development of gene expression systems in

103
Development of gene expression systems in Bifidobacterium bifidum S17 and their application for tumor therapy A dissertation submitted to The Faculty of Natural Science, University of Ulm for partial fulfillment of the degree of Doctor of Sciences (Dr. rer. nat.) presented by Zhongke Sun Born in Shaanxi, China Ulm, July 2014

Upload: others

Post on 23-Jan-2022

3 views

Category:

Documents


0 download

TRANSCRIPT

Development of gene expression systems in

Bifidobacterium bifidum S17 and their

application for tumor therapy

A dissertation submitted to

The Faculty of Natural Science, University of Ulm

for partial fulfillment of the degree of

Doctor of Sciences (Dr. rer. nat.)

presented by

Zhongke Sun

Born in Shaanxi, China

Ulm, July 2014

Amtierender Dekan: Prof. Dr. Steven Jansen

Erstgutachter: Prof. Dr. Bernhard Eikmanns

Zweitgutachter: Prof. Dr. Stefan Binder

Tag der mündlichen Prügung: 30.10.2014

The study was conducted at the Institute of Microbiology and Biotechnology in the Faculty

of Natural Science, University of Ulm.

The work was supported by a PhD fellowship from the German Academic Exchange

Service (DAAD).

1

TABLE OF CONTENTS

TABLE OF CONTENTS .................................................................................................... 1

1 INTRODUCTION ............................................................................................................ 4

1.1 Bifidobacteria: properties and applications.................................................................. 4

1.2 Characteristics of Bifidobacterium bifidum S17 .......................................................... 8

1.3 Expression systems for bifidobacteria ......................................................................... 9

1.3.1 Advantages of bifidobacteria as hosts ................................................................... 9

1.3.2 Plasmids isolated from bifidobacteria ................................................................... 9

1.3.3 Current study of gene expression in bifidobacteria ............................................. 10

1.3.4 Promoters............................................................................................................. 11

1.3.4 Secretion .............................................................................................................. 14

1.4 Bifidobacterial cancer therapy ................................................................................... 15

1.5 The aim of this study.................................................................................................. 17

2 MATERIALS AND METHODS ................................................................................... 18

2.1 Bacterial strains and plasmids.................................................................................... 18

2.1.1 Bacterial strains ................................................................................................... 18

2.1.2 Plasmids and oligonucleotides ............................................................................ 19

2.2 Sample preparation and purification .......................................................................... 19

2.2.1 Preparation of protein samples ............................................................................ 19

2.2.2 Preparation of nucleic acids................................................................................. 20

2.2.3 Purification of nucleic acid samples .................................................................... 22

2.3 Manipulations of nucleic acids .................................................................................. 23

2.3.1 Conventional polymerase chain reaction............................................................. 23

2.3.2 Splicing by overlap extension PCR ..................................................................... 23

2.3.3 Rapid amplification of cDNA ends ..................................................................... 24

2.3.4 Agarose gel electrophoresis................................................................................. 25

2.3.5 DNA restriction and ligation ............................................................................... 26

2.3.6 Transformation of bacteria .................................................................................. 26

2.4 Enzymatic assays ....................................................................................................... 28

2

2.4.1 β-glucuronidase assay.......................................................................................... 28

2.4.2 Phytase assay ....................................................................................................... 28

2.4.3 Nitroreductase assay ............................................................................................ 29

2.5 Growth related assays ................................................................................................ 29

2.5.1 Bacterial growth experiments .............................................................................. 29

2.5.2 Phytate degradation assay.................................................................................... 30

2.5.3 NTR/CB1954 inhibition assay ............................................................................ 30

2.6 Quantification............................................................................................................. 30

2.6.1 BCA protein assay ............................................................................................... 30

2.6.2 2-D Quant protein assay ...................................................................................... 31

2.6.3 DNA/RNA quantification.................................................................................... 31

2.7 Bioinformatic analysis ............................................................................................... 32

3 RESULTS ........................................................................................................................ 33

3.1 Promoter prediction and cloning................................................................................ 33

3.2 Promoter activities in B. bifidum S17 ........................................................................ 35

3.3 Analysis of Pgap .......................................................................................................... 36

3.3.1 Bioinformatic Analysis of Pgap ............................................................................ 36

3.3.2 Experimental determination of TSS .................................................................... 37

3.3.3 Alignment of representatives............................................................................... 38

3.4 Analysis of protein secretion in B. bifidum S17 ........................................................ 40

3.5 Prediction of SPs in B. bifidum S17 ........................................................................... 41

3.7 Construction of a secretion reporter vector................................................................ 44

3.7.1 Clone SP from gene bbif_1734............................................................................ 44

3.7.2 Clone appA as a reporter ..................................................................................... 44

3.7.3 Phytase assay ....................................................................................................... 45

3.8 Comparison of various SPs ........................................................................................ 46

3.9 Secretion of codon adapted ChrR6 in B. bifidum S17 ............................................... 50

4 DISCUSSION .................................................................................................................. 53

4.1 Systems for protein expression and secretion by bifidobacteria ................................ 53

4.1.1 Expression vectors and promoters ....................................................................... 53

4.1.2 Secretion pathway and sorting signal .................................................................. 56

4.1.3 Secretion reporter ................................................................................................ 57

3

4.2 Bifidobacteria as gene delivery vectors in cancer therapy......................................... 60

5 SUMMARY ..................................................................................................................... 63

6 LITERATURES.............................................................................................................. 64

7 APPENDICES ................................................................................................................. 85

7.1 Bacteria strains ........................................................................................................... 85

7.2 Plasmids ..................................................................................................................... 87

7.3 Oligonucleotides ........................................................................................................ 88

7.4 Promoter sequences.................................................................................................... 89

7.5 Putative gap promoters of representative bifidobacteria ........................................... 90

7.6 Reporter gene appA .................................................................................................... 91

7.7 SP sequences .............................................................................................................. 92

7.8 chrR6 sequences......................................................................................................... 93

ABBREVIATIONS ............................................................................................................ 94

PUBLICATIONS ............................................................................................................... 98

ACKNOWLEDGEMENT ................................................................................................ 99

DECLARATION ............................................................................................................. 100

4

1 INTRODUCTION

1.1 Bifidobacteria: properties and applications

Bifidobacterium is a genus of Gram-positive, non-sporulating, non-motile

anaerobic bacteria with irregular shapes. They are endosymbiotic inhabitants of the

gastrointestinal tract (GIT), vagina, and mouth of mammals including humans (Picard et al.,

2005). It was reported that bifidobacteria were first isolated in 1899 by Henry Tissier at

the Pasteur Institute in Paris and the present name was proposed late in 1924 by Orla-

Jensen (Matteuzzi and Sgorbati 1980). Since then, a large number of bifidobacterial strains

were isolated from different habitats. Until April 2014, 48 species were assigned to the

genus Bifidobacterium by German Collection of Microorganisms and Cell Cultures

(DSMZ; http://www.dsmz.de/support/bacterial-nomenclature-up-to-date-downloads.html).

Currently, 37 complete genome sequences of bifidobacteria strains are publically available;

with guanine cytosine (GC) contents ranging from 58% to 63% (see details in Table 1-1).

Among these sequenced strains, the majority are commonly observed in the human GIT

including B. adolencent, B. bifidum, B. breve, and B. longum (Turroni et al., 2009). B.

animalis widely presents in a variety of commercial dairy products distributed worldwide

(Briczinski et al., 2009).

Bifidobacteria are characterized by a special metabolic pathway termed bifidus shunt (de

Vries et al., 1967), with the key enzyme fructose-6-phosphate phosphoketolase (F6PPK)

converting hexoses (Scardovi et al., 1971). One main product of bifidobacteria metabolism

is lactate, while they do not belong to traditional Lactic Acid Bacteria (LAB) due to

obvious phylogenetic discriminations (Mattarelli et al., 2014). Most bifidobacteria colonize

and replicate only in anaerobic conditions even their sensitivity to oxygen may vary in

different strains (Shimamura et al., 1992). The characteristic of oxygen sensitive in

bifidobacteria is primarily proposed to be the production of hydrogen peroxide

(Kawasaki et al., 2009).

5

Table 1-1: Bifidobacterium sp. strains for which complete genome sequences are publically available

Gold stamp Organism Size (kb) GC Content (% )

Gc00470 B. adolescentis ATCC 15703 2090, 1709 orfs 59

Gc02229 B. animalis ATCC 25527 1933, 1597 orfs 60

Gc00888 B .animalis AD011 1934, 1587 orfs 60

Gc01215 B. animalis BB-12 1942, 1706 orfs 60

Gc0042985 B. animalis Bi-07 1939, 1661 orfs 60

Gc01943 B. animalis BLC1 1944, 1622 orfs 60

Gc00988 B. animalis Bl-04, ATCC SD5219 1939, 1631 orfs 60

Gc0051871 B. animalis Bl12 1939, 1607 orfs 60

Gc02353 B. animalis B420 1939, 1625 orfs 60

Gc01890 B. animalis CNCM I-2494 1943, 1724 orfs 60

Gc00987 B. animalis DSM 10140 1938, 1629 orfs 60

Gc0025103 B. animalis subsp. lactis ATCC 27673 1963, 1620 orfs 61

Gc01277 B. animalis V9 1944, 1636 orfs 60

Gc009812 B. asteroides PRL2011 2167, 1731 orfs 60

Gc01507 B. bifidum PRL2010 2215, 1767 orfs 63

Gc02297 B. bifidum BGN4 2224, 1903 orfs 63

Gc01417 B. bifidum S17 2187, 1845 orfs 63

Gc01767 B. breve ACS-071-V-Sch8b 2327, 1890 orfs 59

Gc0071448 B. breve JCM 7017 2289 58.7

Gc0071370 B. breve JCM 7019 2359, 1999 orfs 58.6

Gc0071449 B. breve NCFB 2258 2316 58.7

Gc0018521 B. breve S27 2294, 1898 orfs 59

Gc01883 B. breve UCC2003 (NCIMB8807) 2423, 1914 orfs 59

Gc0071444 B. breve 12L 2245 58.9

Gc0071447 B. breve 689b 2332 58.7

Gc01161 B. dentium Bd1 2636, 2197 orfs 59

(Date: 05/05/2014, available from http://www.genomesonline.org/cgi-b in/GOLD/Search.cg i)

6

Table 1-1 (continued): Bifidobacterium sp. strains for which complete genome sequences are publically

available

Gold stamp Organism Size (kb) GC Content (% )

Gc00886 B. longum ATCC 15697 2833, 2577 orfs 60

Gc01537 B. longum BBMN68 2266, 1870 orfs 60

Gc00811 B. longum DJO10A 2390, 2074 orfs 60

Gc01485 B. longum F8 2385, 1744 orfs 60

Gc01613 B. longum JCM 1222 2829, 2641 orfs 60

Gc01612 B. longum JCM 1217 2385, 2009 orfs 60

Gc01307 B. longum JDM301 2478, 2022 orfs 60

Gc01871 B. longum KACC 91563 2396, 2050 orfs 60

Gc00108 B. longum NCC2705 2260, 1805 orfs 60

Gc01611 B. longum 157F-NC 2409, 2070 orfs 60

Gc0043015 B. thermophilum RBL67 2292, 1904 orfs 60

(Date: 05/05/2014, available from http://www.genomesonline.org/cgi-b in/GOLD/Search.cg i)

Besides the already mentioned characteristics, probably the most interesting feature of

bifidobacteria is that some strains are proposed to have benefits to human health.

According to the official definition, probiotics are microorganisms, which when

administered in adequate amounts confer a health benefit on the host (FAO/WHO, 2001).

Recently, an expert consensus statement confirmed the term of probiotics with minor

grammatical correction and defined the scope as well as appropriate use of probiotics (Hill

et al., 2014). Therefore, some well-studied bifidobacteria strains are entitled as probiotics

after a series of screens. Considering the long application history of these strains, they are

entered into the International Diary Federation (IDF) and European Food and Feed Culture

Association (EFFCA) joint inventory as Microbial Food Cultures (MFC). Most of these

MFC are used qualified presumption of safety (QPS), which is managed by the European

Food Safety Authority (EFSA) (see the updated authorized lists in Table 1-2).

7

Table 1-2: Bifidobacterium sp. authorized by the EFS A used as probiotics in Europe

MFC (IDF-EFFCA, 2012)a QPS (EFS A, 09/2013 updated)

b

B. adolescentis B. adolescentis

B. animalis B. animalis

B. bifidum B. bifidum

B. breve B. breve

B. longum B. longum

B. pseudolongum subsp. pseudolongum

B. thermophilum

MFC, Microbial Food Cultures

IDF-EFFCA, International Diary Federation and European Food and Feed Culture Association

QPS, Qualified Presumption of Safety

EFSA, European Food Safety Authority

a http://www.effca.org/content/inventory-microorganisms

b http://www.efsa.europa.eu/en/efsajournal/doc/3449.pdf

The reported benefits of probiotic bifidobacteria are mainly ameliorations of

gastrointestinal disorders including bacterial or viral induced gastroenteritis, antibiotic-

associated diarrhea, lactose intolerance, constipation, irritable bowel syndrome, and

inflammatory bowel disease (Guyonnet et al., 2007; Guglielmetti et al., 2011; Whelan and

Quigley 2013). Several in vitro or in vivo experimental models provide hints towards

possible explanations for these benefits, such as inhibition of adhesion (Ewaschuk et al.,

2008), immunomodulatory effects (Park et al., 2007; Zhang et al., 2010), production of

antimicrobial substances (Wu et al., 2010), and restoration of the microecology (Relman

2013). Other applications of probiotics are prevention of infectious diseases (Petschow et

al., 2013), obesity (An et al., 2011; Chen et al., 2012), and allergy (Dev et al., 2008).

Despite promising results in animal models, these evidences are not solid enough to

convince related administrative authorities (Katan 2012). For example, the EFSA demands

more systematic studies and well conducted randomized controlled trials to evidence the

efficacy and safety for any health related claim of probiotics (Salminen and van Loveren

2012; Mego et al., 2013; Passariello et al., 2014). Increasing interest of bifidobacteria is

their potential application in cancer gene therapy that will be discussed in one following

part separately.

8

1.2 Characteristics of Bifidobacterium bifidum S17

Even there are 48 species of bifidobacteria and many strains belong to different species are

termed as probiotics, specific benefits or characteristics are frequently strain dependent.

One good example is B. bifidum S17, an isolate from feces of a breast-fed infant. This

strain had been sequenced and studied in detail in our lab. The complete genomic

information of B. bifidum S17 is contained on a single circular chromosome of ~2.18 Mb

with an average GC content of 62% (NCBI accession number: CP002220). A total of 1 782

open reading frames (ORFs), 53 tRNA genes and 3 rrn operons were identified (Zhurina et

al., 2011). Completely annotated genome supplied many interesting targets, like a

potential adhesin BopA (encoded by bbif_0636) and the S-ribosylhomocysteinase LuxS

(encoded by bbif_1299). One study suggested that BopA could improve adhesion of B.

bifidum S17 to intestinal epithelial cells (IECs) as a potential adhesin (Gleinser et al.,

2012). However, this result is questioned by another study that concluded that BopA does

not have a major role in adhesion (Kainulainen et al., 2013). LuxS is responsible for the

production of signal molecules, autoinducer-2 (AI-2) in B. bifidum S17 (Sun et al., 2014).

This raises the question whether S17 can perform LuxS/AI-2 dependent quorum sensing

(QS), an important subject for understanding bifidobacteria inter or cross-species signal

communication. Recently, many fluorescent proteins were expressed in B. bifidum S17 that

provides useful tools for in vivo studies (Grimm et al., 2014).

Along with these molecular studies, many physiological studies were conducted to

investigate potential probiotic properties of B. bifidum S17 as well. B. bifidum S17 does

not induce inflammatory events on HT-29 cells in vitro, but almost completely inhibits

LPS-induced NF-κB activation. Meanwhile, pre-treatment with B. bifidum S17

significantly inhibits transcriptional activation of gene IL-8, TNF-α, COX-2, and ICAM-1

in HT-29 cells challenged with LPS in the presence of 50 ml/l human milk (Riedel et al.,

2006a; Riedel et al., 2006b). These results indicate B. bifidum S17 might be a promising

candidate for probiotic intervention of chronic intestinal inflammation. Furthermore, in line

with above in vitro data, a study confirmed the anti- inflammatory capacity of B. bifidum

S17 for amelioration of murine colitis in vivo and this strain can prevent weight loss in

mice (Philippe et al., 2011). Meanwhile, B. bifidum S17 can adhere to three different IECs

9

better than other tested strains (Preising et al., 2010). This property is extremely attractive

when considering the usage of bifidobacteria as mucosal vaccination vehicles.

1.3 Expression systems for bifidobacteria

1.3.1 Advantages of bifidobacteria as hosts

Due to their probiotic properties, bifidobacteria are promising hosts for expression of

foreign genes for both research and application. Since the status of non-pathogenic or even

probiotic, they are highly acceptable for the production of pharmaceutical proteins (Kullen

and Klaenhammer 2000). Furthermore, the strict anaerobic growth makes them attractive

for cancer therapy as they were shown to specifically translocate to and actively replicate

in hypoxic regions of solid tumors (Yazawa et al., 2000; Fujimori 2006; Cronin et al.,

2010). Thus, there are already several studies using recombinant bifidobacteria as gene

delivery vectors for enzyme/pro-drug therapy (Nakamra et al., 2002; Hamaji et al., 2007;

Hidaka et al., 2007). Moreover, bifidobacteria can induce effective mucosal immunity.

Therefore, recombinant bifidobacteria may be used as live oral vaccines for prevention of

infectious diseases (Takata et al., 2006; Ma et al., 2011). There are other reports use

bifidobacteria as hosts for production of cytokines with the aim at enhancing their

functional properties (Reyes Escogido et al., 2007; Shkoporov et al., 2008).

1.3.2 Plasmids isolated from bifidobacteria

Bacterial expression systems are largely based on the isolation and construction of

plasmids. In 1982, the presence of plasmids in the genus Bifidobacterium was first reported

(Sgorbati et al., 1982). The existence and isolation of plasmids paved the way for further

molecular study of bifidobacteria as they are indispensable tools for molecular

manipulation. According to a recent review, 34 plasmids originated from Bifidobacterium

sp. have been completely sequenced (Sun et al., 2012). More than half of them were

isolated from B. longum, with a GC content ranging from 59.0% to 66.2%. Although many

plasmids were structurally characterized in bifidobacteria, their significance remains

largely unknown, as no obvious phenotypic traits associated with their presence. One

10

exception is a B. bifidum plasmid that was reported to be responsible for production of a

bacteriocin termed bifidocin B (Yildirim et al., 1999). Some of the native bifidobacterial

plasmids or their replicons were exploited for the construction of Escherichia coli (E. coli)

- Bifidobacterium sp. shuttle vectors aiming to overcome the lack of molecular tools in

bifidobacteria (Lee and O’Sullivan 2006).

