipment with potassium transport systems. We binding site (Fig. S1), were cloned into the plasmid pDRIVE (Qiagen,
show that all pathogenic strains, supposed to be exposed to Hilden, Germany) and the sequence of the resulting vectors
challenging environmental limitations and stress conditions, harbor pDRIVE_ktrBA_Cd and pDRIVE_ktrBA_Cj was confirmed (GATC, Kon-
active potassium transport systems in addition to potassium channels. stanz, Germany). The PCR primers were designed in order to
We addressed the impact of the mode of potassium transport by introduce a Strep-tag coding sequence at the 3′ end of the ktrA
active carriers and/or channels on growth and survival from a gene. Subsequently, the ktrBA fragments were cloned into the vector
bioenergetic point of view. Improvement of potassium transport of pEKEX2 mediating the IPTG inducible expression of genes under the
C. glutamicum harboring a functional potassium channel by additional control of the lac promoter [12], resulting in plasmids pEKEX2_ktr-
presence of the transporter KtrAB from C. jeikeium led to an increased BA_Cd and pEKEX2_ktrBA_Cj. After electroporation of the C. glutami-
potassium content and was beneficial for the maintenance of cum cells, the presence of the plasmids was proven by cultivation on
membrane potential and internal pH accompanied by improved kanamycin-containing plates, as well as by PCR. The resulting strains
growth and survival of C. glutamicum under stress conditions. are listed in Table 1. Additionally, expression of the Strep-tagged KtrA
protein was proven by Western blot analysis as described previously
2. Materials and methods [2] using a Strep tag antibody (Sigma).

2.1. Bacterial strains, growth conditions and construction of mutants 2.2. Measurement of external and internal potassium concentration as
well as bioenergetic parameters
C. glutamicum strain ATCC 13032 served as wild type and was grown
either in brain heart infusion (BHI) medium (Becton-Dickenson, Monitoring of both external and internal potassium concentration
Heidelberg, Germany) or in minimal medium MMI [10] at 30 °C in was performed by flame photometry (ELEX 6361; Eppendorf,
Erlenmeyer flasks shaken at 130 rpm or in microtiter plates sealed with a Germany) as described previously [2]. In short, after precultivation

Table 1
Strains, plasmids and primers used in this study.

Strain or plasmid Related genotype or descriptiona Reference

E. coli
DH5αMCR endA1 supE44 recA1 gyr96 relA1 deoR U169 ϕ80ΔlacZΔM15 mcrA Δ Grant et al. [22]
(mrr-hsdRMS-mcrBC)
C. glutamicum strains
ATCC 13032 Wild type Abe et al. [23]
Δkup Deletion of kup (cg0187) in ATCC 13032 Follmann et al. [2]
ΔcglK Deletion of cglK (cg0887) in ATCC 13032 Follmann et al. [2]
ΔcglKΔkup Deletion of kup and cglK in ATCC 13032 Follmann et al. [2]
Cg_p ATCC 13032 harboring plasmid pEKEX2 This study
Cg_pktrBA ATCC 13032 harboring plasmid pEKEX2_ktrBA_Cj This study
Cg_pktrBA ATCC 13032 harboring plasmid pEKEX2_ktrBA_Cd This study
ΔcglKΔkup_pktrBA_Cj ΔkupΔcglK harboring plasmid pEKEX2_ktrBA_Cj This study
Plasmids
pDRIVE A-T cloning vector (KmR, ApR) Qiagen, Hilden, Germany
pDRIVE_cglK_Cd pDRIVE harboring an internal fragment of gene DIP0724 of C. diphtheriae This study
pDRIVE_ktrB_Cd pDRIVE harboring an internal fragment of gene DIP1931 of C. diphtheriae This study
pDRIVE_ktrBA_Cj pDRIVE harboring the ktrBA genomic locus of C. jeikeium This study
pDRIVE_ktrBA_Cd pDRIVE harboring the ktrBA genomic locus of C. diphtheriae This study
pEKEX2 E. coli–C. glutamicum shuttle expression vector (KmR) Eikmanns et al. [12]
pEKEX2_ktrBA_Cj pEKEX2 harboring the ktrBA genomic locus of C. jeikeium This study
pEKEX2_ktrBA_Cd pEKEX2 harboring the ktrBA genomic locus of C. diphtheriae This study
Primerb
Pre500_ktrB_Cjeik_5` 5′-GCCTGCAGGGAGACTCAGCCCGTGCTGCGTTTGC-3′ (PstI) This study
ktrA_CStrep_Cjeik_3′ 5′-GCGAATTCGCCTATTTTTCGAACTGCGGGTGGCTCCAGGAATCCGCAAACTTATCCAGGT-3′ (EcoRI) This study
Pre500_ktrB_Cdiph_5′ 5′-GCCTGCAGGAAACTCAGGCGGTGTTGGCGTTGCTGC-3′ (XhoI) This study
ktrA_CStrep_Cdiph_3′ 5′-CGAATTCGCCTATTTTTCGAACTGCGGGTGGCTCCA-ACCAATGATCAGCCTTTCTACAGG-3′ (EcoRI) This study
a
Abbreviations: ApR, ampicillin resistance; KmR, kanamycin resistance.
b
Letters in bold indicate the recognition site for the restriction enzyme given in parentheses, the stop codon is underlined and the Strep tag-coding sequence is shown in italic
letters.

67
Publications
446 I. Ochrombel et al. / Biochimica et Biophysica Acta 1807 (2011) 444–450

cells were washed three times with MMI medium without potassium channel Kch was applied as query beside the potassium channel KcsA
and then inoculated in MMI medium containing 1 mM KCl at a defined of Streptomyces lividans. Sequences highly similar to the known
pH and an OD600 of 2 in 100 ml of medium. External potassium potassium transport systems were employed for a cluster analysis
concentration was measured after removal of cells by centrifugation including the sequence of the E. coli sodium transporter NhaA as
in the supernatant. For detection of internal potassium concentra- outgroup. As seen in Fig. 1, with either KcsA or CglK all strains harbor
tions, cells of 2 ml culture were collected by centrifugation and the at least one potassium channel protein, while in N. farcinica and S.
pellet was washed twice with potassium-free MMI. Cell disruption coelicolor both types of potassium channels are present. In Gram-
was carried out by ultrasonication 80% (Sonorex Digital DK 512 P;
Badelin, Berlin) at 85 °C in 1.7 ml H2O for 20 min. After centrifugation
the potassium concentration in the supernatant was measured.
The cytoplasmic volume was determined by the distribution of 3H-
labeled H2O (0.55 mCi l− 1) and 14C-labeled inulin (0.14 mCi l− 1)
between the cell pellet and the supernatant to be 1.8 μl mg− 1 cell dry
matter (CDM) as described previously [2]. Processing of samples for
rapid separation of extra- and intracellular fluids was performed by
using silicone oil centrifugation with perchloric acid in the bottom
layer [2]. The membrane potential was determined by measuring the
distribution of tetraphenylphosphonium (TPP) (final concentration
5 μM, specific radioactivity 0.995 Ci mol− 1) as described previously
[2]. Internal pH was determined by measuring the distribution of 14C-
labeled benzoic acid (final concentration 15 μM, specific radioactivity
3.12 Ci mol− 1) as described [2].
Rates of oxygen consumption by C. glutamicum cells were measured
by a Clark-type electrode (Oxygraph; Hansatech, Reutlingen, Germany)
at 30 °C in a total volume of 1 ml minimal medium, pH 6 (250 mM MES)
with cells harvested at the exponential phase and resuspended at an
OD600 of 0.3 to 0.5 in MMI medium. Steady-state oxygen consumption
rates were determined in absence or presence of potassium (50 mM).

2.3. Determination of pH and desiccation tolerance of C. glutamicum

For the drop assays, cells were depleted of potassium by


precultivation in minimal medium without potassium supplementa-
tion. Subsequently, cultures were diluted in minimal medium without
potassium to the OD values indicated. Four microliters of each cell
suspension was dropped onto minimal medium agar plates with a pH
of 7.5 and 7.0 (250 mM MOPS) as well as pH 6.5 and 6.0 (250 mM
MES) and the indicated potassium concentrations. After 48 h of
incubation, results were documented by photography. Desiccation
tolerance was analyzed with cells depleted of potassium diluted in
minimal medium with 5 mM potassium to an OD600 of 1. In a well of a
microtiter plate 100 μl of each cell suspension was exposed to
desiccation by shaking at 1200 rpm at 30 °C. After 24 and 48 h dried
cells were resuspended in 100 μl NaPi -puffer, pH 7.4 and dilutions
were made as indicated. Subsequently, 4 μl of each cell suspension
was dropped onto BHI agar plates containing 100 mM KCl. After 24 h
of incubation, results were documented by photography.

3. Results

3.1. Occurrence of different potassium transport systems in Actinobacteria

Addressing the question whether the existence of active potassium


transport is correlated with a particular bacterial life style, a Fig. 1. Occurrence of different types of potassium transporters in Actinobacteria.
Proteins similar to known potassium transporter subunits in E. coli (Ecoli, Kch, KefB,
bioinformatic analysis of (putative) potassium transport systems in TrkH, KdpA) or S. coelicolor (Scoel, KcsA) were identified by genomicBlast searches
Actinobacteria including the non-pathogenic species C. efficiens and C. against the genomes of Corynebacterium aurimucosum (Cauri), Corynebacterium
glutamicum strain R and the pathogens Corynebacterium aurimucosum, diphtheriae biotype gravis strain NCTC13129 (Cdiph), C. efficiens strain YS-314 (Ceffi),
C. diphtheriae, C. jeikeium, Corynebacterium urealyticum and Coryne- Corynebacterium glutamicum strain ATCC 13032 (Cglut), Corynebacterium glutamicum
strain R (CglutR), Corynebacterium jeikeium K411 (Cjeik), C. urealyticum DSM 7109
bacterium kroppenstedtii was performed. Outside the genus Coryne-
(Curea), C. kroppenstedtii DSM 44385 (Ckrop), Mycobacterium tuberculosis strain H37RV
bacterium, the closely related species M. tuberculosis, Mycobacterium (Mtube), M. abscessus ATCC 19977 (Mabsc), Nocardia farcinica strain IFM 10152 (Nfarc),
abscessus, N. farcinica, P. acnes and Clavibacter michiganensis were Propionibacterium acnes strain KPA171202 (Pacne), S. avermitilis strain MA-4680
included, which are prominent human or plant pathogens as well as (Saver), S. coelicolor strain A3(2) M145 (Scoel), B. longum strain NCC2705 (Blong) and
the non-pathogenic and biotechnologically important strains B. C. michiganensis subsp. michiganensis NCPPB 382 (Cmich). The sequences were grouped
according to a cluster analysis performed by using ClustalX and visualised by TreeView
longum, Streptomyces avermitilis and S. coelicolor [9]. The sequence [24,25]. The sequence of the E. coli sodium transporter NhaA was used as an outgroup.
of (subunits of) the known E. coli potassium transport systems Trk, The (putative) subunit composition and the mode of transport for each subgroup are
Kup and Kdp, the potassium exporter KefB as well as the potassium indicated on the right.

68
Publications
I. Ochrombel et al. / Biochimica et Biophysica Acta 1807 (2011) 444–450 447

positive bacteria homologs of the E. coli Trk transporter, designated or harboring the ktrBA expression plasmid. Comparable rates of
Ktr in this group, and the Kdp-type uptake system are present almost potassium accumulation and comparable internal potassium levels
exclusively in pathogenic species, whereas the Kup-type transporter were observed in both strains within 8 h (data not shown, Fig. 2).
is found in non-pathogenic strains predominantly. The KefB carrier is Obviously, the contribution of the KtrAB transport system to
mostly present in pathogenic strains. In conclusion, non-pathogenic potassium uptake in C. glutamicum is not significant in comparison
strains harbor potassium channels and Kup-type potassium uptake to potassium uptake via the channel CglK under these conditions.
systems whereas pathogenic strains are equipped with Ktr- and Trk- Consequently, we performed experiments under conditions of an
type potassium uptake systems. Because pathogenic strains are increased demand for potassium, namely at neutral and acidic
exposed to numerous stress conditions like acidic pH and/or low medium pH. In comparison to the results obtained at pH 8.5 the
potassium availability the presence of appropriate potassium trans- internal potassium concentration in both C. glutamicum strains was
port systems might be beneficial for survival of pathogenic Actino- maintained at higher levels at pH 7.5 and 6.5 after 8 h of incubation.
bacteria [13,14]. Whereas at pH 8.5 the internal potassium concentration was
We hypothesized that the exclusive presence of a potassium channel comparable in both cell types, at pH 7.5 and 6.5 the internal
may represent a bottleneck for the maintenance of bioenergetic potassium concentration in the recombinant strain was 170 mM
parameters that can be overcome by an additional active transport and 120 mM higher than in wild type cells, respectively (Fig. 2). These
system. To test this hypothesis experimentally C. glutamicum was chosen findings underline the increased potassium requirement of C.
as model system because mutants lacking particular potassium transpor- glutamicum under neutral and acidic conditions and demonstrate
ters are available and a variety of methods is established to address the that potassium accumulation in C. glutamicum is increased by the
impact of potassium transport on bioenergetic parameters and survival. heterologous uptake system KtrAB of C. jeikeium under acidic stress
Our strategy was to improve potassium accumulation in C. glutamicum by conditions.
expression of ktrAB genes from C. diphtheriae or C. jeikeium. Since no The known KtrAB transport systems require sodium ions for
expression of genes derived from C. diphtheriae in C. glutamicum was activity but it is unknown whether sodium activates the transporter
detected (data not shown) we focused on the impact of KtrAB from
C. jeikeium on C. glutamicum.
A
3.2. Expression of functional C. jeikeium ktrBA in C. glutamicum

The Ktr transport system of C. jeikeium putatively consists of two


subunits which are encoded by the genes ktrB (jk0347) and ktrA (jk0348).
For the KtrB subunit eight transmembrane domains, a molecular weight of
49 kDa is predicted, whereas the KtrA subunit is a soluble 23 kDa protein
harboring a putative NAD-binding domain. The presence of the KtrAB
system was demonstrated by detection of the C-terminal Strep-tag of the
KtrA protein in cell extracts of Cg_pktrBA. Upon addition of IPTG to cell
cultures and protein extraction a protein band of approx. 27 kDa was
detected, corresponding to the Strep-tagged version of KtrA (Fig. S1).
Functionality of the KtrAB transport system was addressed by
expression of ktrBA genes in C. glutamicum wild type cells and cells of
the mutant lacking both potassium transporters (CglK and Kup).
Under standard laboratory conditions C. glutamicum does not require
addition of potassium and mutants lacking potassium transporters
grow indistinguishable from wild type cells at alkaline pH [2].
However, potassium accumulation was shown to be crucial for B
survival under acidic conditions. Cells were inoculated at pH 7.5 on
plates containing different potassium concentrations. While in the
presence of 50 mM potassium all strains were able to grow to a
comparable extent, at 5 mM potassium growth of the strain lacking all
potassium uptake systems was impaired. This strain did not grow at
all if no potassium was added (Fig. 5A). Growth of wild type cells was
slightly decreased in presence of low potassium concentrations, but
mutants harboring the ktrBA genes were not affected upon addition of
5 mM or if potassium was lacking in comparison to plates containing
50 mM potassium (Fig. 5A). Moreover, at very low potassium
concentrations the presence of ktrBA genes resulted in slightly better
growth in comparison to wild type cells (Fig. 5A). From these
observations it was concluded that loss of potassium uptake systems
in C. glutamicum can be complemented by the expression of the ktrBA
operon from C. jeikeium indicating that the KtrAB transporter is
Fig. 2. Impact of ktrBA expression on potassium uptake and accumulation in C.
functional in C. glutamicum.
glutamicum and sodium dependence of KtrAB function. (A) Cells of the wild type
harboring a control plasmid (white bars) or the ktrBA expression vector (strain
3.3. Impact of ktrBA expression on potassium uptake and accumulation Cg_pktrBA, black bars) were inoculated in minimal medium of pH 8.5, 7.5 and 6.5 in
presence of 1 mM potassium. The internal potassium content was measured by flame
As a basis for further investigations, potassium uptake and the photometry and the values after 8 h of incubation are shown. (B) Cells of the strains
ΔcglKΔkup and ΔcglKΔkup_pktrBA were grown in liquid media at a pH level of 6.5 at the
cytoplasmic potassium concentration in C. glutamicum cells were indicated potassium concentrations in absence (white bars) or presence (black bars) of
analyzed by flame photometry [2]. Under optimal growth conditions sodium (no addition or addition of 100 mM NaCl), respectively, and growth rates were
(pH 8.5), external potassium was taken up by wild type cells lacking estimated.

69
Publications
448 I. Ochrombel et al. / Biochimica et Biophysica Acta 1807 (2011) 444–450

or KtrAB functions as a K+/Na+ symporter [15]. To test the measured in absence of potassium or immediately after addition of
dependence of the KtrAB transporter of C. jeikeium on sodium, growth 50 mM potassium. In C. glutamicum cells lacking the potassium
experiments in absence or presence of NaCl under potassium limiting transport systems CglK and Kup, a collapse of the pH gradient was
conditions using the ΔcglKΔkup mutant at low pH were performed. observed indicated by low internal pH values of 6.3 and 6.1
Whereas cells of the ΔcglKΔkup mutant were not affected by addition irrespective of the potassium concentration (Fig. 3). Consequently,
of NaCl (data not shown), expression of ktrBA caused significantly high values for the membrane potential were determined in both
increased growth rates of the mutant at low potassium concentrations strains (184 and 183 mV). In contrast, wild type cells maintained a
in the presence of NaCl only (Fig. 2B). At high potassium concentra- more neutral internal pH of 6.7 and a lower membrane potential
tions no difference of ΔcglKΔkup cells harboring or lacking KtrAB was (143 mV) in the presence of potassium. In its absence, however, the
found (Fig. 2B). The data indicate that the demand for potassium of C. internal pH dropped to 6.2 and the membrane potential was found to
glutamicum can be accomplished by KtrAB in a sodium-dependent be increased to 183 mV (Fig. 3). Expression of ktrBA enabled the
manner pointing to the important role of sodium ions for the KtrAB ΔcglKΔkup mutant and, interestingly, even wild type cells to maintain
function. more neutral internal pH values (6.8 and 6.9) and lower values of the
membrane potential (143 and 126 mV), respectively, than the
parental strains (Fig. 3). The improved pH homeostasis was even
3.4. Impact of ktrBA expression on bioenergetic parameters more pronounced at low potassium concentrations resulting in
0.7 units higher internal pH values in ktrBA expression strains
To investigate the impact of the KtrAB-dependent potassium compared to the parental strains. For all strains comparable pmf
transport on the maintenance of bioenergetic homeostasis, mem- values were observed (Fig. 3).
brane potential (ΔΨ), internal pH and proton motive force (pmf) In order to investigate further consequences of the KtrAB-
were measured in cells of the wild type and the ΔcglKΔkup mutant as dependent improvement of pH homeostasis the activity of the
well as the corresponding strains harboring the KtrAB transporter respiratory chain was examined in the same experimental set up.
under acidic stress conditions. Cells were grown in the absence of Respiratory activity in wild type cells without addition of potassium
potassium in minimal medium at pH 7.5 for 3 h in order to deplete the was 38 nmol O 2 min− 1 ml− 1 OD − 1
600 and increased by 50% in
internal potassium concentration, and subsequently the external pH presence of 5 or 50 mM potassium (Fig. 4). For the ΔcglKΔkup
was shifted to 6.0. Simultaneously, bioenergetic parameters were mutant the respiratory activity was significantly lower than in wild
type cells (25 nmol O2 min− 1 ml− 1 OD− 1
600) and did not increase
upon addition of potassium (Fig. 4). In contrast, expression of ktrBA
A in the ΔcglKΔkup mutant resulted in higher rates of oxygen
consumption in comparison to the parental strain. In the wild type
background, however, expression of the ktrBA genes did not
increase oxygen consumption irrespective of the potassium
concentration.

3.5. Impact of ktrBA expression on survival and growth of C. glutamicum


under stress conditions

After proving the functionality of C. jeikeium KtrAB in C.


glutamicum and quantifying the beneficial impact on bioenergetic
homeostasis, we investigated whether growth and survival of C.

Fig. 3. Impact of ktrBA expression on bioenergetic parameters in C. glutamicum. Cells of


the wild type (WT) and the mutant lacking the cglK and kup gene (ΔcglKΔkup) as well
as both strains harboring the ktrBA expression vector (Cg_pktrBA, ΔcglKΔkup_pktrBA) Fig. 4. Impact of ktrBA expression on the activity of the respiratory chain in C.
were exposed to a pH shift in minimal medium from pH 7.5 to pH 6 without addition of glutamicum. Cells of the wild type (grey bars) and the mutant lacking the cglK and kup
potassium (A) or in presence of 50 mM potassium (B). Membrane potential (white gene (white bars) as well as both strains harboring the ktrBA expression vector
bars) and pH gradient (grey bars) were measured and the corresponding pmf (black (hatched bars) were inoculated in minimal medium pH 6 in presence of the indicated
bars) was calculated. All values are presented as potential and are given in mV. On the potassium concentration. Activity of the respiratory chain was measured by
right site the corresponding internal pH is indicated. determining the oxygen consumption rate.

70
Publications
I. Ochrombel et al. / Biochimica et Biophysica Acta 1807 (2011) 444–450 449

glutamicum are affected in a KtrAB-dependent manner under stress 4. Discussion


conditions. C. jeikeium is part of the human skin flora and exposed to
low pH values (around pH 5.5) and desiccation in combination with 4.1. Potassium channels are sufficient for stress-dependent potassium
low potassium concentrations (approx. 10 mM) during colonization uptake
of this habitat. Therefore, the KtrAB-dependent pH and desiccation
tolerance of C. glutamicum were analyzed as an example. The The requirement for potassium under stress conditions was
increased tolerance of C. glutamicum cells expressing the ktrBA observed for bacteria as well as eukaryotic organisms [3]. Potassium
genes towards neutral pH was already described and interestingly accumulation as an immediate response upon an osmotic upshift is
even the pH tolerance of wild type cells was increased in a KtrAB- observed in many bacteria like E. coli, B. subtilis or cyanobacteria and
dependent manner (Fig. 5A). At pH 6.5 even at high potassium was shown to be essential for survival [1,7,16]. During desiccation
concentrations growth of wild type cells harboring the control vector bacterial cells are exposed to physiological challenges comparable to
without potassium transporter genes was strongly impaired osmotic stress situations [17]. In this study, we showed that a CglK
(Fig. 5C). In contrast, cells harboring the KtrAB transporter grew mutant of C. glutamicum was more sensitive towards acidic conditions
even at low potassium concentrations down to 1 mM. At pH 6 growth and desiccation than wild type cells indicating that potassium uptake
of both strains was strongly impaired (Fig. 5C). In comparison, upon is required under these conditions. Also in Helicobacter pylori the
ectopic overexpression of the channel encoding gene cglK growth of potassium channel HpKchA acts as sole potassium uptake system and
C. glutamicum was not improved (Fig. S2). The results demonstrate it was demonstrated that the presence of this channel is required for
that the KtrAB transport system of C. jeikeium confers a higher the persistence of H. pylori in the gastric environment [6]. Conse-
resistance towards potassium limitation under acidic stress condi- quently, supply of cells with potassium seems to be required during
tions to C. glutamicum. various stress conditions and a potassium channel is sufficient to meet
To test the correlation between potassium transport and survival of the demand.
dehydration, wild type and the ΔcglKΔkup mutant harboring the control
vector pEKEX2 or the ktrBA expression plasmid were exposed to 4.2. Potassium uptake represents a bottleneck for bioenergetic homeostasis
desiccation in the presence of 5 mM potassium. After 24 and 48 h survival
of cells was determined by re-suspending the cells and transferring 5 μl of The sole presence of a potassium channel seems not to be
this suspension onto BHI agar plates. The mutant ΔcglKΔkup, lacking all sufficient, however, under potassium limitation as indicated by the
potassium transport systems, was strongly impaired in survival in significantly improved survival of C. glutamicum strains expressing
comparison to wild type cells (Fig. 5B). Expression of the ktrBA genes ktrBA. A plausible reason for this limitation is the sole driving force for
did complement the sensitivity of the ΔcglKΔkup strain and improved the CglK-dependent potassium accumulation, the membrane poten-
survival of wild type cells, respectively (Fig. 5B). In conclusion, under these tial, which was determined in C. glutamicum wild type cells as 143 mV
conditions the presence of potassium and appropriate potassium uptake minimum at low pH. This facilitates a 400-fold internal accumulation
systems are crucial for long-term survival and resistance of C. glutamicum of potassium according to the Nernst equation. Since the internal
towards desiccation stress. potassium concentration in C. glutamicum at acidic pH was around

Fig. 5. Impact of C. jeikeium KtrAB on growth and survival of C. glutamicum during potassium limitation and/or acidic stress as well as desiccation. Cells of the wild type (WT), the
ΔcglKΔkup mutant and the strains Cg_pktrBA and ΔcglKΔkup_pktrBA, harboring the plasmid pEKEX2_ktrBA, were grown on agar plates of pH 7.5 (A) supplemented with 50, 5 mM or
no potassium or exposed to desiccation (B) for 0, 24 and 48 h and survival was determined by the drop assay. The potassium-dependent acidic stress resistance of wild type and
Cg_ktrBA cells was addressed (C) by growth on agar plates of pH 7.0, 6.5 and 6.0 supplemented with 50, 10, 5, 1 mM or no potassium. The OD600 values indicate the cell density of
cultures applied for the assay by transfer of 4 μl onto the plate (A, C) or exposed to desiccation (B). Upon incubation for 24 h documentation by photography was performed.