The first characterized bifidobacterial plasmid was pMB1 isolated from B. longum. With a

size of 1 847 base pair (bp), it contains repA and repB genes (Matteuzzi et al., 1990; Rossi

et al., 1996). Its replicon was used in a number of shuttle vectors, which were successfully

transformed into B. adolescentis and B. longum strains (Li et al., 2003; Xu et al., 2007).

Another frequently used plasmid is pBC1 isolated from B. catenulatum, which encodes a

repB protein with high similarity to repB of pMB1 (Alvarez-Martin et al., 2007). pBC1

based plasmids were used in expression vectors and were able to replicate in B. breve, B.

pseudocatenulatum, and B. longum subsp. infantis strains (Cronin et al., 2008; Alvarez-

Martinet al., 2008; Ma et al., 2011). Both pMB1 and pBC1 replicate via theta replication

as no accumulation of single strand intermediate and containing corresponding theta type

replicase domain (PF03090) (Alvarez-Martin et al., 2007). Plasmid pTB6, pB44, and

pMG1, all isolated from B. longum strains, are widely used in shuttle vector construction as

well. pTB6 replicates in different strains of B. longum, B. animalis, and B. breve with

highest segregational stability and expression of target genes in B. breve (Hidaka et al.,

2007; Hamaji et al., 2007; Yamamoto et al., 2010). pB44 was used for expression of

secreted fibroblast growth factor (FGF-2) in B. breve UCC2003 and yielded lower

productivity compared to the pB80 replicon (Shkoporov et al., 2008). pMG1 was

exclusively used for expression of various target genes in its native host B. longum MG1

(Moon et al., 2005; Rhim et al., 2006; Park et al., 2008a). These plasmids may replicate by

a rolling circle mechanism (RCM) as single strand intermediates were observed for some

of them with close phylogenetic relationship between their replicases (O'Riordan and

Fitzgerald 1999; Park et al., 1999; Park et al., 2003; Park et al., 2008b).

1.3.3 Current study of gene expression in bifidobacteria

Using the above-mentioned plasmids and their replicons, a variety of E. coli -

Bifidobacterium sp. shuttle and expression vectors have been generated. Based on these

11

vectors, heterologous expression of a variety of proteins has been achieved in different

strains of bifidobacteria (Sun et al., 2012). However, the genetic features that determine the

efficiency of expression have not been characterized in detail. The very few reports that

comment on expression levels use indirect measurements of expression by western blot or

functional analysis, suggesting that expression levels are very low.

1.3.4 Promoters

Promoters are DNA elements that constitute binding sites for the RNA polymerase (RNAP)

holoenzyme and transcriptional regulators or other regulatory elements thereby

initiating transcription of downstream genes (Ross et al., 1993). They are usually located

upstream of genes near the transcription start sites (TSS) (Estrem et al., 1999). Due to their

important role in initiation of transcription, promoters are considered as traffic lights in

biotechnology (Patek et al., 2013). Many studies have attempted to identify strong

constitutive promoters driving high- level expression of genes of interest in various

microorganisms including bifidobacteria (Nesvera et al., 2012; Wang et al., 2012).

Constitutive promoters are active in all circumstances and constantly drive transcription

without any regulatory mechanisms. For most biotechnological applications, stable and

efficient production of proteins and simple fermentation processes are considered desirable.

The majority promoters studied for driving gene expression in bifidobacteria are

summarized in table 1-3. The first promoter used for expression of target genes in

bifidobacteria is the promoter region upstream of the hup gene encoding histone- like

DNA-binding protein HU. The cloned DNA fragment containing the hup promoter region

was shown to drive constitutive expression in B. longum (Takeuchi et al., 2002) and was

used for expression of the E. coli cytosine deaminase (CD) for tumors therapy (Nakamura

et al., 2002). The same promoter region was used for expression of Salmonella flagellin

(FliC) in B. animalis ATCC 27536 and B. longum A-105 (Takata et al., 2006; Yamamoto

et al., 2010), and FGF-2 in B. breve UCC2003 (Shkoporov et al., 2008).

12

Table 1-3: Promoters applied for gene expression in bifidobacteria

Promoter Description Target gene Expression host

Phup Promoter of B. longum hup gene cd, fg f-2, IL-10, FliC B. longum,

B. breve,

B. animalis

Pgap Promoter of B. longum gap gene gusA, fgf-2, IL-10,

KatG, SOD

B. longum,

B. breve

PRPL Promoter of bacterial λ phage Endostatin B. adolescentis,

B. longum

P16S rRNA Promoter of B. longum 16S rRNA gusA,

Cholesterol oxidase

Beta-glucosidase

B. breve,

B. longum

B. bifidum

Pamy Promoter of B. longum amylase Pediocin PA -1, CfaB , LTB B. longum (infantis)

Phelp Consensus high expression Listerial

promoter

LuxABCD B. breve

PbetA Promoter of B. longum betA gene, bile

inducible

arabinofuranosidase B. longum,

B. breve,

B. adolescentis,

B. pseudocatenulatum

PbfeUOB Promoter of B. breve bfeUOB operon,

iron inducible

gusA B. breve

ParaBCD Promoter of E. coli araBCD operon,

arabinose inducible

Oxyntmodulin,

IL-10, IL-12

B. longum

Besides the hup promoter region, a number of other bifidobacterial promoters were also

studied. For example, the promoter region together with the signal sequence of the α-

amylase gene of B. adolescentis INT-57 was successfully used for production and secretion

of pediocin PA-1, a class IIa bacteriocin, by B. longum MG1 (Moon et al., 2005). The 16S

rRNA promoter region of B. longum MG1 was used for expression of the E. coli gusA in B.

breve NCIMB 8807 (Sangrador-Vegas et al., 2007), a cholesterol oxidase in B. longum

MG1 (Park et al., 2008a) and three potential beta-glucosidases in B. bifidum BGN4 (Kim

et al., 2012).

Another promoter region widely used in bifidobacteria is the DNA sequence upstream of

the glyceraldehyde-3-phosphate dehydrogenase gene (gap). Microarray data suggests that

13

gap is a housekeeping gene highly expressed in B. longum NCC2705 and thus its promoter

can drive high- level expression. The corresponding DNA sequence was used to drive the

gusA reporter in pMDY23 as a suitable promoter test system (Klijn et al., 2006). Using the

same promoter, levels of FGF-2 expression were achieved comparable to the hup promoter

(Shkoporov et al., 2008). Moreover, a recent paper used this promoter for driving

overexpression of two genes resulting in improved resistance to oxidative stress (Zuo et al.,

2014).

Besides all these originated from bifidobacteria themselves, other promoters (cloned from

other microorganisms or artificially designed) may break the restriction of species and

strength limit of self-driving. At present, two examples can be tracked (do not include

inducible promoter). One is the high expression listerial promoter (help), which was

constructed for high level expression of the luxABCDE operon in Listeria monocytogenes

(Riedel et al., 2007) and subsequently used for the same purpose in B. breve UCC2003

(Cronin et al., 2008). The other is λPRPL, a phage hybrid promoter, which was shown to

drive expression of endostatin in B. adolescentis (Li et al., 2003; Xu et al., 2007).

Promoters for regulated/inducible expression have been successfully established in E. coli

and a wide range of other bacteria, such as the T7 promoter (Tabor and Richardson 1985),

the Lac promoter (Duong et al., 2011), and the Tet promoter (Debowski et al., 2013).

However, there is no good system for regulated expression in bifidobacteria so far. In

many cases, promoters from bifidobacteria are recognized for driving recombinant protein

expression in E. coli (Rhim et al., 2006; Gleinser et al., 2012). This suggests that the

structural consensus of some promoters and their binding sites are consistent between

Bifidobacterium sp. and E. coli. However, most regulated promoters developed in E. coli

have not been effectively used in Gram-positive bacteria in general and bifidobacteria in

particular (Patek et al., 2013; Sun et al., 2012). One exception is the E. coli araBAD

system which was used for arabinose- inducible expression of several targets including

oxyntmodulin, interleukin 10 (IL-10), and IL-12 in B. longum (Long et al., 2010; Yao et al.,

2011; Yu et al., 2011). However, the use of this system in bifidobacteria is limited since

various species of bifidobacteria including B. bifidum S17 do not harbor genes for

arabinose transporters.

14

A starting point for the generation of inducible systems for bifidobacteria is the

identification of bile- and iron- inducible genes or operons. The first inducible gene

characterized in bifidobacteria was betA of B. longum NCC2705, which is highly up

regulated in the presence of bile (Ruiz et al., 2011). A DNA fragment of 469 bp

immediately upstream of betA was found to be the smallest unit that retains bile- inducible

expression. A second study, assayed the promoter region upstream of bfeUOB, a presumed

ferrous iron uptake system of B. breve UCC2003 and found a 2-5 folds increase of reporter

activity with multi-copy plasmid (Cronin et al., 2012). These promoters might be useful for

physiological study but be of limited potential on regulating heterologous gene expression

as they can be strongly affected by other environment factors and their inducible capacity

is very limited.

In summary, there are a number of promoters for expression in bifidobacteria. However, to

date none of these promoters was experimentally characterized and limited information is

available on conserved promoter elements (-10, -35 regions, and ribosome binding site

(RBS)) of bifidobacteria.

1.3.4 Secretion

To perform the intended function properly and to achieve maximal efficacy, it is essential

to ensure that recombinant proteins are expressed at the correct location of a bacterial cell.

In case of extracellular proteins, secretion is necessary for correct folding. Certain

extracellular components produced by probiotic bacteria may be responsible for some

benefits that they exerted on host (Sanchez et al., 2010). Moreover, secretion might be

advantageous in high- level production of recombinant proteins to avoid inclusion body

formation and facilitate purification from the supernatant omitting harvesting strategies

that involve cell disruption (Georgiou and Segatori 2005). Meanwhile, recombinant

bifidobacteria may increase the commercial value of functional foods by secreted

substances, e.g. secondary metabolites or primary proteins/enzymes (Guglielmetti et al.,

2013). For bifidobacteria tumor therapy, specifically, live cells with safe vectors can

actively translocate to hypoxia and necrotic regions. Therefore, the secretion of protein

drugs in these areas may significantly improve the therapeutic effect.

15

In an early study, a genomic library of B. breve UCC2003 strain was screened for

secretion signals using an export-specific nuclease reporter approach and a few putative

signal peptides were identified (MacConail et al., 2001). These identified bifidobacteria

signal peptides contain the AxA consensus cleavage site. Analysis of several

bifidobacterial genomes indicates that these bifidobacteria strains have most

components of the general secretary pathway (Sec) commonly found in Gram-positive

bacteria (Pallen et al., 2003). In contrast, components of the twin arginine translocation

(Tat) secretion machinery are absent in most of the analyzed strains

(http://www.genome.jp/kegg-bin/show_pathway).

Secretion of recombinant proteins by bifidobacteria was first described for fusion of the α-

amylase signal peptide of B. adolescentis INT-57 to the Pediococcus sp. class II

bacteriocin pediocin PA-1. The resulting construct was successfully expressed in B.

longum and found in the culture supernatant (Moon et al., 2005). Other examples are

pB44- and pB80-derived vectors carrying a Sec2 signal peptide-encoding sequence of B.

breve. Functionality of these constructs was tested using human fgf-2 gene (Shkoporov et

al., 2008). Secretion of FGF-2 was verified by Western blot using precipitated supernatants

of the B. breve engineered strains. A series of vectors were constructed using the signal

peptides of protein AmyB of B. breve and ApuB of B. adolescentis for expression and

secretion of recombinant human IL-10 (Khokhlova et al., 2010). Interestingly, the ApuB

signal peptide increased IL-10 secretion in culture medium compared to the Sec2 signal

peptide. Nevertheless, knowledge on protein secretion and secretion signal sequences in

bifidobacteria is still limited.

1.4 Bifidobacterial cancer therapy

A wide range of bacterial species/strains was tested for their antitumor potential, including

Caulobacter, Listeria, Clostridium, Salmonella, Escherichia, Bifidobacterium, Proteus,

and Streptococcus (Forbes 2010). Among them, Salmonella and Clostridium are the most

well studied bacteria for tumor therapy in different mouse models including breast,

pancreatic, prostate, and colon cancer. In theory, bacteria can be utilized for cancer therapy

by either their intrinsic toxicity or by production of antitumor reagents. Spores of

Clostridium sp. have been studied extensively as tumor targeting vectors with more or less

16

encouraging results (Fox et al., 1996; Agrawal et al., 2004; Maletzki et al., 2010). The

major problem of establishing bacteria as tumor therapeutics is safety issues. For some

strains of Salmonella typhimurium, high doses of bacteria are required for therapeutic

efficacy, which is associated with a high risk of systemic toxicity (Dang et al., 2001;

Palumbo et al., 2013). Thus, the use of genetically engineered, non-pathogenic bacteria as

delivery vehicles has come into focus as this approach can overcome the adverse effect.

One representative strain is a virulence attenuated Salmonella typhimurium VNP20009

(Luo et al., 2001). This strain had already been tested in clinical trials but shown limited

efficiency (Toso et al., 2002). Another group of bacteria that has been considered for

cancer therapy is bifidobacteria. These microorganisms have a very good safety record and

were shown can specifically target the hypoxic region of solid tumors in mice (Tangney

2010). Native bifidobacteria have a limited potential to induce tumor regression or inhibit

tumor growth (Kohwi et al., 1978). In consequence, genetic modification of bifidobacteria

is critical for potential tumor therapy (Baban et al., 2010).

One widely used approach for cancer therapy using bacteria as gene delivery vectors is

gene directed enzyme pro-drug therapy (GDEPT, Cheng et al., 2008). In this concept,

therapy is based on expression of an enzyme by a bacterial of viral delivery system

specifically in tumor tissue. This ensures that conversion of a pro-drug that is administered

to the patient systemically is confined to its intended site of action thereby limiting side

effects and increasing efficacy (Chen et al., 2004; Djeha et al., 2005; Mitchell and Minchin

2008; Warmann et al., 2009). Several enzyme/pro-drug systems were frequently reported.

These include cytosine deaminase/5-fluorocytosine (CD/5-FC, Huber et al., 1994), purine

nucleoside phosphorylase/6-Methylpurine-deoxyriboside (MePdR/PNP, Hughes et al.,

1995; Stritzker et al., 2008), β-glucuronidase/9-aminocamptothecin glucuronide (βG/9-

ACG, Cheng et al., 2008), nitroreductase/5-aziridin- l-yl-2,4-dinitrobenzamide

(NTR/CB1954, Barak et al., 2006; Vass et al., 2009); and herpes simplex virus thymidine

kinase/ganciclovir (HSV-TK/GCV, Immonen et al., 2004). Some of these systems have

been used for generation of recombinant bifidobacteria for tumor therapy (Yi et al., 2005;

Tang et al., 2009; Yin et al. , 2013). However, most of these genes are expressed at unknown

level and intracellularly. Thus, there is a need for further development of more efficient

bifidobacterial expression and secretion systems to enhance the therapeutic effect.

17

1.5 The aim of this study

The aim of this study was to establish a system for efficient expression and secretion of a

pro-drug converting enzyme and to use this system engineering a recombinant

Bifidobacterium sp. strain as a gene delivery vector for cancer therapy. This goal was

approached in three consecutive parts: (i) identification and characterization of a strong

constitutive promoter for high- level expression, (ii) screening of a sorting signal for

efficient secretion of proteins, and (iii) construction of a recombinant strain of B. bifidum

S17 that produces and secretes a pro-drug converting enzyme.

18

2 MATERIALS AND METHODS

2.1 Bacterial strains and plasmids

2.1.1 Bacterial strains

All bacterial strains used in this study were listed in appendices (Table 7-1). Storage and

propagation of these strains were conducted according to standard operation procedures.

For long term storage, 10 ml of a culture incubated for 16 h were harvested by

centrifugation and the bacterial pellet was resuspended in 0.5 ml fresh sterile medium,

mixed with an equal volume of 30% sterilized glycerol and store at -80°C. For short-term

storage, 0.5 ml of an overnight culture was mixed with 0.5 ml 30% glycerol and store at -

20°C for up to 3 months.

E. coli strains used in this study are E. coli DH5 and E. coli DH10B. Both strains were

routinely cultivated in Luria-Bertani (LB) broth (10 g/l peptone, 5 g/l yeast extract, 5 g/l

sodium chloride) or LB agar (additionally 12 g/l agar) at 37 °C. Medium was supplemented

with 100 μg/ml spectinomycin (Spc) when necessary. For liquid culture, glass tubes

containing 5 ml LB were agitated at 170 revolutions per minute (rpm); for solid culture, 9

cm agar plates were placed in an incubator.

Bifidobacteria were routinely grown by inoculating 100 μl of a glycerol stock into 10 ml

pre-boiled MRSc (Lactobacilli MRS medium (BD DifcoTM, USA) supplemented with 0.5

g/l L-cysteine HCl) and grown at 37°C overnight (16-24 h) under anaerobic conditions.

Anaerobic conditions were created by placing CO2 producing AnaeroGenTM sachets (Oxoid,

Germany) in a sealed jar (Anaerocult, Merck, Germany). For growth experiments,

overnight pre-cultures were used to inoculate fresh medium to an initial optical density at

600nm (OD600) of 0.1. Medium was supplemented with 100 μg/ml Spc where appropriate.

For solid media, 12 g/l agar was added.

19

2.1.2 Plasmids and oligonucleotides

All plasmids used in this study were listed in appendices (Table 7-2). Plasmids were

routinely maintained in and isolated from E. coli DH10B grown in LB broth with 100

μg/ml Spc. Pure plasmid DNA was dissolved in 10 mM Tris-HCl, pH8.5, and was stored at

-20°C.

All oligonucleotides used in this study were listed in appendices (Table 7-3). All primers

were designed with primer3plus (http://primer3plus.com/cgi-bin/dev/primer3plus.cgi)

using public available genome sequences (www.ncbi.nlm.nih.gov), and synthesized a t high

purity salt free (HPSF) grade by a commercial supplier (Eurofins MWG, Germany). With

the exception of the GSPR primers, which were dissolved in nuclease- free water, all other

primers were dissolved in double distilled water (ddH2O) at a concentration of 100 μM

(stocks) and stored at -20°C. For frequently used primers, stocks were mixed to

corresponding primer pairs at a final concentration of 10 μM (each oligonucleotide) and

stored at 4°C.

2.2 Sample preparation and purification

2.2.1 Preparation of protein samples

Sample preparation for β-glucuronidase assays

Bacterial cells were harvested at different time points by centrifugation (4 226 relative

centrifuge force (rcf), 10 min, 4°C) of 10 ml cultures grown in MRSc containing 100 μg/ml

Spc under anaerobic condition. Cell pellets were washed with ddH2O twice and then

resuspended in 1 ml GUS buffer (50 mM phosphate sodium, pH7.0; 1 mM

ethylenediaminetetraacetic acid (EDTA); 5 mM dithiothreitol (DTT), 0.1% Triton X-100).

The suspensions were transferred to 2 ml screw cap tubes containing 250 μl glass beads

(diameter 0.1 mm) and bacteria were disrupted in a Precellys24 (Peqlab, Germany) during

2 cycles of 35 s at 6 500 rpm). Cell crude extract was prepared by collection of the

supernatant after centrifugation (17 949 rcf, 15 min, 4°C). The cell crude extract containing

glucuronidase was stored at 4°C and enzymatic assays were conducted within three days.

20

Total protein in the cell crude extract was measured by a 2-D Quant protein assay kit (see

2.6.2).

Sample preparation for phytase assays

For phytase assays, special attention is required to deplete potential phosphate

contaminations. All reagents and media were prepared in ultrapure water (18 MΩ·cm H2O,

Millipore, USA). Supernatants and cell pellets containing phytase protein were collected at

different time points by centrifugation (17 949 rcf, 5 min, 4°C) of recombinant B. bifidum

S17 strains grown in reinforced clostridia medium (RCM, Oxoid) containing 100 μg/ml

Spc under standard conditions. Bacterial pellets were rinsed twice in 0.2 M sodium citrate

buffer, pH5.5, and then resuspended in 500 μl of the same buffer for disruption in

Precellys24 as described above. Both supernatant (containing extracellular phytase) and

cell lysate (containing intracellular phytase) were stored at 4°C and phytase assays were

conducted within one week. Total protein in cell lysate was quantified by a BCA protein

assay kit (see 2.6.1).

Sample preparation for nitroreductase assays

For nitroreductase assays, supernatants and bacterial pellets of recombinant B. bifidum S17

strains were collected by centrifugation (17 949 rcf, 5 min, 4°C) after growth for 16 h in 10

ml RCM containing 100 μg/ml Spc under standard condition. Pellets were rinsed twice in

10 mM Tris-HCl buffer, pH7.0, and then resuspended in 500 μl same buffer disrupted as

described above. Both supernatant (containing extracellular nitroreductase) and cell lysate

(containing intracellular nitroreductase) were stored at 4°C and enzymatic assays were

conducted within three days. Total protein in cell lysate was quantified by a BCA protein

assay kit (see 2.6.2).

2.2.2 Preparation of nucleic acids

Preparation of genomic DNA

Template DNA for PCR cloning of target DNA fragments, such as promoter, reporter, and

SP, was prepared by centrifugation (14 549 rcf, 5 min) of 1 ml culture fluid. The pellet was

rinsed twice in 1 ml ddH2O, and then resuspended in 100 μl ddH2O. After heat treatment at

21

98°C for 10 min, cell debris was spin down (14 549 rcf for 5 min). Supernatant containing

genomic DNA was stored at -20°C for up to 3 month and used as template DNA for PCR.

Preparation of plasmid DNA

Plasmid DNA was prepared based on the principle of alkaline-sodium dodecyl sulfate

(SDS) lysis and ethanol precipitation, using E.Z.N.A. Plasmid Mini Kit I according to

manufacturer’s guideline (Omega, USA). 2 ml overnight culture of recombinant E. coli

DH10B was collected for plasmid isolation. Elution of DNA from the columns of the kit

was generally performed in 40 μl elution buffer (10 mM Tris-HCl, pH 8.5). After elution, 2

μl plasmids were resolved on 1% agarose to estimate its size and another 2 μl were used for

quantification (see 2.6.3). Plasmids were stored at -20°C for further application.

Preparation of total RNA

Total RNA of bifidobacteria was prepared as described previously after phenol-chloroform

extraction of proteins from crude extracts obtained by mechanical disruption as described

above (Gleinser et al., 2012). 10 ml of an overnight culture were mixed with 40 ml

quenching buffer (mixture of 16 ml 66.7 mM HEPES pH 6.5 and 24 ml 100% methanol) in

a 50 ml falcon tube and stored at -80°C for 30 min. Bacteria were harvested by

centrifugation (4 226 rcf, 20 min, 4°C) and resuspended in 200 μ l ddH2O. The suspension

was transferred into screw cap tubes filled with 0.5 ml glass beads (diameter 0.1 mm), 30

µl sodium acetate (3 M, pH 5.2), and 30 µl SDS (10% weight/volume (w/v)). Then, 400 µl

phenol and 100 µl chloroform were added. Cells were disrupted by spinning the tubes in a

Precellys24 for 2 cycles at 6500 rpm for 35 s. Aqueous and organic phases were formed by

centrifugation (17 949 rcf, 15min, 4°C). The aqueous phase was carefully transferred to a

sterile 1.5 ml Eppendorf (EP) tube and mixed with 1.5 volumes of 100% ethanol. The

mixture was incubated at -20°C over night for precipitation. The next day, precipitated

samples were centrifuged and washed with 70% ethanol (17 949 rcf, 30 min, 4°C). RNA

pellets were air dried by placing the tube under the airflow in a clean cabinet for 15-20 min.

Dried pellets were dissolved in 200 μ l 65°C pre-warmed nuclease-free water. Then,

dissolved RNA samples were treated with 200 U DNase I in 1 × DNase I reaction buffer at

37°C for 60 min (Thermo Scientific, Germany). DNase I was denatured by adding 15 µl 50

mM EDTA and incubation at 65°C for 10 min. 8 μ l of DNAse I-treated sample were

analyzed by agarose gel electrophoresis (see 2.3.4) and RNA concentration was quantified

22

(see 2.6.3). RNA samples were used immediately for downstream applications or stored at

-80°C until use.

2.2.3 Purification of nucleic acid samples

DNA fragments produced by PCR or enzymatic restriction were purified by using

NucleoSpin Extract II Kit according to the user manual (Macherey-Nagel, Germany). For

isolation of vector DNA or PCR products from agarose gels, the corresponding bands were

excised from the gel after electrophoresis, weighted, dissolved in two volumes (w/v) NTI

buffer at 50°C for 5 min, and then purified according to the Gel Extract protocol. To verify

successful purification of target DNA, samples were analyzed by agarose gel

electrophoresis (see 2.3.4) and quantified spectrophotometrically (see 2.6.3).

Ligation products

To achieve efficient transformation, ligation mixes were concentrated by ethanol

precipitation. Briefly, 10-20 μl of a ligation reaction were mixed with 10%

(volume/volume, v/v) 3 M sodium acetate, pH5.2 and precipitated by 2.5 volumes 98%

ethanol at -20°C for 1 h. Precipitated DNA was collected by centrifugation (14 549 rcf, 10

min) and washed twice with 70% ethanol. Then, DNA pellet was air dried by incubation at

37°C for 15 min and the dried pellet was dissolved in 2-5 μl 65°C pre-warmed ddH2O.

RNA

RNA samples were purified using RNeasy Mini Kit (Qiagen, Germany) according the

manufacturer’s instructions. Briefly, samples were mixed with an equal volume of 70%

ethanol and loaded onto an RNeasy Mini spin column by centrifugation. Columns were

rinsed twice with RPE buffer and RNA eluted in 30 µl of RNase-free H2O with incubation

for 10 min before centrifugation.

23

2.3 Manipulations of nucleic acids

2.3.1 Conventional polymerase chain reaction

Polymerase chain reaction (PCR) is a basic technique used for amplification of specific

DNA fragments (Bartlett et al., 2003). In this study, PCR was employed for general

cloning of target genes or screening of transformants for cloned inserts. Phusion DNA

polymerase was used for amplification according to supplier’s instruction (Thermo

Scientific, Germany). The composition of a typical PCR reaction was:

Template DNA

1-100 ng

Primer mixture 50-500 nM

dNTPs 200 nM

5 × HF buffer (containing 7.5mM MgCl2) 1 ×

Phusion DNA polymerase

0.02 U/μl

*DMSO 0-10%

*optional, DMSO was added only for amplification of complex templates

Thermo cycling was performed on a FlexCycler (Analytik Jena, Germany) by pre-

denaturation at 98°C for 2 min followed by 25-35 cycles of denaturation (95°C for 10 s),

annealing (50-70°C for 10 s), and elongation (68-72°C for 30-60 s), and a final elongation

(72°C for 5 min). PCR products were stored at 4°C until use. Annealing temperature was

optimized for each primer pair.

2.3.2 Splicing by overlap extension PCR

Splicing by overlap extension PCR (SOE-PCR) is used to fuse two (or more) PCR

products without the use of restriction enzymes and DNA ligase. In a first round, PCR is

performed to generate two gene segments with overlapping ends introduced by internal

primers with complementary 5’ ends. Then, these two PCR products are used together as

template DNA for the second round PCR to create a full- length chimerical product. A

standard procedure for SOE-PCR was described (Heckman and Pease 2007). In both

24

rounds of PCR, composition of the SOE-PCR reaction and parameters for cycling was

identical to conventional PCR (see 2.3.1) with optimization of annealing temperature.

2.3.3 Rapid amplification of cDNA ends

The protocol used for rapid amplification of cDNA 5’-end (5’-RACE) was described

previously (Scotto-Lavino, et al., 2006). The 2-Generation RACE Kit (Roche, Switzerland),

which is based on this principle as shown in figure 2-1 (Fig. 2-1) was used. Briefly, first-

strand cDNA was synthesized from 2 μg total RNA using a gene-specific primer (GSPR1).

Synthesized single strand cDNA was purified with a High-Pure PCR Clean-up Kit (Roche,

Switzerland) before appending a homopolymeric tail using terminal deoxynucleotide

transferase (TdT) and deoxyadenosine triphosphate (dATP). The tailed cDNA was directly

amplified by PCR using the oligo dT-anchor primer and the target-specific primer GSPR3.

A second round of PCR was conducted with anchor primer and the target-specific primer

GSPR4 to increase specificity. The PCR products were resolved on 1.5 % (w/v) agarose

gel by electrophoresis. Specific target band was cut out of the ge l and purified from the

agarose gel as described above. Isolated DNA fragments were cloned into pJET1.2

(Thermo Scientific, Germany) and sequenced with pJet1_PF by Sanger sequencing (GACT

biotech, Germany).

25

Fig. 2-1: Schematic representation of the procedure for identification of transcriptional start sites by

5’-RACE including reverse transcription, cDNA purification, poly A appending, two rounds of PCR,

and blunt end cloning.

2.3.4 Agarose gel electrophoresis

Agarose gel electrophoresis is used to analyze DNA molecules for detection, size

determination, or purification. In this study, agarose concentrations of 1-3 % (w/v) were

used for resolving nucleic acid fragments with different sizes. Briefly, 1-3% agarose was

dissolved in 1× TAE buffer (40 mM Tris-HCl, 10 mM acetic acid, 1 mM EDTA, pH 8.0)

by heating in microwave. Gels were casted using standard electrophoresis apparatus

(Peqlab, Germany). Samples were mixed with 6 × loading dye (Thermo Scientific,

Germany) and loaded to the slots on gel. Electrophoresis was carried out at constant

voltage (100 V) for 30-60 min. When bromophenol blue in loading dye had migrated to

approximate 0.5 mm from the bottom of the gel, electrophoresis was terminated and

agarose gel was stained for 5-10 min in ethidium bromide staining solution (1 µg/ml in

dH2O). Gels were rinsed with distilled water (dH2O) to remove excess ethidium bromide

and captured under exposure to UV light (302 nm) with a photo documentation system

(MWG-Biotech, Germany). Under these conditions, DNA stained by ethidium bromide

26

produces bright orange fluorescence. For determination of the size of DNA fragments,

GeneRulerTM 1 kb DNA Ladder or 50 bp DNA Ladder (Thermo Scientific, Germany) was

run in a separate lane on every gel.

2.3.5 DNA restriction and ligation

DNA restriction is a method to produce complementary DNA ends between that

effectively ligation can be achieved. All restriction enzymes were purchased from Thermo

Scientific, Germany. In this study, DNA restriction was performed using two different

restriction enzymes simultaneously. For each combination of enzymes, optimal buffers

were selected according to the recommendation of the supplier

(http://www.thermoscientificbio.com/webtools/doubledigest/; Thermo Scientific,

Germany). Digestion was carried out at 37°C for 3-20 h in a total volume of 40 µl in sterile

PCR tubes. Typical digestion reaction was composed of:

DNA sample (vector or insert)

1-2 µg

Restriction enzyme 1 10 U

Restriction enzyme 2 10 U

Recommended compatible buffer

1 ×

Digested insert DNA fragments were purified using NucleoSpin II columns (Machery-

Nagel, Germany) according to the PCR Extract protocol. Digested Vector DNA was

purified from agarose gels. All recovered DNA was analyzed by agarose gel

electrophoresis. For ligation, vector and insert DNA were mixed at molar ratios between 1 :

3 and 1 : 10 in a total volume of 10-20 µl containing 5 U T4 DNA ligase and 1 × T4 DNA

ligase buffer (Thermo Scientific, Germany). For successful ligation, a minimum of 0.5

pmol insert (calculated, 660 pg/pmol) was always used. Ligation reaction was carried out

by incubation at 22°C for 15-60 min.

2.3.6 Transformation of bacteria

Transformation of E. coli DH10B

27

Competent cell of E. coli DH10B was prepared as described (Gonzales et al., 2013). E. coli

DH10B was transformed by electroporation of 50 µl competent cells and 2 µl ligation

mixtures in an ice-cold electroporation cuvette (electrode distance 1mm; Peqlab, Germany).

After incubation for 5 min on ice, electroporation was performed in a Gene Pulser XcellTM

system (Bio-Rad, Germany) with following settings: 1 500 V, 200 Ω, and 25 µF.

Immediately after the pulse, 950 µl ice cold LB broth were added followed by incubation

on ice for 2 min. Then, cell suspension was transferred into a sterile 1.5 ml EP tube and

incubated for 60 min at 37°C with agitation (150 rpm). Finally, bacteria were collected by

centrifugation and spread on two LB agar (corresponding to 100 and 900 µl of regenerated

bacterial suspension) supplemented with the appropriate antibiotic and incubated overnight

at 37°C aerobically.

Transformation of B. bifidum S17

For efficient transformation, B. bifidum S17 competent cells were always prepared freshly

as described elsewhere (Serafini et al., 2012). Briefly, an overnight pre-culture was used

to inoculate 100 ml pre-warmed MRSc supplemented with 16 % (w/v) Actilight® fructo-

oligosaccharides (Beghin Meiji, France) to an initial OD600 of 0.1. Cultures were grown

under standard conditions (anaerobic, 37°C) for 6-8 h to an OD600 of ~ 0.5. Bacteria were

harvested by centrifugation (4°C, 4 226 rcf, 10 min) and washed three times with ice cold

citrate sucrose buffer (CSB, 1 mM citric acid, 0.5 M sucrose, pH 5.8). After the final

washing step, the pellet was resuspended in 1 ml CSB and used immediately as competent

cells. An aliquot of 100 µl of competent cells were mixed with 2 µl plasmid (100-500 ng)

in an ice-cold electroporation cuvette (1 mm electrode distance; Peqlab, Germany) and

incubated on ice for 5-10 min for transformation. Bacteria and plasmid mixture were

pulsed in a Gene Pulser XcellTM system with the following settings: 1 750 V, 200 Ω, 25

µF. After the pulse, bacteria was resuspended in 900 µl ice cold RCM broth immediately

and kept on ice for 2 min. The bacterial suspension was transferred to a sterile 1.5 ml EP

tube and incubated for 2.5-3 h at 37°C under anaerobic condition. Regenerated bacteria

were centrifuged (14 549 rcf, 2 min), pellets resuspended in two aliquots corresponding to

100 and 900 µl of the regenerated suspension were spread on two RCM agar plates

supplemented with 100 µg/ml Spc and incubated for 48-72 h at 37 °C under anaerobic

conditions.

28

2.4 Enzymatic assays

2.4.1 β-glucuronidase assay

Promoter activities were assayed using the gusA reporter gene of pMDY23 (Klijn et al.,

2006). The method for determination of GusA β-glucuronidase activity is described

elsewhere (Grimm et al., 2014). Briefly, bacterial crude extract corresponding to 1-5 μg

total protein were mixed with 25 μL 4-Methylumbelliferyl-β-D-glucuronide (4-MUG)

substrate solution (5 mM stock) and brought to a total volume of 100 μl GUS buffer ( 50

mM disodium hydrogen phosphate pH 7, 1 mM EDTA, 0.1% Triton X-100, 5 mM

dithiothreitol (DTT)) in the wells on a 96-well plate. After incubation for 30 min at 37°C,

the reaction was stopped and 5 μl transferred into another 96 well plate containing 195 μl

stop buffer (0.2 M Sodium carbonate in ddH2O, pH 9.5). Fluorescence of the product 4-

methylumbelliferone (4-MU) was measured in triplicate using an Infinite M200 multimode

microplate reader (Tecan, Switzerland) with excitation at 388 nm and emission at 480 nm.

The β-glucuronidase activity was defined as fluorescence units per microgram protein

(FU/μg). B. bifidum S17 harboring the promoter- less pMDY23 plasmid was included as a

negative control.

2.4.2 Phytase assay

Phytase assay was adapted from the Phytex method described elsewhere (Kim and Lei

2005). Phytase can catalyze the release of inorganic phosphate from phytate. Free

phosphate reacts with ammonium molybdate and forms stable blue complex, which has a

specific absorbance. Briefly, 100 μl sample (supernatant or crude extract) were incubated

at 37°C for 5 min and then mixed with 100 μl 10.8 mM sodium phytate (50% phytic acid

diluted in 0.2 M sodium citrate buffer, Sigma, Switzerland). The reaction was carried out at

37°C for 15 min, and then stopped by adding 200 μl 15% tricholoroacetic acid (TCA).

After centrifugation at 14549 rcf for 2min, an aliquot of 20 μl was mixed with 480 μl 18

MΩ·cm H2O and 500 μl color solution (mix of 1M sulfuric acid, 2.5% (w/v) ammonium

molybdate in ddH2O, and 10% (w/v) ascorbic acid in ddH2O at a ratio of 3:1:1). The

mixture was incubated at 50°C for 15 min, and 100 μl were transferred to a transparent 96-

29

well plate (Thermo Scientific, Germany). Absorbance at 820 nm was measured in triplicate

using an Infinite M200 multimode microplate reader (Tecan, Switzerland). Phytase activity

was calculated according to a standard curve produced by a serial dilution of 9 mM

potassium dihydrogen phosphate in aqueous solution. One phytase unit (FTU) was defined

as the amount of enzyme that catalyzes the release of 1 μmol of inorganic phosphate per

minute from 5.4 mM sodium phytate under above conditions at 37°C. Enzyme activities

were expressed as FTU/ml in supernatant and FTU/mg protein in cell lysate.

2.4.3 Nitroreductase assay

Nitroredutase assay was performed with CB1954 as substrate and β-nicotinamide adenine

dinucleotide reduced (NADH) as cofactor (Sigma, Switzerland), as described elsewhere

(Race et al., 2007) with minor modifications. In brief, NADH was dissolved in 0.01N

sodium hydroxide to give a stock concentration of 5 mM and prevented from light.