71
Publications
450 I. Ochrombel et al. / Biochimica et Biophysica Acta 1807 (2011) 444–450

600 mM an external potassium concentration of at least 1.5 mM is Supplementary materials related to this article can be found online
required. If the potassium availability is lower, lower levels of internal at doi:10.1016/j.bbabio.2011.01.008.
potassium can be maintained causing the collapse of internal pH
homeostasis under acidic stress conditions. Overexpression of the Acknowledgements
homologous potassium channel encoding gene cglK did not lead to
improved survival under acidic conditions and did not cause an We would like to thank the Bundesministerium für Bildung und
improved maintenance of bioenergetic parameters [2]. The potassium Forschung and the Deutsche Forschungsgemeinschaft for financial
potential at the cytoplasmic membrane might be adjusted faster upon support (BMBF SysMap program (0313704) and SFB796, B5).
overexpression of cglK because more channels mediate a higher
potassium flux, but at thermodynamical equilibrium the same References
internal potassium concentration will be achieved. Consequently,
[1] D. McLaggan, J. Naprstek, E.T. Buurman, W. Epstein, Interdependence of K+ and
survival of cells depends on the availability of potassium and efficient glutamate accumulation during osmotic adaptation of Escherichia coli, J. Biol.
transport systems are required for growth and survival [6]. This Chem. 269 (1994) 1911–1917.
conclusion is in agreement with the exclusive occurrence of Ktr- and [2] M. Follmann, M. Becker, I. Ochrombel, V. Ott, R. Krämer, K. Marin, Potassium
transport in Corynebacterium glutamicum is facilitated by the putative channel
Trk-type potassium transport systems in representatives of the protein CglK, which is essential for pH homeostasis and growth at acidic pH, J.
Actinobacteria that are supposed to be challenged by such stress Bacteriol. 191 (2009) 2944–2952.
conditions. [3] W. Epstein, The roles and regulation of potassium in bacteria, Prog. Nucleic Acid
Res. Mol. Biol. 75 (2003) 293–320.
The presence of the C. jeikeium potassium transporter KtrAB could [4] Y. Jiang, A. Lee, J. Chen, M. Cadene, B.T. Chait, R. MacKinnon, Crystal structure and
complement the loss of homologous potassium transport systems in mechanism of a calcium-gated potassium channel, Nature 417 (2002) 515–522.
C. glutamicum and, moreover, was found to be beneficial even for [5] A.N. Thompson, D.J. Posson, P.V. Parsa, C.M. Nimigean, Molecular mechanism of
pH sensing in KcsA potassium channels, Proc. Natl Acad. Sci. USA 105 (2008)
C. glutamicum wild type cells. Cells harboring the KtrAB transporter can
6900–6905.
accumulate higher levels of internal potassium which causes an improved [6] K. Stingl, S. Brandt, E.M. Uhlemann, R. Schmid, K. Altendorf, C. Zeilinger, C.
homeostasis of the internal pH and the membrane potential in particular Ecobichon, A. Labigne, E.P. Bakker, H. de Reuse, Channel-mediated potassium
under conditions of potassium limitation. The internal pH in ktrBA uptake in Helicobacter pylori is essential for gastric colonization, EMBO J. 26
(2007) 232–241.
expressing cells was kept 0.7 units higher upon application of acidic stress. [7] A.M. Whatmore, R.H. Reed, Determination of turgor pressure in Bacillus subtilis: a
A decrease of the internal pH by 0.7 units under the same conditions possible role for K+ in turgor regulation, J. Gen. Microbiol. 136 (1990) 2521–2526.
caused the reduction of growth rate by 50% in wild type cells [11]. This fact [8] R.G. Kroll, I.R. Booth, The role of potassium transport in the generation of a pH
gradient in Escherichia coli, Biochem. J. 198 (1981) 691–698.
seems to be the basic advantage of ktrBA expressing cells over wild type [9] T.M. Embley, E. Stackebrandt, The molecular phylogeny and systematics of the
cells. In agreement with the more neutral internal pH during acidic stress actinomycetes, Annu. Rev. Microbiol. 48 (1994) 257–289.
the membrane potential was found to be not increased in C. glutamicum [10] H. Kase, K. Nakayama, Production of L-threonin by analog-resistant mutants,
Agric. Biol. Chem. 36 (1972) 1611–1621.
cells harboring the KtrAB system and the respiratory activity can be [11] M. Follmann, I. Ochrombel, R. Krämer, C. Trötschel, A. Poetsch, C. Rückert, A. Hüser,
increased in a KtrAB-dependent manner. M. Persicke, D. Seiferling, J. Kalinowski, K. Marin, Functional genomics of pH
homeostasis in Corynebacterium glutamicum revealed novel links between pH
response, oxidative stress, iron homeostasis and methionine synthesis, BMC
4.4. The presence of active potassium transport systems improves Genomics 10 (2009) 621.
[12] B.J. Eikmanns, E. Kleinertz, W. Liebl, H. Sahm, A family of Corynebacterium
survival under stress conditions glutamicum/Escherichia coli shuttle vectors for cloning, controlled gene expres-
sion, and promoter probing, Gene 102 (1991) 93–98.
We showed that the maintenance of bioenergetic parameters in C. [13] C. Ben Salem, H. Hmouda, K. Bouraoui, Drug-induced hypokalaemia, Curr. Drug
Saf. 4 (2009) 55–61.
glutamicum upon expression of the ktrBA genes of C. jeikeium was [14] M. Denda, J. Hosoi, Y. Asida, Visual imaging of ion distribution in human
improved and this fact was correlated to the significantly enhanced epidermis, Biochem. Biophys. Res. Commun. 272 (2000) 134–137.
growth at acidic pH and survival during desiccation stress. The [15] N. Tholema, M. Vor der Bruggen, P. Maser, T. Nakamura, J.I. Schroeder, H.
Kobayashi, N. Uozumi, E.P. Bakker, All four putative selectivity filter glycine
increased survival of C. glutamicum cells harboring KtrAB was more
residues in KtrB are essential for high affinity and selective K+ uptake by the
pronounced under potassium limitation and in the presence of KtrAB system from Vibrio alginolyticus, J. Biol. Chem. 280 (2005) 41146–41154.
sodium, which is in agreement with the properties of KtrAB [16] R.H. Reed, S.R.C. Warr, D.L. Richardson, W.D.P. Stewart, Multiphasic osmotic
transporters. Interestingly, C. jeikeium is exposed to both types of adjustment in a euryhaline cyanobacterium, FEMS Microbiol. Lett. 28 (1985)
225–229.
stress conditions during colonization of the human skin. The pH on [17] D. Billi, M. Potts, Life and death of dried prokaryotes, Res. Microbiol. 153 (2002)
dry skin is approx. 5.5 or can drop to 5.0 in sweat and low potassium 7–12.
(5 mM) and high sodium concentrations (80 mM) on skin or in sweat [18] R.M. Morgan, M.J. Patterson, M.A. Nimmo, Acute effects of dehydration on sweat
composition in men during prolonged exercise in the heat, Acta Physiol. Scand.
can be found [18]. This is in agreement with the hypothesis that the 182 (2004) 37–43.
KtrAB system is of relevance for acclimatization of Actinobacteria [19] M. Potts, Desiccation tolerance of prokaryotes, Microbiol. Rev. 58 (1994) 755–805.
towards low pH and desiccation stress. In agreement, desiccation [20] R.R. Facklam, D.N. Lawrence, F.O. Sottnek, Modified culture technique for
Corynebacterium diphtheriae isolation from desiccated swabs, J. Clin. Microbiol.
tolerance was concluded for C. diphtheriae which was isolated after 7 (1978) 137–138.
9 weeks of desiccation in silica gel-dried swabs. It was also shown for [21] K.I. Racher, R.T. Voegele, E.V. Marshall, D.E. Culham, J.M. Wood, H. Jung, M. Bacon,
M. tuberculosis that can remain viable in dust for up to 120 days and M.T. Cairns, S.M. Ferguson, W.J. Liang, P.J. Henderson, G. White, F.R. Hallett,
Purification and reconstitution of an osmosensor: transporter ProP of Escherichia
upon storage under vegetable oils for approx. 2 years [19,20]. The coli senses and responds to osmotic shifts, Biochemistry 38 (1999) 1676–1684.
comparison of potassium transport systems in these strains revealed [22] S.G. Grant, J. Jessee, F.R. Bloom, D. Hanahan, Differential plasmid rescue from
that in contrast to C. glutamicum mycobacteria harbor genes encoding transgenic mouse DNAs into Escherichia coli methylation-restriction mutants,
Proc. Natl Acad. Sci. USA 87 (1990) 4645–4649.
the high-affinity potassium ATPase Kdp and C. diphtheriae carries a
[23] S. Abe, K. Takayama, S. Kinoshita, Taxonomical studies on glutamic acid-producing
KtrAB transport system in addition to a CglK-type channel. The bacteria, J. Gen. Microbiol. 13 (1967) 279–301.
correlation between desiccation tolerance and effective potassium [24] J.D. Thompson, T.J. Gibson, F. Plewniak, F. Jeanmougin, D.G. Higgins, The
uptake is underlined by the presence of genes encoding the KtrAB and CLUSTAL_X windows interface: flexible strategies for multiple sequence align-
ment aided by quality analysis tools, Nucleic Acids Res. 25 (1997) 4876–4882.
Kdp carrier in the genome of the highly desiccation-tolerant [25] R.D. Page, TreeView: an application to display phylogenetic trees on personal
Deinococcus radiodurans R1 [21]. computers, Comput. Appl. Biosci. 12 (1996) 357–358.

72
Publications

3.5 Evaluation of invertebrate infection models for pathogenic


corynebacteria (Ott et al, 2012a)

73
Publications

RESEARCH ARTICLE

Evaluation of invertebrate infection models for pathogenic


corynebacteria
Lisa Ott1, Ashleigh McKenzie2, Maria Teresa Baltazar2, Sabine Britting1,2, Andrea Bischof1,2,
Andreas Burkovski1 & Paul A. Hoskisson2
1
Lehrstuhl für Mikrobiologie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany; and 2Strathclyde Institute of Pharmacy and
Biomedical Sciences, University of Strathclyde, Glasgow, UK

Correspondence: Andreas Burkovski, Abstract


Lehrstuhl für Mikrobiologie, Friedrich-
IMMUNOLOGY & MEDICAL MICROBIOLOGY

Alexander-Universität Erlangen-Nürnberg, For several pathogenic bacteria, model systems for host–pathogen interactions
Staudtstr. 5, 91058 Erlangen, were developed, which provide the possibility of quick and cost-effective high
Germany. Tel.: +49 9131 85 28086; throughput screening of mutant bacteria for genes involved in pathogenesis. A
Fax: +49 9131 85 28082; e-mail: number of different model systems, including amoeba, nematodes, insects, and
aburkov@biologie.uni-erlangen.de
fish, have been introduced, and it was observed that different bacteria respond
in different ways to putative surrogate hosts, and distinct model systems might
Received 7 October 2011; revised 8 March
2012; accepted 9 March 2012.
be more or less suitable for a certain pathogen. The aim of this study was to
develop a suitable invertebrate model for the human and animal pathogens
DOI: 10.1111/j.1574-695X.2012.00963.x Corynebacterium diphtheriae, Corynebacterium pseudotuberculosis, and Coryne-
bacterium ulcerans. The results obtained in this study indicate that Acanth-
Editor: Nicholas Carbonetti amoeba polyphaga is not optimal as surrogate host, while both Caenorhabtitis
elegans and Galleria larvae seem to offer tractable models for rapid assessment
Keywords of virulence between strains. Caenorhabtitis elegans gives more differentiated
Acanthamoeba; Caenorhabtitis
results and might be the best model system for pathogenic corynebacteria,
elegans; Corynebacterium diphtheriae;
given the tractability of bacteria and the range of mutant nematodes available
Corynebacterium pseudotuberculosis;
Corynebacterium ulcerans; Galleria to investigate the host response in combination with bacterial virulence. Never-
mellonella. theless, Galleria will also be useful in respect to innate immune responses to
pathogens because insects offer a more complex cell-based innate immune sys-
tem compared with the simple innate immune system of C. elegans.

best-investigated pathogenic corynebacterium; however,


Introduction
even for this species, only a few virulence factors have
The genus Corynebacterium belongs to the class of been characterized. Beside the diphtheria toxin, these
Actinobacteria (high G+C gram-positive bacteria) and include mainly pili and other adhesion factors (for
comprises a collection of morphologically similar, irregu- reviews, see Collier, 2001; Rogers et al., 2011). Already
lar-shaped, or club-shaped microorganisms (Ventura these data show a high degree of variation in different
et al., 2007; Zhi et al., 2009). To date, more than 80 isolates, for example, in respect to pili formation (Ott
species were taxonomically classified into the genus et al., 2010). Molecular data on infection are even scarcer
Corynebacterium including industrially important bacteria, for two closely related species, Corynebacterium ulcerans
commensals of animals and humans as well as pathogens. and Corynebacterium pseudotuberculosis, which are separate
A well-known member of the pathogenic species is Cory- species within a distinct cluster of the genus Corynebacte-
nebacterium diphtheriae, which is also the type species of rium but share the ability to harbor diphtheria toxin-
the whole genus. encoding lysogenic corynephage (Riegel et al., 1995).
Corynebacterium diphtheriae is the etiological agent of Corynebacterium ulcerans has been detected as a
diphtheria, a localized toxaemic infection of respiratory commensal in domestic and wild animals, which may
tract and skin that can be fatal (Hadfield et al., 2000). serve as reservoirs for zoonotic infections. During the last
Owing to its medical importance, C. diphtheriae is the decade, the frequency and severity of human infections

FEMS Immunol Med Microbiol && (2012) 1–9 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved

74
Publications

2 L. Ott et al.

associated with C. ulcerans appear to be increasing in var- moeba polyphaga, which is especially applied to investigate
ious countries. Respiratory diphtheria-like illnesses caused adhesion and phagocytosis (Mattos-Guaraldi et al., 2008),
by toxigenic C. ulcerans strains are increasingly reported the nematode Caenorhabtitis elegans, which has been
from various industrialized countries (Tiwari et al., 2008) widely used and often serves as an oral infection route
and have become more common than C. diphtheriae model (for review see Dorer & Isberg, 2006; Waterfield
infections in the United Kingdom (Wagner et al., 2010). et al., 2008; O’Callaghan & Vergunst, 2010), and larvae of
Infections of humans with toxigenic C. ulcerans can be the greater wax moth (Galleria mellonella), an established
fatal in unvaccinated patients. They usually occur in model which represents the more complex insect immune
adults who have consumed raw milk or had close contact system (Waterfield et al., 2008). Bacterial strains were
with domestic animals (Bostock et al., 1984; Hart, 1984; selected based on their previous characterization and the
Wagner et al., 2010). Beside respiratory diphtheria-like availability of experimental data and sequence informa-
illnesses, C. ulcerans can also cause extrapharyngeal infec- tion.
tions in humans, including severe pulmonary infections
(Dessau et al., 1995; Nureki et al., 2007; Cosson & Sold-
Materials and methods
ati, 2008).
Corynebacterium pseudotuberculosis is the etiological
Strains and growth conditions
agent of caseous lymphadenitis, which is prevalent in
sheep and goat herds throughout the world (Dorella Strains used in this study are listed in Table 1. Corynebac-
et al., 2006; Baird & Fontaine, 2007). Infections due to terium diphtheriae, C. ulcerans, and C. pseudotuberculosis
C. pseudotuberculosis are rare in humans, but this patho- were grown in heart infusion (HI) broth at 37 °C, Cory-
gen is occasionally recovered from cases of suppurative nebacterium glutamicum was grown in brain heart infu-
lymphadenitis in patients with a classical risk exposure of sion (BHI) broth at 30 °C, and Escherichia coli OP50 was
close contact with sheep. Corynebacterium pseudotubercu- grown in Luria broth (LB) (Sambrook et al., 1989) at
losis is a facultative intracellular pathogen that is able to 37 °C. If appropriate, kanamycin was added (25 lg mL!1
survive and grow in macrophages, thus escaping the for C. glutamicum and C. pseudotuberculosis, 30 lg mL!1
immune response of the host (McKean et al., 2005; for E. coli, 50 lg mL!1 for C. diphtheriae and C. ulcer-
Dorella et al., 2006). ans). Caenorhabtitis elegans N2, used as the wild type
As mentioned earlier, the three described species are strain, were maintained and propagated on E. coli OP50
not very well characterized on a molecular level, despite as described (Brenner, 1974).
the fact that genome sequences became available for
C. diphtheriae (Cerdeno-Tarraga et al., 2003), C. ulcerans
Infection of Acanthamoeba polyphaga
(Trost et al., 2011), and C. pseudotuberculosis (Trost
et al., 2010; Cerdeira et al., 2011a, b; Silva et al., 2011) For a amoebae-bacteria co-culture assay, A. polyphaga
during the last years. This might be partly due to the lack cultures were grown to exponential phase (3–5 days) in
of a suitable, easy-to-handle infection model, which Peptone Yeast Glucose medium at 21 °C (Rowbotham,
would allow fast identification of virulence factors in 1980). Cells were harvested by centrifugation and re-sus-
large-scale mutant pools. Besides the classical guinea-pig pended in phosphate-buffered saline (PBS), and live cell
model already used by Loeffler, a mouse model for septic titers (visualized by trypan blue exclusion) were carried
arthritis was successfully established for C. diphtheriae out to assess the number of amoebae for each assay (nor-
(Puliti et al., 2006); however, nonmammalian model mally 2 9 105 amoebae mL!1). Corynebacteria were
systems might offer several advantages over mammals, for grown overnight in HI medium, diluted to an OD600 nm
example, in respect to cost effectiveness, handling, and of 0.1 and grown to an OD600 nm of 0.6 and compared
ethical aspects. with a standard curve of viable numbers for infection
The aim of this study was to identify a nonmammalian assays at a range of multiplicity of infection (MOI). The
infection model for pathogenic corynebacteria with amoebae and bacteria were combined in a 24-well plate
potential for high throughput screening and testing of at a MOI of 1 : 1 or 1 : 10. Plates were incubated at 21 °
mutants. A number of different model systems, including C, with cells harvested for microscopy at the indicated
amoeba, nematodes, insects, and fish, have been intro- time intervals.
duced to study host–pathogen interactions (for recent For a phagocytic plaque assay of amoebae killing, the
reviews, see Dorer & Isberg, 2006; Kurz & Ewbank, 2007, method of Froquet et al. (2009) was adapted for assaying
O’Callaghan & Vergunst, 2010). Here, we present data for the interaction of A. polyphaga and Corynebacterium spe-
three invertebrate models and their suitability as surrogate cies. Aliquots (0.75 mL) of HI agar in varying concentra-
hosts of pathogenic corynebacteria, the amoeba Acantha- tions (10–100% of the manufacturer’s recommended

ª 2012 Federation of European Microbiological Societies FEMS Immunol Med Microbiol && (2012) 1–9
Published by Blackwell Publishing Ltd. All rights reserved

75
Publications

Corynebacterium infection model systems 3

Table 1. Strains, plasmids and cell lines used in this study

Strains Description Reference/Source

Corynebacterium diphtheriae
DSM 43988 Nontoxigenic, isolated from throat culture DSMZ, Braunschweig, Germany
DSM 43989 tox+, unknown source DSMZ, Braunschweig, Germany
ISS3319 C. diphtheriae var. mitis, nontoxigenic, isolated Bertuccini et al. (2004)
from patient affected by pharyngitis/tonsillitis
Tn5-46 ISS 3319 carrying a Tn5 insertion in DIP1546 Laboratory strain collection
(encoding a hypothetical protein)

Corynebacterium glutamicum
ATCC 13032 Type-strain, nonpathogenic Abe et al. (1967)

Corynebacterium pseudotuberculosis
FRC41 Isolated from the inguinal lymph node of a Join-Lambert et al. (2006)
12-year-old girl with necrotizing lymphadenitis

Corynebacterium ulcerans
809 Bronchoalveolar lavage sample from an elderly Trost et al. (2011)
woman with a fatal pulmonary infection
BR-AD22 Nasal sample of an asymptomatic female dog Trost et al. (2011)
ELHA1 Phospholipase D-deficient BR-AD22 mutant strain E. Hacker (pers. commun.)

Escherichia coli
OP50 Uracil auxotrophic E. coli B strain (Brenner, 1974)

Plasmids
pEPR1-p45gfp P45, gfpuv, KmR, rep, per, T1, T2 Knoppova et al. (2007)
pXMJ19mCherry ori colE1, oricg, ptac, mCherry, CmR M. Höller (pers. commun.)

Cell lines
Detroit 562 Human hypopharyngeal carcinoma cells Peterson et al. (1968)

concentration) were placed in each well of a 24-well plate plates at 21 °C for 24 h. Subsequently, C. elegans was
and dried. Diluting the media equalizes the conditions for transferred to plates with 200 lL of unlabeled E. coli OP50
amoeba and bacterial growth and establishes the appro- for a further 24 h, to allow the gut to clear of fluorescent
priate conditions for plaque formation (Froquet et al., organisms. Worms were assessed each day following infec-
2009). Corynebacterial cultures were prepared as described tion, and the dead worms were counted. Transfer back to
earlier and 50 lL of the culture was pipetted out onto the E. coli OP50 is essential following infection as corynebacte-
surface of the agar and allowed to dry. Amoebal cells were ria are not a preferred prey source for C. elegans and con-
harvested as described earlier and diluted to a concentra- tinued feeding results in the nematodes attempting to leave
tion of 200 9 104, 20 9 104, 2 9 104 or 0.2 9 104 live the culture plates. Nematodes were paralyzed with 0.6%
cells mL!1 with PBS. Amoebal suspension (5 lL) was pip- 2-phenoxy-2-propanol (Sigma; Wormbook.org), mounted
etted into the centre of each well. Plates were incubated at onto agar pads, and photographed using a Leica DMR
room temperature for 2–5 days and observed daily for signs fluorescence microscope.
of plaque formation.
Infection of Galleria mellonella larvae
Infection of C. elegans
Aliquots (20 mL) of HI broth were inoculated with bacte-
Caenorhabtitis elegans N2 were maintained on E. coli OP50 ria from an overnight culture to an OD600 nm of 0.1, and
for 6–7 days until the worms become starved, indicated by the cultures were grown until an OD600 nm of 0.6 was
clumping behaviour (de Bono & Bargmann, 1998). The reached. Bacteria were harvested by centrifugation
nematodes were infected with different Corynebacterium (10 min, 4500 g) and resuspended in 10 mM MgSO4 to
wild-type strains, transformed with pEPR1-p45gfp as well an OD600 nm of 10 (approximately 3 9 109 CFU mL!1).
as E. coli OP50 transformed with pEPR1-p45gfp. Infection For infection, a 50-lL Hamilton syringe was used to
of 20 L4 stage larval worms was carried out with 200 lL of inject 5 lL aliquots into G. mellonella larvae via the hind-
each bacteria strain (from an overnight culture) on NGM most left proleg (Jander et al., 2000). For each biological

FEMS Immunol Med Microbiol && (2012) 1–9 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved

76
Publications

4 L. Ott et al.

replicate, five larvae were infected with each strain and inside the acid vacuole of A. polyphaga (data not shown).
incubated at 25 °C for 48 h. Although the amoebae were able to predate corynebacte-
ria, in co-culture experiments, the number of dead
amoeba increased depending on the presence of bacteria
Adhesion assays
(data not shown). The reason for this is unclear; however,
Detroit 562 cells were seeded in 24-well plates (bio-one these results suggest that the A. polyphaga model was not
Cellstar; Greiner, Frickenhausen, Germany) at a density optimal for characterization of corynebacterial strains,
of 2 9 105 cells per well 48 h prior to infection. Bacteria and further experiments with C. diphtheriae, C. pseudotu-
were subcultured (OD600 nm of 0.1 from overnight cul- berculosis, and C. ulcerans strains were abandoned.
tures) in HI broth for 3.5 h and adjusted to an OD600 nm
of 0.2. A master mix of the inoculum was prepared in
Caenorhabtitis elegans
DMEM without penicillin/streptomycin at a MOI of 200
(viable counts experiments). The plates were centrifuged Caenorhabtitis elegans has been extensively used to study
for 5 min at 500 g to synchronize infection and subse- bacterial virulence and offers many advantages such as its
quently incubated for 1.5 h. The cells were washed with ease of culture, microscopic tractability, extensive genetic
PBS nine times, detached with 500 lL trypsin solution resources, mutant libraries, and ease of manipulation
(0.12% trypsin, 0.01% EDTA in PBS) per well (5 min, (Mahajan-Miklos et al., 1999). We colonized C. elegans
37 °C, 5% CO2, 90% humidity), and lysed with 0.025% with a range of GFP-labeled corynebacterial strains and
Tween 20 for 5 min at 37 °C. Serial dilutions were made subsequently transferred to plates with unlabelled E. coli
in prechilled 19 PBS and plated out on blood agar plates. OP50 before microscopy (Fig. 1) and quantifying survival
After overnight incubation, the number of colony form- (Fig. 2). Worms infected with different strains of coryne-
ing units (CFU) was determined. bacteria, exhibiting a range of virulence properties,
showed different characteristics in the kill curve reflecting
the differences in virulence between strains (Fig. 2), with
Results and discussion
the clinical C. diphtheriae strain ISS 3319 resulting in
The use of multiple, disparate lower eukaryotic hosts, has faster worm death rates than strain DSM 43988. Infecting
been used to model bacterial pathogenesis in mammals C. elegans with GFP-labeled E. coli or C. glutamicum
and is well established for some organisms (Mahajan- followed by transfer to unlabeled OP50 E. coli resulted in
Miklos et al., 2000). The application and comparison of clearance of GFP-expressing strains, suggesting that these
such models in corynebacteria to indicate strain-specific bacteria were not colonizing the worms pathogenically. In
differences has not been demonstrated before; however, it the case of C. diphtheriae strain ISS 3319, foregut (buccal
is clear that some of these models offer utility in the cavity and pharynx) and midgut were colonized by the
study of pathogenic corynebacteria. bacteria, while C. diphtheriae DSM 43988 colonized solely
the midgut. Toxigenic C. diphtheriae DSM 43989 as well
as C. ulcerans 809 colonized the hindgut of the worm,
Acanthamoeba polyphaga
while C. ulcerans BR-AD22 and C. pseudotuberculosis
Amoebae species are well-established model systems for a FRC41 spread all over the body. These data suggest that
number of pathogenic bacteria; however, some bacteria different virulence properties and mechanisms used by
respond in different ways to phagocytosis, and it has been the wide range of strains we tested are reflected in the
shown that some bacteria, such as Listeria monocytogenes, localization and the killing kinetics of the experiments.
do not survive ingestion by Acanthamoeba (Akya et al., Investigation of the time required to colonize the worms
2010). Amoebae models have been used extensively for indicated that a minimum of 6 h feeding on pathogenic
studies on phagocytosis of mycobacteria as surrogate corynebacteria was required for successful colonization
macrophages to study vacuole survival and adaptation (data not shown). Interestingly, the colonization of the
(Mattos-Guaraldi et al., 2008). We tested different nematodes in the foregut reflects the natural niche of
C. diphtheriae strains using a modified version of the these organisms in their mammalian hosts, rather than
method of Froquet et al. (2009), plaques on C. diphthe- the disperse colonization of the gut observed in Pseudo-
riae lawns appeared on average after three to five days of monas or Serratia (Mahajan-Miklos et al., 1999; Kurz
incubation, indicating that the corynebacteria tested were et al., 2003). In summary, in contrast to the nonpatho-
not able to resist predation by A. polyphaga (data not genic C. glutamicum and E. coli, pathogenic corynebacte-
shown). This observation was further supported by co- ria were able to colonize C. elegans in a strain-specific
culture experiments and visualization by fluorescence manner and were tractable by microscopic studies. The
microscopy, which indicated digestion of the bacteria availability of innate immune mutant C. elegans strains