CB1954 was dissolved in pure dimethylsulfoxide (DMSO, Merck, Germany) at a stock

concentration of 10 mM. Cofactor and substrate stocks were stored at 4°C and used within

one week. In a 1 ml reaction, 100 μl NADH stock (final 500 μM) and 10μl CB1954 stock

(final 100 μM) were mixed with 100 μl 100 mM Tris-HCl pH7.0 (final 10 mM)). Reaction

was started by adding 100 μl crude extract or 500 μl spent culture supernatant. Volumes

were brought to 1 ml with ddH2O and reactions were carried out for 10 min at room

temperature. Then absorbance was measured at 340 nm in a plastic cuvette with 10 mm

light path (Thermo Scientific, Germany) using a Nanophotometer (Implen, Germany). One

NTR unit (U) was defined as the amount of enzyme that catalyzes the conversion of 1

μmol NADH to NAD per minute under the described conditions. Enzyme activities were

expressed as mU/ml in culture supernatants or mU/mg protein in crude extracts.

2.5 Growth related assays

2.5.1 Bacterial growth experiments

Cell growth was monitored by reading OD600 spectrophotometrically using either an

Infinite M200 multimode microplate reader (Tecan, Switzerland) with 200 μl culture fluids

30

in a transparent 96-well plates (Nuc, Thermo Scientific, Germany) or a Nanophotometer

(Implen, Germany) with 1mL 1:10 diluted cultures in a plastic cuvette with 10 mm light

path (Thermo Scientific, Germany).

2.5.2 Phytate degradation assay

As a first indication for phytase secretion of recombinant B. bifidum S17 strains, a phytate

degradation assay was employed. Strains were cultured on a solid phytase screen medium

(PSM) using calcium phytate as substrate (Quan et al., 2001). For the PSM medium, RCM

agar containing 100 μg/ml Spc was supplemented with 0.5% calcium phytate. A defined

volume of 2 μl of the strains to be tested was spotted on the PSM agar and incubated

anaerobically at 37°C for 72 h in a sealed jar. Ability of recombinant strains to hydrolyze

calcium phytate is indicated by clear zones on the agar surrounding bacterial colonies.

2.5.3 NTR/CB1954 inhibition assay

An initial screen for expression of ChrR6 by recombinant B. bifidum S17 was performed

by a growth inhibition assay. Strains expressing NTR can convert CB1954 into the

cytotoxic drug, which inhibits cell growth. Briefly, recombinant bifidobacteria were grown

in 10 ml RCM broth containing 100 μg/ml Spc and different concentrations of CB1954.

After 24 h anaerobic incubation at 37°C, OD600 was assayed and compared. NTR

expressing strains showed significantly lower final OD600.

2.6 Quantification

2.6.1 BCA protein assay

For quantification of the protein in crude extracts used for phytase and NTR assays, BCA

Protein Assay Kit (Thermo Scientific, Germany) was used according to the manufacturer’s

instructions. The assay is based on bicinchoninic acid (BCA) for the colorimetric

quantification of total protein. It combines the reduction of Cu+2 by protein in alkaline

31

solution with colorimetric detection of Cu+1 using a reagent containing BCA. The purple

reaction product of this assay is formed by chelating of two molecules of BCA with one

Cu+1 ion. The absorbance of this water-soluble product directly correlates to protein

concentrations over a broad working range (linear range 20-2000 μg/ml). The method was

adapted small sample volumes (10-25 μl). Absorbance at 562 nm was measured for each

sample in triplicate using an Infinite M200 multimode microplate reader (Tecan,

Switzerland).

2.6.2 2-D Quant protein assay

For quantification of the protein in crude extracts used for glucuronidase assay, the 2-D

Quant Kit (GE Healthcare, Germany) was used according to the manufacturer’s

instructions. This kit is developed primarily to accurately quantify protein samples

prepared for two-dimensional electrophoresis as well as any sample that contains

substances interfering with common protein quantification. The procedure uses a

combination of 500 μl patented precipitant and 500 μl co-precipitant to precipitate up to 50

μg in a total volume of 1-50 μl protein sample while leaving interfering contaminants in

solution. Precipitated protein is then collected by centrifugation and resuspended in 100 μl

alkaline copper solution. 1 ml colorimetric agent that reacts with unbound cupric ions is

added. The color density is inversely related to the concentration of protein in the sample.

Since cupric ions bind to the polypeptide backbones of any protein without dependence on

amino acid composition, protein concentration can be accurately estimated by comparison

to a bovine serum albumin (BSA) standard curve. Absorbance was measured for each

sample in duplicate using an Infinite M200 multimode microplate reader at 480 nm (Tecan,

Switzerland).

2.6.3 DNA/RNA quantification

Nucleic acids were quantified by measuring absorbance at 230, 260, and 280 nm of 2 μl

samples spectrophotometrically using a quartz-based NanoQuant Plate and an Infinite

M200 multimode microplate reader (Tecan, Switzerland) and the DNA or RNA mode of

the Tecan i-control™ Software. Wells on the plate were rinsed for three times with ddH2O

32

every time before loading new samples or blanks. Blanking was always conducted for each

well individually twice.

2.7 Bioinformatic analysis

The prediction of core promoter regions of hup (bl_1798), 16S rRNA (blr_01), gap

(bbif_0612) and luxS (bbif_1299) was predicted using the BPROM online software tool

(http://linux1.softberry.com/BPROM) and Neural Network Promoter Prediction (NNPP,

www.fruitfly .org/seq_tools /promoter .html ) using complete inter-region sequences

upstream of the respective genes (for full sequences see appendices 7.4). Potential SPs

were identified in the N terminal 60 amino acid residues of deduced protein sequences

using SignalP4.0 (www.cbs.dtu.dk/services/SignalP/). Corresponding DNA sequences of

predicted SPs were listed in appendices 7.7. Proteins with predicted SPs were searched in

ePSORTb database for their annotation to check whether they are experimentally

confirmed as extracellular proteins. The 8 signal peptides cloned in this study were also

analyzed by TatP (http://www.cbs.dtu.dk/services/TatP/) for potential twin arginine

translocation motifs. To confirm correct open reading frames and to rule out mutations all

cloned constructs were sequenced and obtained sequences aligned online with the Basic

Local Alignment Search Tool (BLAST, www.ncbi.nlm.nih.gov/BLAST/). CLC workbench

(CLC Bio, Denmark) was used for local alignment of 5’-RACE sequences and

representative Pgap sequences (see appendices 7.5). Codon optimization for gene synthesis

was performed according to B. bifidum S17 codon usage table provided by a commercial

service provider (Eurofins MWG, Germany, see appendices 7.8).

33

3 RESULTS

3.1 Promoter prediction and cloning

The range of promoters tested included the promoter regions upstream of the gap gene

(bbif_0612) encoding glyceraldehyde-3-phosphate dehydrogenase (Pgap) and the luxS

(bbif_1299) gene for the methylthioadenosine/S-adenosyl-homocysteine (MTA/SAH)

nucleosidase LuxS (PluxS) of B. bifidum S17. Furthermore, the sequence upstream of the

gene bl_1798 for histone-like HU protein (Phup) and the sequence upstream (P16S rRNA) of

16S rRNA gene (blr_01) of B. longum NCC2705 were included. The intergenic regions

upstream of these four genes (for complete sequences see appendices 7.4) were used to

predict potential bacterial promoters using BPROM, which was developed for bacterial

promoter prediction based on the recognition of E. coli σ70 type sigma factors. This

analysis yielded single promoters with relatively high linear discrimination function (LDF)

in front of gap, hup, and 16S rRNA when the default unchangeable threshold value equals

0.2. By contrast, no predicted promoter was found in front of luxS.

Table 3-1: Predicted promoters core regions by BPROM

Gene *Core sequence (50bp, 5’3’) LDF Potential TF

gap GTACAGACATATTTGTTAGCGTTAACGAAATA

TGGCCGTTTTATGCTCAA

3.37 rpoS17,

soxS

hup ACCCTTATAAAACGCGGGTTTTCGCAGAAACA

TGCGCTAGTATCATTGAT

5.18 rpoD16,

rpoD15

16S rRNA TCGTCAATTTTTGTTTTGAGA GTCATCTATTCG

GATGCTTTTCATGAAGT

2.40 rpoD15,

rpoD17

*Red nucleotides are proposed -35 regions; blue nucleotides are proposed -10 regions. LDF: linear

discrimination function; TF, transcription factor

The promoter elements of the gap and hup promoters predicted by BPROM are not ideal

because the spacer length between predicted -10 and -35 sequences was considerably

shorter (11 and 13 bp, respectively) than in consensus promoter (17 ± 1 bp; Lisser and

34

Margalit 1993). Since expression of LuxS has been reported in different bifidobacteria

(Yuan et al., 2008; Sun et al., 2014), further in silico promoter predictions were performed

using Neural Network Promoter Prediction (NNPP). This prediction produced several

possible core promoter regions with potential TSS for gap, hup, and luxS (Table 3-2).

Table 3-2: Some predicted core regions of promoters by NNPP

Gene Core promoter sequences (50bp, 5’3’) Scores

gap TATTTGTTAGCGTTAACGAAATATGGCCGTTTTATGCTCAAAGCAAGCGC

TCGCATCGAATCGCCGCAGGCTGTACAGACATATTTGTTA GCGTTAACGA

ACACCGTTGCTCTAGTACAGACGGCGCATTACAGTAGACACTGTTGGTAA

0.92

0.46

0.22

luxS TATAGCCAAGAATGCGGCGAGACGCGCGCGATAGTGAGACAATGGTCGAT

ACCGGACTCGCCTCGTGCCGTATATGCCGTATATGAGGAA CGCCTATAGC

AGAATGCGGCGAGACGCGCGCGATAGTGAGACAATGGTCGATGGAATGCC

0.55

0.23

0.20

hup CATGAAGTGGCTTGACAAGCATAATCTTGTCTGATTCGTCTATTTTCATA

CGTCTATTTTCATACCCCCTTCGGGGAAATAGATGTGAAAACCCTTATAA

GCTTGACAAGCATAATCTTGTCTGATTCGTCTATTTTCATA CCCCCTTCG

0.89

0.78

0.73

Potential TSS is underlined

In order to probe the functionality of these possible promoters, the reporter vector

pMDY23 was used (Fig. 3-1). PluxS and Pgap were amplified from the genome of B. bifidum

S17 and Phup and P16S rRNA were amplified from the genome of B. longum NCC2705 by

PCR introducing XhoI and BglII restriction sites. These restriction sites were used for

directional cloning of the promoter fragments in front of the gusA reporter gene in

pMDY23 as exact transcriptional fusions. Resulting plasmids were verified for absence of

any mutations and transformed into B. bifidum S17.

35

Fig. 3-1: Schematic representation of promoter fragments cloned into pMDY23. Four promoter

regions are amplified py PCR and cloned using BglII and XhoI restriction sites into the reporter vector

pMDY23 in front of gusA (repA and repB: bi fidobacterial origin of replication; spc: s pectinomycin

resistance gene).

3.2 Promoter activities in B. bifidum S17

The transcriptional activities of the four putative promoters were assayed by β-

glucuronidase activities in crude extracts of cloned B. bifidum S17 derivatives containing

the respective plasmids. Preliminary time course experiments reveal measurable β-

glucuronidase activity in B. bifidum S17/pMDY23-Pgap and B. bifidum S17/pMDY23-Phup

grown in MRSc with the highest level in early exponential phases, i.e. 8 h growth (Fig. 3-

2). Transcriptional activity of Pgap was significantly higher than Phup. Unexpectedly, no β-

glucuronidase activity above background levels (i.e. those observed with the empty

pMDY23 plasmid) could be detected with the luxS and 16S rRNA promoters. Monitor of

OD600 shown there is no difference in growth of all recombinant B. bifidum S17 strains

excluding any effect of growth on β-glucuronidase activity (data unshown).

36

Fig. 3-2: β-glucuronidase activity of crude extracts of recombinant B. bifidum S17 strains containing

di fferent promoter probe constructs derived from pMDY23. Samples were prepared from bacteria

grown 8 h in MRSc, i.e. in exponential growth phase. Values are fluorescence units per µg protein

(FU/µg) and are mean ± standard deviation of three independent cultures each measured in triplicate.

Since the promoter region of the gap gene of B. bifidum S17 yielded highest transcriptional

activity among these promoters tested. This region was used in all further experiments and

analysis.

3.3 Analysis of Pgap

3.3.1 Bioinformatic Analysis of Pgap

Complete sequence upstream of gap gene was shown in Fig. 3-3. As predicted in Table 3-1

and 3-2, and marked in this figure, potential -35 region, -10 region, and TSS were proposed

by online software. In silico, analysis suggested a putative bacterial promoter in the cloned

gap promoter region with a predicted TSS 114 bp upstream of the gap ATG start codon.

The 5’-untranslated region upstream of the ATG start codon of gap contains a putative

RBS as well.

37

Fig. 3-3 : Sequence of the DNA fragment upstream of gap of B. bifidum S17 cloned for reporter analysis.

The sequence contains a putative bacterial promoter predicted independently with two software tools.

The predicted transcription start site (position 1), -10, and -35 regions, as well as the putative

ribosome-binding site are indicated by black boxes.

During initiation of translation, the RBS interacts with a sequence motif in the 3’-end of

the 16S rRNA termed anti-Shine-Dalgarno sequence (anti-SD). The anti-SD of B. bifidum

S17 is highly complementary with only one mismatched base pair to the anti-SD (Fig. 3-4)

indicative of efficient translation initiation.

Fig. 3-4: Base pairing of the putative RBS of the Pgap promoter (RBS Pgap) with the anti-Shine-Dalgarno

sequence (Anti -SD) at the 3’-end of the 16S rRNA of B. bifidum S17

3.3.2 Experimental determination of TSS

To confirm the TSS of the gap gene of B. bifidum S17 experimentally, 5’-RACE was

conducted using RNA samples prepared from two independent cultures of B. bifidum S17

grown on MRSc to late exponential growth phase. For each RNA sample, inserts of two

randomly selected, positive E. coli clones containing pJet1.2-derivatives were sequenced.

Alignment of all four inserts equivocally indicated a thymidine (T) residue 62 bp upstream

of the ATG start codon of the gap open reading frame as TSS (Fig. 3-5), which is

considerable different to the predicted TSS (Table 3-2 and Fig. 3-3).

38

Fig. 3-5: Sequences of four inserts obtained from 5’-RACE experiments performed on two independent

RNA samples were aligned to the genomic region of the putative Pgap promoter. The experimentally

determined transcriptional start site (TSS), presumable ribosome binding site (RBS), and the ATG

start codon (Start) of the gap mRNA are indicated by grey boxes. The gene s pecific primer used for the

amplification of the gap transcripts is marked by a black box. Sequences of the oligo dT16 anchor

primer upstream of the TSS and vector sequences up and downstream of the two primers were clipped.

3.3.3 Alignment of representatives

Upon visual analysis, no sequences motifs with obvious homology to the consensus -10

(TATAAT) or -35 (TTGACA) sequences of σA-dependent promoters of Bacillus subtilis,

i.e. the prototype housekeeping promoter of Gram-positive bacteria, could be identified. In

order to identify potential -35 and -10 regions, the Pgap sequence of B. bifidum S17 was

aligned to the DNA sequences upstream of the gap genes of five other Bifidobacterium sp..

The results suggest a -35 region with the sequence TTGCTC, a spacer of 17 bases, and a -

10 region consisting of the nucleotides TACAGT for Pgap of B. bifidum S17 (Fig. 3-6).

39

Fig. 3-6: Alignment of the DNA sequences immediately upstream of the gap genes from 5

representative strains of Bifidobacterium. The degree of conservation is indicated on a grey scale with

black indicating highest conservation and conserved residues are shown as sequence logo. Presumable

or determined transcription start sites (TSS), corres ponding -10 and -35 regions are indicated by black

boxes. For orientation, the predicted TSS of bbif_0612 is also indicated. B. longum E18 (blong_1334), B.

breve UCC2003 (bbr_1233), B. adolescentis ATCC 15703 (bad_1079), B. dentium Bd1 (bdp_1514), B.

lactis ATCC 27673 (blac_0592), B. bifidum S17 (bbif_0612)

The consensus -35 and -10 sequences of the analyzed gap promoters of bifidobacteria are

TTGCCN and TANAGT, respectively, with a spacer of 17-19 bases (Fig. 3-6). Compared

to the canonical -35 (TTGACA) and -10 (TATAAT) regions of σ70- and σA-dependent

promoters of E. coli (Lisser and Margalit 1993) and Bacillus subtilis (Helmann 1995), the

bifidobacterial Pgap consensus -35 and -10 sequences each differ in two nucleotides (Fig. 3-

7).

Fig. 3-7: Alignment of the consensus sequences of bifidobacterial gap promoters and σA

-dependent

housekeeping promoters of Gram-positive bacteria.

40

3.4 Analysis of protein secretion in B. bifidum S17

As a basis for protein secretion in bifidobacteria, the genome sequences of two strains

frequently used in our labs were analyzed for genes of protein export systems. A number of

homologues of the Sec-dependent protein secretion are present in both B. bifidum S17 and

B. longum E18 (Table 3-3). An integral membrane complex composed by SecY, SecE and

SecG is the core component of Sec translocon. YajC plays a part in stabilizing and

regulating secretion through the SecYEG via SecA, which is an ATPase supplying energy.

This pathway is responsible for the secretion of the majority of extracellular proteins in

both Gram-negative and Gram-positive bacteria (Pugsley 1993; van Roosmalen et al.,

2004). Additionally, a complete twin arginine translocon operon was found in the genome

of B. longum E18 (BLONG_0088: TatB; BLONG_0089: TatC; BLONG_0090: TatA),

suggesting that this strain can secrete folded protein by the twin-arginine translocation (Tat)

pathway.

Table 3-3: Some components of Sec translocon in B. bifidum S17 and B. longum E18

Sec components B. bifidumS17 B. longum E18

SecA BBIF_1223 BLONG_1273

SecY BBIF_1484 BLONG_1793

SecE BBIF_0279 BLONG_2073

SecG BBIF_0980 BLONG_1190

YajC BBIF_0957 BLONG_1111

On the genomes of B. bifidum S17 and B. longum E18, the type I SPases (LepB) are

encoded by bbif_0379 and blong_1040 that recognize type I SP. BBIF_0514 and

BLONG_1429 are type II SPase (LspA) that response for cleavage of lipoprotein. The type

III SPs are prepilin- like SPs, that differ from the former two SPs as they do not contain any

hydrophobic regions. They also called type IV prepilin SP because they are recognized by

type IV prepilin peptidases. Type IV pili structure and function had been studied in B.

breve UCC2003 (Turroni et al., 2013), and the corresponding SPase annotated in the

genomes of many bifidobacteria strains. BBIF_0835 (annotated as hypothetical protein in

B. bifidum S17) has 58% identity to B. longum JDM301 type IV prepilin peptidase

(BLJ_0873).

41

3.5 Prediction of SPs in B. bifidum S17

In theory, proteins with SPs can be composed by three main groups: extracellular proteins,

cell wall proteins, and some proteins may have multiple localization sites. All proteins

containing SPs are potential candidates for cloning of a SP used for sorting protein

secretion. To screen reliable SPs, a complete list of proteins with SPs of B. bifidum S17

was compiled by in silico analysis using cPSORTb database (Table 3-4). Meanwhile, the

most N terminal 60 amino acids of these proteins were analyzed one by one with SignalP,

which yielded either positive SPs with potential cleavage sites or negative results.

Furthermore, both cPSORTb and SignalP positive proteins were searched in ePSORTb

database. Proteins existed in ePSORTb were underlined in this table, which had been

confirmed as extracellular proteins in E. coli.

42

Table 3-4: List of proteins with signal peptide in B. bifidum S17 predicted by cPSORTb

Gene tag Annotation E-Score SignalP 4.1 (cleavage site) D-score

extracellular proteins

BBIF_0022 Alpha-L-arabinofuranosidase 9.26 pos. 36 - 37: AYA-AD 0.718

BBIF_0048 1,4-beta-N-acetylmuramidase 9.98 pos. 30 - 31: AYA-LQ 0.732

BBIF_0246 peptidylprolylisomerase, FKBP-type 9.26 pos. 36 - 37: SGS-SS 0.556

BBIF_0285 hypothetical protein containing multip le

sugar recognition domains

9.76 pos. 27 - 28: ASA-AP 0.886

BBIF_0483 conserved protein with the pectin lyase

fold domain

9.13 pos. 30 - 31: AAA-ST 0.723

BBIF_0507 beta-galactosidaseBbgIII 7.74 pos. 32 - 33: AWA-VE 0.665

BBIF_1193 serine/cysteine peptidase 9.13 pos. 30 - 31: ASA-SP 0.836

BBIF_1317 alpha-L-fucosidase 9.97 pos. 37 - 38: AVA-AN 0.714

BBIF_1380 hypothetical protein BBIF_1380 9.13 pos. 27 - 28: ANA-AD 0.831

BBIF_1426 hypothetical protein with NlpC/P60

domain

9.73 pos. 25 - 26: AAA-AV 0.633

BBIF_1461 beta-N-acetylglucosamin idase 9.76 pos. 29 - 30: AYA-AG 0.76

BBIF_1576 beta-N-acetylglucosamin idase 9.98 pos. 34 - 35: AMA-AS 0.68

BBIF_1732 sialidase 9.97 pos. 39 - 40: ANA-AD 0.792

BBIF_1734 sialidase 9.98 pos. 35 - 36: ASA-AS 0.862

BBIF_1740 Alkaline phosphatase 9.73 pos. 29 - 30: AFA-AS 0.776

Cell wall proteins

BBIF_0215 cell wall protein containing Ig-like

domains (group2 - 3)

0.61 pos. 41 - 42: AAA-SD 0.681

BBIF_0301 hypothetical protein containing von

Willebr- factor type A domain

0 pos. 26 - 27: AIA-AN 0.884

BBIF_0302 hypothetical protein with Cna B-type

domain

0 pos. 29 - 30: ANA-AE 0.815

E-score, extracellu lar score, is calculated by cPSORTb; Pos., position of amino acid residues. Potential

cleavage sites are predicted by SignalP 4.1; D-Score, discrimination score. Proteins existed in ePSORTb are

underlined.

43

Table 3-4 (continued): List of proteins with signal peptide in B. bifidum S17 predicted by cPSORTb

Gene tag Annotation E-Score SignalP 4.1 (cleavage site) D-score

Cell wall proteins

BBIF_0522 hypothetical protein with CHAP domain 0.78 pos. 36 - 37: EQA-AA 0.716

BBIF_0592 peptide/nickel transport system,

substrate-binding protein

0.78 pos. 28 - 29: VKD-AN 0.612

BBIF_1382 hypothetical protein containing bacterial

Ig-like domain (group 2)

0.63 pos. 34 - 35: ASA-AD 0.768

BBIF_1452 hypothetical protein with the 5'-

nucleotidase domain

0.61 pos. 27 - 28: VTA-AY 0.837

BBIF_1607 hypothetical protein BBIF_1607 0.78 pos. 34 - 35: AGA-NP 0.652

BBIF_1648 hypothetical protein containing CnaB

domain - LPXTG-anchor

0.06 pos. 29 - 30: ANA-QV 0.729

BBIF_1761 Cell surface protein with gram positive

anchor - Cna protein B-type domains

0 pos. 31 - 32: ANA-AD 0.879

Proteins may have multiple localization sites

BBIF_0449 ATPase associated with various cellular

activities, AAA3

5.15 No SP 0.374

BBIF_0636 peptide/nickel transport system,

extracellular solute-binding protein

4.87 pos. 34 - 35: GSA-GN 0.644

BBIF_1059 hypothetical protein BBIF_1059 0.02 pos. 19 - 20: ALA-AV 0.473

BBIF_1257 hypothetical protein containing bacterial

Ig-like domain (group 2)

4.87 pos. 37 - 38: AQA-AG 0.641

BBIF_1681 subtilysin family peptidase (lactocepin) 4.87 pos. 28 - 29: ALA-AP 0.798

BBIF_1769 hypothetical protein with collagen-

binding domain

5.15 pos. 36 - 37: GVA-AR 0.54

E-score, extracellu lar score, is calculated by cPSORTb; Pos., position of amino acid residues. Potential

cleavage sites are predicted by SignalP 4.1; D-Score, discrimination score. Proteins existed in ePSORTb are

underlined.

44

3.7 Construction of a secretion reporter vector

3.7.1 Clone SP from gene bbif_1734

Amongst all SPs detected in silico, the SP of protein BBIF_1734 (S0 in short) has the

highest D-score. BBIF_1734 is annotated as a putative extracellular sialidase. In addition,

the ePSORTb database indicates that the E. coli sialidase is expressed as extracellular

protein. The BBIF_1734 SP was further confirmed by SignalP (Fig. 3-8) and TatP

prediction (data unshown). To test its potential as a sorting signal on heterologous protein

secretion, it was cloned by PCR amplification with primer S0F/S0R. To preserve the

predicted cleavage site between A35 and A36, two extra amino acids were included (A36S37).

Fig. 3-8: Prediction of the SP sequence of BBIF_1734 by SignalP4.1 using the N-terminal 60 amino

acids. The violet line is the default cutoff (score = 0.5 ). SignalP produces three scores: C-score (raw

cleavage site score), S-score (signal peptide score), and Y-score, which is a combination of the C-score

and the slope of the S-score.

3.7.2 Clone appA as a reporter

To test the utility of S0 as a sorting signal, a suitable reporter is needed. appA gene

encoding a phytase was amplified from the genome of E. coli DH10B by PCR and fused

with S0 by SOE-PCR using primers S0F/S0R, P0F/PhyR, and S0F/PhyR. The SOE-PCR

45

product (S0P) was ligated into pMDY23-Pgap to replace the gusA gene and the resulting

vector named pMgapS0P (Fig. 3-9, panel A). Meanwhile, appA was amplified using

primers PhyF and PhyR and cloned into pMDY23-Pgap to replace gusA by appA. The

resulting vector named pMgapP was used as a control to express phytase intracellularly

(Fig. 3-9, panel B). Positive constructs were confirmed by PCR with primers PMF/PhyR

and Sanger sequencing. Then, they were introduced into B. bifidum S17 by electroporation

for heterologous phytase expression.

Fig. 3-9: Plas mid map of pMgapS0P (A) and pMgapP (B). On pMgapS0P, gusA was replaced by S0

fused mature appA between Xho I and Hind III. S0 is standing for the predicted SP of gene bbif_1734.

Pgap is gap promoter. On pMgapP, gusA was replaced by mature appA between Xho I and Hind III

with an extra ATG start codon.

3.7.3 Phytase assay

Both plasmids were transformed into B. bifidum S17 and phytase activity was measured at

various time points during growth in crude extracts (intracellular) and culture supernatants

(extracellular). Both strains displayed almost identical growth ruling (data unshown).

Phytase activity markedly increased over time in supernatants of B. bifidum

S17/pMgapS0P whereas little to no activity was measured for the control strain without S0

before 12 h (B. bifidum S17/pMgapP, Fig. 3-10, panel A). By contrast, throughout growth

phytase activities were higher in crude extracts of B. bifidum S17/pMgapP. The relative

levels of extra- and intracellular phytase activity in both strains suggests efficient yet not

complete secretion mediated by the S0 (Fig. 3-10, panel B)

46

Fig. 3-10: Phytase activi ty in the supernatants (A) and crude extracts (B) of B. bifidum S17

recombinant constructs. B. bifidum S17/pMgapS0P and the SP-less control strain B. bifidum

S17/pMgapP were grown in RCM containing 100 μg/ml Spc. Supernatants and crude extracts were

collected at indicated time points. FTU is the standard unit of phytase activi ty that was defined as the

amount of enzyme needed for catalyzing the release of 1 μmol phosphate from phytate per minute.

Values are FTU per ml culture supernatant (A) or FTU per mg protein (B) and are mean ± standard

deviation of three independent cultures each measured in triplicate.

3.8 Comparison of various SPs

Following the successful establishment of an appA-based reporter vector and assay for S0

in B. bifidum S17, this system was used to test various other SPs of bifidobacteria selected

from three strains, exactly, B. bifidum S17, B. longum E18, and B. breve S27 (Table 3-5).

For B. longum E18 and B. breve S27, key words “secreted” and “secreted protein” were

searched from the annotation of genomic datasets. This yielded several candidates that

might be secreted proteins. Further prediction of all these candidates by SiganlP excluded

some of them as they are SP negative (Gram-positive model, D-score cutoff = 0.5). Some

representatives of them with relative high D-scores were selected as they might secreted

through different pathways according to functional annotation or report in literatures. For

example, BLONG_0223 and BLONG_1620 were annotated as Tat secreted proteins.

47

Table 3-5: 7 selected proteins and their signal peptides

Gene function Gene tag (NCBI) D-score Signal peptide

Tat-secreted glycosidase blong_0223 0.865 S1, 34 aa

Putative tat-secreted pectin lyase-like protein blong_1620 0.772 S2, 32 aa

Hypothetical secreted protein with NlpC/P60 domain blong_1728 0.891 S3, 31 aa

Conserved hypothetical secreted protein with CHAP

domain

blong_0476 0.838 S4, 36 aa

subtilisin family peptidase (lactocepin) bbif_1681 0.798 S5, 30 aa

surface protein with gram positive anchor and Cna

protein B-type domains

bbif_1761 0.879 S6, 33 aa

Conserved hypothetical secreted protein with CHAP

domain

S27_0508

0.823 S7, 36 aa

Analysis of the amino acids composition of these predicted SPs indicates they are type I SP

as possessing the typical AxA cleavage site (Table 3-6). Although BLONG_0223 (S1) and

BLONG_1620 (S2) were annotated as Tat secreted proteins on the genome B. longum E18,

the Tat signal motif (RRxFLK) was not detected by TatP. It seems no type II SPs among

them as the conserved feature structure is absent. These potential SPs (S1-S7) range from

30 to 36 amino acids (aa), which is typical in Gram-positive bacteria (Paetzel et al., 2002).

Table 3-6: Amino acid sequences of the seven predicted signal peptides

Signal peptide Sequence

S1 MHQSTRKRW LASIGAVAAVATLATGGAVTAQA*AD

S2 MTSRQGRQAIAATAAMGVAVALALPTAAFA*QS

S3 MKTKTVASSIVAAIVSCACLFMFVPTATA*AE

S4 MTNVRVIKPALAALVAAAACVGGLAFSSAQPAQA*DT

S5 MANKQWPRW VAAVAALAMAGSLPGTALA*AP

S6 MKSLMKKVFAAAAAIATVFGLAATTVATANA*AD

S7 MTKAKTMRPVLAACAASAICVAGLLASPVQPARA*NT

*indicates predicted cleavage sites; amino acids of conserved AxA motifs are shown in red.

48

To test the functionality of these predicted SPs, they were fused to the appA reporter gene

by SOE-PCR, cloned into the pMDY23-Pgap plasmid replacing the gusA gene and

introduced into B. bifidum S17 by electroporation. Monitoring of growth on RCM

indicated that all recombinant strains show the same growth characteristics (data not

shown). Moreover, preliminary temporal analysis indicated that for all strains phytase

mainly accumulated in supernatants in stationary phase. Thus in all further experiments,

samples were collected from cultures grown for 16 h for phytase assays. Phytase activity

was detected in the supernatants of all recombinant strains of B. bifidum S17 harboring

predicted SPs. B. bifidum S17 strains introduced plasmids pMgapS1P, pMgapS4P,

pMgapS6P, or pMgapS7P shown comparable or higher activities than B. bifidum

S17/pMgapS0P. In contrast, strains harboring pMgapS2P, pMgapS3P, and pMgapS5P

show lower activity than B. bifidum S17/pMgapS0P. B. bifidum S17/pMgapP, which does

not encode an SP, has no obvious activity in supernatant (Fig. 3-11).

Fig. 3-11: Phytase activi ties in the supernatants of different B. bifidum S17 constructs. Samples were

prepared from RCM derived s pent medium containing 100 μg/ml S pc after inoculation for 16 h. FTU

is the standard unit of phytase activity that was defined as the amount of enzyme needed for catalyzing

the release of 1 μmol phos phate from phytate per minute. Values are FTU per ml supernatant (FTU/ml)

and are mean ± standard deviation of three independent cultures each measured in triplicate.

In cell crude extracts, B. bifidum S17/pMgapP, which expressing appA intracellularly,

showed highest phytase activity. All strains with SP-containing constructs showed

markedly lower activity than B. bifidum S17/pMgapP as expected (Fig. 3-12).

49

Fig. 3-12: Phytase activi ties in the cell lysate of different B. bifidum S17 constructs. Samples were

prepared from 16 h incubated cells grown in RCM containing 100 μg/ml S pc. FTU is the standard unit

of phytase activity that was defined as the amount of enzyme needed for catalyzing the release of 1

μmol phos phate from phytate per minute. Total protein is quantified with a BCA kit. Values are FTU

per mg protein (FTU/mg) and are mean ± standard deviation of three independent cultures each

measured in triplicate.

To confirm the expression and secretion of phytase in B. bifidum S17 visually, a

phenotypic assay based on degradation of insoluble calcium phytate by extracellular

phytase in solid medium was used. Clear zones of degradation were observed for B.

bifidum S17 strains harboring pMgapS0P, pMgapS1P, pMgapS4P, pMgapS6P, and

pMgapS7P whereas strains carrying pMgapS2P, pMgapS3P, and pMgapS5P respectively

showed low or only weak calcium phytate degradation (Fig. 3-13). The degradation

potentials of these constructs are consistent with their enzymatic assay results. Collectively,

this data suggests that S6 is the most effective protein secretion signal.

Fig. 3-13: Calcium phytate degradation by recombinant B. bifidum S17 strains. P, B. bifidum

S17/pMgapP; S0-S7, B. bifidum S17/pMgapS0P-pMgapS7P; N, B. bifidum S17/pMDY23-Pgap. One

representative of three independent experiments is shown.

50

3.9 Secretion of codon adapted ChrR6 in B. bifidum S17

The ultimate goal of this thesis was the generation of a recombinant bifidobacterium

expression a prodrug-converting enzyme for cancer therapy using a suitable promoter

combined with protein secretion. As a therapeutic approach, nitroreductase (NTR)

converting the non-toxic pro-drug 5-aziridin- l-yl-2,4-dinitrobenzamide (CB1954) to the

tumor therapeutic drug 4-hydroxylamine (4-HX), which intercalates with DNA thereby

inhibiting proliferation, was selected. To construct a recombinant B. bifidum S17 for

conversion of CB1954, the gene encoding ChrR6 was synthesized codon-optimized for B.

bifidum S17. The ChrR6 is an artificial NTR created by mutation of the E. coli YieF and

shows faster enzyme kinetics than its native form (Thorne et al., 2009). The codon-

optimized chrR6 gene was sub-cloned under control of Pgap and containing the S6 secretion

signal thereby creating pMgapS6C for secretion (Fig. 3-14).

Fig. 3-14: Plasmid map of pMgapS6C. Pgap, in red, the gap promoter; S6, in green, SP of gene

bbif_1671; chrR6, codon optimized chromate reductase gene, also a nitroreductase gene.

Expression and secretion of ChrR6 in recombinant B. bifidum S17 was measured by NTR

enzymatic assay of culture supernatant and crude extracts. No NTR activity was observed

in empty controls (fresh medium or Tris-HCl buffer, data not shown). In crude extracts of

B. bifidum S17/pMgapS6C, about 240 mU/mg NTR activity were measured (Fig. 3-15,

panel A). In the negative vector control strain B. bifidum S17/pMDY23-Pgap, NTR activity

was markedly lower yet still detectable (~50 mU/mg), possibly due to the intrinsic NTR of

51

the strain. Similarly, NTR levels were by far higher in culture supernatants of B. bifidum

S17/pMgapS6C compared to B. bifidum S17/pMDY23-Pgap (Fig. 3-15, panel B).

Fig. 3-15: NTR activity in crude extracts (A) and culture supernatant (B) of B. bifidum S17/pMgapS6C

or B. bifidum S17/pMDY23-Pgap (empty vector control) grown for 16 h in RCM containing 100 μg/ml

Spc. 1U is the enzyme required for oxidizing 1 μmol NADH per minute. Values are NTR activity in

mU/ml supernatant (A) or mU/mg protein in crude extracts and are mean ± standard deviation of

three independent experiments each measured in triplicate.

Additionally, the enzymatic activity of Chr6R on CB1954 conversion was indirectly

demonstrated by growth inhibition experiment. For this purpose, OD600 was measured of B.

bifidum S17 derivatives grown for 24 h in the presence of different concentrations of

CB1954. The control strain B. bifidum S17/pMDY23-Pgap was highly resistant to CB1954

with supplementation of as much as 500 μM CB1954 only reduced the final OD600 to about

90%. By contrast, ChrR6 expressing B. bifidum S17/pMgapS6C was by far more sensitive

to CB1954 and growth of B. bifidum S17/pMgapS6C was inhibited by CD1954 in a dose-

dependent pattern (Fig. 3-16) indicating effective conversion of CB1954 to cytotoxic 4-HX.

52

Fig. 3-16: Growth inhibition of recombinant B. bifidum S17 strains by adding CB1954. B. bifidum

S17/pMgapS6C and B. bifidum S17/pMDY23 -Pgap (empty vector control) were grown for 24 h in RCM

broth containing 100 μg/ml S pc and different concentrations CB1954. Values are final OD600 relative to

the culture without CB1954 and are mean ± standard deviation of three independent experiments

assayed in triplicate.

53

4 DISCUSSION

4.1 Systems for protein expression and secretion by

bifidobacteria

4.1.1 Expression vectors and promoters

In this study, a series of E. coli - Bifidobacterium sp. shuttle vectors were constructed

based on the reporter vector pMDY23, which is based on the cryptic plasmid pNCC293

(Klijn et al., 2006). pNCC293 has 98.8% identity to the 1 847 bp plasmid pMB1, which is

believed to replicate by a theta mechanism. The plasmid pMDY23 harbors an E. coli gusA

gene as transcriptional reporter , is 100% stable in B. longum NCC2705 for at least 10

generations without antibiotic pressure, and has a medium copy number around 10 (Klijn,

et al., 2006). This plasmid was shown to stable replicate in B. bifidum S17 (Gleinser et al.,

2012), and was used previously by our group as a promoter probe vector in this strain

(Grimm et al., 2014).