ª 2012 Federation of European Microbiological Societies FEMS Immunol Med Microbiol && (2012) 1–9
Published by Blackwell Publishing Ltd. All rights reserved

77
Publications

Corynebacterium infection model systems 5

a1 a2 b1 b2

c1 c2 d1 d2

e1 e2 f1 f2

g1 g2 h1 h2

Fig. 1. Infection of Caenorhabtitis elegans with different corynebacteria. Infection of worms was carried out with 200 lL of each bacterial strain
on NGM plates at 21 °C for 24 h. Nematodes were mounted onto agar pads, paralyzed with 0.6% 2-phenoxy-2-propanol (Sigma), and
photographed using a Leica DMR fluorescent microscope (1 bright field, 2 fluorescence). In each of at least five independent experiments,
approximately 20 worms were infected. Pictures show representative results. Caenorhabtitis elegans N2 fed with (a) Escherichia coli OP50, (b)
Corynebacterium glutamicum ATCC 13032, (c) Corynebacterium diphtheriae ISS3319, (d) C. diphtheriae DSM 43988, (e) C. diphtheriae DSM
43989, (f) Corynebacterium ulcerans 809, (g) C. ulcerans BR-AD22, (h) Corynebacterium pseudotuberculosis FRC41. Tails of nematodes are
indicated by arrows.

would offer attractive possibilities for further studying While control larvae showed a pale whitish colour and
host–corynebacterium interactions. were highly active, injection of C. glutamicum, a non-
pathogenic Corynebacterium species of biotechnological
importance, led to the development of small melanized
Galleria mellonella
spots. The melanization of larvae is associated with path-
The greater wax moth (Galleria mellonella) model system ogen killing and phagocytosis in lepidopteran larvae
has also been successfully used to characterize bacterial (Nappi & Christensen, 2005; Senior et al., 2011). The
virulence in a range of strains, such as in Pseudomonas effects on the larvae became more severe in response to
aeruginosa and Burkholderia to illustrate inter-strain varia- injection of nontoxigenic C. diphtheriae strain DSM
tion in pathogenicity (Jander et al., 2000). The larvae 43988, which led to brown larvae, and toxigenic C. diph-
show a high sensitivity towards these pathogenic bacteria theriae strain DSM 43989, which caused even stronger
resulting in melanization in response to infection, and a melanization, with all infected larvae being immobile
50% lethal dose (LD50) of < 10 bacteria was reported without external stimulation. The strongest effects on the
when these were injected into the hemolymph (Jander larvae were obtained for C. pseudotuberculosis and C. ulc-
et al., 2000; Wand et al., 2011). In general, distinct differ- erans strains, which exhibited high degrees of melaniza-
ences in the response of the larvae were detected when tion, immobility, and rapid death. These data suggest that
different corynebacterial strains were injected (Fig. 3). the C. pseudotuberculosis and C. ulcerans strains are more

FEMS Immunol Med Microbiol && (2012) 1–9 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved

78
Publications

6 L. Ott et al.

Buffer control (a)


100
white, active

80
% Nematode survival

60
Infection with (b)
40 C. glutamicum
white, small black
spots in the belly,
20 active

0 (c)
Infection with
C. diphtheriae
0 20 40 60 80 100 120
ISS3319
Time (h)
white, black spots in
the belly, decreased
Fig. 2. Nematode survival following infection with Corynebacterium
activity
diphtheriae. Control – Escherichia coli OP50 (▼); C. diphtheriae
ISS3319 (▲); C. diphtheriae DSM 43988 (d); C. diphtheriae DSM Infection with (d)
43989 (&). Data are the mean of three experiments, and the error C. diphtheriae
bars represent the standard deviation of the data. DSM43988
completely brown,
decreased activity
virulent in this model, perhaps reflecting their wider host
range when compared to C. diphtheriae; a phenomenon Infection with (e)
observed in the opportunistic broad host range pathogen C. diphtheriae
P. aeruginosa (Jander et al., 2000). DSM43989
brown with black
belly, decreased
Application of model systems activity

Infection with (f)


In addition to the already obvious strain-specific differ- C. pseudotuber-
ences detectable in the C. elegans and Galleria model sys- culosis FRC41
tems, two mutant strains were tested as further proof of black, dead
principle (Fig. 4). When the C. diphtheriae wild-type ISS
3319 was compared with a corresponding transposon
mutant Tn5-46, carrying an insertion in a gene encoding a Infection with (g)
hypothetical protein (DIP1546), the mutant was drastically C. ulcerans 809
black, dead
impaired in gut colonization in the C. elegans model, while
in the Galleria system, no difference of wild type and corre-
sponding mutant was observed upon injection, indicating
that the DIP1546 protein is influencing adhesion and host
Infection with (h)
colonization. In fact, in cell culture experiments, Tn5-46
C. ulcerans
showed a decreased adhesion rate to Detroit 562 cells. BR-AD22
In contrast, the C. ulcerans phospholipase D-deficient black, dead
strain ELHA1 showed unaltered colonization behaviour
compared with the parental strain BR-AD22 in the
C. elegans system, while lack of this virulence factor causes
a less severe response in the Galleria model. In this case, Fig. 3. Infection of Galleria mellonella with different corynebacteria.
the adhesion rates of wild type and mutant were identical. A 50-lL Hamilton syringe was used to inject 5 lL aliquots of the
different bacteria into G. mellonella larvae via the hindmost left
proleg. Five larvae were infected with each strain at 25 °C for 48 h.
Conclusions Infection was carried out with (a) buffer control (10 mM MgSO4),
(b) Corynebacterium glutamicum ATCC 13032, (c) Corynebacterium
The results obtained in this study indicate that A. polyph- diphtheriae ISS3319, (d) C. diphtheriae DSM 43988, (e) C. diphtheriae
aga is not optimal as surrogate host for the study and DSM 43989, (f) Corynebacterium pseudotuberculosis FRC41, (g)
characterization of pathogenic corynebacteria. Both Corynebacterium ulcerans 809, (h) C. ulcerans BR-AD22.

ª 2012 Federation of European Microbiological Societies FEMS Immunol Med Microbiol && (2012) 1–9
Published by Blackwell Publishing Ltd. All rights reserved

79
Publications

Corynebacterium infection model systems 7

Wild type Mutant Wild type Mutant


2.00

1.50

Adhesion [%]
ISS3319

1.00

0.50

0.00
ISS3319 TN5-46

80.00

Adhesion [%]
60.00
BRAD22

40.00

20.00

0.00
BRAD22 ELHA-1

Fig. 4. Application of model systems. Caenorhabtitis elegans and Galleria larvae were infected with mCherry-labelled corynebacteria (details as
described in legends to Figs 1 and 3), and adhesion rates to Detroit 562 cells were determined for the corresponding strains (experiments carried
out in at least in triplicate). Upper panel: Corynebacterium diphtheriae strain ISS 3319 and corresponding mutant Tn5-46; lower panel:
Corynebacterium ulcerans strain BR-AD22 and phospholipase D-deficient mutant ELHA1. Tails of C. elegans are indicated by arrows.

C. elegans and Galleria larvae seem to offer attractive support of R. Palmisano (Friedrich-Alexander-Universität
models for rapid assessment of virulence differences Erlangen-Nürnberg) in respect to fluorescence microscopy
between strains. While the Galleria larvae are very easy to is gratefully acknowledged.
handle, C. elegans gives the more differentiated results,
has extensive genetic tools as a host, is microscopically
References
tractable, and might represent the best model system for
pathogenic corynebacteria, especially when factors respon- Abe S, Takayama K & Kinoshita S (1967) Taxonomical studies
sible for colonization are screened. Based on our data, on glutamic acid-producing bacteria. J Gen Appl Microbiol
both of these hosts, however, will be useful given the 13: 279–301.
differences detectable in the Galleria system in response Akya A, Pointon A & Thomas C (2010) Listeria monocytogenes
to absence or presence of toxins (diphtheria toxin) and does not survive ingestion by Acanthamoeba polyphaga.
other virulence factors (phospholipase D). Furthermore, Microbiology 156: 809–818.
Baird GJ & Fontaine MC (2007) Corynebacterium
insects such as Galleria offer the possibility to study a
pseudotuberculosis and its role in ovine caseous
more complex cell-based innate immune system com-
lymphadenitis. J Comp Pathol 137: 179–210.
pared with the simple innate immune system of C. ele-
Bertuccini L, Baldassarri L & von Hunolstein C (2004)
gans (Mahajan-Miklos et al., 2000). Internalization of non-toxigenic Corynebacterium diphtheriae
by cultured human respiratory epithelial cells. Microb Pathog
Acknowledgements 37: 111–118.
Bostock AD, Gilbert FR, Lewis D & Smith DC (1984)
The Burkovski laboratory is supported by the Deutsche Corynebacterium ulcerans infection associated with untreated
Forschungsgemeinschaft in frame of SFB 796 (project B5). milk. J Infect 9: 286–288.
The work in the Hoskisson laboratory is supported by Brenner S (1974) The genetics of Caenorhabditis elegans.
Medical Research Scotland (Grant FRG-422) and by a Genetics 77: 71–94.
Society for General Microbiology Vacation Studentship Cerdeira LT, Pinto AC, Schneider MP et al. (2011a) Whole-
grant to A.M. Corynebacterium ulcerans phospholipase D genome sequence of Corynebacterium pseudotuberculosis
mutant ELHA1 was kindly provided by E. Hacker and PAT10 strain isolated from sheep in Patagonia, Argentina.
plasmid pXMJ19mCherry by M. Höller (both Friedrich- J Bacteriol 193: 6420–6421.
Cerdeira LT, Schneider MP, Pinto AC et al. (2011b) Complete
Alexander-Universität Erlangen-Nürnberg), C. elegans N2
genome sequence of Corynebacterium pseudotuberculosis
was a gift from J. Pettitt (University of Aberdeen). The

FEMS Immunol Med Microbiol && (2012) 1–9 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved

80
Publications

8 L. Ott et al.

strain CIP 52.97, isolated from a horse in Kenya. J Bacteriol Mattos-Guaraldi AL, Sampaio JL, Santos CS et al. (2008) First
193: 7025–7026. detection of Corynebacterium ulcerans producing a
Cerdeno-Tarraga AM, Efstratiou A, Dover LG et al. (2003) diphtheria-like toxin in a case of human with pulmonary
The complete genome sequence and analysis of infection in the Rio de Janeiro metropolitan area, Brazil.
Corynebacterium diphtheriae NCTC13129. Nucleic Acids Res Mem Inst Oswaldo Cruz 103: 396–400.
31: 6516–6523. McKean S, Davies J & Moore R (2005) Identification of
Collier RJ (2001) Understanding the mode of action of macrophage induced genes of Corynebacterium
diphtheria toxin: a perspective on progress during the 20th pseudotuberculosis by differential fluorescence induction.
century. Toxicon 39: 1793–1803. Microbes Infect 7: 1352–1363.
Cosson P & Soldati T (2008) Eat, kill or die: when amoeba Nappi AJ & Christensen BM (2005) Melanogenesis and
meets bacteria. Curr Opin Microbiol 11: 271–276. associated cytotoxic reactions: applications to insect innate
de Bono M & Bargmann CI (1998) Natural variation in immunity. Insect Biochem Mol Biol 35: 443–459.
a neuropeptide Y receptor homolog modifies social Nureki S, Miyazaki E, Matsuno O, Takenaka R, Ando M,
behavior and food response in C. elegans. Cell 94: 679–689. Kumamoto T, Nakano T, Ohkusu K & Ezaki T (2007)
Dessau RB, Brandt-Christensen M, Jensen OJ & Tonnesen P Corynebacterium ulcerans infection of the lung mimicking
(1995) Pulmonary nodules due to Corynebacterium ulcerans. the histology of Churg–Strauss syndrome. Chest 131: 1237–
Eur Respir J 8: 651–653. 1239.
Dorella FA, Pacheco LG, Oliveira SC, Miyoshi A & Azevedo V O’Callaghan D & Vergunst A (2010) Non-mammalian animal
(2006) Corynebacterium pseudotuberculosis: microbiology, models to study infectious disease: worms or fly fishing?
biochemical properties, pathogenesis and molecular studies Curr Opin Microbiol 13: 79–85.
of virulence. Vet Res 37: 201–218. Ott L, Höller M, Rheinlaender J, Schäffer TE, Hensel M &
Dorer MS & Isberg RR (2006) Non-vertebrate hosts in the Burkovski A (2010) Strain-specific differences in pili
analysis of host–pathogen interactions. Microbes Infect 8: formation and the interaction of Corynebacterium
1637–1646. diphtheriae with host cells. BMC Microbiol 10: 257.
Froquet R, Lelong E, Marchetti A & Cosson P (2009) Peterson WD Jr, Stulberg CS, Swanborg NK & Robinson AR
Dictyostelium discoideum: a model host to measure bacterial (1968) Glucose-6-phosphate dehydrogenase isoenzymes in
virulence. Nat Protoc 4: 25–30. human cell cultures determined by sucrose-agar gel and
Hadfield TL, McEvoy P, Polotsky Y, Tzinserling VA & cellulose acetate zymograms. Proc Soc Exp Biol Med 128:
Yakovlev AA (2000) The pathology of diphtheria. J Infect 772–776.
Dis 181(Suppl 1): S116–S120. Puliti M, von Hunolstein C, Marangi M, Bistoni F & Tissi L
Hart RJ (1984) Corynebacterium ulcerans in humans and cattle (2006) Experimental model of infection with non-toxigenic
in North Devon. J Hyg (Lond) 92: 161–164. strains of Corynebacterium diphtheriae and development of
Jander G, Rahme LG & Ausubel FM (2000) Positive septic arthritis. J Med Microbiol 55: 229–235.
correlation between virulence of Pseudomonas aeruginosa Riegel P, Ruimy R, de Briel D, Prevost G, Jehl F, Christen R &
mutants in mice and insects. J Bacteriol 182: 3843–3845. Monteil H (1995) Taxonomy of Corynebacterium diphtheriae
Join-Lambert OF, Ouache M, Canioni D, Beretti JL, Blanche S, and related taxa, with recognition of Corynebacterium
Berche P & Kayal S (2006) Corynebacterium ulcerans sp. nov. nom. rev. FEMS Microbiol Lett 126: 271–276.
pseudotuberculosis necrotizing lymphadenitis in a twelve- Rogers EA, Das A & Ton-That H (2011) Adhesion by
year-old patient. Pediatr Infect Dis J 25: 848–851. pathogenic corynebacteria. Adv Exp Med Biol 715: 91–103.
Knoppova M, Phensaijai M, Vesely M, Zemanova M, Nesvera Rowbotham TJ (1980) Preliminary report on the pathogenicity
J & Patek M (2007) Plasmid vectors for testing in vivo of Legionella pneumophila for freshwater and soil amoebae.
promoter activities in Corynebacterium glutamicum and J Clin Pathol 33: 1179–1183.
Rhodococcus erythropolis. Curr Microbiol 55: 234–239. Sambrook J, Fritsch EF & Maniatis T 1989. Molecular cloning.
Kurz CL & Ewbank JJ (2007) Infection in a dish: high- A laboratory manual. Cold Spring Harbor Laboratory Press,
throughput analyses of bacterial pathogenesis. Curr Opin Cold Spring Harbor, NY.
Microbiol 10: 10–16. Senior NJ, Bagnall MC, Champion OL, Reynolds SE, La
Kurz CL, Chauvet S, Andres E et al. (2003) Virulence factors Ragione RM, Woodward MJ, Salguero FJ & Titball RW
of the human opportunistic pathogen Serratia marcescens (2011) Galleria mellonella as an infection model for
identified by in vivo screening. EMBO J 22: 1451–1460. Campylobacter jejuni virulence. J Med Microbiol 60: 661–669.
Mahajan-Miklos S, Tan MW, Rahme LG & Ausubel FM Silva A, Schneider MP, Cerdeira L et al. (2011) Complete
(1999) Molecular mechanisms of bacterial virulence genome sequence of Corynebacterium pseudotuberculosis I19,
elucidated using a Pseudomonas aeruginosa–Caenorhabditis a strain isolated from a cow in Israel with bovine mastitis.
elegans pathogenesis model. Cell 96: 47–56. J Bacteriol 193: 323–324.
Mahajan-Miklos S, Rahme LG & Ausubel FM (2000) Tiwari TS, Golaz A, Yu DT et al. (2008) Investigations of 2
Elucidating the molecular mechanisms of bacterial virulence cases of diphtheria-like illness due to toxigenic
using non-mammalian hosts. Mol Microbiol 37: 981–988. Corynebacterium ulcerans. Clin Infect Dis 46: 395–401.

ª 2012 Federation of European Microbiological Societies FEMS Immunol Med Microbiol && (2012) 1–9
Published by Blackwell Publishing Ltd. All rights reserved

81
Publications

Corynebacterium infection model systems 9

Trost E, Ott L, Schneider J et al. (2010) The complete genome Wand ME, Müller CM, Titball RW & Michell SL (2011)
sequence of Corynebacterium pseudotuberculosis FRC41 Macrophage and Galleria mellonella infection models reflect
isolated from a 12-year-old girl with necrotizing the virulence of naturally occurring isolates of B.
lymphadenitis reveals insights into gene-regulatory networks pseudomallei, B. thailandensis and B. oklahomensis. BMC
contributing to virulence. BMC Genomics 11: 728. Microbiol 11: 11.
Trost E, Al-Dilaimi A, Papavasiliou P et al. (2011) Waterfield NR, Sanchez-Contreras M, Eleftherianos I et al.
Comparative analysis of two complete Corynebacterium (2008) Rapid Virulence Annotation (RVA): identification of
ulcerans genomes and detection of candidate virulence virulence factors using a bacterial genome library and
factors. BMC Genomics 12: 383. multiple invertebrate hosts. P Natl Acad Sci USA 105:
Ventura M, Canchaya C, Fitzgerald GF, Gupta RS & van 15967–15972.
Sinderen D (2007) Genomics as a means to understand Zhi XY, Li WJ & Stackebrandt E (2009) An update of the
bacterial phylogeny and ecological adaptation: the case of structure and 16S rRNA gene sequence-based definition of
bifidobacteria. Antonie Van Leeuwenhoek 91: 351–372. higher ranks of the class Actinobacteria, with the proposal
Wagner KS, White JM, Crowcroft NS, De Martin S, Mann G of two new suborders and four new families and emended
& Efstratiou A (2010) Diphtheria in the United Kingdom, descriptions of the existing higher taxa. Int J Syst Evol
1986-2008: the increasing role of Corynebacterium ulcerans. Microbiol 59: 589–608.
Epidemiol Infect 138: 1519–1530.

FEMS Immunol Med Microbiol && (2012) 1–9 ª 2012 Federation of European Microbiological Societies
Published by Blackwell Publishing Ltd. All rights reserved

82
Publications

3.6 Contour and persistence length of Corynebacterium diphtheriae


pili by atomic force microscopy (Rheinlaender et al, 2012)

83
Publications
Eur Biophys J
DOI 10.1007/s00249-012-0818-4

ORIGINAL PAPER

Contour and persistence length of Corynebacterium diphtheriae


pili by atomic force microscopy
Johannes Rheinlaender • Anna Gräbner •
Lisa Ott • Andreas Burkovski • Tilman E. Schäffer

Received: 10 January 2012 / Revised: 5 April 2012 / Accepted: 21 April 2012


! European Biophysical Societies’ Association 2012

Abstract Many bacteria are characterized by nanoscale proved to be more precise and resulted in persistence
ultrastructures, for example S-layers, flagella, fimbriae, or lengths in the narrow range of 220–280 nm, with no sig-
pili. The last two are especially important for attachment to nificant strain-specific variations. This is small compared
different abiotic and biotic surfaces and for host–pathogen with some other bacterial polymers, for example type IV
interactions. In this study, we investigated the geometric pili, F-pili, or flagella.
and elastic properties of pili of different Corynebacterium
diphtheriae strains by atomic force microscopy (AFM). We Keywords Bacteria ! AFM ! Contour length ! Persistence
performed quantitative contour-length analysis of bacterial length ! Stiffness
pili and found that the visible contour length of the pili can
be described by a log-normal distribution. Our data
revealed significant strain-specific variations in the mean Introduction
visible contour length of the pili, ranging from 260 to
1,590 nm. To estimate their full contour length, which is The first crucial steps of interaction of pathogens with their
not directly accessible from the AFM images, we devel- corresponding hosts are host recognition and binding to the
oped a simple correction model. Using this model, we host’s surface. For this purpose, many bacteria possess
determined the mean full contour length as 510–2,060 nm. fimbriae and pili, which facilitate attachment to different
To obtain the persistence length we used two different abiotic and biotic surfaces.
methods of analysis, one based on the end-to-end distance Corynebacterium diphtheriae is the etiological agent of
of the pili and one based on the bending angles of short diphtheria, a localized infection of the upper respiratory
segments. In comparison, the bending angle analysis tract and skin (cutaneous diphtheria), which can be fatal,
because of the potent diphtheria toxin. Adhesion of dif-
Electronic supplementary material The online version of this ferent C. diphtheriae strains to lung, pharyngeal, and lar-
article (doi:10.1007/s00249-012-0818-4) contains supplementary yngeal epithelial cells and to erythrocytes has been
material, which is available to authorized users. reported (Colombo et al. 2001; Bertuccini et al. 2004;
Hirata et al. 2002, 2004). Although it is known that
J. Rheinlaender ! A. Gräbner ! T. E. Schäffer
Institute of Applied Physics, University of Erlangen-Nuremberg, both cell-wall-linked surface proteins and fibrous protein
Erlangen, Germany polymers, for example fimbriae and pili, are of major
importance in the adhesion process for many types of
L. Ott ! A. Burkovski
Gram-positive bacteria, many aspects of the underlying
Chair for Microbiology, University of Erlangen-Nuremberg,
Erlangen, Germany mechanisms remain unclear (Rogers et al. 2011). For
C. diphtheriae the effect of pili on recognition of and
Present Address: adhesion to host cells have recently been characterized on
T. E. Schäffer (&)
the genetic level. Analysis of mutants has revealed that
Institute of Applied Physics, University of Tübingen,
Auf der Morgenstelle 10, 72076 Tübingen, Germany C. diphtheriae type strain NCTC13129 is able to assemble
e-mail: Tilman.Schaeffer@uni-tuebingen.de pili from different pilin subunits on its surface. Pili are