In silico prediction by BPROM of putative promoters using complete inter-region

sequences yielded potential promoters for all promoter regions except for the fragment

upstream of luxS. Moreover, in the fragments with a positive hit for a bacterial promoter,

potential binding sites for RpoS and RpoD were detected. This indicates that these

promoters may dependent on σA (encoded by rpoD) consistent with a role as promoters of

housekeeping genes (Nesvera et al., 2012; Patek et al., 2013). Using BLAST analysis, a

gene (bbif_1216) was identified in the genome of B. bifidum S17. The deduced amino acid

sequence of BBIF_1216 shows high homology to the prototype σA of Bacillus subtilis

(Haldenwang 1995). The protein contains Sigma-70 factor regions 1.2, 2, 3, and 4

(residues 205-240, 272-342, 351-427 and 440-493, respectively) with a helix-turn-helix

motif for DNA binding and a motif for interaction with the RNA polymerase (RNAP) β’

core subunit in region 2. Together with the high conservation of the -35 and -10 promoter

elements, this suggests that the gene product of bbif_1216 is likely to be the housekeeping

sigma factor σA of B. bifidum S17 directing the RNAP to Pgap to initiate transcription.

54

Using pMDY23, a number of DNA fragments containing potential bifidobacterial

promoters were analyzed for transcriptional activity. Pgap and Phup were already used for

protein expression in bifidobacteria by different groups (Sun et al., 2012). PluxS was

assumed to bear transcriptional activity based on the detection of autoinducer-2 activity in

culture supernatants of B. longum NCC2705 (Sun et al., 2014).

Among the promoter fragments tested, the gap promoter of B. bifidum S17 and the hup

promoter of B. longum NCC2705 yielded detectable GusA reporter. This is in line with the

suspected role of gap and hup as housekeeping genes. The results also consistent with

previous study by our group showing higher Pgap-driven reporter activity in B. bifidum S17

compared to other promoters in all growth phases tested (Grimm et al., 2014). Furthermore,

several other groups also report on high transcriptional activity of bifidobacterial gap

promoters and these promoters have been used successfully for expression in bifidobacteria

(Klijn et al., 2006; Khokhlova et al., 2010; Sun et al., 2014). The gap genes encode the

enzyme glyceraldehyde-3-phosphate dehydrogenase (GAPDH, EC 1.2.1.12), which

catalyses the conversion of glyceraldehyde 3-phosphate to D-glycerate 1,3-bisphosphate

(Nagradova 2001). Although GAPDH is primarily know for its role in glycolysis, it is also

required for formation of lactate from glyceraldehyde 3-phosphate and, in consequence,

efficient energy conservation from substrates that are fermented via the bifidus shunt. Thus,

it seems reasonable to hypothesize that the gap gene is constitutively expressed at high

levels in a wide range of bacteria including bifidobacteria.

Histone- like HU proteins are ubiquitous in the bacterial kingdom (Chouayekh et al., 2009).

In E. coli, HU proteins are encoded by two genes, hupA and hupB (Claret and Rouviere-

Yaniv, 1997). In most other bacteria including bifidobacteria, only hupB was annotated.

HU proteins are able to bend and condense DNA and play an important role in bacterial

nucleotide organization (Czapla et al., 2008) and are involved in numerous cellular

processes, such as replication, transcription and gene regulation (Kamashev et al., 2008;

Mukherjee et al., 2009). Again, this indicates that the hup genes are constitutively

expressed.

The results indicate that Pgap has higher transcriptional activity under the conditions tested

than Phup. This contradicts previous studies showing comparable expression of FGF-2 in B.

breve using the Pgap and Phup sequences of B. longum (Shkoporov et al., 2008). The

55

difference may due to better recognition of Pgap by the RNAP holoenzyme based on

differences in the sequences (Fig. 3-6). The luxS and 16S rRNA promoters did not show

any transcriptional activity under the conditions tested although other studies have reported

considerable activity of the 16S rRNA promoter of B. longum (Park et al., 2008a).

Although several putative promoters have been successfully applied in bifidobacteria to

facilitate gene expression, none of these promoters’ elements has been experimentally

confirmed. Two important features that have an impact on promoter activity are the -10

and -35 regions. AT rich elements in the -10 region are involved in formation of

transcription bubbles (Feklistov and Darst 2011). Using 5’-RACE experiments, the

transcription start site (TSS) of Pgap was shown to be a thymidine (T) residue 62 bp

upstream of the translation start site. This is different from the TSS prediction by NNPP.

This suggests that bifidobacterial housekeeping promoters may be somewhat different to

those used for software-based promoter prediction.

Following the identification of the TSS, the -35 and -10 regions of Pgap can be localized by

alignment of other putative gap promoters of bifidobacteria. Alignment of five

representative potential Pgap sequences originated from different species/strains using the

core sequences from the ATG start codon to 50 bp upstream of the TSS suggested

conserved -10 and -35 regions (Fig. 3-6). Notably, nucleotides near the TSS are highly

conserved. This indicates that Pgap of B. bifidum S17 is likely to show transcriptional

activity in other bifidobacteria as well. In fact, this has been demonstrated by successful

expression of different proteins in B. bifidum S17, B. breve S27 and B. longum E18

(Grimm et al., 2014, Gleinser et al., 2012). In addition, Pgap is able to drive high- level gusA

reporter activity in E. coli (data not shown).

The 5’-untranslated region upstream of the ATG start codon of gap contains a putative

ribosome-binding site (RBS, Fig. 3-3). During initiation of translation, the RBS interacts

with a sequence motif in the 3’-end of the 16S rRNA termed anti-Shine-Dalgarno sequence

(anti-SD) (Laursen et al., 2005). The anti-SD of B. bifidum S17 is identical to that of other

Gram-positive bacteria including Bacillus subtilis and Lactococcus lactis (Wegmann et al.,

2013). Moreover, the RBS of Pgap and anti-SD of B. bifidum S17 are highly complementary

with only one mismatched base pair to the anti-SD (Fig. 3-4) indicative of efficient

translation initiation (Makrides 1996).

56

4.1.2 Secretion pathway and sorting signal

Generally, bacteria possess three different pathways for protein secretion: the Sec-

dependent (Sec), Sec-independent, and the twin arginine translocation (Tat) pathway with

the Sec pathway being the most common secretion machinery (Dautin and Bernstein 2007;

Tseng et al., 2009; Jongbloed et al., 2002). Proteins secreted via the Sec-dependent

pathway utilize a common machinery consisting of the integral membrane complex

SecYEG and other accessory and regulatory proteins including SecA, SecB, YajC

(Kostakioti et al., 2005). Tat pathway is responsible for secretion of readily folded proteins

(Jongbloed et al., 2004). Search of corresponding components in Bifidobacterium (NCBI

taxid: 1678) indicates that a large number of bifidobacteria possess a Sec-dependent

pathway, whereas Tat systems are only presented in a few strains of B. longum. Sec-

independent pathways allow direct export from the cytoplasm to the extracellular

environment in one-step. These pathways may include ABC (ATP-binding cassette)

exporters (Holland et al., 2005; Schneewind and Missiakas 2012) and the type III secretion

systems (T3SS, Hueck 1998). However, there is no study of Sec- independent proteins

secretion in bifidobacteria at present.

In biotechnology, protein secretion is required for a number of applications, when the

protein exerts its function outside the bacterial cell, e.g. in recombinant vaccine strains or

for downstream purification (Quax 1997). Secretion of recombinant protein by

bifidobacteria was first achieved by testing various signal peptides (SPs) with a nuclease

reporter using B. breve UCC2003 as host (MacConaill et al., 2003). Later, fusion of the α-

amylase SP to pediocin PA-1 leads to active protein secretion in B. longum (Moon et al.,

2005). Other examples are pB44- and pB80-derived vectors carrying a sec2 SP, cloned

from B. breve for secretion of FGF-2 (Shkoporov et al., 2008).

In this thesis, eight SPs were cloned from three strains of bifidobacteria. In a first set of

experiments, The SP of sialidase (encoded by bbif_1734) termed S0 was used to establish a

reporter system for protein secretion in B. bifidum S17 since this SP has the highest D

score and the corresponding protein was experimentally confirmed to be secreted in other

organisms (Corfield 1992; Crennell et al., 1993). The E. coli appA gene encoding phytase

57

was selected as reporter. This reporter gene was successfully expressed in bacteria

(Shouldice et al., 2010), plant (Zimmermann et al., 2003), and animal (Golovan et al.,

2001). The S0 SP was fused to the phytase gene and cloned downstream of Pgap in the

pMDY23 backbone lacking gusA. Following transformation of this construct into B.

bifidum S17, phytase activity was measurable in culture supernatants of the recombinant

strain and this activity increased whereas specific phytase activity in crude extracts

decreased during growth. These results demonstrate that S0 is a functional SP and appA

can be used in combination with the pMDY23 backbone and Pgap as a reporter for protein

secretion in bifidobacteria.

In a second step, several selected SPs that may direct protein secretion through different

pathways were cloned into the reporter plasmid and assayed for protein secretion. S1 and

S2 are thought to be Tat type secretion signals as corresponding proteins were annotated as

Tat secreted proteins. However, S1 mediates phytase secretion in B. bifidum S17 that lacks

Tat secretion system indicating it is in fact not a Tat SP. Of the other SPs used, only S4, S6

and S7 result in marked protein secretion. S4 and S7 are signal peptides of two

homologues (conserved hypothetical protein with CHAP domain) of B. longum E18 and B.

breve S27. Their functionality in B. bifidum S17 suggests that SP maybe recognized by

different strains/species within the genus Bifidobacterium. S5 and S6 are SPs of B. bifidum

S17 proteins with S5 being predicted to mediate secretion by the Sec-dependent and S6 by

the Sec-independent pathway. Interestingly S6, SP of a surface protein with Gram-positive

anchor and Cna protein B-type domains was the most efficient SP amongst those tested. In

all, comparison of these SPs in the same host highlights the importance of choosing sorting

signal in protein secretion. Finally, the relatively efficient S6 was selected for targeting

prodrug converting enzymes in following part as its obvious supper performance on sorting

phytase (Fig. 3-10).

4.1.3 Secretion reporter

Reporter systems are widely used in molecular biology. The most frequently used reporters

are enzymes, e.g. β-galactosidase (lacZ), chloramphenicol acetyltransferase (Cat), and β-

glucuronidase (gusA) (Poyart and Trieu-Cuot 1997; Palmano et al., 2001; Jefferson et al.,

1987). Another group of reporters is fluorescent proteins like green fluorescent protein

58

(GFP, Chary et al., 2005) or luciferases such as the Photorhabdus luminescence

luxCDABE (Hakkila et al., 2002; Waidmann et al., 2011). All these reporter systems are

convenient tools for assessment of promoter activities. However, naturally occurring

fluorescence can lead to high background levels during measurements (Greer and Szalay

2002). Moreover, enzymatic assays that involve cell disruption and addition of specific

substrates are usually more accurate (Kim et al., 2006).

A number of factors determine successful application of a reporter. Two important criteria

are the absence of similar activities in the host and the availability of simple and sensitive

methods for quantitative assays. In the extracellular environment, there are many

substances including proteases, acids and ions that might have an impact on the activity of

a secreted protein reporter. For bifidobacteria at laboratory scale, some specific

characteristics must be considered, e.g. producing large amount acid and growth under

anaerobic conditions in complex medium. Many common reporters designed for

intracellular expression in neutral pH are inactive in the complex and ac idic extracellular

environment and are thus undetectable.

In the presented thesis, the gusA reporter was used for promoter probing experiments and

its enzymatic activity is assayed with bacteria cell lysate at near neutral pH as it was widely

used a reporter (Jefferson et al., 1987). The Michaelis constant (Km) for one substrate, p-

nitrophenyl-beta-D-glucuronide (PNPG) was found to be 0.22 mM (Kim et al., 1995). The

cytosolic enzyme has an optimal pH 6-7 and culture supernatants of bifidobacteria easily

reach pH below 4.5 (Sun et al., 2014, supplementary material). Moreover, the active form

of this enzyme is composed of four subunits with a molecular weight of approximately

290-kilo Dalton (KD) (Kim et al., 1995). Thus, other authors have concluded that the (E.

coli) β-glucuronidase is not a good reporter enzyme to assay protein secretion (Kim et al.,

2006; Kavita and Burma 2008). This is confirmed by results of the present study showing

that β-glucuronidase activity cannot be detected in culture supernatants of recombinant B.

bifidum S17 containing constructs with S0 fused to the gusA reporter under the control of

Pgap (data unshown).

A number of genes were studied as secretion reporters. One is the secreted alkaline

phosphatase of E. coli, which was successfully used in bacteria and yeast (Bina et al., 1997;

Nallaseth and Anderson 2013). In eukaryotic organisms a similar protein, secreted

59

embryonic alkaline phosphatase (SEAP) is widely used as secreted reporter (Berger et al.,

1988; Christou and Parks 2011). Another example is the non-specific, class C family acid

phosphatase of Staphylococcus aureus, which is used as a secretion reporter in both Gram-

positive and Gram-negative bacteria (du Plessis et al., 2007). However, these phosphatases

are limited in their application due to presence of both alkaline phosphatase and non-

specific acid phosphatase in a wide range of bacteria resulting in high background levels. B.

bifidum S17 harbors two acid phosphatases (gene bbif_1161 and bbif_1680) and one

alkaline phophatase (bbif_1740). Moreover, the protein encoded by bbif_1740 contains a

type I SP with extracellular sub- location and the same protein in E. coli was experimentally

confirmed as extracellular protein (see Table 3-4). Thus, this protein is expected to

interfere with a phosphatase reporter.

Other secretion reporters include the luciferase from the marine copepod Gaussia princeps

(Gluc) and nuclease (Nuc) from Staphylococcus aureus. Gluc was used in Salmonella

enteric (Wille et al., 2012) and Mycobacterium smegmatis (Andreu et al., 2010). However,

high luminescence was detected in supernatants of Salmonella and Mycobacterium

smegmatis regardless of the presence or absence of a secretion signal. Nuc, a heat stable

phosphodiesterase is secreted as an active small polypeptide that undergoes proteolytic

cleavage to produce its active form (Shortle 1983, Poquet et al., 1998). Nuc is efficiently

secreted by numerous Gram-positive bacteria when its native signal sequence is replaced

by the SP of exported proteins (Downing et al., 1999; MacConaill et al., 2003; Chouayekh

et al., 2009; Karlskas et al., 2014). Of note, Nuc is the first and the only secretion reporter

used in bifidobacteria (MacConaill et al., 2003). However, similar to Gluc, considerable

nuclease activity is also observed in the absence of a functional SP. It has been suggested

that the release of the staphylococcal nuclease to the extracellular environment was not due

to cell lysis but by an unknown SP-independent pathway (Recchi et al., 2002).

Considering all advantages and disadvantages, these reporters might be unsuitable to

investigate SPs in bifidobacteria. Thus, the E. coli appA gene encoding a histidine acid

phytase (myo-inositol hexakisphosphate phosphohydrolase) was selected as a secretion

reporter. This enzyme catalyzes the stepwise removal of phosphates from phytic acid

(inositol hexakisphosphate, IP6) or its salt phytate and other phosphate donors. It is active

in a broad of pH profile and resistant to protease (Lei et al., 2013). Ideally, most

bifidobacteria including B. bifidum S17 do not harbor a phytase homologue. Exceptions are

60

B. pseudocatenulatum ATCC 27919 and B. longum subsp. infantis ATCC 15697, which

were reported to harbor phytase genes (Haros et al., 2007; Haros et al., 2009). Moreover,

this single subunit enzyme can be easily assayed with high specific activity in both cell

lysate and culture supernatants. For E. coli appA encoded phytase, the Km = 0.13 μM, and

Kcat/Km = 4.476 × 107 (Greiner et al., 1993). In fact, phytase were successfully secreted by

different host strains including Lactobacillus plantarum (Kerovuo and Tynkkynen 2000), B.

longum (Park et al., 2005), Pichiapastoris (Xiong et al., 2003) and Arxula adeninivorans

(Boer et al., 2009).

There are several methods for determining phytase activity based on similar chemical

principles. In this work, the Phytex procedure was employed, which was developed

specifically for E. coli derived phytase (Kim and Lei 2005; Weaver et al., 2009). In all

experiments, RCM other than MRS medium was used for cultivation of bacteria prior to

phytase assays since MRS contains 2 g/l sodium phosphate, which interfere the assay

(Olstorpe et al., 2009).

4.2 Bifidobacteria as gene delivery vectors in cancer

therapy

Cancer is one of the most challenging diseases worldwidely. Despite many approaches for

treatment, resistance to chemotherapy and radiotherapy is still a major obstacle limiting

therapeutic success (de Groot et al., 2011; Quispe-Tintaya et al., 2013). Alternative and

supplementary therapies such as bacterial tumor targeting have been studied for many

years (Hoption Cann et al., 2003). However, this approach only recently has attracted

increasing interest owing to advances in genetic engineering of promising bacterial strains

that so far have been difficult to access, e.g. clostridia and bifidobacteria (Forbes 2006).

The most frequently used facultative or anaerobic bacterial vectors for delivery of

therapeutic genes are Salmonella, Clostridium, and Bifidobacterium (Forbes 2010). The

therapeutic approaches include expression of toxins, cytokines, ligands and pro-drug

converting enzymes by these bacteria (see reviews, Patyar et al., 2011; Zu and Wang 2014).

Solid tumors are characterized by hypoxic and necrotic regions (Dewhirst et al., 2008), and

this microenvironment provides suitable conditions for the colonization of facultative and

61

obligate anaerobes (Fujimori et al., 2002). Anaerobic bacteria including bifidobacteria and

clostridia were shown to localize to these regions in solid tumors in mouse models

selectively (Cronin et al., 2008; Cronin et al., 2010; Lambin et al., 1998). The main

advantage of therapeutic proteins produced in situ in tumors by live bacteria is high local

concentrations while at the same time reducing toxicity at other sites. To date, two classes

of therapeutic proteins have been successfully expressed in bifidobacteria. One class of

proteins have direct cytotoxic effects such as toxins, cytokines, antibodies and antigens (Li

et al., 2003; Zhu et al., 2009; Li et al., 2012; Yin et al., 2012). The other class is enzymes

that convert non-toxic prodrugs into their active derivatives (Hamaji et al., 2007; Tang, et

al., 2009; Yin et al., 2013).