123
84
Publications
Eur Biophys J

important for bacterium–host cell contact and host cell on a glass slide or freshly cleaved mica substrate by drying,
preference (Gaspar and Ton-That 2006; Swierczynski and by use of compressed air.
Ton-That 2006; Mandlik et al. 2007), making them an
interesting focus of research to understand host–pathogen Atomic force microscopy
interaction. Recently, it has been reported that the expression
of different pilin subunit proteins in C. diphtheriae affects pili The samples were imaged by AFM (MFP-3D; Asylum
length qualitatively (Swierczynski and Ton-That 2006; Research, Santa Barbara, USA) using standard silicon
Mandlik et al. 2008; Guttilla et al. 2009). However, a cantilever probes (NCH-W; Nanosensors, Neuchatel,
comprehensive statistical analysis of the pili, e.g. giving Switzerland) in tapping mode under ambient laboratory
the mean contour length, has not yet been reported for conditions. All images are unprocessed except for line-by-
C. diphtheriae. Also, the persistence length, which is an line polynomial background removal (‘‘flattening’’). All
important measure of the mechanical stiffness of filamen- data analysis was performed in IgorPro (Wavemetrics,
tous polymers, has not yet been reported for C. diphtheriae Lake Oswego, OR, USA) by use of custom-written soft-
pili. ware and least-squares fitting routines.
Pili and other bacterial cell appendices and surface
structures are often studied by electron microscopy, which
requires staining or labeling procedures. Atomic force
Theoretical background
microscopy (AFM) can image the topography of unstained
samples and has already been intensively applied to the
Persistence length
study of bacteria (Anselmetti et al. 2007; Dupres et al.
2009; Wright et al. 2010) and to bacterial pili (Touhami
To determine the persistence length of the pili, we used
et al. 2006; Miller et al. 2006; Fälker et al. 2008; Arce et al.
the worm-like chain (WLC) model, which describes a
2009). The contour length of the pili can vary by one order
semi-flexible polymer chain in thermodynamic equilib-
of magnitude within a single cell (Yanagawa and Honda
rium (Landau et al. 1980). The WLC model has been
1976; Touhami et al. 2006). However, when imaged while
applied to many filamentous biomolecules and biostruc-
being attached to the cell body, the contour length of the
tures, for example actin (Nagashima and Asakura 1980;
pili can be systematically underestimated.
Yanagida et al. 1984), microtubules (Gittes et al. 1993),
Here we describe comprehensive characterization of pili
DNA (Rivetti et al. 1996), bacterial flagella (Trachtenberg
ultrastructure for five different C. diphtheriae strains, giv-
and Hammel 1992), type F-pili (Silverman and Clarke
ing two important quantities: the mean full contour length
2010), and type IV pili (Skerker and Berg 2001). In this
and the persistence length. For this purpose, we develop a
model, the polymer is described as a homogenous elastic
simple model for estimating the mean full contour length
rod with a circular cross-section. The energy U required
from the experimentally accessible mean visible contour
to bend a polymer segment of length DL by an angle # is
length in high resolution microscopy images. To obtain
given by:
persistence length of the pili we used two different analysis
methods: one based on the end-to-end distance and one EI 2
U ð# Þ ¼ # ; ð1Þ
based on the bending angles of short pili segments. Finally, 2DL
we relate the strain-specific variations in pili formation to where E is the stiffness (Young’s modulus) and I = pr4/4
the different abilities for adhesion and invasion of host is the polymer’s cross sectional area moment of inertia,
cells found in previous studies and to genetic information where r is the radius of the cross section (Landau et al.
(Swierczynski and Ton-That 2006; Mandlik et al. 2007; 1986). In thermodynamic equilibrium, the probability p
Guttilla et al. 2009; Ott et al. 2010). that the segment is bent by a specific angle is given by
Boltzmann’s law:
U ð# Þ 1P
pð#Þ / e$ kB T ¼ e$2DL# :
2
Methods ð2Þ
Consequently, the standard deviation r# of the bending
Growth of bacteria and sample preparation
angles is given by r2# = h#2i = DL/P and the moments of
the bending angle distribution fulfill h#4i/h#2i2 = 3. The
Corynebacterium diphtheriae overnight cultures grown in
persistence length of the polymer:
20 ml HI broth were washed five times in 20 ml ice-cold
distilled water and were resuspended in 10 ml ice-cold EI
P¼ ; ð3Þ
distilled water. Five microliters of each sample was fixed kB T

123
85
Publications
Eur Biophys J

can be interpreted as the distance along the polymer at Experimental results


which its angular orientation becomes uncorrelated. For a
long polymer of contour length L that is confined within a Characterization of the ultrastructure of pili
plane, the WLC model predicts the mean squared end-to-
end distance as (Rivetti et al. 1996): AFM topography images of C. diphtheriae strains revealed
% $& pili on the substrate extending from the cell bodies
! 2" 2P # L
R ¼ 4PL 1 " 1 " e"2P : ð4Þ (Fig. 1a–e, arrows). Strain-specific differences in pili
L
number and morphology were apparent. For example,
Log-normal distribution whereas DSM44123 cells had only a small number of sub-
micrometer long, relatively straight pili (Fig. 1b), ISS4749
Growth processes with randomly fluctuating growth rates cells had many pili, which were much longer and at first
often have log-normal distributions (Koch 1966). Hence, sight seemed more curved (Fig. 1e). DSM43988 cells had
many quantities in biology, but also in many other fields, no pili (Fig. 1f).
for example medicine, social sciences, and economics, are To obtain quantitative, strain-specific differences among
log-normally distributed (Limpert et al. 2001). For a log- length and curvature, we performed a systematic study of
normally distributed random variable L, the variable’s the pili. First, we recorded many images of pili for the
natural logarithm x = ln(L) is normally distributed. So the different strains at high resolution. In offline analysis, we
probability density function / for x is: then manually traced the pili by selecting close-spaced
! points along their contour (Fig. 2a). Only pili that were
1 ð x " lÞ 2 clearly identifiable along their entire contour on the sub-
/l;r ð xÞ ¼ pffiffiffiffiffi exp " ; ð5Þ
2pr 2r2 strate and with a visible length of at least 100 nm were
selected for analysis. Sufficient statistical precision was
where l and r are the arithmetic mean and the standard
achieved by analyzing approximately 30 individual pili for
deviation, respectively, of the distribution. Consequently,
each strain which originated from 6 to 17 different cells.
the probability density function of L is:
To investigate the effect of the substrate on persistence
!
dx 1 ðlnðLÞ " lÞ2 length, cells prepared on both glass and mica were ana-
f ðLÞ ¼ /l;r ð xÞ ¼ pffiffiffiffiffi exp " ð6Þ lyzed. Finally, we converted the sets of points into smooth
dL 2prL 2r2
contour paths by cubic spline interpolation (Fig. 2b).
and the cumulative distribution function is: Interpolation was necessary because contour length esti-
mation of a curved structure from a discrete set of points
ZL lnðLÞ
Z can lead to significant discrepancies between actual and
0 0
F ðLÞ ¼ f ðL Þ dL ¼ /l;r ðx0 Þ dx0 determined contour length (Dorst and Smeulders 1987).
0 ( "1 ) These discrepancies can be reduced to less than 1 % by
1 lnðLÞ " l cubic spline interpolation, as was demonstrated for AFM
¼ erfc " pffiffi : ð7Þ
2 2r images of DNA molecules (Rivetti and Codeluppi 2000).
Here, ‘‘erfc’’ denotes the complementary error function. We also estimated the diameters of the pili by analyzing
For a log-normal distribution the geometric mean L! is their cross sections in the high-resolution AFM images;
they were 1–2 nm for all strains.
related to the arithmetic mean l of x by L! ¼ expðlÞ. The
standard deviation of x, r, can be transformed to the geo-
Visible contour length
metric standard deviation of L, rGSD = exp(r). For the
normally distributed x the interval l ± r ranges from
As is apparent in the AFM images (Fig. 1), the visible
l - r to l ? r and covers approximately 68.3 % of the
contour length of the pili for a given C. diphtheriae strain
data. For the log-normally distributed L the corresponding
! GSD to L! 9 rGSD. The varies. We therefore statistically analyzed the measured
interval exp(l ± r) ranges from L/r
visible contour lengths L of the imaged pili by generating a
95 % confidence interval of the mean after N measure-
pffiffiffiffi cumulative histogram for each strain (Fig. 3a, b, and Suppl.
ments, which ranges from l - t 9 r/ N to l ? t 9 Fig. S1, black curves). The log-normal distribution
pffiffiffiffi
r/ N for a normally distributed x, is transformed accord- describes the data well, as can be seen by the close match
pffiffiffiffi
!
ingly, resulting in a range from L/exp[t 9 r/ N ] to between the measured cumulative histograms and the
pffiffiffiffi
L! 9 exp[t 9 r/ N ] for a log-normally distributed L. The corresponding cumulative distribution functions, using
expression t = t0.975,N-1 denotes the 97.5th percentile of the respective L! and rGSD of the data (solid gray curves).
the Student’s t-distribution with N - 1 degrees of freedom. More formally, we confirmed this close match by a

123
86
Publications
Eur Biophys J

Fig. 1 a–f AFM topography a DSM43989 b DSM44123 c ISS3319


images of bacteria from
different C. diphtheriae strains.
Pili of different number, length,
and curvature appear as fine
lines extending from the cell
bodies (blue arrows). For
example, DSM44123 cells
(b) have only a small number of
sub-micrometer long pili,

height
whereas ISS4749 cells (e) have
many pili up to several d ISS4746 e ISS4749 f DSM43988
micrometers long. DSM43988
cells (f) do not have pili. Scan
size 4 lm 9 4 lm, height data
range 500 nm, color scale range
2 nm

Full contour length


a Topography b Contour paths

The pili in the AFM images could be identified on the


substrate but not on the cell bodies, owing to the relatively
large surface roughness (several nanometers) of the cell
bodies. Therefore, the full contour length of a pilus is
longer than its visible contour length by the part that lies on
the cell body (Fig. 3d). The consequence is that the mea-
cell body sured, visible contour lengths (Fig. 3c and Table 1)
underestimate the respective full contour lengths. To esti-
mate the full contour lengths we developed a simple, one-
height
dimensional model using four simplifications.
Fig. 2 Analysis of pili ultrastructure. a High-resolution AFM topog-
raphy image of the edge of a DSM44123 cell. Pili contours were 1. Because C. diphtheriae cells are rod-shaped and
traced by manually selecting several points along each pilus elongated the statistical effect of the pili at the cell
(markers). Only pili that were clearly identifiable along their entire poles is small. We therefore excluded them from the
contour on the substrate and with a visible length of at least 100 nm
were selected for analysis. b A smooth contour path was generated
model.
from selected points along each pilus by cubic spline interpolation, 2. A pilus is anchored in the cell surface at a random
enabling an accurate determination of the visible contour length L and position.
the visible end-to-end distance R. Scan size 500 nm 9 500 nm, color 3. Because the persistence length is similar in size to the
scale range 2 nm
width of the body of a typical cell, the pili were
modeled as straight.
4. For simplicity we assumed the pili were of equal
Kolmogorow–Smirnow test using a significance level length for each strain and were oriented perpendicular
a = 0.05. This justifies the use of the log-normal to the long cell axis.
distribution. We now consider the two limiting cases:
The results for the mean visible contour lengths for the 1. short pili with a full contour length smaller than the
different strains are plotted in Fig. 3c and are listed in upper perimeter B of the cell body (Fig. 4a); and
Table 1. The differences between the mean visible contour 2. long pili with a full contour length larger than the
lengths for the five strains were statistically significant as upper perimeter B of the cell body (Fig. 4b).
confirmed by Student’s t tests applied to the logarithmi-
cally transformed data (Limpert and Stahel 2011; p val- For the short pili, some of the pili reach the substrate and
ues \ 0.01), except for the difference between DSM43989 become visible. On average their visible contour length is
and ISS4746 (p value = 0.025) and the difference between half their full length: L! ! L!full =2. For long pili all pili reach
DSM44123 and ISS3319 (p value = 0.083). the substrate and, on average, the visible contour length

123
87
Publications
Eur Biophys J

a c
1.0 = 28

(µm)
relative frequency
1.5

mean visible contour length


0.5 = 28
ISS3319 1.0 = 29
= 256 nm
= 1.60
0.5 = 29
0.0 = 27
0.0 0.2 0.4 0.6 0.8

0.0

19

46

49
b

89

3
12

33

47

47
39
1.0

44

ISS

ISS

ISS
M4

M
relative frequency

DS

DS
d
0.5
ISS4746 AFM tip cell body
= 940 nm
= 1.31 pilus
full
0.0
0.0 0.5 1.0 1.5 2.0
visible contour length (µm)

Fig. 3 Contour length analysis. a, b Cumulative histograms of shown in Suppl. Fig. S1. c Strain-specific visible contour lengths
measured visible contour lengths of pili a from strain ISS3319 and (geometric mean, error bars represent the 95 % confidence intervals).
b from strain ISS4746 (black curves) with respective geometric means Statistical significance of the differences (Student’s t test):
L! and geometric standard deviations rGSD. The corresponding log- p value \ 0.01 for all pairs except for the pair DSM43989—
normal cumulative distribution functions (Eq. 7) (solid gray curves) ISS4746 (p value = 0.025) and the pair DSM44123—ISS3319
match the experimental data well. Also shown are the corresponding (p value = 0.083). d Schematic diagram illustrating the difference
probability density functions (Eq. 6) (not normalized) (dashed gray between the visible (L) and the full (Lfull) contour length of a pilus.
curves). The cumulative histograms for the remaining strains are B denotes the upper perimeter of the cell body

Table 1 Measured mean visible contour length (geometric mean L),! mean full contour length from the correction model, and persistence length
from method 2 (segment bending angles) for the pili from five different C. diphtheriae strains
Strain Mean visible contour Mean full contour Persistence length
length (nm) length (nm) (nm) (method 2)

DSM43989 780 [680, 880] 1,230 [1,070, 1,400] 220 [190, 250]
DSM44123 320 [270, 380] 640 [530, 760] 280 [230, 340]
ISS3319 260 [210, 310] 510 [430, 620] 280 [220, 350]
ISS4746 940 [840, 1,040] 1,390 [1,250, 1,540] 240 [210, 270]
ISS4749 1,590 [1,400, 1,790] 2,040 [1,800, 2,300] 260 [230, 280]
The values in parentheses indicate the respective 95 % confidence interval

differs from the full contour length by half of the upper Persistence length
perimeter of the cell body: L!full ! L! " B=2. In summary,
the mean full contour length can be approximated from the To obtain the persistence length P of the pili we used two
mean visible contour length by: different methods of analysis. The first method is based on
! the end-to-end distance (‘‘method 1’’) and has previously
! L\B=2
L; !
L!full " L! þ : ð8Þ been applied to biopolymers such as DNA (Rivetti et al.
B=2; L! $ B=2
1996; Mantelli et al. 2011), flagella (Trachtenberg and
Here we used B = 1 lm as a typical upper perimeter of the Hammel 1992), F-pili (Silverman and Clarke 2010), and
cell body for all strains analyzed. The results of the cor- filamentous bacteriophages (Wang et al. 2006). Using this
rection based on Eq. (8) are shown in Fig. 4c and Table 1. method we plotted, for each strain, the mean squared

123
88
Publications
Eur Biophys J

short pili ( Lfull < B ) N = 28

mean full contour length Lfull (µm)


a c
cell surface
2.0
pilus
N = 28
N = 29
L
L full 1.0
N = 29
N = 27
B

long pili ( Lfull > B )


b 0.0

89

23

19

46

49
39

41

33

47

47
M4

M4

ISS

ISS

ISS
DS

DS
Fig. 4 One-dimensional model for estimating the mean full contour upper perimeter B of the cell body. All pili reach the substrate and on
length of the pili. a Short pili with a full contour length L!full smaller average the visible contour length differs from the full contour length
than the upper perimeter B of the cell body. Only some of the pili by half of the upper perimeter B: L!full ! L! $ B=2. c Strain-specific
reach the substrate and become visible in the AFM images. On mean full contour lengths (geometric mean, error bars indicate the
average their visible contour length is half their full contour length: 95 % confidence intervals)
L! $ L!full =2. b Long pili with a full contour length L!full larger than the

visible end-to-end distance R2 of each pilus vs. its visible the bending angle, cos(#), were considered. However, as
contour length L. This is shown in Fig. 5a for strain can be seen from Eq. (2), the persistence length can also be
DSM44123 as an example; the measured values of R2 and directly obtained from the standard deviation of the bend-
L are labeled with different markers for a preparation on a ing angles, r#. This has particular benefits compared with
glass (‘‘?’’) or mica (‘‘9’’) as substrate. We then fitted method 1:
Eq. (4) to these data (Fig. 5a, solid line) to obtain the
persistence length P of the pili. For strain DSM44123, – first, because much more information is used, the sta-
using method 1, we obtained P = 520 ± 160 nm (fit tistical precision is much higher for the same number of
coefficient ±95 % confidence interval). The persistence polymers; and
lengths of the other strains were obtained similarly (for the – second, the statistical uncertainty follows directly from
other strains see Suppl. Fig. S2). The resulting persistence the statistics of the standard deviation.
lengths based on the end-to-end distance range from
The 95 % confidence interval of r2# after measuring Ns
280 to 520 nm and are plotted in Fig. 5b.
polymer segments is given by:
However, two problems arise from this method when
" #
applied to polymers of different contour length: first, long Ns ! 1 2 Ns ! 1 2
polymers have an overly high effect on the fit and hence on the s ; s : ð9Þ
v2Ns !1;0:975 # v2Ns !1;0:025 #
resulting persistence length. Second, the width of the confi-
dence interval obtained is only a lower estimate, because the Therefore, the 95 % confidence interval of the resulting
statistical fluctuation of R2 is not included. Furthermore, persistence length P is:
because only one value per polymer (the end-to-end distance) " #
is measured the statistical sample is relatively small. As can be DL v2Ns !1;0:025 DL v2Ns !1;0:975
; : ð10Þ
seen in Fig. 5b the differences obtained between strain-spe- s2# Ns ! 1 s2# Ns ! 1
cific persistence lengths are high (approx. a factor of two),
nevertheless they are not statistically significant (Student’s t Here, s2# is the measured sample variance of the bending
tests give p values [ 0.01 for all pairs). angle, v2Ns !1; 0:025 and v2Ns !1; 0:975 denote the 2.5th and 97.5th
To improve the statistical precision we therefore applied percentiles of the chi-squared distribution with Ns - 1
a second method that is based on the bending angle of short degrees of freedom.
polymer segments (‘‘method 2’’). This was possible Figure 5c shows the histogram of bending angles
because the paths of the pili were traced digitally. Similar using pili segments of length DL = 50 nm for strain
methods have been applied to DNA (Bednar et al. 1995; DSM44123 as an example (for the other strains see
Abels et al. 2005), for which the statistics of the cosine of Suppl. Fig. S3). The mean bending angle is zero within

123
89
Publications
Eur Biophys J

Fig. 5 Two methods for


persistence length analysis, both Method 1: Method 2:
shown for strain DSM44123 as end-to-end distance R segment bending angles
example (for the other strains
see Suppl. Figs. S2 and S3).
a Method 1: Squared visible
end-to-end distance R2 versus L
visible contour length L. The a c
persistence length P (±95 % 0.3
confidence interval) is DSM44123 DSM44123
determined by fitting Eq. (4) P = (520±160) nm 100 P = 280 [230,340] nm
(solid line). The dotted line 0.2

frequency
R 2 (µm2)

indicates the limit of infinitely


stiff pili, corresponding to
50
R2 = L2. Different markers 0.1
represent preparations on glass
(plus) or mica (cross) as
substrates. b Strain-specific
0.0 0
persistence lengths based on R2
0.0 0.1 0.2 0.3 0.4 0.5 -2.0 -1.0 0.0 1.0 2.0
versus L data. c Method 2:
Histogram of bending angles visible contour length L (µm) bending angle (rad)
using segments of length
b d
DL = 50 nm. The standard
deviation of the bending angles 0.8 N = 28 0.8
r# = 0.42 [0.35, N = 29
0.52] corresponds to a N = 28
0.6 0.6
persistence length of
P = 280 [230, 340] nm.
P (µm)

N = 29 N = 27
The gray trace shows the 0.4 0.4 Ns = 210 Ns = 137
Ns = 464 Ns = 531 Ns = 913
corresponding Gaussian
distribution from Eq. (2). 0.2 0.2
d Strain-specific persistence
lengths based on the segment
bending angles. Error bars 0.0 0.0
indicate the 95 % confidence
89

23

19

46

49
89

23

19

46

49

39

41

33

47

47
39

41

33

47

47

intervals
M4

M4

ISS

ISS

ISS
M4

M4

ISS

ISS

ISS

DS

DS
DS

DS

the measurement precision, h#i = -0.03 [-0.09, 0.03], Discussion


and the standard deviation of the bending angles is
r# = 0.42 [0.35, 0.52]. The values in brackets indicate Pili are crucial for adhesion to host cells, and for different
the corresponding 95 % confidence intervals based on C. diphtheriae strains this process varies substantially (Ott
Eq. (9). Furthermore, the distribution is Gaussian, et al. 2010). Surprisingly, there seems to be no direct
as indicated by the close match with the correspond- correlation between adhesion efficiency and pili number or
ing distribution from Eq. (2) (gray trace) (for the other length. Whereas ISS4746 and ISS4749 have long pili
strains see Suppl. Fig. S3). The standard deviation (Fig. 3c) and strong adhesion, DSM43989 has long pili but
of the bending angles corresponds to a persistence adheres much less strongly, and DSM44123 and ISS3319
length of P = DL/r2# = 280 [230, 340] nm. The persistence have short pili but moderate adhesion.
lengths based on the bending angle statistics (method 2) are One important question is whether features of the bac-
summarized in Fig. 5d and are listed in Table 1. It can be terial phenotype, for example pili contour length, can be
seen that the strain-specific persistence lengths from related to genetic information. Pili subunits SpaA and
method 2 are within a narrow range of 220–280 nm and SpaH have been identified as the main shaft proteins of
that the confidence intervals are much smaller than those C. diphtheriae, and their existence or overproduction have
for method 1. Nevertheless, the differences between the been found to increase the pili contour length
persistence lengths from the different strains are still not (Swierczynski and Ton-That 2006; Mandlik et al. 2007;
significant (Student’s t tests give p values [ 0.05 for all Guttilla et al. 2009). Strains ISS4746 and ISS4749 have a
pairs). When we applied method 2 to the data from the large pili contour length (Fig. 3c) and high expression of
preparations on glass and mica separately, no significant SpaA and SpaH (Ott et al. 2010). On the other hand, strains
effect of the substrate on the measured persistence length DSM44123 and ISS3319, which have short pili, and
was detected. express no SpaH and comparatively little SpaA. Therefore,

123
90
Publications
Eur Biophys J

increased expression of SpaA and SpaH in combination or analysis is the assumption that the pili thermally equilibrate
alone might result in increased pili length. in the substrate plane (Eq. 4) before they are immobilized
To analyze the mechanical properties of the pili, we by the drying process (Wang et al. 2006; Silverman and
applied the WLC model to determine the persistence Clarke 2010). This is in contrast with another possible
length. We used two different methods of analysis, based scenario in which the pili are ‘‘kinetically trapped’’ on the
on the end-to-end distance (method 1, Fig. 5b) and on the substrate, i.e. are immobilized immediately upon initial
bending angles of short segments (method 2, Fig. 5d). In contact with the substrate. For polymers without a charge
comparison, two differences can be seen between the or with a low charge, for example C. diphtheriae pili,
persistence lengths when using method 1 and when using however, this scenario is less likely (Bezanilla et al. 1995).
method 2. From the AFM images we estimated the diameter of the
– First, the confidence intervals from method 2 are pili as 1–2 nm. This is smaller than the value obtained by
smaller than those from method 1. electron microscopy (2.5–3.5 nm; Yanagawa and Honda
– Second, on average, the persistence lengths from 1976). A possible explanation of this discrepancy is that
method 2 seem to be smaller. AFM might underestimate the diameter because of the non-
zero imaging force that indents soft samples (Jiao and
There are three main reasons for these differences.
Schäffer 2004). Electron microscopy, on the other hand,
might overestimate the diameter because of the conductive
– First, in method 1 only one value per pili is used. The
coating required before imaging. From the crystal structure
statistical sample of approximately 30 pili per strain is
of the pili subunit SpaA (Kang et al. 2009), we read off a
therefore much smaller than for method 2 in which
diameter of 2.5 nm. In the following discussion, we assume
approximately 100–900 segments per strain are
a diameter of 2.5 nm to estimate the stiffness of the pili.
analyzed.
In combination with the persistence length, the pili
– Second, because the persistence length is positive by
diameter can be used to estimate the stiffness (Young’s
definition, noise will, on average, increase the resulting
modulus) of the pili by rearranging Eq. (3) and assuming
persistence length.
the pili are homogeneous, isotropic cylindrical rods of
– Third, method 1 is more sensitive to directional bias.
diameter 2.5 nm. This gives a stiffness of 400–600 MPa
These reasons imply that method 2 is both more accurate for the analyzed C. diphtheriae strains. In general, the
and more precise. Therefore, we only consider the results from stiffness of C. diphtheriae pili is greater than that of DNA,
method 2 (Table 1) in the following discussion. which is approximately 100–300 MPa, depending on base
By estimating statistical error we found that differences in composition (Hogan et al. 1983) and on the composition
persistence length between the strains were not significant (Wang et al. 1997) and ionic strength (Baumann et al.
(Student’s t test p values [ 0.05). In general, the persistence 1997) of the surrounding buffer solution. But it is smaller
length of C. diphtheriae pili seems to be significantly lower than that of some other proteins, for example microtubules
than the persistence lengths of other bacterial polymers, for (E & 1.2 GPa; Gittes et al. 1993) or actin filaments
example type IV pili (P & 5 lm; Skerker and Berg 2001), (E & 2 GPa; Kojima et al. 1994; Huxley et al. 1994). It is
F-pili (P & 5 lm; Silverman and Clarke 2010), or bacterial also smaller than that of electrically conductive microbial
flagella (P & 20–40 lm; Trachtenberg and Hammel 1992). nanowire pili (E & 1 GPa; Leung et al. 2011), which were
C. diphtheriae pili are also much more flexible than actin discovered recently (Reguera et al. 2005).
(P & 20 lm) or microtubules (P & 5 mm; Gittes et al. Our results also enable us to draw conclusions about the
1993), but they are stiff in comparison with DNA mechanical properties of the pili subunits. The pili of
(P & 50 nm; Taylor and Hagerman 1990; Rivetti et al. strains ISS4746 und ISS4749 consist of SpaA and SpaH
1996). Interestingly, the persistence length of filamentous subunits whereas the pili of the other strains in this study
bacteriophages (P & 1 lm) is similar to that of C. diph- consist of SpaH only (Ott et al. 2010). Because the pili
theriae pili (Wang et al. 2006). persistence lengths of all investigated strains were similar
One limitation possibly affecting our measurements and (within the precision of our measurement), we can con-
those previously obtained by electron microscopy is the use clude that SpaA and SpaH must contribute approximately
of dried samples. It is known for electrically charged equally to the persistence length.
polymers, for example DNA, that changing local electro-
static interactions have an effect on persistence length
(Baumann et al. 1997; Mantelli et al. 2011). C. diphtheriae Conclusion
pili, however, are probably uncharged (Kang et al. 2009),
making the persistence length of the pili insensitive to We have performed comprehensive AFM analysis of pili
electrostatic conditions. Another possible limitation of our contour length and persistence length for different

123
91
Publications
Eur Biophys J

C. diphtheriae strains. Measurements revealed significant Corynebacterium diphtheriae surface proteins as adhesins to
strain-specific differences among mean visible contour human erythrocytes. FEMS Microbiol Lett 197(2):235–239. doi:
10.1111/j.1574-6968.2001.tb10609.x
length. Because each pilus lay partially on the cell body, Dorst L, Smeulders AWM (1987) Length estimators for digitized
on which it could not be visualized, its visible contour contours. Comput Vis Graph Image Process 40(3):311–333
length underestimated its full contour length. By use of a Dupres V, Alsteens D, Pauwels K, Dufrêne YF (2009) In vivo
simple model we estimated mean full contour lengths for imaging of S-layer nanoarrays on Corynebacterium glutamicum.
Langmuir 25(17):9653–9655. doi:10.1021/la902238q
the different strains as 510–2,060 nm, with significant Fälker S, Nelson AL, Morfeldt E, Jonas K, Hultenby K, Ries J,
strain-specific variation. We also measured the persistence Melefors Ö, Normark S, Henriques-Normark B (2008) Sortase-
lengths of the pili—as far as we are aware the first such mediated assembly and surface topology of adhesive pneumococcal
analysis for type III pili. We used two different methods pili. Mol Microbiol 70(3):595–607. doi:10.1111/j.1365-2958.2008.
06396.x
of analysis—one based on the end-to-end distance and Gaspar AH, Ton-That H (2006) Assembly of distinct pilus structures
one based on the bending angles of short segments. The on the surface of Corynebacterium diphtheriae. J Bacteriol
bending angle analysis proved to be more precise. 188(4):1526–1533. doi:10.1128/jb.188.4.1526-1533.2006
The persistence lengths did not vary significantly among Gittes F, Mickey B, Nettleton J, Howard J (1993) Flexural rigidity of
microtubules and actin filaments measured from thermal fluctu-
the strains investigated, and were in the range ations in shape. J Cell Biol 120(4):923–934. doi:10.1083/jcb.
220–280 nm, which is relatively low compared with other 120.4.923
bacterial polymers, for example type IV pili, F-pili, or Guttilla IK, Gaspar AH, Swierczynski A, Swaminathan A, Dwivedi P,
flagella. Das A, Ton-That H (2009) Acyl enzyme intermediates in
sortase-catalyzed pilus morphogenesis in gram-positive bacteria.
J Bacteriol 191(18):5603–5612. doi:10.1128/jb.00627-09
Acknowledgments ISS strains were kindly provided by Hirata R Jr, Souza SMS, Rocha de Souza CM, Andrade AF,
C. v. Hunolstein (Istituto Superiore di Sanita, Rome). LO and AB are Monteiro-Leal LH, Formiga LCD, Mattos-Guaraldi AL (2004)
grateful for financial support of the Deutsche Forschungsgemeinschaft Patterns of adherence to HEp-2 cells and actin polymerisation by
(SFB796, B5). We thank Yves Muller (Biotechnology Division, toxigenic Corynebacterium diphtheriae strains. Microb Pathog
University of Erlangen-Nuremberg) for assistance with the Protein Data 36(3):125–130. doi:10.1016/j.micpath.2003.10.002
Bank. We thank Asylum Research and Atomic Force F&E for support. Hirata R Jr, Napoleao F, Monteiro-Leal LH, Andrade AFB, Nagao
PE, Formiga LCD, Fonseca LS, Mattos-Guaraldi AL (2002)
Intracellular viability of toxigenic Corynebacterium diphtheriae
strains in Hep-2 cells. FEMS Microbiol Lett 215(1):115–119.
References doi:10.1111/j.1574-6968.2002.tb11379.x
Hogan M, LeGrange J, Austin B (1983) Dependence of DNA helix
Abels JA, Moreno-Herrero F, van der Heijden T, Dekker C, Dekker flexibility on base composition. Nature 304(5928):752–754. doi:
NH (2005) Single-molecule measurements of the persistence 10.1038/304752a0
length of double-stranded RNA. Biophys J 88(4):2737–2744. Huxley HE, Stewart A, Sosa H, Irving T (1994) X-ray diffraction
doi:10.1529/biophysj.104.052811 measurements of the extensibility of actin and myosin filaments in
Anselmetti D, Hansmeier N, Kalinowski J, Martini J, Merkle T, contracting muscle. Biophys J 67(6):2411–2421. doi:10.1016/s0006-
Palmisano R, Ros R, Schmied K, Sischka A, Toensing K (2007) 3495(94)80728-3
Analysis of subcellular surface structure, function and dynamics. Jiao Y, Schäffer TE (2004) Accurate height and volume measure-
Anal Bioanal Chem 387(1):83–89. doi:10.1007/s00216-006- ments on soft samples with the atomic force microscope.
0789-3 Langmuir 20(23):10038–10045. doi:10.1021/la048650u
Arce FT, Carlson R, Monds J, Veeh R, Hu FZ, Stewart PS, Lal R, Kang HJ, Paterson NG, Gaspar AH, Ton-That H, Baker EN (2009)
Ehrlich GD, Avci R (2009) Nanoscale structural and mechanical The Corynebacterium diphtheriae shaft pilin SpaA is built of
properties of nontypeable Haemophilus influenzae biofilms. tandem Ig-like modules with stabilizing isopeptide and disulfide
J Bacteriol 191(8):2512–2520. doi:10.1128/jb.01596-08 bonds. Proc Natl Acad Sci USA 106(40):16967–16971. doi:
Baumann CG, Smith SB, Bloomfield VA, Bustamante C (1997) Ionic 10.1073/pnas.0906826106
effects on the elasticity of single DNA molecules. Proc Natl Koch AL (1966) The logarithm in biology 1. Mechanisms generating
Acad Sci USA 94(12):6185–6190 the log-normal distribution exactly. J Theoret Biol
Bednar J, Furrer P, Katritch V, Stasiak A, Dubochet J, Stasiak A 12(2):276–290. doi:10.1016/0022-5193(66)90119-6
(1995) Determination of DNA persistence length by cryo- Kojima H, Ishijima A, Yanagida T (1994) Direct measurement of
electron microscopy. Separation of the static and dynamic stiffness of single actin filaments with and without tropomyosin
contributions to the apparent persistence length of DNA. J Mol by in vitro nanomanipulation. Proc Natl Acad Sci USA 91(26):
Biol 254(4):579–594. doi:10.1006/jmbi.1995.0640 12962–12966
Bertuccini L, Baldassarri L, von Hunolstein C (2004) Internalization Landau LD, Lifshits EM, Pitaevskiı̌ LP (1980) Statistical physics.
of non-toxigenic Corynebacterium diphtheriae by cultured Course of theoretical physics, 3rd edn. Butterworth-Heinemann,
human respiratory epithelial cells. Microb Pathog 37(3): Oxford
111–118. doi:10.1016/j.micpath.2004.06.002 Landau LD, Lifshitz EM, Kosevich AM, Pitaevskiı̌ LP (1986) Theory
Bezanilla M, Manne S, Laney DE, Lyubchenko YL, Hansma HG of elasticity. Course of theoretical physics, 3rd edn. Butterworth-
(1995) Adsorption of DNA to mica, silylated mica, and minerals: Heinemann, Oxford
characterization by atomic force microscopy. Langmuir Leung KM, Wanger G, Guo Q, Gorby Y, Southam G, Lau WM, Yang
11(2):655–659. doi:10.1021/la00002a050 J (2011) Bacterial nanowires: conductive as silicon, soft as
Colombo AV, Hirata R Jr, de Souza CM, Monteiro-Leal LH, Previato polymer. Soft Matter 7(14):6617–6621. doi:10.1039/c1sm0
JO, Formiga LC, Andrade AF, Mattos-Guaraldi AL (2001) 5611e

123
92
Publications
Eur Biophys J

Limpert E, Stahel WA (2011) Problems with using the normal Rogers EA, Das A, Ton-That H (2011) Adhesion by pathogenic
distribution—and ways to improve quality and efficiency of data corynebacteria. In: Linke D, Goldman A (eds) Bacterial
analysis. PLoS ONE 6(7):e21403. doi:10.1371/journal.pone. adhesion, vol 715. Advances in experimental medicine and
0021403 biology. Springer, the Netherlands, pp 91–103. doi:10.1007/978-
Limpert E, Stahel WA, Abbt M (2001) Log-normal distributions 94-007-0940-9_6
across the sciences: keys and clues. Bioscience 51(5):341–352. Silverman PM, Clarke MB (2010) New insights into F-pilus structure,
doi:10.1641/0006-3568(2001)051[0341:LNDATS]2.0.CO;2 dynamics, and function. Integr Biol 2(1):25–31. doi:10.1039/b917
Mandlik A, Swierczynski A, Das A, Ton-That H (2007) Corynebac- 761b
terium diphtheriae employs specific minor pilins to target human Skerker JM, Berg HC (2001) Direct observation of extension and
pharyngeal epithelial cells. Mol Microbiol 64(1):111–124. doi: retraction of type IV pili. Proc Natl Acad Sci USA 98(12):
10.1111/j.1365-2958.2007.05630.x 6901–6904. doi:10.1073/pnas.121171698
Mandlik A, Das A, Ton-That H (2008) The molecular switch that Swierczynski A, Ton-That H (2006) Type III pilus of corynebacteria:
activates the cell wall anchoring step of pilus assembly in gram- pilus length is determined by the level of its major pilin subunit.
positive bacteria. Proc Natl Acad Sci USA 105(37):14147– J Bacteriol 188(17):6318–6325. doi:10.1128/jb.00606-06
14152. doi:10.1073/pnas.0806350105 Taylor WH, Hagerman PJ (1990) Application of the method of phage T4
Mantelli S, Muller P, Harlepp S, Maaloum M (2011) Conformational DNA ligase-catalyzed ring-closure to the study of DNA structure:
analysis and estimation of the persistence length of DNA using II. NaCl-dependence of DNA flexibility and helical repeat. J Mol
atomic force microscopy in solution. Soft Matter 7(7):3412– Biol 212(2):363–376. doi:10.1016/0022-2836(90)90131-5
3416. doi:10.1039/c0sm01160f Touhami A, Jericho MH, Boyd JM, Beveridge TJ (2006) Nanoscale
Miller E, Garcia T, Hultgren S, Oberhauser AF (2006) The characterization and determination of adhesion forces of Pseu-
mechanical properties of E. coli type 1 pili measured by atomic domonas aeruginosa pili by using atomic force microscopy.
force microscopy techniques. Biophys J 91(10):3848–3856. doi: J Bacteriol 188(2):370–377. doi:10.1128/jb.188.2.370-377.2006
10.1529/biophysj.106.088989 Trachtenberg S, Hammel I (1992) The rigidity of bacterial flagellar
Nagashima H, Asakura S (1980) Dark-field light microscopic study of filaments and its relation to filament polymorphism. J Struct Biol
the flexibility of F-actin complexes. J Mol Biol 136(2):169–182 109(1):18–27. doi:10.1016/1047-8477(92)90063-g
Ott L, Höller M, Rheinlaender J, Schäffer TE, Hensel M, Burkovski A Wang MD, Yin H, Landick R, Gelles J, Block SM (1997) Stretching
(2010) Strain-specific differences in pili formation and the DNA with optical tweezers. Biophys J 72(3):1335–1346. doi:
interaction of Corynebacterium diphtheriae with host cells. 10.1016/s0006-3495(97)78780-0
BMC Microbiol 10:257. doi:10.1186/1471-2180-10-257 Wang YA, Yu X, Overman S, Tsuboi M, Thomas GJ Jr, Egelman EH
Reguera G, McCarthy KD, Mehta T, Nicoll JS, Tuominen MT, Lovley (2006) The structure of a filamentous bacteriophage. J Mol Biol
DR (2005) Extracellular electron transfer via microbial nanowires. 361(2):209–215. doi:10.1016/j.jmb.2006.06.027
Nature 435(7045):1098–1101. doi:10.1038/nature03661 Wright CJ, Shah MK, Powell LC, Armstrong I (2010) Application of
Rivetti C, Codeluppi S (2000) Accurate length determination of DNA AFM from microbial cell to biofilm. Scanning 32(3):134–149.
molecules visualized by atomic force microscopy: evidence for a doi:10.1002/sca.20193
partial B- to A-form transition on mica. Ultramicroscopy Yanagawa R, Honda E (1976) Presence of pili in species of human
87(1–2):55–66. doi:10.1016/s0304-3991(00)00064-4 and animal parasites and pathogens of the genus corynebacte-
Rivetti C, Guthold M, Bustamante C (1996) Scanning force rium. Infect Immun 13(4):1293–1295
microscopy of DNA deposited onto mica: Equilibration versus Yanagida T, Nakase M, Nishiyama K, Oosawa F (1984) Direct
kinetic trapping studied by statistical polymer chain analysis. observation of motion of single F-actin filaments in the presence
J Mol Biol 264(5):919–932. doi:10.1006/jmbi.1996.0687 of myosin. Nature 307(5946):58–60. doi:10.1038/307058a0

123
93
Publications

3.7 Induction of the NFĸB signal transduction pathway in response


to Corynebacterium diphtheriae infection (Ott et al, 2012b)

94
Publications

Induction of the NF-B signal transduction pathway in response to Corynebacterium

diphtheriae infection

Running title: NF-B induction by C. diphtheriae

Lisa Ott1), Brigitte Scholz2), Martina Höller1), Kristin Hasselt3), Armin Ensser2) and Andreas

Burkovski1)*

1)
Friedrich-Alexander-Universität Erlangen-Nürnberg, Lehrstuhl für Mikrobiologie, Staudtstr.

5, 91058 Erlangen, Germany


2)
Klinische und Molekulare Virologie, Virologisches Institut des Universitätsklinikums

Erlangen, Schlossgarten 4, 91054, Erlangen, Germany


3)
BioCer Entwicklungs-GmbH, Ludwig-Thoma-Str. 36c, 95447 Bayreuth, Germany

* Corresponding author: Andreas Burkovski, Lehrstuhl für Mikrobiologie, Friedrich-Alexander-

Universität Erlangen-Nürnberg, Staudtstr. 5, 91058 Erlangen, Germany. Phone: +49 9131 85

28086. Fax: +49 9131 85 28082. E-mail: aburkov@biologie.uni-erlangen.de

Keywords: host-pathogen interaction, internalization, invasion, MAMP, PAMP

95
Publications

Summary (Abstract)

Corynebacterium diphtheriae, the causative agent of diphtheria, has been thoroughly studied

with respect to toxin production and pili formation, while knowledge on host responses to C.

diphtheriae infection is limited. In this study, we studied adhesion and invasion of epithelial

cells by different C. diphtheriae isolates. When NF-B reporter cell lines were used to monitor

the effect of C. diphtheriae infection on human cells, strain-specific differences were

observed. While adhesion to host cells had no effect, a correlation of invasion rate with NF-

B induction was found, which indicates that internalization of bacteria is crucial for NF-B

induction. Immune-fluorescence microscopy experiments used to support the reporter

assays showed that translocation of p65, as a hallmark of NF-B induction, was only

observed in association with cell invasion by C. diphtheriae. Our data indicate that the

response of epithelial cells to C. diphtheriae infection is determined by internalization of

bacteria and that invasion into these cells is an active process; tetracycline-treated C.

diphtheriae were still able to attach to host cells, but lost their ability to invade into the

cytoplasm. Recognition of pathogen-associated molecular patterns such as pili subunits by

membrane-bound receptors facing the outside of the cell is not sufficient for NF-B induction.

96
Publications

Introduction

Corynebacterium diphtheriae is the etiologic agent of diphtheria, a toxaemic infection of

respiratory tract or skin. While diphtheria has almost been eradicated by vaccination programs

in many industrialized countries (Galazka, 2000; Hadfield et al., 2000; von Hunolstein et al.,

2003), the disease is still a global medical challenge and especially dangerous in many

resource poor countries. Furthermore, it has become evident that C. diphtheriae infections can

cause other manifestations besides diphtheria, such as bacteraemia, endocarditis, pneumonia,

osteomyelitis, septic arthritis and spleenic abscesses (Puliti et al., 2006 and references

therein; Hirata et al., 2008; Farfour et al., 2012). These systemic infections indicate that this

pathogen is not only able to attach to host epithelial cells, but must also be able to gain access

to deeper tissues by unknown portals of entry and to persist in these tissues. A possible

indication with regard to mechanisms of persistence of C. diphtheriae within the human body

came from comparative investigations of adherence and invasion of toxigenic and non-

toxigenic strains. Using a combination of gentamicin protection assays and thin-section

electron microscopy, Hirata and co-workers (Hirata et al., 2002) showed that toxigenic C.

diphtheriae were not only able to adhere to laryngeal HEp-2 cells, but also enter these cells

and survive after internalization. Similar observations were made for non-toxigenic strains,

showing that pharyngeal Detroit562 cells can be also invaded by C. diphtheriae (Bertuccini et

al., 2004; Ott et al., 2010a; Ott et al., 2010b).

With exception of the well documented effect of the diphtheria toxin on cellular

metabolism, responses of the human host to C. diphtheriae infection are almost not

investigated. Only a limited number of virulence factors important for host recognition and

attachment are known (for example see (Mandlik et al., 2007; Mattos-Guaraldi et al., 2000; Ott

et al., 2010a; Ott et al., 2010b). Mice were infected by intravenous injection of bacteria in order

to establish an experimental model of C. diphtheriae infection and development of septic

arthritis. Besides mortality and arthritogenic potential of different strains, cytokine production

was determined in this study (Puliti et al., 2006). Strain-specific differences were observed for

the production of interleukins and interferons, i.e. IL-1, IL-6, IFN- and TNF. On the one

97
Publications

hand, production of these cytokines is known to be involved in NF-B signalling (Perkins,

2007; Tato & Hunter, 2002), a pathway which is frequently activated upon invasion of

pathogens. On the other hand it is known that pathogens have different strategies to interfere

with the NF-B pathway and some are even able to inhibit NF-B activation (Tato & Hunter,

2002). Since data were lacking for C. diphtheriae, we started an approach to investigate NF-

B induction in epithelial cells in response to C. diphtheriae infection, which is presented here.

Materials and methods

Bacterial strains and growth

Strains used in this study are listed in Table 1. C. diphtheriae was grown in Heart Infusion (HI)

broth at 37°C, Escherichia coli DH5aMCR were grown in Luria Broth (LB) (Sambrook et al.,

1989) at 37°C. If appropriate, kanamycin was added (30 μg ml-1 for E. coli; 50 μg ml-1 for C.

diphtheriae).

Cell cultures

Detroit562 cells were cultured in DMEM (Dulbecco’s  modified  Eagle’s  medium), high glucose

with L-glutamine and sodium pyruvate (PAA Laboratories, Austria), supplemented with 120 µg

ml-1 penicillin, 120 µg ml-1 streptomycin and 10 % heat inactivated fetal calf serum (FCS) in a

CO2 incubator. Cells were passaged at a ratio of 1:20 twice per week. HeLa cells were

cultured in DMEM, high glucose with L-glutamine (PAA Laboratories, Austria) supplemented

with 100 µg ml-1 gentamicin and 12 µg ml-1 ciprofloxacin and 10 % heat inactivated FCS in a

CO2 incubator. Cells were passaged at a ratio of 1:3 twice per week.

Scanning electron microscopy (SEM)

Infected monolayer cultures were first fixed with 0.1 % glutaraldehyde, 2 % paraformaldehyde,

5 % sucrose in 0.2 M sodium cacodylate solution for one hour at 37°C. Subsequently, samples

were fixed with 0.3 % glutaraldehyde, 3 % paraformaladehyde in 0.2 M sodium cacodylate

98
Publications

solution for 1 h at 37°C. Samples were subsequently dehydrated using a graded series of

acetone, critical point dried and gold sputtered and examined using a FEI Quanta 200

scanning electron microscope.

Adhesion assays

Detroit562 and HeLa cells were seeded in 24 well plates (Nunc, Roskilde, Denmark) at a

density of 1 x 105 and 5 x 104 cells per well, respectively, 48 h prior to infection. Bacteria were

inoculated to an OD600 of 0.1 from overnight cultures and grown in HI broth to an OD600 of 0.4

to 0.6. Subsequently, the bacteria were harvested by centrifugation and cell density was

adjusted to an OD600 of 0.2. A master mix of the inoculum with a multiplicity of infection (MOI)

of 200 was prepared in DMEM and 500 µl per well were used to infect the cells. The plates

were centrifuged for 5 min at 500 rpm to synchronize infection and subsequently incubated for

90 min. The cells were washed with PBS three times, detached with 500 µl trypsin solution

(0.12 % trypsin, 0.01 % EDTA in PBS) per well (5 min, 37°C, 5 % CO 2, 90 % humidity) and

lysed with 0.025 % Tween 20 for 5 min at 37°C. Serial dilutions were made in pre-chilled 1 x

PBS and plated on Columbia agar with sheep blood (Oxoid, Wesel, Germany) to determine

the number of colony forming units (cfu).

Epithelial cell invasion model

Detroit562 and HeLa cells were seeded in 24 well plates (Nunc, Roskilde, Denmark) at a

density of 1 x 105 and 5 x 104 cells, respectively, per well 48 h prior to infection. Overnight

cultures grown in HI were re-inoculated to an OD600 of 0.1 in fresh medium and grown to an

OD600 of 0.4 to 0.6. An inoculum with a MOI of 200 was prepared in DMEM without antibiotics

and 500 µl per well were used to infect the cells. The plates were centrifuged for 5 min at 500

rpm to synchronize infection and subsequently incubated for 90 min (37°C, 5 % CO2, 90 %

humidity). The cells were washed three times with PBS to remove planctonic and loosely

attached bacteria. Subsequently, the cells were incubated for two hours in DMEM (500 µl per

well), containing 100 µg ml-1 gentamicin to kill remaining extracellular bacteria. After this

99
Publications

incubation, the cell layers were washed three times with PBS, detached by adding 500 µl

trypsin solution (0.12 % trypsin, 0.01 % EDTA in PBS) per well (5 min, 37°C, 5 % CO2, 90 %

humidity), and lysed for 5 min at 37°C with 0.025 % Tween 20 to liberate the intracellular

bacteria. Serial dilutions of the inoculums and the lysates were plated out on Columbia agar

with sheep blood (Oxoid, Wesel, Germany) to determine the number of cfu.

Construction of an NF-B reporter cell line

The lentiviral NF-B reporter construct was generated as following: a fragment including an

eukaryotic elongation factor 1 alpha promoter (EF1)-driven puromycin resistance gene and the

Woodchuck hepatitis virus posttranscriptional regulatory element (WPRE) was excised from

the vector pCDH-CMV-MCS-EF1-Puro  (System  Biosciences)  with  restriction  enzymes  NotI  (5’  

overhang filled in with Klenow DNA Polymerase) and KpnI. This fragment was ligated into the

vector pTRH-3xSIE-Luc (based on pTRH-MCS-Luc, System Biosciences) that has been cut

with   XbaI   (5’   overhang   filled   in   with   Klenow   DNA   Polymerase)   and   KpnI.   The   3X   SIE  

responsive element was then replaced with a synthetic linker containing 4x NFB responsive

elements. Correct insertion was verified restriction digests and by DNA sequencing. Lentiviral

vector stocks were produced in 293T cells (DSMZ ACC 635, Bennett & Pearson, 1993) that

were seeded in 80 cm2 flasks and transfected using Lipofectamine 2000 (Invitrogen) according

to  manufacturer’s  instructions  when  the  cells  were  60  %  confluent.  The  pTRH  reporter  vector  

plasmid  was  cotransfected  together  with  psPAX2  and  pMD2.G  in  a  ratio  of  4:3:1,  in  total  15  μg  

plasmid  with  12.5  μl  Lipofectamine  2000  per  80  cm²  flask.   pMD2G encodes for the vesicular

stomatitis virus G protein (VSV-G protein), a protein which mediates fusion of viral particles

with cellular membranes, ensuring a broad cell tropism. The plasmid psPAX2 encodes for the

HIV gag, pol, tat and rev genes, but is deleted for the env, vpr, vpu and nef genes.

Transfection of the different plasmids into 293T cells led to generation of VSV-G-pseudotyped

replication-defective viral particles that were released into the culture supernatant. The cell

free supernatants were further concentrated by ultracentrifugation and used for infection of T

100
Publications

cells. Detroit562 and HeLa cells were transduced with the reporter lentivirus and selected with

1 µg ml-1 puromycin (Sigma) for 3 weeks, resulting in the corresponding NFB cell lines.

Luciferase activity measurements

Detroit562-NFB and HeLa-NFB cells were seeded in 48 well plates (Greiner bio-one

Cellstar, Frickenhausen, Germany) at a density of 5 x104 cells per well 24 h prior to infection.

Overnight cultures grown in HI were re-inoculated to an OD600 of 0.1 in fresh medium and

grown to an OD600 of 0.4 to 0.6. To test if dead bacteria can activate eukaryotic signalling

pathways, 10 ml of the overnight cultures were incubated for 3.5 h with 8 µg ml-1 tetracycline.