Gene directed enzyme prodrug therapy (GDEPT) is considered as one of the most

promising strategies since prodrug activation and the associated cytotoxicity are co nfined

to certain cells or regions (Palmer et al., 2003; Sasaki et al., 2006). In bifidobacteria,

several GDEPT approaches have been pursued. Herpes simplex virus thymidine kinase

(HSV-TK) expression by B. infantis was used to activate ganciclovir (GCV) and was

shown to result in antitumor activity in a rat model of bladder cancer (Tang et al., 2009).

However, the highly-polar, phosphorylated GCV derivative, which is the active drug,

shows limited diffusion across cell membranes (Mesnil et al., 2000) and two phase III

clinical trials using this approach had contradictory results (Rainov 2000; Immonen et al.,

2004).

At present, the mostly well-studied prodrug-converting enzyme system in bifidobacteria is

cytosine deaminase/5-Fluorocytosine (CD/5-FC, Hidaka et al., 2007; Hamaji et al., 2007).

Mammalian cells are CD negative and thus CD expressed by bacterial vectors can be used

to convert 5-FC into the toxic nucleotide analogue 5-fluorouracil (5-FU) (Kuriyama et al.,

1999; Longley et al., 2003). In contrast to HSV-TK/GSV system, the CD/5-FC system has

much stronger bystander effect, i.e. diffusion of the active compound to neighboring cells

without direct cell-cell contact (Ichikawa et al., 2004). However, the efficiency of 5-FC

penetration into bacterial cells is limited and hepatic metabolism of 5-FC is associated with

moderate systemic toxicity (Hooiveld et al., 2004; van Kuilenburg 2004).

Nitroreductase (NTR) combined with nitro aromatic pro-drugs is attractive as these drugs

are toxic to both replicative and non-replicative cells (Blackwood et al., 2001), and the

62

prodrug CB1954 can freely diffuse across membranes (Christofferson and Wilkie 2009).

NTR converts CB1954 into the tumor drug 4-hydroxylamine (4-HX), which alkylates

DNA (Vass et al., 2009). To date, this system has not been tested in bifidobacteria. The

efficiency of enzyme/pro-drug therapy is largely confined by the actual activity of the

converting enzyme in vivo. Of note, activities of wild type NTRs are quiet low, e.g. E. coli

ChrR Km = 260 μM (Ackerley et al., 2004). To test this approach for bifidobacterial tumor

targeting, a codon-optimized nitroreductase gene (chrR6) was cloned under Pgap for high-

level heterologous expression in B. bifidum S17. ChrR6, also named as Y6, is a mutant

version of ChrR with much higher enzyme kinetics to CB1954 (Barak et al., 2006).

Moreover, the S6 SP was fused to the ChrR6 coding sequence for efficient secretion to

enhance efficiency as well as decrease cytotoxity to the expression host. The presented

preliminary results indicate the secretion of active ChrR6 into medium as shown by

transformation of substrate CB1954 and NADH by culture supernatants of the recombinant

B. bifidum S17 strain. The potential of ChrR6 secretion by recombinant bifidobacteria for

cancer therapy will be tested in tumor models in vitro and in vivo in the future.

63

5 SUMMARY

Based on their beneficial properties and their generally recognized as safe status,

bifidobacteria are used as probiotics in food and pharmaceutical industries. In the future,

recombinant strains with enhanced and/or new beneficial properties may become an option

for alternative or supplementary treatment of various diseases including inflammatory

bowel disease or cancer. The aim of this thesis was to establish an efficient system for

production of secreted proteins with therapeutic potential using recombinant bifidobacteria.

This was achieved by following work.

1. Four putative promoter sequences were analyzed in silico and in vitro using a

glucuronidase reporter system in B. bifidum S17. This revealed that the DNA

sequences upstream of the gap gene yielded highest levels of GusA reporter activity

amongst the promoters tested.

2. The transcription start site of the gap promoter (Pgap) was determined by 5’-RACE and

was found to be a thymidine residue 62 bp upstream of the translational start site.

Using bioinformatics comparison to other putative gap promoters of bifidobacteria, the

-10, -35 and ribosome-binding site of Pgap were identified. The Pgap core regions are

highly conserved in all five Bifidobacterium sp. analyzed indicating that Pgap of B.

bifidum S17 is functional across these species.

3. Using the E. coli appA gene and the predicted signal peptide of a sialidase

(BBIF_1734), a reporter vector for analysis of signal peptides in B. bifidum S17 was

generated. This system was used to analyze the efficacy of other seven predicted signal

peptides (S1-S7) to mediate protein secretion. One of these signal peptides showing

best performance was chosen in further experiments.

4. The most efficient signal peptide were fused to a codon adapted gene encoding for a

nitroreductase, which is a prodrug converting enzyme (ChrR6), and cloned under

control of Pgap. The resulting vector was transformed into B. bifidum S17 and this

recombinant strain exhibited high nitroreductase activity in culture supernatants. The

engineered strain can efficiently convert the prodrug CB1954 to its active derivative.

64

6 LITERATURES

1. Ackerley DF, Gonzalez CF, Keyhan M, Blake R 2nd, Matin A (2004) Mechanism of

chromate reduction by the Escherichia coli protein, NfsA, and the role of different

chromate reductases in minimizing oxidative stress during chromate reduction. Environ

Microbiol 6: 851-860.

2. Agrawal N, Bettegowda C, Cheong I, Geschwind JF, Drake CG, Hipkiss EL, Tatsumi M,

Dang LH, Diaz LA, Jr., Pomper M, Abusedera M, Wahl RL, Kinzler KW, Zhou S,

Huso DL, Vogelstein B (2004) Bacteriolytic therapy can generate a potent immune

response against experimental tumors. Proc Natl Acad Sci U S A 101: 15172-15177.

3. Alvarez-Martin P, Belen Florez A, Margolles A, del Solar G, Mayo B (2008) Improved

cloning vectors for bifidobacteria, based on the Bifidobacterium catenulatum pBC1

replicon. Appl Environ Microbiol 74: 4656-4665.

4. An HM, Park SY, Lee do K, Kim JR, Cha MK, Lee SW, Lim HT, Kim KJ, Ha NJ (2011)

Antiobesity and lipid- lowering effects of Bifidobacterium spp. in high fat diet- induced

obese rats. Lipids Health Dis 10: 116.

5. Andreu N, Zelmer A, Fletcher T, Elkington PT, Ward TH, Ripoll J, Parish T, Bancroft

GJ, Schaible U, Robertson BD, Wiles S (2010) Optimisation of bioluminescent

reporters for use with mycobacteria. PLoS One 5: e10777.

6. Baban CK, Cronin M, O'Hanlon D, O'Sullivan GC, Tangney M (2010) Bacteria as

vectors for gene therapy of cancer. Bioeng Bugs 1: 385-394.

7. Barak Y, Thorne SH, Ackerley DF, Lynch SV, Contag CH, Matin A (2006) New

enzyme for reductive cancer chemotherapy, YieF, and its improvement by directed

evolution. Mol Cancer Ther 5: 97-103.

8. Bartlett JM, Stirling D (2003) A short history of the polymerase chain reaction. Methods

Mol Biol 226: 3-6.

9. Berger J, Hauber J, Hauber R, Geiger R, Cullen BR (1988) Secreted placental alkaline

phosphatase: a powerful new quantitative indicator of gene expression in eukaryotic

cells. Gene 66: 1-10.

65

10. Bina JE, Nano F, Hancock RE (1997) Utilization of alkaline phosphatase fusions to

identify secreted proteins, including potential efflux proteins and virulence factors from

Helicobacter pylori. FEMS Microbiol Lett 148: 63-68.

11. Blackwood L, O'Shaughnessy PJ, Reid SW, Argyle DJ (2001) E. coli

nitroreductase/CB1954: in vitro studies into a potential system for feline cancer gene

therapy. Vet J 161: 269-279.

12. Boer E, Piontek M, Kunze G (2009) Xplor 2--an optimized transformation/expression

system for recombinant protein production in the yeast Arxula adeninivorans. Appl

Microbiol Biotechnol 84: 583-594.

13. Briczinski EP, Loquasto JR, Barrangou R, Dudley EG, Roberts AM, Roberts RF (2009)

Strain-specific genotyping of Bifidobacterium animalis subsp lactis by using single-

nucleotide polymorphisms, insertions, and deletions. Appl Environ Microbiol 75:

7501-7508.

14. Chary VK, Busuioc M, Renye JA, Jr., Piggot PJ (2005) Vectors that facilitate the

replacement of transcriptional lacZ fusions in Streptococcus mutans and Bacillus

subtilis with fusions to gfp or gusA. FEMS Microbiol Lett 247: 171-176.

15. Chen JJ, Wang R, Li XF, Wang RL (2012) Bifidobacterium adoiescentis

supplementation ameliorates visceral fat accumulation and insulin sensitivity in an

experimental model of the metabolic syndrome. Brit J Nutr 107: 1429-1434.

16. Chen MJ, Green NK, Reynolds GM, Flavell JR, Mautner V, Kerr DJ, Young LS,

Searle PF (2004) Enhanced efficacy of Escherichia coli nitroreductase/CB1954 prodrug

activation gene therapy using an E1B-55K-deleted oncolytic adenovirus vector. Gene

Ther 11: 1126-1136.

17. Cheng CM, Lu YL, Chuang KH, Hung WC, Shiea J, Su YC, Kao CH, Chen BM,

Roffler S, Cheng TL (2008) Tumor-targeting prodrug-activating bacteria for cancer

therapy. Cancer Gene Ther 15: 393-401.

18. Chouayekh H, Serror P, Boudebbouze S, Maguin E (2009) Highly efficient production

of the staphylococcal nuclease reporter in Lactobacillus bulgaricus governed by the

promoter of the hlbA gene. FEMS Microbiol Lett 293: 232-239.

19. Christofferson A, Wilkie J (2009) Mechanism of CB1954 reduction by Escherichia coli

nitroreductase. Biochem Soc Trans 37: 413-418.

66

20. Christou C, Parks RJ (2011) Rational design of murine secreted alkaline phosphatase

for enhanced performance as a reporter gene in mouse gene therapy preclinical studies.

Hum Gene Ther 22: 499-506.

21. Claret L, Rouviere-Yaniv J (1997) Variation in HU composition during growth of

Escherichia coli: the heterodimer is required for long term survival. J Mol Biol 273: 93-

104.

22. Corfield T (1992) Bacterial sialidases--roles in pathogenicity and nutrition.

Glycobiology 2: 509-521.

23. Crennell SJ, Garman EF, Laver WG, Vimr ER, Taylor GL (1993) Crystal structure of a

bacterial sialidase (from Salmonella typhimurium LT2) shows the same fold as an

influenza virus neuraminidase. Proc Natl Acad Sci U S A 90: 9852-9856.

24. Cronin M, Morrissey D, Rajendran S, El Mashad SM, van Sinderen D, O'Sullivan GC,

Tangney M (2010) Orally administered bifidobacteria as vehicles for delivery of agents

to systemic tumors. Mol Ther 18: 1397-1407.

25. Cronin M, Sleator RD, Hill C, Fitzgerald GF, van Sinderen D (2008) Development of a

luciferase-based reporter system to monitor Bifidobacterium breve UCC2003

persistence in mice. BMC Microbiology 8.

26. Cronin M, Zomer A, Fitzgerald GF, van Sinderen D (2012) Identification of iron-

regulated genes of Bifidobacterium breve UCC2003 as a basis for controlled gene

expression. Bioeng Bugs 3: 157-167.

27. Czapla L, Swigon D, Olson WK (2008) Effects of the nucleoid protein HU on the

structure, flexibility, and ring-closure properties of DNA deduced from Monte Carlo

simulations. J Mol Biol 382: 353-370.

28. Dang LH, Bettegowda C, Huso DL, Kinzler KW, Vogelstein B (2001) Combination

bacteriolytic therapy for the treatment of experimental tumors. Proc Natl Acad Sci U S

A 98: 15155-15160.

29. Dautin N, Bernstein HD (2007) Protein secretion in gram-negative bacteria via the

autotransporter pathway. Annu Rev Microbiol 61: 89-112.

30. de Groot JF, Lamborn KR, Chang SM, Gilbert MR, Cloughesy TF, Aldape K, Yao J,

Jackson EF, Lieberman F, Robins HI, Mehta MP, Lassman AB, Deangelis LM, Yung

WK, Chen A, Prados MD, Wen PY (2011) Phase II study of aflibercept in recurrent

67

malignant glioma: a North American Brain Tumor Consortium study. J Clin Oncol 29:

2689-2695.

31. de Vries W, Gerbrandy SJ, Stouthamer AH (1967) Carbohydrate metabolism in

Bifidobacterium bifidum. Biochim Biophys Acta 136: 415-425.

32. Debowski AW, Verbrugghe P, Sehnal M, Marshall BJ, Benghezal M (2013)

Development of a tetracycline- inducible gene expression system for the study of

Helicobacter pylori pathogenesis. Appl Environ Microbiol 79: 7351-7359.

33. Dev S, Mizuguchi H, Das AK, Matsushita C, Maeyama K, Umehara H, Ohtoshi T,

Kojima J, Nishida K, Takahashi K, Fukui H (2008) Suppression of histamine signaling

by probiotic Lac-B: a possible mechanism of its anti-allergic effect. J Pharmacol Sci

107: 159-166.

34. Dewhirst MW, Cao Y, Moeller B (2008) Cycling hypoxia and free radicals regulate

angiogenesis and radiotherapy response. Nat Rev Cancer 8: 425-437.

35. Djeha HA, Todryk SM, Pelech S, Wrighton CJ, Irvine AS, Mountain A, Lipinski KS

(2005) Antitumor immune responses mediated by adenoviral GDEPT using

nitroreductase/CB1954 is enhanced by high- level coexpression of heat shock protein

70. Cancer Gene Ther 12: 560-571.

36. Downing KJ, McAdam RA, Mizrahi V (1999) Staphylococcus aureus nuclease is a

useful secretion reporter for mycobacteria. Gene 239: 293-299.

37. du Plessis E, Theron J, Berger E, Louw M (2007) Evaluation of the Staphylococcus

aureus class C nonspecific acid phosphatase (SapS) as a reporter for gene expression

and protein secretion in gram-negative and gram-positive bacteria. Appl Environ

Microbiol 73: 7232-7239.

38. Duong T, Miller MJ, Barrangou R, Azcarate-Peril MA, Klaenhammer TR (2011)

Construction of vectors for inducible and constitutive gene expression in Lactobacillus.

Microb Biotechnol 4: 357-367.

39. Estrem ST, Ross W, Gaal T, Chen ZW, Niu W, Ebright RH, Gourse RL (1999)

Bacterial promoter architecture: subsite structure of UP elements and interactions with

the carboxy-terminal domain of the RNA polymerase alpha subunit. Genes Dev 13:

2134-2147.

40. Ewaschuk JB, Diaz H, Meddings L, Diederichs B, Dmytrash A, Backer J, Looijer-van

68

Langen M, Madsen KL (2008) Secreted bioactive factors from Bifidobacterium infantis

enhance epithelial cell barrier function. Am J Physiol Gastrointest Liver Physiol 295:

G1025-1034.

41. Feklistov A, Darst SA (2011) Structural basis for promoter-10 element recognition by

the bacterial RNA polymerase sigma subunit. Cell 147: 1257-1269.

42. Forbes NS (2006) Profile of a bacterial tumor killer. Nat Biotechnol 24: 1484-1485.

43. Forbes NS (2010) Engineering the perfect (bacterial) cancer therapy. Nat Rev Cancer

10: 785-794.

44. Fox ME, Lemmon MJ, Mauchline ML, Davis TO, Giaccia AJ, Minton NP, Brown JM

(1996) Anaerobic bacteria as a delivery system for cancer gene therapy: in vitro

activation of 5-fluorocytosine by genetically engineered clostridia. Gene Ther 3: 173-

178.

45. Fujimori M (2006) Genetically engineered bifidobacterium as a drug delivery system

for systemic therapy of metastatic breast cancer patients. Breast Cancer 13: 27-31.

46. Fujimori M, Amano J, Taniguchi S (2002) The genus Bifidobacterium for cancer gene

therapy. Curr Opin Drug Di De 5: 200-203.

47. Georgiou G, Segatori L (2005) Preparative expression of secreted proteins in bacteria:

status report and future prospects. Curr Opin Biotechnol 16: 538-545.

48. Gleinser M, Grimm V, Zhurina D, Yuan J, Riedel CU (2012) Improved adhesive

properties of recombinant bifidobacteria expressing the Bifidobacterium bifidum-

specific lipoprotein BopA. Microb Cell Fact 11.

49. Golovan SP, Hayes MA, Phillips JP, Forsberg CW (2001) Transgenic mice expressing

bacterial phytase as a model for phosphorus pollution control. Nat Biotechnol 19: 429-

433.

50. Gonzales MF, Brooks T, Pukatzki SU, Provenzano D (2013) Rapid protocol for

preparation of electrocompetent Escherichia coli and Vibrio cholerae. J Vis Exp. 80:

50684

51. Greer LF, 3rd, Szalay AA (2002) Imaging of light emission from the expression of

luciferases in living cells and organisms: a review. Luminescence 17: 43-74.

52. Greiner R, Konietzny U, Jany KD (1993) Purification and characterization of two

69

phytases from Escherichia coli. Arch Biochem Biophys 303: 107-113.

53. Grimm V, Gleinser M, Neu C, Zhurina D, Riedel CU (2014) Expression of fluorescent

proteins in bifidobacteria for analysis of host-microbe interactions. Appl Environ

Microbiol 80: 2842-2850.

54. Guglielmetti S, Mayo B, Alvarez-Martin P (2013) Mobilome and genetic modification

of bifidobacteria. Benef Microbes 4: 143-166.

55. Guglielmetti S, Mora D, Gschwender M, Popp K (2011) Randomised clinical trial:

Bifidobacterium bifidum MIMBb75 significantly alleviates irritable bowel syndrome

and improves quality of life - a double-blind, placebo-controlled study. Aliment Pharm

Therap 33: 1123-1132.

56. Guyonnet D, Chassany O, Ducrotte P, Picard C, Mouret M, Mercier CH, Matuchansky

C (2007) Effect of a fermented milk containing Bifidobacterium animalis DN-173 010

on the health-related quality of life and symptoms in irritable bowel syndrome in adults

in primary care: a multicentre, randomized, double-blind, controlled trial. Aliment

Pharm Therap 26: 475-486.

57. Hakkila K, Maksimow M, Karp M, Virta M (2002) Reporter genes lucFF, luxCDABE,

gfp, and dsred have different characteristics in whole-cell bacterial sensors. Analytical

Biochemistry 301: 235-242.

58. Haldenwang WG (1995) The sigma factors of Bacillus subtilis. Microbiol Rev 59: 1-30.

59. Hamaji Y, Fujimori M, Sasaki T, Matsuhashi H, Matsui-Seki K, Shimatani-Shibata Y,

Kano Y, Amano J, Taniguchi S (2007) Strong enhancement of recombinant cytosine

deaminase activity in Bifidobacterium longum for tumor-targeting enzyme/prodrug

therapy. Biosci Biotech Bioch 71: 874-883.