An inoculum with a MOI of 200 was prepared in DMEM and 300 µl per well were used to infect

the cells. In addition to the infected cells, a negative control (uninfected reporter cells) as well

as a positive control (reporter cells induced with 20 ng ml-1 Phorbol 12-myristate 13-acetate in

DMSO (1 mg ml-1), diluted 1:100 in DMEM) and 200 ng ml-1 calcimycin (1 mg ml-1 in DMSO

and diluted 1:100 in DMEM) was carried along. The plates were centrifuged for 5 min at 500

rpm to synchronize infection and subsequently incubated for 3 h (37°C, 5 % CO 2, 90 %

humidity). Afterwards cells were washed and DMEM supplemented with 8 µg ml-1 tetracycline

was added to avoid bacteria overgrowing the cells. Samples were taken after different time

points, cells were lysed with lysis buffer (100 mM potassium phosphate buffer (pH 7.8)

containing 0.1 % (v/v) Triton-X-100) for 20 min on a seesaw under light exclusion.

To determine the luciferase activity 100 µl luciferase buffer (15 mM potassium phosphate

buffer (pH 8.0), 25 mM glycylglycine, 4 mM EGTA, 15 mM MgSO 4, 2 mM ATP, 1 mM DTT, 75

µM D-luciferin) were added to 10 µl cell extract automatically and the luminal-dependent

chemiluminescence was measured using a Luminometer (Orion, Berthold, Pforzheim). The

unit of chemiluminescent activity is the relative light unit and was expressed as relative light

units per second (RLU sec-1). A Bradford protein assay was used to determine protein

concentrations in the cell extract. For this purpose 2 µl of the cell extract were added to 240 ml

deionised water and 60 ml Bradford reagent (Roti®-Quant, Roth, Karlsruhe) (10 min, room

temperature). The absorption at 595 nm was determined using a Spectra Fluor Plus (TECAN,

101
Publications

Crailsheim). A calibration curve was prepared using bovine serum albumin (BSA) as reference

solution. The protein concentration was used to normalize the RLUs (RLU µg-1).

Immuno-fluorescence microscopy

HeLa cells were seeded on round coverslips in 24-well plates and after 24 h infected at a MOI

of 250 with GFP-expressing C. diphtheriae for two hours. Infected or uninfected HeLa cells

were fixed with 1 % paraformaldehyde at room temperature, permeabilized with 0.1 % Triton

X-100 in phosphate buffered saline without calcium and magnesium ions and stained. Primary

rabbit polyclonal anti-NFkB p65 antibody (C-20, sc-372; Santa Cruz Biotechnology) was used

at a dilution 1:250; this was detected with Alexa 647-labelled goat anti-rabbit secondary

antibody diluted 1:500 (Life Technologies). Cells were counterstained with the fixable dye

Pacific blue succimidyl ester (0.4 µg ml-1), visualized on a Leica TCS SP5 confocal microscope

and analyzed with the LAS software suite.

Cell viability assay

C. diphtheriae strains DSM43988 and DSM43989 were inoculated in 200 ml DMEM

(Dulbecco’s   modified   Eagle’s   medium,   PAA;;   high  glucose,   10   %   FCS,   2  mM   glutamine)   and  

cultivated overnight, at 37°C in an orbital shaker. Subsequently, the bacterial suspensions

were centrifuged (4500 x g, 10 min, 4°C) and the supernatants were filtered (syringe filter

Ministart, 0.45 µm pore size, Sartorius, Göttingen, Germany) to clear it from remaining

bacterial cells. 200 ml supernatant were concentrated to a volume of 15 ml by centrifugal filter

devices  (Ultracel  10K  membrane,  Merck  Millipore,  Darmstadt,  Germany)  after  manufacturer’s  

protocol and stored at 4°C. Detroit562 and HeLa cells were seeded in 96-well plates (bio-one

Cellstar, Greiner, Frickenhausen, Germany) at a density of 5 x 103 cells per well directly in 190

µl DMEM containing secreted proteins of DSM43988 and DSM43989, respectively. After 20 h

of incubation at 37°C, 5 % CO2 and 95 % humidity, alamar blue (AbD Serotec, Oxford, UK)

was added (10 % in culture medium) to each well and fluorescence was measured at 560 nm

102
Publications

(excitation wavelength) and 590 nm (emission wavelength) using a microplate reader (Tecan

Infinite M200 Pro) at different time points.

Results

Adhesion of C. diphtheriae strains to epithelial cells

C. diphtheriae strains cannot only be isolated from human hosts but also from various animals,

for example from domestic cats (Hall et al., 2010), cows (Corboz et al., 1996) and horses

(Henricson et al., 2000; Leggett et al., 2010). Furthermore, adhesion of this bacterium to

several human cell lines was reported, e. g. pharyngeal Detroit562 (Bertuccini et al., 2004;

Mandlik et al., 2007; Swaminathan et al., 2007), laryngeal HEp-2 (Bertuccini et al., 2004;

Hirata et al., 2002; Hirata et al., 2008; Hirata et al., 2004; Mandlik et al., 2007), lung A596

(Mandlik et al., 2007), human U-937 macrophages (dos Santos et al., 2010) and erythrocytes

(Colombo et al., 2001), indicating that C. diphtheriae is able to bind to different cells as would

be expected based on the different host organisms and different clinical manifestations, i.e.

diphtheria of upper respiratory tract, cutaneous diphtheria or systemic infections.

Adherence assays with different C. diphtheriae isolates carried out before, showed the

highest adhesion to Detroit562 cells with a rate of about 8 % for strains ISS4746 and ISS4749

(Figure 1A, Ott et al., 2010b). While this cell line is close to the natural situation at least for the

upper respiratory tract diphtheria, HeLa cells are the most widespread model in host-pathogen

interaction studies used for a wide variety of organisms (for recent manuscripts, see Bose et

al., 2011; Corbett et al., 2011; Revez et al., 2011) and were therefore tested in this study. In

fact, C. diphtheriae was able to bind to this epithelial cell line and up to tenfold higher adhesion

rates compared to Detroit562 cells were observed (Ott et al., 2010a; Ott et al., 2010b). The

highest numbers of adherent bacteria to HeLa cells corresponding to adhesion rates of 50 to

70 % were observed for strains ISS3319, ISS4060 and DSM43988. The lowest adhesion with

a rate of only 1.25 ± 0.14 percent was observed for the toxin-producer DSM43989 (Figure 1B).

Interestingly, the ability to adhere to the host cells was not only strain- but also cell line-

specific: strains such as ISS4746 and ISS4749 carrying numerous pili showed always the

103
Publications

higher adhesion rates to Detroit562 cells compared to the pili-free strain ISS4060. In contrast,

ISS4060 showed higher adhesion rates compared to these strains to HeLa cells.

Figure 1. Adhesion of C. diphtheriae wild type strains to epithelial cells.


Adhesion of C. diphtheriae strains to Detroit562 (A) and HeLa cell layers (B). Cell lines were
infected with different C. diphtheriae wild type strains, washed with PBS, detached with trypsin
solution, lysed with Tween 20, and the number of colony forming units (cfus) was determined.
Adhesion is expressed as percentage of the inoculum, showing means and standard
deviations of three independent measurements (biological replicates) with three samples each
(technical replicates). Statistically relevant differences between the strains (based on students
TTEST values below 0.05) are indicated by letters above columns.

The observed adhesion behaviour of the different strains to HeLa cells was validated

qualitatively by scanning electron microscopy (SEM) experiments. All samples of non-toxigenic

strains showed numerous bacteria attached to the HeLa cells (Figure 2A-E and 2G), while in

104
Publications

case of the toxin producer, bacteria were hardly detectable (Figure 2F). Furthermore, in case

of infection with tox+ strain DSM43989 most HeLa cells were characterized by a ball-like shape

instead of the flat, extended form in case of infections with other strains, indicating that the

cells were undergoing apoptosis or necrosis (Figure 2F).

A B

C D

E F

Figure 2. Scanning electron microscopy of C. diphtheriae and HeLa cells.


Interaction of C. diphtheriae strains with HeLa cells viewed by scanning electron microscopy at
90 min post-infection. Infected monolayer cultures were fixed, dehydrated sputtered with gold
and examined using a FEI Quanta 200 scanning electron microscope. (A) ISS3319, (B)
ISS4060, (C) ISS4746, (D) ISS4749, (E) DSM43988, (F) DSM43989 (toxigenic) and (G)
DSM44123.

105
Publications

Internalization of C. diphtheriae in eukaryotic cells

When gentamicin protection assays were carried out to analyze the number of invading

bacteria in Detroit562 cells, previous studies revealed strain-specific differences: for the pili-

free strains ISS4060 and DSM43988 the highest invasion efficiencies were observed (Figure

3A, see also Ott et al., 2010b). Strains ISS4060 and DSM43988 also showed the highest

invasion rates into HeLa cells with 3.97 ± 1.84 and 2.72 ± 1.13 %, followed by ISS3319 and

DSM44123 with invasion rates of 0.9 ± 0.07 % and 1.08 ± 0.35 %, respectively. For strains

ISS4746, ISS4749 and for the toxin-producer DSM43989 only negligible invasion rates were

observed (Figure 3B). The invasion rates obtained with HeLa cells were significantly higher

compared to those obtained with Detroit562 cells reported before (Ott et al., 2010b). A

correlation between adhesion and invasion rate was not apparent.

106
Publications

Figure 3. Invasion of C. diphtheriae wild type strains into eukaryotic cells.


Detroit562 (A) and HeLa cells (B) were infected with different C. diphtheriae wild type strains,
washed, and incubated for two hours with 100 μg ml-1 gentamicin. Subsequently, cells were
washed, detached with trypsin solution, lysed with Tween 20, and the number of intracellular
cfus was determined. Invasion is shown as percentage of the inoculum internalized (means
and standard deviations of three independent biological replicates with three samples each
(technical replicates). Statistically relevant differences between the strains (based on students
TTEST values below 0.05) are indicated by letters above columns.

Analysis of NF-B reporter cell lines

Detroit562-NFB and HeLa-NFB reporter cells were used to study activation after exposure

to equivalent doses of the various C. diphtheriae strains at different time points post infection.

When these reporter cell lines were infected with different isolates of C. diphtheriae, strain-

specific differences in the cellular response were observed eight hours after infection by

luciferase activity measurements (Figure 4). For Detroit562-NFB cells only weak activation of

the NFB pathway was observed during infection with different C. diphtheriae strains (Figure

4A). The highest luciferase signals were detected when cells were infected with strains

ISS3319 and ISS4060. Infection of HeLa- NFB cells resulted in much stronger reporter

activation (Figure 4B). Interestingly, the strength of activation determined in the reporter assay

correlated with the invasion rates shown above (Figure 3, compare also data for Detroit562).

Strains ISS3319, ISS4060, ISS4749, DSM43988 and DSM44123 showed luciferase activity

significantly above background. Strongest light intensities were measured when the reporter

cells were infected with strain ISS4060 or DSM43988, which showed the highest invasion

rates. Luciferase activity was lower in the less invasive strains ISS3319 and DSM44123, while

no induction of luciferase in the NF-B reporter cell lines were observed in case of non-

invasive strains ISS4746 and DSM43989. The only exception observed was strain ISS4749,

which was also not invasive but showed significant luciferase activity for unknown reasons.

The luciferase signal declined below background level when HeLa cells were infected by the

toxigenic strain DSM43989 for 20 hours, indicating that this strain has a detrimental effect on

HeLa cells.

107
Publications

3000 a, d, e, A
g, h
d, f,
2500 g, h
b, c, c b, d, h
c f, h b, c, h b, c, d,
2000
f, g

1500

1000

500

a b c d e f g h
rel. luciferase units

a, b, d,
4500 e, g, h B
4000
a, d, e,
3500 g, h
a, c,
3000
d, g
2500 a, c, d, a, c, d,
f, g f, g
2000 a, b, c, e,
f, g, h b, c, d,
1500 b, c, d,
e, f, h e, f, h
1000

500
a b c d e f g h
0

Strains
Figure 4. Activation of the NF-B pathway in eucaryotic cells after 8 h of infection with
C. diphtheriae wild type strains.
Detroit562-NFkB (A) and HeLa-NFkB cells (B) were infected with different C. diphtheriae wild
type strains (DSM43989 tox+, all others are non-toxigenic) for three hours, washed and
incubated for 5 h with DMEM containing 8 µg ml-1 tetracycline. Black bar: uninfected reporter
cells, grey bars: reporter cells infected with C. diphtheriae, white bars: reporter cells infected
with C. diphtheriae inactivated by tetracycline. Measurements of the luciferase units were
carried out on a Luminometer (Orion, Berthold, Pforzheim), expressed in RLU µg-1. Statistically
relevant differences between the strains (living bacteria, grey bars) (based on students TTEST
values below 0.05) are indicated by letters above columns.

When bacteria were killed by three hours of treatment with 8 µg ml-1 tetracycline

(Figure 4) or by 15 min of UV radiation (data not shown) no NF-B induction was observed.

Interestingly, the correspondingly treated bacteria were still found to be attached to the host

cells, when specimens were investigated for control by SEM (Figure 5), showing that adhesion

is a passive process. In addition, the inactivated tox+ strain DSM43989 was not able to kill the

108
Publications

host cells (Figure 5B) as it was the case during infection with living bacteria (Figure 2F).

Furthermore, tetracycline-treated DSM43989 adhered in higher amounts to HeLa cells (Figure

5B) compared to the active strain (Figure 2F).

A B

Figure 5. Scanning electron microscopy of dead C. diphtheriae and HeLa cells.


Interaction of inactivated C. diphtheriae strains (incubation with 8 µg ml-1 tetracycline for 3.5 h)
with HeLa cells viewed by scanning electron microscopy at 90 min post-infection. (A) ISS4746
(non-toxigenic), (B) DSM43989 (toxigenic).

Investigation of NF-B induction by immuno-fluorescence microscopy

The increase of luciferase activity in the HeLa reporter strain upon invasion of C. diphtheriae

indicated an induction of the NF-B signal pathway. This was further studied using immuno-

fluorescence microscopy. Translocation of the NF-B subunit p65 into the nucleus served as a

marker for NF-B induction; this was tested using a p65-specific antibody and bacteria tagged

by GFP expression (Figure 6). In these experiments, p65 was labelled in red, the DNA within

the nucleus stained in blue and bacteria in green (Figure 6A). When the pictures of the

different fluorescence channels were merged, translocation of p65 was visualized by a change

of the nucleus colour from blue to pink due to translocation of p65 from the cytoplasm into the

nucleus. When bacteria were clearly inside the cell, as shown by orthogonal z stacks, a

translocation of p65 from the cytoplasm into the nucleus was detectable; bacteria attached to

the cell surface did not induce p65 translocation (Figure 6B) further supporting the idea that

invasion of C. diphtheriae is a prerequisite for NF-B induction.

109
Publications

Figure 6. Investigation of NF-B induction by immuno-fluorescence microscopy.


(A) Overview: HeLa cells incubated 2 hours with invasive C. diphtheriae strain ISS4060
expressing a GFP marker protein (green) show nuclear translocation of NFB p65.
Permeabilized cells were counterstained with Pacific blue (blue), NFB p65 subunit (anti NFB
p65, C-20, 1:250) was detected with anti-rabbit Alexa 647 (1:500, red). Translocation of p65 is
indicated by pink coloring of the nucleus. (B) Non-adherent bacteria were removed by washing
and confocal image stacks were acquired on a Leica TCS SP5. Upper panel: representative
HeLa cell with intracellular C. diphtheriae DSM49388 pEPR1-P45gfp and nuclear translocation
of NFB p65 (arrow). Lower panel: HeLa cell with extracellular C. diphtheriae ISS4060

110
Publications

pEPR1-P45gfp and predominantly cytoplasmatic localization of NFB p65. Left: DAPI, GFP
and NFB; Middle: GFP and NFB; Right: orthogonal sections of image stacks in x and y axis.

Effects of diphtheria toxin in eukaryotic cell cultures

As described above, the luciferase signal declined below background level when HeLa cells

were infected by the tox+ strain DSM43989 for 20 h. This led to the hypothesis that the

production of diphtheria toxin might have a detrimental effect on the cells after prolonged

incubation in this assay. To address this idea, cell viability assays based on mitochondrial

enzyme activity were performed (Figure 7). For this purpose Detroit562 and HeLa cells were

exposed to DMEM containing secreted proteins from strain DSM43988 and the toxin-

producing strain DSM43989, respectively. After 20 h alamar blue was added. In case of

undergoing apoptosis or necrosis, a redox reaction, in which alamar blue is reduced by the

cells, can be measured by fluorescence of 560 nm (excitation) and 590 nm (emission). Indeed,

the data revealed a cytotoxic effect of DMEM containing secreted proteins of the toxigenic

strain DSM43989. The fluorescence values of all cells incubated with this media declined to

that of cells lysed by 0.1 % Triton-X-100. This was not the case for cells exposed to the

supernatant of the non-toxigenic strain DSM43988, which values are up to five-fold higher than

the negative control. Our data indicate that strain DSM43989 might be able to produce the

toxin under cell culture conditions, which should be the reason for luciferase values declined

below background level, when cells were incubated for 20 h with the toxigenic strain.

111
Publications

Detroit562 HeLa
50000 50000

45000 45000

Fluorescence 560/590nm
40000 40000

35000 35000

30000 30000

25000 25000

20000 20000

15000 15000

10000 10000

5000 5000

0 0
0 20 40 60 80 0 20 40 60 80

Time [h]

Figure 7. Cell viability assay of different eukaryotic cells exposed to diphtheria toxin.
Detroit562 and HeLa cells were seeded in 96-well plates at a density of 510 x 103 cells per
well directly in 190 µl DMEM containing secreted proteins of DSM43988 and DSM43989,
respectively. After 20 h of incubation at 37°C, 5 % CO2 and 95 % humidity, alamar blue was
added (10 % in culture medium) to each well and absorbance was measured at 560 nm
(excitation wavelength) and 590 nm (emission wavelength) using a microplate reader (Tecan
Infinite M200 Pro) at different time points. negative control: 0.1 % Triton-X-100, positive
control: cells without treatment, supernatant DSM43988, supernatant DSM43989
(toxigenic).

Discussion

In this study, we showed that HeLa cells are recognized by C. diphtheriae with extremely high

efficiency. This result might also explain, why HEp-2 cells are preferentially recognized

compared to Detroit562 cells in other studies (see for example Mandlik et al., 2007), since this

cell line carries HeLa markers according to the American Type Culture Collection (ATCC) and

the National Collection of Type Cultures (NCTC).

As mentioned above, adhesion of C. diphtheriae was described before for a variety of

human cell lines. All in all, this indicates that a general property or component of cells is

recognized by C. diphtheriae, besides the fact that minor pili subunits are involved in cell

specificity at least in vitro (Mandlik et al., 2008), and might explain the broad host and tissue

specificity of the bacterium.

The effect of C. diphtheriae infection on host cell signalling is not well understood and

besides this study only one further manuscript is available. Puliti and co-workers showed

strong differences in cytokine secretion in a mouse model in response to infection with various

112
Publications

C. diphtheriae strains and it was concluded that the bacterial strains studied interact with the

host in a different manner (Puliti et al., 2006). The NF-B signalling pathway is involved in the

regulation of innate and adaptive immune responses, inflammation and various other cellular

processes (O'Dea & Hoffmann, 2009). It has a crucial role for resistance to infections (Tato &

Hunter, 2002) and controls cytokine excretion and consequently the fate of a pathogen inside

the host. During evolution, some pathogens have found mechanisms to inhibit NF-B

activation, while others even utilize and manipulate the pathway for their benefit (Tato &

Hunter, 2002).

In the study presented here, the influence of seven C. diphtheriae isolates on NF-B

induction in human epithelial cells was investigated. Recognition of pathogen-associated

molecular patterns such as pili subunits by membrane-bound receptors facing the outside of

the cell, as often found for pathogen host interaction (Hayden et al., 2006; Li & Verma, 2002),

was not crucial for NF-B induction. Piliation of the different (invading) strains and luciferase

expression were not correlated and also killed bacteria were not able to induce the NF-B

pathway despite adhesion. As shown recently (Ott et al., 2010b) the bold strain ISS4060 was a

strong inducer in this study, while DSM43989, a poor NF-B inducer, shows strong pili

formation. It might be argued that formation of diphtheria toxin and consequent inactivation of

protein synthesis might prevent luciferase expression, but also the non-toxigenic strain

ISS4749, which showed a high number of pili, caused only minor NF-B induction. In fact,

strong pili formation seems to have a negative effect on invasion into host cells (Ott et al.,

2010b) and subsequently on NF-B induction.

Our data indicate that the response of human epithelial cells to C. diphtheriae infection

is determined by the internalization of bacteria and that invasion into these cells is an active

process of the bacteria, since tetracycline-treated C. diphtheriae were still able to attach to

host cells, but lost their ability to invade into the cytoplasm. Since we found this correlation

between invasion and NF-B induction, it would be tempting to either manipulate an invasive

strain to become non-invasive or vice versa and to analyze NF-B induction in these mutants;

113
Publications

unfortunately, the molecular background of C. diphtheriae internalization is almost unknown.

Only recently, in vitro experiments with protein-coated latex beads indicated that DIP0733,

encoding 67-72p hemagglutinin, contributes to invasion in HEp-2 cells (Sabbadini et al., 2012).

Once inside the cell, either a cell wall component of the internalized bacteria might be

recognized or a protein secreted by C. diphtheriae into the cytosol of the host cell. As shown

by a proteomics approach (Hansmeier et al., 2006), C. diphtheriae is able to export more than

one hundred proteins to the outside of the bacterium. These are prime candidates for putative

signals sensed by the host cell and future mutagenesis studies of the corresponding genes

might help to further understand the interaction of pathogen and host on a molecular level.

Acknowledgements

The  authors  wish  to  thank  C.  v.  Hunolstein  (Istituto  Superiore  di  Sanita’,  Rome)  for  providing  

strains ISS3319, ISS4060, ISS4746 and ISS4749. Lentiviral packaging plasmids were a

generous gift from D. Trono, Geneva. The financial support of AB, AE and LO by the Deutsche

Forschungsgemeinschaft for in frame of SFB796 (projects B1 and B5) is gratefully

acknowledged.

References

Bennett, K. L. & Pearson, G. D. (1993). Sequence conversion during postreplicative

adenovirus overlap recombination. Proc Natl Acad Sci U S A 90, 1397-1401.

Bertuccini, L., Baldassarri, L. & von Hunolstein, C. (2004). Internalization of non-toxigenic

Corynebacterium diphtheriae by cultured human respiratory epithelial cells. Microb Pathog 37,

111-108.

Bose, R., Thinwa, J., Chaparro, P., Zhong, Y., Bose, S., Zhong, G. & Dube, P. H. (2011).

MAP-kinase-dependent IL-1alpha intracrine signaling is modulated by YopP during Y.

enterocolitica infection. Infect Immun.

114
Publications

Colombo, A. V., Hirata, R., Jr., de Souza, C. M., Monteiro-Leal, L. H., Previato, J. O.,

Formiga, L. C., Andrade, A. F. & Mattos-Guaraldi, A. L. (2001). Corynebacterium

diphtheriae surface proteins as adhesins to human erythrocytes. FEMS Microbiol Lett 197,

235-239.

Corbett, D., Wang, J., Schuler, S., Lopez-Castejon, G., Glenn, S., Brough, D., Andrew, P.

W., Cavet, J. S. & Roberts, I. S. (2011). Two Zinc Uptake Systems Contribute to the Full

Virulence of Listeria monocytogenes During Growth in vitro and in vivo. Infect Immun.

Corboz, L., Thoma, R., Braun, U. & Zbinden, R. (1996). [Isolation of Corynebacterium

diphtheriae subsp. belfanti from a cow with chronic active dermatitis]. Schweiz Arch Tierheilkd

138, 596-599.

dos Santos, C. S., dos Santos, L. S., de Souza, M. C. & other authors (2010). Non-opsonic

phagocytosis of homologous non-toxigenic and toxigenic Corynebacterium diphtheriae strains

by human U-937 macrophages. Microbiol Immunol 54, 1-10.

Farfour, E., Badell, E., Zasada, A., Hotzel, H., Tomaso, H., Guillot, S., & Guiso, N. (2012).

Characterization and comparison of invasive Corynebacterium diphtheriae isolates from

France and Poland. J Clin Microbol 50,173-175.

Galazka, A. (2000). The changing epidemiology of diphtheria in the vaccine era. J Infect Dis

181 Suppl 1, S2-S9.

Gey, G. O., Coffmann, W. D. & Kubicek, M. T. (1952). Tissue culture studies of the

proliferative capacity of cervical carcinoma and normal epithelium. Cancer Research 12, 264-

265.

115
Publications

Grant, S. G., Jessee, J., Bloom, F. R. & Hanahan, D. (1990). Differential plasmid rescue

from transgenic mouse DNAs into Escherichia coli methylation-restriction mutants. Proc Natl

Acad Sci U S A 87, 4645-4649.

Hadfield, T. L., McEvoy, P., Polotsky, Y., Tzinserling, V. A. & Yakovlev, A. A. (2000). The

pathology of diphtheria. J Infect Dis 181 Suppl 1, S116-S120.

Hall, A. J., Cassiday, P. K., Bernard, K. A. & other authors (2010). Novel Corynebacterium

diphtheriae in domestic cats. Emerg Infect Dis 16, 688-691.

Hansmeier, N., Chao, T. C., Kalinowski, J., Pühler, A. & Tauch, A. (2006). Mapping and

comprehensive analysis of the extracellular and cell surface proteome of the human pathogen

Corynebacterium diphtheriae. Proteomics 6, 2465-2476.

Hayden, M. S., West, A. P. & Ghosh, S. (2006). NF-B and the immune response. Oncogene

25, 6758-6780.

Henricson, B., Segarra, M., Garvin, J., Burns, J., Jenkins, S., Kim, C., Popovic, T., Golaz,

A. & Akey, B. (2000). Toxigenic Corynebacterium diphtheriae associated with an equine

wound infection. J Vet Diagn Invest 12, 253-257.

Hirata, R., Napoleao, F., Monteiro-Leal, L. H., Andrade, A. F., Nagao, P. E., Formiga, L.

C., Fonseca, L. S. & Mattos-Guaraldi, A. L. (2002). Intracellular viability of toxigenic

Corynebacterium diphtheriae strains in HEp-2 cells. FEMS Microbiol Lett 215, 115-119.

Hirata, R., Pereira, G. A., Filardy, A. A., Gomes, D. L., Damasco, P. V., Rosa, A. C.,

Nagao, P. E., Pimenta, F. P. & Mattos-Guaraldi, A. L. (2008). Potential pathogenic role of

116
Publications

aggregative-adhering Corynebacterium diphtheriae of different clonal groups in endocarditis.

Braz J Med Biol Res 41, 986–991

Hirata, R., Jr., Souza, S. M., Rocha-de-Souza, C. M., Andrade, A. F., Monteiro-Leal, L. H.,

Formiga, L. C. & Mattos-Guaraldi, A. L. (2004). Patterns of adherence to HEp-2 cells and

actin polymerisation by toxigenic Corynebacterium diphtheriae strains. Microb Pathog 36, 125-

130.

Leggett, B. A., De Zoysa, A., Abbott, Y. E., Leonard, N., Markey, B. & Efstratiou, A.

(2010). Toxigenic Corynebacterium diphtheriae isolated from a wound in a horse. Vet Rec

166, 656-657.

Li, Q. & Verma, I. M. (2002). NF-B regulation in the immune system. Nat Rev Immunol 2,

725-734.

Mandlik, A., Swierczynski, A., Das, A. & Ton-That, H. (2007). Corynebacterium diphtheriae

employs specific minor pilins to target human pharyngeal epithelial cells. Mol Microbiol 64,

111-124.

Mandlik, A., Swierczynski, A., Das, A. & Ton-That, H. (2008). Pili in Gram-positive bacteria:

assembly, involvement in colonization and biofilm development. Trends Microbiol 16, 33-40.

Mattos-Guaraldi, A. L., Duarte Formiga, L. C. & Pereira, G. A. (2000). Cell surface

components and adhesion in Corynebacterium diphtheriae. Microbes Infect 2, 1507-1512.

O'Dea, E. & Hoffmann, A. (2009). NF-B signaling. Wiley Interdiscip Rev Syst Biol Med 1,

107-115.

117
Publications

Ott, L., Höller, M., Gerlach, R. G., Hensel, M., Rheinlaender, J., Schäffer, T. E. &

Burkovski, A. (2010a). Corynebacterium diphtheriae invasion-associated protein (DIP1281) is

involved in cell surface organization, adhesion and internalization in epithelial cells. BMC

Microbiol 10:2.

Ott, L., Höller, M., Rheinlaender, J., Schäffer, T. E., Hensel, M. & Burkovski, A. (2010b).

Strain-specific differences in pili formation and the interaction of Corynebacterium diphtheriae

with host cells. BMC Microbiol 10, 257.

Ott, L., McKenzie, A., Baltazar, M. T., Britting, S., Bischof, A., Burkovski, A. & Hoskisson,

P. A. (2012). Evaluation of invertebrate infection models for pathogenic corynebacteria. FEMS

Immunol Med Microbiol.

Perkins, N. D. (2007). Integrating cell-signalling pathways with NF-B and IKK function. Nat

Rev Mol Cell Biol 8, 49-62.

Peterson, W. D., Jr., Stulberg, C. S., Swanborg, N. K. & Robinson, A. R. (1968). Glucose-

6-phosphate dehydrogenase isoenzymes in human cell cultures determined by sucrose-agar

gel and cellulose acetate zymograms. Proc Soc Exp Biol Med 128, 772-776.

Puliti, M., von Hunolstein, C., Marangi, M., Bistoni, F. & Tissi, L. (2006). Experimental

model of infection with non-toxigenic strains of Corynebacterium diphtheriae and development

of septic arthritis. J Med Microbiol 55, 229-235.

Rheinlaender, J., Gräbner, A., Ott, L., Burkovski, A. & Schäffer, T. E. (2012). Nanoscale

analyses of Corynebacterium diphtheriae pili by atomic force microscopy. Eur Biophys J 41,

561-570.

118
Publications

Revez, J., Rossi, M., Ellstrom, P., de Haan, C., Rautelin, H. & Hanninen, M. L. (2011).

Finnish Campylobacter jejuni strains of multilocus sequence type ST-22 complex have two

lineages with different characteristics. PLoS One 6, e26880.

Sabbadini, P. S., Assis, M. C., Trost, E., Gomes, D. L., Moreira, L. O., Dos Santos, C. S.,

Pereira, G. A., Nagao, P. E., Azevedo, V. A., Hirata, Junior R., Dos Santos, A. L., Tauch,

A. & Mattos-Guaraldi, A.L. (2012). Corynebacterium diphtheriae 67-72p hemagglutinin,

characterized as the protein DIP0733, contributes to invasion and induction of apoptosis in

HEp-2 cells Microb Pathog 52, 165-176.

Sambrook, J., Fritsch, E. F. & Maniatis, T. (1989). 2nd ed edn. Cold Spring Harbor, N.Y.:

Cold Spring Harbor Laboratory Press.

Scherer, W. F., Syverton, J. T. & Gey, G. O. (1953). Studies on the propagation in vitro of

poliomyelitis viruses. IV. Viral multiplication in a stable strain of human malignant epithelial

cells (strain HeLa) derived from an epidermoid carcinoma of the cervix. J Exp Med 97, 695-

710.

Swaminathan, A., Mandlik, A., Swierczynski, A., Gaspar, A., Das, A. & Ton-That, H.

(2007). Housekeeping sortase facilitates the cell wall anchoring of pilus polymers in

Corynebacterium diphtheriae. Mol Microbiol 66, 961-974.

Tato, C. M. & Hunter, C. A. (2002). Host-pathogen interactions: subversion and utilization of

the NF- Infect Immun 70, 3311-3317.

von Hunolstein, C., Alfarone, G., Scopetti, F. & other authors (2003). Molecular

epidemiology and characteristics of Corynebacterium diphtheriae and Corynebacterium

ulcerans strains isolated in Italy during the 1990s. J Med Microbiol 52, 181-188.

119
Publications

Table 1. Bacterial strains and eukaryotic cell lines used in this study.

Strains Description Reference

C. diphtheriae

DSM43988 non-toxigenic, isolated from throat DSMZ, Braunschweig,

culture, no pili (Ott et al., 2010b, Germany

Rheinlaender et al., 2012)

DSM43989 tox+, unknown source, medium pili length DSMZ, Braunschweig,

and number (Ott et al., 2010b, Germany

Rheinlaender et al., 2012)

DSM44123 non-toxigenic isolate, type-strain, DSMZ, Braunschweig,

unknown source, medium number short Germany

spike-like pili (Ott et al., 2010b,

Rheinlaender et al., 2012)

ISS3319 C. diphtheriae var. mitis, non-toxigenic, (Bertuccini et al., 2004)

isolated from patients affected by

pharyngitis/tonsillitis, medium pili length

and number (Ott et al., 2010b,

Rheinlaender et al., 2012)

ISS4060 C. diphtheriae var. gravis, non-toxigenic, (Bertuccini et al., 2004)

isolated from patients affected by

pharyngitis/tonsilitis, no pili (Ott et al.,

2010b, Rheinlaender et al., 2012)

ISS4746 C. diphtheriae var. gravis, non-toxigenic, (Puliti et al., 2006)

isolated from patients affected by

pharyngitis/tonsillitis, medium number of

extended pili (Ott et al., 2010b,

Rheinlaender et al., 2012)

120
Publications

ISS4749 C. diphtheriae var. gravis, non-toxigenic, (Puliti et al., 2006)

isolated from patients affected by

pharyngitis/tonsillitis, high number of

extended pili (Ott et al., 2010b,

Rheinlaender et al., 2012)

DSM43988 pEPR1- DSM43988 transformed with pEPR1- (Ott et al., 2012)

P45gfp P45gfp

ISS4060 pEPR1- C. diphtheriae var. gravis transformed (Ott et al., 2012)

P45gfp with pEPR1 carrying P45, gfpuv, KmR,

rep, per, T1, T2

E. coli

DH5MCR endA1 supE44 thi-1 - recA1 gyrA96 (Grant et al., 1990)

relA1 deoR (lacZYA-argF) U196

80lacZ M15mcrA (mmr

hsdRMSmcrBC)

Cell lines

Detroit562 human hypopharyngeal carcinoma cells (Peterson et al., 1968)

HeLa human cervical carcinoma cells (Gey et al., 1952;

Scherer et al., 1953)

Detroit-NFB reporter lentiviral transduced Detroit562 cells with This study

cells stable integrated NF-B reporter gene

construct pTRH-NFB FL-Luc-Puro

HeLa-NFB reporter lentiviral transduced HeLa cells with This study

cells stable integrated NF-B reporter gene

construct pTRH-NFB FL-Luc-Puro

121
Summary/Zusammenfassung

4 Summary/Zusammenfassung
4.1 Summary

Corynebacterium diphtheriae, the causative agent of respiratory diphtheria was generally


considered as an extracellular colonizer, with the diphtheria toxin as the main virulence factor.
But non-toxigenic strains are also able to adhere to the host cell and to internalize into it. To
date, not much is known about the factors, which are crucial for colonization of the host.
In the framework of this study, the characterization of the interaction of C. diphtheriae
with host cells revealed strain-specific differences in adhesion and invasion rates of seven wild
type strains and that adhesion and invasion are not strictly coupled processes. Atomic force
microscopic analysis showed that the studied strains differ significantly in their macromolecular
surface structure, especially in pili formation and length and number of pili structures (Ott et al,
2010b). Additionally, these pili structures show different biophysical properties which were
investigated in collaboration with Reinlaender and co-workers (Rheinlaender et al, 2012).
Interestingly, there is no correlation between pili formation, adhesion and invasion of the
different wild type strains. Furthermore, we were able to show that one of the three known pili
cluster (spaDEF) for C. diphtheriae is absent in the strains investigated. The work proved that
there must be other factors which are crucial for colonization if the host besides pili (Ott et al,
2010b).
To investigate the invasion process at a more molecular level, we generated insertion
mutants in gene DIP1281, whose gene product was annotated as an invasion-associated
protein. The resulting mutant strains were not able to separate the daughter cells after cell
division, to adhere to the host cell or to invade the cell. Further investigations of the mutants
demonstrated that this phenotype is an effect of rearrangements of the outer surface of
bacteria, and DIP1281 seems to be predominantly involved in the organization of the outer
surface protein layer (Ott et al, 2010a).
For further identification of virulence factors, we established an invertebrate infection
model for pathogenic corynebacteria for high throughput screening of mutant strains. The
nematode Caenorhabditis elegans gives differentiated results with regard to colonization of the
host, while the wax moth larvae Galleria mellonella will be useful with respect to innate
immune responses to pathogens because it offers a more complex cell-based immune system
compared to C. elegans (Ott et al, 2012a).
When we studied the host immune response during infection with C. diphtheriae we
were able to show that C. diphtheriae is capable of activating the  NFĸB  transduction  pathway  
in eukaryotic cells in a strain-specific manner; induction only occurs during incubation with
living bacteria and requires invasion of the bacteria into the epithelial cells (Ott et al, 2012b).

122
Summary/Zusammenfassung

In cooperation with Ochrombel and co-workers, we investigated potassium uptake


systems in corynebacteria (Ochrombel et al, 2011) and found that pathogenic corynebacteria,
such as C. diphtheriae and C. jeikeium, exhibit more potassium transporters than non-
pathogens, such as C. glutamicum. Furthermore, C. diphtheriae K+ transporter mutants were
eliminated faster by macrophages and are less stress-resistant, which make K+ uptake
systems of pathogenic corynebacteria potential virulence factors with regard to survival in an
acid environment, like the inside of macrophages.
In collaboration with Trost and co-workers, we participated in data evaluation of the
genome sequence of C. pseudotuberculosis FRC41 with the idea of a comparative analysis of
pathogenic corynebacteria. This led to the identification of an extended number of virulence
factors, indicating that the human isolate C. pseudotuberculosis FRC41 is equipped with a
distinct gene set promoting its survival under unfavorable environmental conditions (Trost et
al, 2010b).

123
Summary/Zusammenfassung

4.2 Zusammenfassung

Als Hauptvirulenzfaktor des Gram-positiven, aeroben Bakteriums Corynebacterium diphtheriae


wurde bisher das hochwirksame Diphtherietoxin betrachtet. Doch auch nicht-toxigene Stämme
sind in der Lage, sich an Wirtszellen anzuheften, in diese einzudringen und sie zu schädigen.
Über die Virulenzfaktoren, die dafür verantwortlich sind, ist noch wenig bekannt.
Im Rahmen dieser Arbeit wurde die Interaktion von C. diphtheriae mit eukaryotischen
Zellen näher charakterisiert. Adhäsions- und Invasionsanalysen von sieben verschiedenen
Wildtypstämmen an Epithelzellen ergaben, dass Adhäsion und Invasion keine strikt
gekoppelten Prozesse darstellen. Atomic force mikroskopische Untersuchungen zeigten, dass
sich die Stämme stark in der makromolekularen Oberflächenstruktur, hinsichtlich der
Ausbildung und Länge von Pili-Strukturen, voneinander unterscheiden (Ott et al, 2010b).
Diese Pili-Strukturen haben außerdem verschiedene biophysikalische Eigenschaften, was
durch Arbeiten in Kooperation mit der Arbeitsgruppe um Rheinlaender gezeigt werden konnte
(Rheinlaender et al, 2012). Interessanterweise konnte keine Korrelation zwischen Pili-
Formation, Adhäsion und Invasion gefunden werden. Es konnte auch gezeigt werden, dass
den untersuchten Stämmen eines von drei für C. diphtheriae bekannten Pili-Clustern
(spaDEF), wider Erwarten, fehlt. Diese Ergebnisse machen deutlich, dass es weitere Faktoren
neben den Pili geben muss, die für Adhäsion an Wirtszellen verantwortlich sind (Ott et al,
2010b).
Um die Invasion auf molekularer Ebene zu untersuchen, wurden Insertionsmutanten im
Gen DIP1281 hergestellt, dessen Genprodukt als ein Invasions-assoziiertes Protein annotiert
ist. Die daraus resultierenden Mutanten waren nicht mehr in der Lage, sich nach der
Zellteilung von einander zu separieren, an Wirtszellen anzuheften oder in diese zu invadieren.
Weiterführende Studien der Mutanten zeigten, dass dieser Phänotyp auf eine Umgestaltung
der Oberfläche der Bakterien zurückzuführen ist. DIP1281 scheint daher vorwiegend in die
Organisation der äußeren Proteinschicht involviert zu sein (Ott et al, 2010a).
Für zukünftige Arbeiten konnte ein Infektionsmodell für pathogene Corynebakterien
etabliert werden, welches ermöglicht Mutanten-Banken von C. diphtheriae in großem Maßstab
hinsichtlich der Virulenz zu testen. Dabei erhält man mittels Infektion des Nematoden
Caenorhabditis elegans detaillierte Ergebnisse bezüglich der Wirtskolonisation von C.
diphtheriae, während die Wachsmottenlarve Galleria mellonella hilfreich bei der Aufklärung der
Immunantwort gegenüber Pathogenen ist, da sie über ein komplexeres Immunsystem verfügt
als C. elegans (Ott et al, 2012a).
Außerdem konnte gezeigt werden, dass C. diphtheriae den  NFĸB-Signalweg in HeLa-
Zellen auslösen kann und dass dafür die Invasion der Bakterien notwendig ist. Die bloße

124
Summary/Zusammenfassung

Adhäsion der Bakterien an die Wirtszellen führte zu keiner intrazellulären Wirtsantwort (Ott et
al, 2012b).
In Kooperation mit der Arbeitsgruppe um Ochrombel wurden unterschiedliche Kalium-
Aufnahmesysteme in Corynebakterien untersucht. Es konnte gezeigt werden, dass pathogene
Corynebakterien wie C. diphtheriae und C. jeikeium über mehr Kaliumtransporter verfügen als
nicht-pathogene. Außerdem werden K+-Transportermutanten von C. glutamicum und C.
diphtheriae schneller von Makrophagen eliminiert und sind weniger Stress-resistent
(Ochrombel et al, 2011; Ott, unveröffentlicht).
Für eine vergleichende Analyse pathogener Corynebakterien, habe ich mich in einem
weiteren Kooperationsprojekt an der Datenauswertung der Genom-Entschlüsselung des
humanen Isolats Corynebacterium pseudotuberculosis FRC41 maßgeblich beteiligt. Dies
führte zur Identifikation einer großen Anzahl von Virulenzfaktoren, die zeigen, dass C.
pseudotuberculosis mit einem enormen Genset ausgestattet ist, welches sein Überleben in
einer nachteiligen Umgebung sichert (Trost et al, 2010b).

125
References

5 References

Akira S, Uematsu S, Takeuchi O (2006) Pathogen recognition and innate immunity. Cell 124: 783-801

Anantharaman V, Aravind L (2003) Evolutionary history, structural features and biochemical diversity of
the NlpC/P60 superfamily of enzymes. Genome Biol 4: R11

Baird GJ, Fontaine MC (2007) Corynebacterium pseudotuberculosis and its role in ovine caseous
lymphadenitis. J Comp Pathol 137: 179-210

Barakett V, Morel G, Lesage D, Petit JC (1993) Septic arthritis due to a nontoxigenic strain of
Corynebacterium diphtheriae subspecies mitis. Clin Infect Dis 17: 520-521

Barksdale L (1970) Corynebacterium diphtheriae and its relatives. Bacteriol Rev 34: 378-422

Bayan N, Houssin C, Chami M, Leblon G (2003) Mycomembrane and S-layer: two important structures
of Corynebacterium glutamicum cell envelope with promising biotechnology applications. J Biotechnol
104: 55-67

Beesley ED, Brubaker RR, Janssen WA, Surgalla MJ (1967) Pesticins. 3. Expression of coagulase and
mechanism of fibrinolysis. J Bacteriol 94: 19-26

Bernard K (2012) The genus corynebacterium and other medically relevant coryneform-like bacteria. J
Clin Microbiol 50: 3152-3158

Bertuccini L, Baldassarri L, von Hunolstein C (2004) Internalization of non-toxigenic Corynebacterium


diphtheriae by cultured human respiratory epithelial cells. Microb Pathog 37: 111-108

Bortner CD, Cidlowski JA (2002) Apoptotic volume decrease and the incredible shrinking cell. Cell
Death Differ 9: 1307-1310

Bostock AD, Gilbert FR, Lewis D, Smith DC (1984) Corynebacterium ulcerans infection associated with
untreated milk. J Infect 9: 286-288

Burkovski A (2012) Diphtheria. In The Prokaryotes, 4th ed., E. Rosenberg, E.F. DeLong, F. Thompson,
S. Lory, Stackebrandt E (eds). New York: Springer

Cerdeira LT, Pinto AC, Schneider MP, de Almeida SS, dos Santos AR, Barbosa EG, Ali A, Barbosa MS,
Carneiro AR, Ramos RT, de Oliveira RS, Barh D, Barve N, Zambare V, Belchior SE, Guimarães LC, de
Castro Soares S, Dorella FA, Rocha FS, de Abreu VA, Tauch A, Trost E, Miyoshi A, Azevedo V, Silva A

126
References

(2011a) Whole-genome sequence of Corynebacterium pseudotuberculosis PAT10 strain isolated from


sheep in Patagonia, Argentina. J Bacteriol 193: 6420-6421

Cerdeira LT, Schneider MP, Pinto AC, de Almeida SS, dos Santos AR, Barbosa EG, Ali A, Aburjaile FF,
de Abreu VA, Guimarães LC, Soares Sde C, Dorella FA, Rocha FS, Bol E, Gomes de Sa PH, Lopes
TS, Barbosa MS, Carneiro AR, Juca Ramos RT, Coimbra NA, Lima AR, Barh D, Jain N, Tiwari S, Raja
R, Zambare V, Ghosh P, Trost E, Tauch A, Miyoshi A, Azevedo V, Silva A (2011b) Complete genome
sequence of Corynebacterium pseudotuberculosis strain CIP 52.97, isolated from a horse in Kenya. J
Bacteriol 193: 7025-7026

Cerdeno-Tarraga AM, Efstratiou A, Dover LG, Holden MT, Pallen M, Bentley SD, Besra GS, Churcher
C, James KD, De Zoysa A, Chillingworth T, Cronin A, Dowd L, Feltwell T, Hamlin N, Holroyd S, Jagels
K, Moule S, Quail MA, Rabbinowitsch E, Rutherford KM, Thomson NR, Unwin L, Whitehead S, Barrell
BG, Parkhill J (2003) The complete genome sequence and analysis of Corynebacterium diphtheriae
NCTC13129. Nucleic Acids Res 31: 6516-6523

Cianciotto NP, Groman NB (1996) Extended host range of a beta-related corynebacteriophage. FEMS
Microbiol Lett 140: 221-225

Colombo AV, Hirata R, Jr., de Souza CM, Monteiro-Leal LH, Previato JO, Formiga LC, Andrade AF,
Mattos-Guaraldi AL (2001) Corynebacterium diphtheriae surface proteins as adhesins to human
erythrocytes. FEMS Microbiol Lett 197: 235-239

D'Onofrio C, Paradisi F (1983) The influence of bacterial exotoxins and endotoxins on the phagocytic
activity of human macrophages in culture. Infection 11: 137-143

Dixon B (2010) Sick as a dog. Lancet Infect Dis 10: 73

Dorella FA, Pacheco LG, Oliveira SC, Miyoshi A, Azevedo V (2006) Corynebacterium
pseudotuberculosis: microbiology, biochemical properties, pathogenesis and molecular studies of
virulence. Vet Res 37: 201-218

dos Santos CS, dos Santos LS, de Souza MC, dos Santos Dourado F, de Souza de Oliveira Dias AA,
Sabbadini PS, Pereira GA, Cabral MC, Hirata Junior R, de Mattos-Guaraldi AL (2010) Non-opsonic
phagocytosis of homologous non-toxigenic and toxigenic Corynebacterium diphtheriae strains by
human U-937 macrophages. Microbiol Immunol 54: 1-10

Dramsi S, Caliot E, Bonne I, Guadagnini S, Prevost MC, Kojadinovic M, Lalioui L, Poyart C, Trieu-Cuot
P (2006) Assembly and role of pili in group B streptococci. Mol Microbiol 60: 1401-1413

127
References

Duthie ES (1954) Evidence for two forms of staphylococcal coagulase. J Gen Microbiol 10: 427-436

Fischetti VA, Pancholi V, Schneewind O (1990) Conservation of a hexapeptide sequence in the anchor
region of surface proteins from gram-positive cocci. Mol Microbiol 4: 1603-1605

Groman N, Schiller J, Russell J (1984) Corynebacterium ulcerans and Corynebacterium


pseudotuberculosis responses to DNA probes derived from corynephage beta and Corynebacterium
diphtheriae. Infect Immun 45: 511-517

Guran P, Mollaret HH, Chatelain R, Gropman M, Prigent F, Beal G (1979) [Septic arthritis due to a
nontoxigenic diphtheria bacillus]. Arch Fr Pediatr 36: 926-929

Hadfield TL, McEvoy P, Polotsky Y, Tzinserling VA, Yakovlev AA (2000) The pathology of diphtheria. J
Infect Dis 181 Suppl 1: 116-120

Hansmeier N, Chao TC, Daschkey S, Müsken M, Kalinowski J, Pühler A, Tauch A (2007) A


comprehensive proteome map of the lipid-requiring nosocomial pathogen Corynebacterium jeikeium
K411. Proteomics 7: 1076-1096

Hansmeier N, Chao TC, Kalinowski J, Pühler A, Tauch A (2006a) Mapping and comprehensive analysis
of the extracellular and cell surface proteome of the human pathogen Corynebacterium diphtheriae.
Proteomics 6: 2465-2476

Hansmeier N, Chao TC, Pühler A, Tauch A, Kalinowski J (2006b) The cytosolic, cell surface and
extracellular proteomes of the biotechnologically important soil bacterium Corynebacterium efficiens YS-
314 in comparison to those of Corynebacterium glutamicum ATCC 13032. Proteomics 6: 233-250

Hart RJ (1984) Corynebacterium ulcerans in humans and cattle in North Devon. J Hyg (Lond) 92: 161-
164

Hermann T, Finkemeier M, Pfefferle W, Wersch G, Krämer R, Burkovski A (2000) Two-dimensional


electrophoretic analysis of Corynebacterium glutamicum membrane fraction and surface proteins.
Electrophoresis 21: 654-659

Hirata R, Jr., Souza SM, Rocha-de-Souza CM, Andrade AF, Monteiro-Leal LH, Formiga LC, Mattos-
Guaraldi AL (2004) Patterns of adherence to HEp-2 cells and actin polymerisation by toxigenic
Corynebacterium diphtheriae strains. Microb Pathog 36: 125-130

128
References

Hirata R, Napoleao F, Monteiro-Leal LH, Andrade AF, Nagao PE, Formiga LC, Fonseca LS, Mattos-
Guaraldi AL (2002) Intracellular viability of toxigenic Corynebacterium diphtheriae strains in HEp-2 cells.
FEMS Microbiol Lett 215: 115-119

Hirata R, Pereira GA, Filardy AA, Gomes DL, Damasco PV, Rosa AC, Nagao PE, Pimenta FP, Mattos-
Guaraldi AL (2008) Potential pathogenic role of aggregative-adhering Corynebacterium diphtheriae of
different clonal groups in endocarditis. Braz J Med Biol Res 41: 986–991

Honma Y, Yoshii Y, Watanabe Y, Aoki N, Komiya T, Iwaki M, Arai H, Arakawa Y, Takahashi M, Kimura
H (2009) A case of afebrile pneumonia caused by non-toxigenic Corynebacterium diphtheriae. Jpn J
Infect Dis 62: 327-329

Isaac-Renton JL, Boyko WJ, Chan R, Crichton E (1981) Corynebacterium diphtheriae septicemia. Am J
Clin Pathol 75: 631-634

Kim S, Oh DB, Kwon O, Kang HA (2010) Identification and functional characterization of the NanH
extracellular sialidase from Corynebacterium diphtheriae. J Biochem 147: 523-533

Lehmann KBN, R. (1896) Atlas und Grundriss der Bakteriologie und Lehrbuch der speziellen
bakteriologischen Diagnostik. München: Lehmanns

Lopes T, Silva A, Thiago R, Carneiro A, Dorella FA, Rocha FS, dos Santos AR, Lima AR, Guimaraes
LC, Barbosa EG, Ribeiro D, Fiaux KK, Diniz CA, de Abreu VA, de Almeida SS, Hassan SS, Ali A,
Bakhtiar SM, Aburjaile FF, Pinto AC, Soares Sde C, Pereira Ude P, Schneider MP, Miyoshi A, Edman J,
Spier S, Azevedo V (2012) Complete genome sequence of Corynebacterium pseudotuberculosis strain
Cp267, isolated from a llama. J Bacteriol 194: 3567-3568

Mandlik A, Swierczynski A, Das A, Ton-That H (2007) Corynebacterium diphtheriae employs specific


minor pilins to target human pharyngeal epithelial cells. Mol Microbiol 64: 111-124

Marienfeld S, Uhlemann EM, Schmid R, Krämer R, Burkovski A (1997) Ultrastructure of the


Corynebacterium glutamicum cell wall. Antonie Van Leeuwenhoek 72: 291-297

Mattos-Guaraldi AL, Duarte Formiga LC, Pereira GA (2000) Cell surface components and adhesion in
Corynebacterium diphtheriae. Microbes Infect 2: 1507-1512

Mattos-Guaraldi AL, Formiga LC, Camello TC, Pereira GA, Hirata R, Jr., Halpern M (2001)
Corynebacterium diphtheriae threats in cancer patients. Rev Argent Microbiol 33: 96-100

129
References

McKean S, Davies J, Moore R (2005) Identification of macrophage induced genes of Corynebacterium


pseudotuberculosis by differential fluorescence induction. Microbes Infect 7: 1352-1363

Moreira Lde O, Andrade AF, Vale MD, Souza SM, Hirata R, Jr., Asad LM, Asad NR, Monteiro-Leal LH,
Previato JO, Mattos-Guaraldi AL (2003) Effects of iron limitation on adherence and cell surface
carbohydrates of Corynebacterium diphtheriae strains. Appl Environ Microbiol 69: 5907-5913

Moreira LO, Mattos-Guaraldi AL, Andrade AF (2008) Novel lipoarabinomannan-like lipoglycan (CdiLAM)
contributes to the adherence of Corynebacterium diphtheriae to epithelial cells. Arch Microbiol 190: 521-
530

Muttaiyah S, Best EJ, Freeman JT, Taylor SL, Morris AJ, Roberts SA (2011) Corynebacterium
diphtheriae endocarditis: a case series and review of the treatment approach. Int J Infect Dis 15: 584-
588

Navarre WW, Schneewind O (1999) Surface proteins of gram-positive bacteria and mechanisms of their
targeting to the cell wall envelope. Microbiol Mol Biol Rev 63: 174-229

Niederweis M, Danilchanka O, Huff J, Hoffmann C, Engelhardt H (2010) Mycobacterial outer


membranes: in search of proteins. Trends Microbiol 18: 109-116

Ochrombel I, Ott L, Kramer R, Burkovski A, Marin K (2011) Impact of improved potassium accumulation
on pH homeostasis, membrane potential adjustment and survival of Corynebacterium glutamicum.
Biochim Biophys Acta 1807: 444-450

Ofek I, Goldhar J, Keisari Y, Sharon N (1995) Nonopsonic phagocytosis of microorganisms. Annu Rev
Microbiol 49: 239-276

Ott L, Höller M, Gerlach RG, Hensel M, Rheinlaender J, Schäffer TE, Burkovski A (2010a)
Corynebacterium diphtheriae invasion-associated protein (DIP1281) is involved in cell surface
organization, adhesion and internalization in epithelial cells. BMC Microbiol 10:2

Ott L, Höller M, Rheinlaender J, Schäffer TE, Hensel M, Burkovski A (2010b) Strain-specific differences
in pili formation and the interaction of Corynebacterium diphtheriae with host cells. BMC Microbiol 10:
257

Ott L, McKenzie A, Baltazar MT, Britting S, Bischof A, Burkovski A, Hoskisson PA (2012a) Evaluation of
invertebrate infection models for pathogenic corynebacteria. FEMS Immunol Med Microbiol

130
References

Ott L, Scholz B, Höller M, Hasselt K, Ensser A, Burkovski A (2012b) Induction of NFkB transduction
pathway in response to Corynebacterium diphtheriae infection. Microbiology, accepted

Patey O, Bimet F, Riegel P, Halioua B, Emond JP, Estrangin E, Dellion S, Alonso JM, Kiredjian M,
Dublanchet A, Lafaix C (1997) Clinical and molecular study of Corynebacterium diphtheriae systemic
infections in France. J Clin Microbiol 35: 441-445

Pethick FE, Lainson AF, Yaga R, Flockhart A, Smith DG, Donachie W, Cerdeira LT, Silva A, Bol E,
Lopes TS, Barbosa MS, Pinto AC, Dos Santos AR, Soares SC, Almeida SS, Guimaraes LC, Aburjaile
FF, Abreu VA, Ribeiro D, Fiaux KK, Diniz CA, Barbosa EG, Pereira UP, Hassan SS, Ali A, Bakhtiar SM,
Dorella FA, Carneiro AR, Ramos RT, Rocha FS, Schneider MP, Miyoshi A, Azevedo V, Fontaine MC
(2012a) Complete genome sequence of Corynebacterium pseudotuberculosis strain 1/06-A, isolated
from a horse in North America. J Bacteriol 194: 4476

Pethick FE, Lainson AF, Yaga R, Flockhart A, Smith DG, Donachie W, Cerdeira LT, Silva A, Bol E,
Lopes TS, Barbosa MS, Pinto AC, dos Santos AR, Soares SC, Almeida SS, Guimaraes LC, Aburjaile
FF, Abreu VA, Ribeiro D, Fiaux KK, Diniz CA, Barbosa EG, Pereira UP, Hassan SS, Ali A, Bakhtiar SM,
Dorella FA, Carneiro AR, Ramos RT, Rocha FS, Schneider MP, Miyoshi A, Azevedo V, Fontaine MC
(2012b) Complete genome sequences of Corynebacterium pseudotuberculosis strains 3/99-5 and
42/02-A, isolated from sheep in Scotland and Australia, respectively. J Bacteriol 194: 4736-4737

Puliti M, von Hunolstein C, Marangi M, Bistoni F, Tissi L (2006) Experimental model of infection with
non-toxigenic strains of Corynebacterium diphtheriae and development of septic arthritis. J Med
Microbiol 55: 229-235

Rheinlaender J, Grabner A, Ott L, Burkovski A, Schaffer TE (2012) Contour and persistence length of
Corynebacterium diphtheriae pili by atomic force microscopy. Eur Biophys J 41: 561-570

Rivera J, Vannakambadi G, Hook M, Speziale P (2007) Fibrinogen-binding proteins of Gram-positive


bacteria. Thromb Haemost 98: 503-511

Rosenshine I, Duronio V, Finlay BB (1992) Tyrosine protein kinase inhibitors block invasin-promoted
bacterial uptake by epithelial cells. Infect Immun 60: 2211-2217

Sabbadini PS, Assis MC, Trost E, Gomes DL, Moreira LO, Dos Santos CS, Pereira GA, Nagao PE,
Azevedo VA, Hirata Junior R, Dos Santos AL, Tauch A, Mattos-Guaraldi AL (2012) Corynebacterium
diphtheriae 67-72p hemagglutinin, characterized as the protein DIP0733, contributes to invasion and
induction of apoptosis in HEp-2 cells. Microb Pathog 52: 165-176

131
References

Sabbadini PS, Genovez MR, Silva CF, Adelino TL, Santos CS, Pereira GA, Nagao PE, Dias AA,
Mattos-Guaraldi AL, Hirata Junior R (2010) Fibrinogen binds to nontoxigenic and toxigenic
Corynebacterium diphtheriae strains. Mem Inst Oswaldo Cruz 105: 706-711

Saelinger C, Bonventre PF, Imhoff J (1975) Interaction of toxin of Corynebacterium diphtheriae with
phagocytes from susceptible and resistant species. J Infect Dis 131: 431-438

Sangal V, Tucker NP, Burkovski A, Hoskisson PA (2012a) Draft genome sequence of Corynebacterium
diphtheriae biovar intermedius NCTC 5011. J Bacteriol 194: 4738

Sangal V, Tucker NP, Burkovski A, Hoskisson PA (2012b) The draft genome sequence of
Corynebacterium diphtheriae bv. mitis NCTC 3529 reveals significant diversity between the primary
disease-causing biovars. J Bacteriol 194: 3269

Schneewind O, Fowler A, Faull KF (1995) Structure of the cell wall anchor of surface proteins in
Staphylococcus aureus. Science 268: 103-106

Schneewind O, Model P, Fischetti VA (1992) Sorting of protein A to the staphylococcal cell wall. Cell 70:
267-281

Schröder J, Glaub A, Schneider J, Trost E, Tauch A (2012a) Draft genome sequence of


Corynebacterium bovis DSM 20582, which causes clinical mastitis in dairy cows. J Bacteriol 194: 4437

Schröder J, Maus I, Meyer K, Wordemann S, Blom J, Jaenicke S, Schneider J, Trost E, Tauch A


(2012b) Complete genome sequence, lifestyle, and multi-drug resistance of the human pathogen
Corynebacterium resistens DSM 45100 isolated from blood samples of a leukemia patient. BMC
Genomics 13: 141

Schröder J, Maus I, Trost E, Tauch A (2011) Complete genome sequence of Corynebacterium variabile
DSM 44702 isolated from the surface of smear-ripened cheeses and insights into cheese ripening and
flavor generation. BMC Genomics 12: 545

Schuhegger R, Schoerner C, Dlugaiczyk J, Lichtenfeld I, Trouillier A, Zeller-Peronnet V, Busch U,


Berger A, Kugler R, Hormansdorfer S, Sing A (2009) Pigs as source for toxigenic Corynebacterium
ulcerans. Emerg Infect Dis 15: 1314-1345

Silva A, Schneider MP, Cerdeira L, Barbosa MS, Ramos RT, Carneiro AR, Santos R, Lima M,
D'Afonseca V, Almeida SS, Santos AR, Soares SC, Pinto AC, Ali A, Dorella FA, Rocha F, de Abreu VA,
Trost E, Tauch A, Shpigel N, Miyoshi A, Azevedo V (2011) Complete genome sequence of

132
References

Corynebacterium pseudotuberculosis I19, a strain isolated from a cow in Israel with bovine mastitis. J
Bacteriol 193: 323-324

Sykes JE, Mapes S, Lindsay LL, Samitz E, Byrne BA (2010) Corynebacterium ulcerans
bronchopneumonia in a dog. J Vet Intern Med 24: 973-976

Tauch A, Kaiser O, Hain T, Goesmann A, Weisshaar B, Albersmeier A, Bekel T, Bischoff N, Brune I,


Chakraborty T, Kalinowski J, Meyer F, Rupp O, Schneiker S, Viehoever P, Pühler A (2005) Complete
genome sequence and analysis of the multiresistant nosocomial pathogen Corynebacterium jeikeium
K411, a lipid-requiring bacterium of the human skin flora. J Bacteriol 187: 4671-4682

Tauch A, Schneider J, Szczepanowski R, Tilker A, Viehoever P, Gartemann KH, Arnold W, Blom J,


Brinkrolf K, Brune I, Gotker S, Weisshaar B, Goesmann A, Dröge M, Pühler A (2008a) Ultrafast
pyrosequencing of Corynebacterium kroppenstedtii DSM44385 revealed insights into the physiology of
a lipophilic corynebacterium that lacks mycolic acids. J Biotechnol 136: 22-30

Tauch A, Trost E, Tilker A, Ludewig U, Schneiker S, Goesmann A, Arnold W, Bekel T, Brinkrolf K,


Brune I, Gotker S, Kalinowski J, Kamp PB, Lobo FP, Viehoever P, Weisshaar B, Soriano F, Dröge M,
Pühler A (2008b) The lifestyle of Corynebacterium urealyticum derived from its complete genome
sequence established by pyrosequencing. J Biotechnol 136: 11-21

Tiley SM, Kociuba KR, Heron LG, Munro R (1993) Infective endocarditis due to nontoxigenic
Corynebacterium diphtheriae: report of seven cases and review. Clin Infect Dis 16: 271-275

Ton-That H, Schneewind O (2003) Assembly of pili on the surface of Corynebacterium diphtheriae. Mol
Microbiol 50: 1429-1438

Trost E, Al-Dilaimi A, Papavasiliou P, Schneider J, Viehoever P, Burkovski A, Soares SC, Almeida SS,
Dorella FA, Miyoshi A, Azevedo V, Schneider MP, Silva A, Santos CS, Santos LS, Sabbadini P, Dias
AA, Hirata R, Jr., Mattos-Guaraldi AL, Tauch A (2011) Comparative analysis of two complete
Corynebacterium ulcerans genomes and detection of candidate virulence factors. BMC Genomics 12:
383

Trost E, Blom J, Soares Sde C, Huang IH, Al-Dilaimi A, Schroder J, Jaenicke S, Dorella FA, Rocha FS,
Miyoshi A, Azevedo V, Schneider MP, Silva A, Camello TC, Sabbadini PS, Santos CS, Santos LS,
Hirata R, Jr., Mattos-Guaraldi AL, Efstratiou A, Schmitt MP, Ton-That H, Tauch A (2012) Pangenomic
study of Corynebacterium diphtheriae that provides insights into the genomic diversity of pathogenic
isolates from cases of classical diphtheria, endocarditis, and pneumonia. J Bacteriol 194: 3199-3215

133
References

Trost E, Götker S, Schneider J, Schneiker-Bekel S, Szczepanowski R, Tilker A, Viehoever P, Arnold W,


Bekel T, Blom J, Gartemann KH, Linke B, Goesmann A, Pühler A, Shukla SK, Tauch A (2010a)
Complete genome sequence and lifestyle of black-pigmented Corynebacterium aurimucosum ATCC
700975 (formerly C. nigricans CN-1) isolated from a vaginal swab of a woman with spontaneous
abortion. BMC Genomics 11: 91

Trost E, Ott L, Schneider J, Schröder J, Jaenicke S, Goesmann A, Husemann P, Stoye J, Dorella FA,
Rocha FS, Soares Sde C, D'Afonseca V, Miyoshi A, Ruiz J, Silva A, Azevedo V, Burkovski A, Guiso N,
Join-Lambert OF, Kayal S, Tauch A (2010b) The complete genome sequence of Corynebacterium
pseudotuberculosis FRC41 isolated from a 12-year-old girl with necrotizing lymphadenitis reveals
insights into gene-regulatory networks contributing to virulence. BMC Genomics 11: 728

Tsuge Y, Ogino H, Teramoto H, Inui M, Yukawa H (2008) Deletion of cgR_1596 and cgR_2070,
encoding NlpC/P60 proteins, causes a defect in cell separation in Corynebacterium glutamicum R. J
Bacteriol 190: 8204-8214

Ventura M, Canchaya C, Tauch A, Chandra G, Fitzgerald GF, Chater KF, van Sinderen D (2007)
Genomics of Actinobacteria: tracing the evolutionary history of an ancient phylum. Microbiol Mol Biol
Rev 71: 495-548

Vimr ER (1992) Microbial sialidases: does bigger always mean better? Trends Microbiol 2: 271-277

von Hunolstein C, Scopetti F, Efstratiou A, Engler K (2002) Penicillin tolerance amongst non-toxigenic
Corynebacterium diphtheriae isolated from cases of pharyngitis. J Antimicrob Chemother 50: 125-128

Wagner KS, White JM, Crowcroft NS, De Martin S, Mann G, Efstratiou A (2010) Diphtheria in the United
Kingdom, 1986-2008: the increasing role of Corynebacterium ulcerans. Epidemiol Infect 138: 1519-
1530

Zhi XY, Li WJ, Stackebrandt E (2009) An update of the structure and 16S rRNA gene sequence-based
definition of higher ranks of the class Actinobacteria, with the proposal of two new suborders and four
new families and emended descriptions of the existing higher taxa. Int J Syst Evol Microbiol 59: 589-608

134
Curriculum Vitae

6 Curriculum Vitae

Lisa Ott
Schronfeld 61
91054 Erlangen
0177 3885373
lisaott@biologie.uni-erlangen.de

Persönliches

geboren am 05.01.1982 in Nürnberg


ledig, keine Kinder

Ausbildung

Doktorarbeit
1/2009 - 2012 Titel: “Interaction  of  Corynebacterium diphtheriae with  host  cells“
Lehrstuhl für Mikrobiologie, Friedrich-Alexander Universität
Erlangen-Nürnberg, Prof. Dr. A. Burkovski

Mitglied des Sonderforschungsbereichs 796 und des integrierten


Graduiertenkollegs des SFB796

Diplomarbeit
03/2008 - 12/2008 Titel: „Adhäsion  und  Invasion  von  Corynebacterium diphtheriae“  
Lehrstuhl für Mikrobiologie, Friedrich-Alexander Universität
Erlangen-Nürnberg, Prof. Dr. A. Burkovski

Diplom
10/2003 - 01/2008 Biologie, Friedrich-Alexander Universität Erlangen-Nürnberg
Diplomprüfungen in: Mikrobiologie (Hauptfach), Molekulare
Pflanzenphysiologie, Humangenetik und Biochemie

Vordiplomprüfungen in: Bio I, Bio II, Bio III, Bio IV, Physikalische
Chemie, Grundlagen der anorganischen und organischen Chemie

Abitur
09/1992 - 07/2001 Ohm-Gymnasium Erlangen

135
Curriculum Vitae

Lehrerfahrung

seit 01/2009 Durchführung  und  Betreuung  des  Mastermoduls  „Pathogenität“  und  


des  Fachmoduls  „Mikrobiologie“  sowie  Betreuung  von  Studenten  in  
Bachelor- und Masterarbeiten

Publikationen

2012 Ott L., Scholz B., Höller M., Hasselt K., Ensser A., and Burkovski A.
(2012). Induction of the NFk-B signal transduction pathway in
response to Corynebacterium diphtheriae infection, akzeptiert.

2012 Rheinlaender, J., Gräbner, A., Ott, L., Burkovski, A. & Schäffer, T. E.
(2012). Contour and persistence length of Corynebacterium
diphtheriae pili by atomic force microscopy. Eur. Biophys. J. 41, 561-
570.

2012 Ott, L., McKenzie, A., Baltazar, A. T., Britting, S., Bischof, A.,
Burkovski, A. & Hoskisson, P. A. (2012). Evaluation of invertebrate
infection models for pathogenic corynebacteria. FEMS Immunol.
Med. Microbiol. 65, 413-421.

2011 Ochrombel, I., Ott, L., Krämer, R., Burkovski, A. and Marin, K. (2011).
Impact of improved potassium accumulation on pH homeostasis,
membrane potential adjustment and survival of Corynebacterium
glutamicum. Biochim Biophys Acta. 1807, 444-450.

2010 Trost, E., Ott, L., Schneider, J., Schröder, J., Jaenicke, S., Goesmann,
A., Husemann, P., Stoye, J., Alves Dorella, F., Rocha, F. S., de Castro
Soares,  S.,  D‘Afonseca,  V.,  Miyoshi,  A.,  Ruiz,  J.,  Silva,  A.,  Azevedo,  V.,  
Burkovski, A., Guiso, N., Join-Lambert, O. F., Kayal, S. and Tauch, A.
(2010). The complete genome sequence of Corynebacterium
pseudotuberculosis FRC41 isolated from a 12-year-old girl with
necrotizing lymphadenitis reveals insights into gene-regulatory
networks contributing to virulence. BMC Genomics 11, 728.

2010 Ott, L., Höller, M., Rheinlaender, J., Schäffer, T. E., Hensel, M. and
Burkovski, A. (2010). Strain-specific differences in pili formation and
the interaction of Corynebacterium diphtheriae with host cells. BMC
Microbiol 10:257.

2010 Ott, L., Höller, M., Gerlach, R. G., Hensel, M., Rheinländer, J.,
Schäffer, T. E. and Burkovski, A. (2010). Corynebacterium diphtheriae
invasion-associated protein (DIP1281) is involved in cell surface
organization, adhesion and internalization in epithelial cells. BMC
Microbiol 10:2.

136
Curriculum Vitae

2008 Amon, J., Bräu, T., Grimrath, A., Hänßler, E., Hasselt K., Höller, M.,
Jessberger, N., Ott, L., Szököl, J., Titgemeyer, F. and Burkovski, A.
(2008). Nitrogen control in Mycobacterium smegmatis: nitrogen-
dependent expression of ammonium transport and assimilation
proteins depends on the OmpR-type regulator GlnR. J Bacteriol 190,
7108-7116.

Buchkapitel

2012 Corynebacterium diphtheriae and related toxigenic species.


A. Burkovski (ed.), Springer, Dortrecht, NL
Kapitel: Adhesion, Invasion and Host Response, in Vorbereitung.

Präsentationen

07/2012 4th Annual Retreat, Erlangen School of Molecular Communication,


Kloster Banz, Bad Staffelstein
Vortrag:  “Evaluation  of  invertebrate  infection  models  for  pathogenic  
corynebacteria”

03/2012 Jahrestagung der Vereinigung für Allgemeine und Angewandte


Mikrobiologie, Tübingen
Poster: “Evaluation  of  invertebrate  infection  models  for  pathogenic  
corynebacteria”

07/2011 3rd Annual Retreat, Erlangen School of Molecular Communication,


Kloster Banz, Bad Staffelstein
Vortrag und Poster:  “Interaction  of  Corynebacterium diphtheriae
with  host  cells”

05/2011 Mol Micro Meeting, Würzburg


Poster:  “Strain-specific differences in pili-formation and interaction
of Corynebacterium diphtheriae with  host  cells”

04/2011 Jahrestagung der Vereinigung für Allgemeine und Angewandte


Mikrobiologie, Karlsruhe
Poster:  “Strain-specific differences in pili-formation and interaction
of Corynebacterium diphtheriae with  host  cells”

09/2010 2nd Annual Retreat, Erlangen School of Molecular Communication,


Kloster Banz, Bad Staffelstein, Germany
Vortrag:  “Adhesion  and  Invasion  of  Corynebacterium diphtheriae”

03/2010 3th joint Conference DGHM and VAAM, Hannover, Germany


Poster: “Strain-specific differences in the interaction of
Corynebacterium diphtheriae with  host  cells”

137
Curriculum Vitae

09/2009 First Annual Retreat, Erlangen School of Molecular Communication,


Schloss Atzelsberg, Atzelsberg, Germany
Vortrag: “Adhesion  and  Invasion  of  Corynebacterium diphtheriae“

Forschungsreisen

05/2011 Strathclyde Institute of Pharmacy and Biomedical Sciences,


Glasgow, Scottland; Infektion von Caenorhabditis elegans und
Galleria mellonella mit pathogenen Bakterien

138
Danksagung

7 Danksagung
Mein größter Dank gilt Prof. Dr. Andreas Burkovski. Ohne seine uneingeschränkte
Unterstützung für mein Projekt wäre die Arbeit in diesem Umfang nicht möglich gewesen. Ich
danke Andreas für das entgegengebrachte Vertrauen, die wichtigen Impulse für meine Arbeit
und dass ich immer meine Ideen verwirklichen durfte. Auch für die stets ausführlichen
Korrekturvorschläge und die immer offene Tür möchte ich mich ganz herzlich bei ihm
bedanken. Die Zusammenarbeit war auf wissenschaftlicher sowie persönlicher Ebene immer
fair und unkompliziert und hat mir sehr viel Spaß gemacht.

Weiterhin möchte ich Prof. Dr. Uwe Sonnewald für die Übernahme des Zeitgutachtens für
diese Arbeit danken.

An dieser Stelle möchte ich mich auch bei der besten und nettesten Sekretärin der Welt
bedanken: Frau Donata Wehr! Sie ist eine sehr liebenswürdige und zuverlässige Person!

Mein Dank gilt weiterhin allen Mitgliedern des Burki-Labs: Nadine, Kristin, Nadja, Eva und
Elena: ihr alle habt mir die Zeit im Labor extrem angenehm und kurzweilig gemacht und ich bin
sehr froh, dass daraus so tolle Freundschaften entstanden sind! Eva danke ich ganz
besonders für die unendliche Geduld und Unterstützung, wenn ich mal wieder an Word, Clone
manager oder Vector NTI verzweifelt bin. Ich danke auch meinen beiden Lieblingsmasteretten
Susi und Kerstin für die schöne und lustige Zeit.

Zu guter Letzt möchte ich mich bei meinem Freund Daniel, meiner Familie und ganz
besonders bei meinen Eltern bedanken. Sie haben mich immer bei meinen Plänen unterstützt
und mir vollstes Vertrauen entgegengebracht.

139

You might also like