60. Haros M, Bielecka M, Honke J, Sanz Y (2007) Myo-inositol hexakisphosphate

degradation by Bifidobacterium infantis ATCC 15697. Int J Food Microbiol 117: 76-84.

61. Haros M, Carlsson NG, Almgren A, Larsson-Alminger M, Sandberg AS, Andlid T

(2009) Phytate degradation by human gut isolated Bifidobacterium pseudocatenulatum

ATCC27919 and its probiotic potential. Int J Food Microbiol 135: 7-14.

62. Heckman KL, Pease LR (2007) Gene splicing and mutagenesis by PCR-driven overlap

extension. Nat Protoc 2: 924-932.

70

63. Helmann JD (1995) Compilation and analysis of Bacillus subtilis sigma A-dependent

promoter sequences: evidence for extended contact between RNA polymerase and

upstream promoter DNA. Nucleic Acids Res 23: 2351-2360.

64. Hidaka A, Hamaji Y, Sasaki T, Taniguchi S, Fujimori M (2007) Exogenous cytosine

deaminase gene expression in Bifidobacterium breve I-53-8w for tumor-targeting

enzyme/prodrug therapy. Biosci Biotechnol Biochem 71: 2921-2926.

65. Hill C, Guarner F, Reid G, Gibson GR, Merenstein DJ, Pot B, Morelli L, Canani RB,

Flint HJ, Salminen S, Calder PC, Sanders ME (2014) Expert consensus document: The

International Scientific Association for Probiotics and Prebiotics consensus statement

on the scope and appropriate use of the term probiotic. Nat Rev Gastroenterol Hepatol.

[Epub ahead of print] PMID: 24912386

66. Holland IB, Schmitt L, Young J (2005) Type 1 protein secretion in bacteria, the ABC-

transporter dependent pathway (review). Mol Membr Biol 22: 29-39.

67. Hooiveld EA, van Kuilenburg AB, Haanen JB, Westermann AM (2004) Severe

toxicity after treatment with capecitabine and fluorouracil due to partial

dihydropyrimidine dehydrogenase deficiency. Ned Tijdschr Geneeskd 148: 626-628.

68. Hoption Cann SA, van Netten JP, van Netten C (2003) Dr William Coley and tumour

regression: a place in history or in the future. Postgrad Med J 79: 672-680.

69. Huber BE, Austin EA, Richards CA, Davis ST, Good SS (1994) Metabolism of 5-

fluorocytosine to 5-fluorouracil in human colorectal tumor cells transduced with the

cytosine deaminase gene: significant antitumor effects when only a small percentage of

tumor cells express cytosine deaminase. Proc Natl Acad Sci U S A 91: 8302-8306.

70. Hueck CJ (1998) Type III protein secretion systems in bacterial pathogens of animals

and plants. Microbiol Mol Biol Rev 62: 379-433.

71. Hughes BW, Wells AH, Bebok Z, Gadi VK, Garver RI, Jr., Parker WB, Sorscher EJ

(1995) Bystander killing of melanoma cells using the human tyrosinase promoter to

express the Escherichia coli purine nucleoside phosphorylase gene. Cancer Research 55:

3339-3345.

72. Ichikawa W, Takahashi T, Suto K, Nihei Z, Shirota Y, Shimizu M, Sasaki Y, Hirayama

R (2004) Thymidylate synthase and dihydropyrimidine dehydrogenase gene expression

in relation to differentiation of gastric cancer. Int J Cancer 112: 967-973.

71

73. Immonen A, Vapalahti M, Tyynela K, Hurskainen H, Sandmair A, Vanninen R,

Langford G, Murray N, Yla-Herttuala S (2004) AdvHSV-tk gene therapy with

intravenous ganciclovir improves survival in human malignant glioma: a randomised,

controlled study. Mol Ther 10: 967-972.

74. Jefferson RA, Kavanagh TA, Bevan MW (1987) GUS fusions: beta-glucuronidase as a

sensitive and versatile gene fusion marker in higher plants. EMBO J 6: 3901-3907.

75. Jongbloed JD, Antelmann H, Hecker M, Nijland R, Bron S, Airaksinen U, Pries F,

Quax WJ, van Dijl JM, Braun PG (2002) Selective contribution of the twin-arginine

translocation pathway to protein secretion in Bacillus subtilis. J Biol Chem 277: 44068-

44078.

76. Jongbloed JD, Grieger U, Antelmann H, Hecker M, Nijland R, Bron S, van Dijl JM

(2004) Two minimal Tat translocases in Bacillus. Mol Microbiol 54: 1319-1325.

77. Kainulainen V, Reunanen J, Hiippala K, Guglielmetti S, Vesterlund S, Palva A,

Satokari R (2013) BopA does not have a major role in the adhesion of Bifidobacterium

bifidum to intestinal epithelial cells, extracellular matrix proteins, and mucus. Appl

Environ Microbiol 79: 6989-6997.

78. Kamashev D, Balandina A, Mazur AK, Arimondo PB, Rouviere-Yaniv J (2008) HU

binds and folds single-stranded DNA. Nucleic Acids Res 36: 1026-1036.

79. Karlskas IL, Maudal K, Axelsson L, Rud I, Eijsink VG, Mathiesen G (2014)

Heterologous protein secretion in lactobacilli with modified pSIP vectors. PLoS One 9:

e91125.

80. Katan MB (2012) Why the European Food Safety Authority was right to reject hea lth

claims for probiotics. Benef Microbes 3: 85-89.

81. Kavita P, Burma PK (2008) A comparative analysis of green fluorescent protein and

beta-glucuronidase protein-encoding genes as a reporter system for studying the

temporal expression profiles of promoters. J Biosci 33: 337-343.

82. Kawasaki S, Sakai Y, Takahashi T, Suzuki I, Niimura Y (2009) O2 and reactive

oxygen species detoxification complex, composed of O2-responsive NADH:rubredoxin

oxidoreductase-flavoprotein A2-desulfoferrodoxin operon enzymes, rubperoxin, and

rubredoxin, in Clostridium acetobutylicum. Appl Environ Microbiol 75: 1021-1029.

83. Kerovuo J, Tynkkynen S (2000) Expression of Bacillus subtilis phytase in

72

Lactobacillus plantarum 755. Lett Appl Microbiol 30: 325-329.

84. Khokhlova EV, Efimov BA, Kafarskaia LI, Shkoporov AN (2010) Heterologous

expression of secreted biologically active human interleukin-10 in Bifidobacterium

breve. Arch Microbiol 192: 769-774.

85. Kim DH, Jin YH, Jung EA, Han MJ, Kobashi K (1995) Purification and

characterization of beta-glucuronidase from Escherichia coli HGU-3, a human

intestinal bacterium. Biol Pharm Bull 18: 1184-1188.

86. Kim JY, Wang Y, Park SJ, Ji GE, Park MS (2012) Cloning and expression of beta-

glucosidases from Bifidobacterium lactis AD011. Food Sci Biotechnol 21: 731-738.

87. Kim KW, Franceschi VR, Davin LB, Lewis NG (2006) Beta-glucuronidase as reporter

gene: advantages and limitations. Methods Mol Biol 323: 263-273.

88. Kim TW, Lei XG (2005) An improved method for a rapid determination of phytase

activity in animal feed. J Anim Sci 83: 1062-1067.

89. Klijn A, Moine D, Delley M, Mercenier A, Arigoni F, Pridmore RD (2006)

Construction of a reporter vector for the analysis of Bifidobacterium longum promoters.

Appl Environ Microbiol 72: 7401-7405.

90. Kohwi Y, Imai K, Tamura Z, Hashimoto Y (1978) Antitumor effect of Bifidobacterium

infantis in mice. Gann 69: 613-618.

91. Kostakioti M, Newman CL, Thanassi DG, Stathopoulos C (2005) Mechanisms of

protein export across the bacterial outer membrane. J Bacteriol 187: 4306-4314.

92. Kullen MJ, Klaenhammer TR (2000) Genetic modification of intestinal lactobacilli and

bifidobacteria. Curr Issues Mol Biol 2: 41-50.

93. Kuriyama S, Kikukawa M, Masui K, Okuda H, Nakatani T, Sakamoto T, Yoshiji H,

Fukui H, Ikenaka K, Mullen CA, Tsujii T (1999) Cytosine deaminase/5-fluorocytosine

gene therapy can induce efficient anti-tumor effects and protective immunity in

immunocompetent mice but not in athymic nude mice. Int J Cancer 81: 592-597.

94. Lambin P, Theys J, Landuyt W, Rijken P, van der Kogel A, van der Schueren E,

Hodgkiss R, Fowler J, Nuyts S, de Bruijn E, Van Mellaert L, Anne J (1998)

Colonisation of Clostridium in the body is restricted to hypoxic and necrotic areas of

tumours. Anaerobe 4: 183-188.

73

95. Laursen BS, Sorensen HP, Mortensen KK, Sperling-Petersen HU (2005) Initiation of

protein synthesis in bacteria. Microbiol Mol Biol Rev 69: 101-123.

96. Lee JH, O'Sullivan DJ (2006) Sequence analysis of two cryptic plasmids from

Bifidobacterium longum DJO10A and construction of a shuttle cloning vector. Appl

Environ Microbiol 72: 527-535.

97. Lee JH, O'Sullivan DJ (2010) Genomic insights into bifidobacteria. Microbiol Mol Biol

Rev 74: 378-416.

98. Lei XG, Weaver JD, Mullaney E, Ullah AH, Azain MJ (2013) Phytase, a new life for

an “old” enzyme. Annu Rev Anim Biosci 1: 283-309.

99. Li X, Fu GF, Fan YR, Liu WH, Liu XJ, Wang JJ, Xu GX (2003) Bifidobacterium

adolescentis as a delivery system of endostatin for cancer gene therapy: selective

inhibitor of angiogenesis and hypoxic tumor growth. Cancer Gene Ther 10: 105-111.

100. Li ZJ, Zhu H, Ma BY, Zhao F, Mao SH, Liu TG, He JP, Deng LC, Yi C, Huang Y

(2012) Inhibitory effect of Bifidobacterium infantis-mediated sKDR prokaryotic

expression system on angiogenesis and growth of Lewis lung cancer in mice. Bmc

Cancer 12.

101. Lisser S, Margalit H (1993) Compilation of E. coli mRNA promoter sequences.

Nucleic Acids Res 21: 1507-1516.

102. Long RT, Zeng WS, Chen LY, Guo J, Lin YZ, Huang QS, Luo SQ (2010)

Bifidobacterium as an oral delivery carrier of oxyntomodulin for obesity therapy:

inhibitory effects on food intake and body weight in overweight mice. Int J Obesity 34:

712-719.

103. Longley DB, Harkin DP, Johnston PG (2003) 5-fluorouracil: mechanisms of action

and clinical strategies. Nat Rev Cancer 3: 330-338.

104. Luo X, Li Z, Lin S, Le T, Ittensohn M, Bermudes D, Runyab JD, Shen SY, Chen J,

King IC, Zheng LM (2001) Antitumor effect of VNP20009, an attenuated Salmonella,

in murine tumor models. Oncol Res 12: 501-508.

105. Ma Y, Luo Y, Huang X, Song F, Liu G (2011) Construction of Bifidobacterium

infantis as a live oral vaccine that expresses antigens of the major fimbrial subunit

(CfaB) and LTB from Enterotoxigenic Escherichia coli. Microbiology 158(Pt 2):498-

504.

74

106. MacConaill LE, Fitzgerald GF, van Sinderen D (2003) Investigation of protein export

in Bifidobacterium breve UCC2003. Appl Environ Microbiol 69: 6994-7001.

107. Makrides SC (1996) Strategies for achieving high- level expression of genes in

Escherichia coli. Microbiol Rev 60: 512-538.

108. Maletzki C, Gock M, Klier U, Klar E, Linnebacher M (2010) Bacteriolytic therapy of

experimental pancreatic carcinoma. World J Gastroenterol 16: 3546-3552.

109. Mattarelli P, Holzapfel W, Franz CM, Endo A, Felis GE, Hammes W, Pot B, Dicks L,

Dellaglio F (2014) Recommended minimal standards for description of new taxa of the

genera Bifidobacterium, Lactobacillus and related genera. Int J Syst Evol Microbiol 64:

1434-1451.

110. Matteuzzi D, Brigidi P, Rossi M, Di D (1990) Characterization and molecular cloning

of Bifidobacterium longum cryptic plasmid pMB1. Lett Appl Microbiol 11: 220-223.

111. Matteuzzi D, Sgorbati B (1980) Taxonomy of the Genus Bifidobacterium. Ann Inst

Pasteur Mic B131: 92-92.

112. Mego M, Holec V, Drgona L, Hainova K, Ciernikova S, Zajac V (2013) Probiotic

bacteria in cancer patients undergoing chemotherapy and radiation therapy.

Complement Ther Med 21: 712-723.

113. Mesnil M, Yamasaki H (2000) Bystander effect in herpes simplex virus-thymidine

kinase/ganciclovir cancer gene therapy: role of gap-junctional intercellular

communication. Cancer Research 60: 3989-3999.

114. Mitchell DJ, Minchin RF (2008) E. coli nitroreductase/CB1954 gene-directed enzyme

prodrug therapy: role of arylamine N-acetlytransferase 2. Cancer Gene Ther 15: 758-

764.

115. Moon GS, Pyun YR, Park MS, Ji GE, Kim WJ (2005) Secretion of recombinant

pediocin PA-1 by Bifidobacterium longum, using the signal sequence for

bifidobacterial alpha-amylase. Appl Environ Microbiol 71: 5630-5632.

116. Mukherjee S, Maity S, Roy S, Ghorai S, Chakrabarti M, Agarwal R, Dutta D, Ghosh

AK, Das AK (2009) Cloning, overexpression, purification, crystallization and

preliminary X-ray diffraction analysis of glyceraldehyde-3-phosphate dehydrogenase

from Antheraea mylitta. Acta Crystallogr Sect F Struct Biol Cryst Commun 65: 937-

940.

75

117. Nagradova NK (2001) Study of the properties of phosphorylating D-glyceraldehyde-

3-phosphate dehydrogenase. Biochemistry (Mosc) 66: 1067-1076.

118. Nakamura T, Sasaki T, Fujimori M, Yazawa K, Kano Y, Amano J, Taniguchi S (2002)

Cloned cytosine deaminase gene expression of Bifidobacterium longum and

application to enzyme/pro-drug therapy of hypoxic solid tumors. Biosci Biotechnol

Biochem 66: 2362-2366.

119. Nallaseth FS, Anderson S (2013) A screen for over-secretion of proteins by yeast

based on a dual component cellular phosphatase and immuno-chromogenic stain for

exported bacterial alkaline phosphatase reporter. Microb Cell Fact 12: 36.

120. Nesvera J, Holatko J, Patek M (2012) Analysis of Corynebacterium glutamicum

Promoters and Their Applications. Subcell Biochem 64: 203-221.

121. Olstorpe M, Schnurer J, Passoth V (2009) Screening of yeast strains for phytase

activity. FEMS Yeast Res 9: 478-488.

122. O'Riordan K, Fitzgerald GF (1999) Molecular characterisation of a 5.75-kb cryptic

plasmid from Bifidobacterium breve NCFB2258 and determination of mode of

replication. Fems Microbiol Lett 174: 285-294.

123. Paetzel M, Karla A, Strynadka NC, Dalbey RE (2002) Signal peptidases. Chem Rev

102: 4549-4580.

124. Pallen MJ, Chaudhuri RR, Henderson IR (2003) Genomic analysis of secretion

systems. Curr Opin Microbiol 6: 519-527.

125. Palmano S, Kirkpatrick BC, Firrao G (2001) Expression of chloramphenicol

acetyltransferase in Bacillus subtilis under the contro l of a phytoplasma promoter.

FEMS Microbiol Lett 199: 177-179.

126. Palmer DH, Milner AE, Kerr DJ, Young LS (2003) Mechanism of cell death induced

by the novel enzyme-prodrug combination, nitroreductase/CB1954, and identification

of synergism with 5-fluorouracil. Br J Cancer 89: 944-950.

127. Palumbo MO, Kavan P, Miller WH, Jr., Panasci L, Assouline S, Johnson N, Cohen V,

Patenaude F, Pollak M, Jagoe RT, Batist G (2013) Systemic cancer therapy:

achievements and challenges that lie ahead. Front Pharmacol 4: 57.

128. Park M, Seo J, Kim J, Ji GN (2005) Heterologous gene expression and secretion in

76

Bifidobacterium longum. Lait 85: 1-8.

129. Park MS, Kim MJ, Ji GE (2007) Assessment of lipopolysaccharide-binding activity of

bifidobacterium and its relationship with cell surface hydrophobicity, autoaggregation,

and inhibition of interleukin-8 production. J Microbiol Biotechnol 17: 1120-1126.

130. Park MS, Kwon B, Shim JJ, Huh CS, Ji GE (2008a) Heterologous expression of

cholesterol oxidase in Bifidobacterium longum under the control of 16S rRNA gene

promoter of bifidobacteria. Biotechnol Lett 30: 165-172.

131. Park MS, Moon HW, Ji GE (2003) Molecular characterization of plasmid from

Bifidobacterium longum. J Microbiol Biotechnol 13: 457-462.

132. Park MS, Shin DW, Lee KH, Ji GE (1999) Sequence analysis of plasmid pKJ50 from

Bifidobacterium longum. Microbiology 145 (Pt 3): 585-592.

133. Park YS, Kim KH, Park JH, Oh IK, Yoon SS (2008b) Isolation and molecular

characterization of a cryptic plasmid from Bifidobacterium longum. Biotechnol Lett 30:

145-151.

134. Passariello A, Agricole P, Malfertheiner P (2014) A critical appraisal of probiotics (as

drugs or food supplements) in gastrointestinal diseases. Curr Med Res Opin 30: 1055-

1064.

135. Patek M, Holatko J, Busche T, Kalinowski J, Nesvera J (2013) Corynebacterium

glutamicum promoters: a practical approach. Microb Biotechnol 6: 103-117.

136. Patyar S, Joshi R, Byrav DS, Prakash A, Medhi B, Das BK (2010) Bacteria in cancer

therapy: a novel experimental strategy. J Biomed Sci 17: 21.

137. Petschow B, Dore J, Hibberd P, Dinan T, Reid G, Blaser M, Cani PD, Degnan FH,

Foster J, Gibson G, Hutton J, Klaenhammer TR, Ley R, Nieuwdorp M, Pot B, Relman

D, Serazin A, Sanders ME (2013) Probiotics, prebiotics, and the host microbiome: the

science of translation. Ann N Y Acad Sci 1306: 1-17.

138. Philippe D, Heupel E, Blum-Sperisen S, Riedel CU (2011) Treatment with

Bifidobacterium bifidum 17 partially protects mice from Th1-driven inflammation in a

chemically induced model of colitis. Int J Food Microbiol 149: 45-49.

139. Picard C, Fioramonti J, Francois A, Robinson T, Neant F, Matuchansky C (2005)

Review article: bifidobacteria as probiotic agents -- physiological effects and clinical

77

benefits. Aliment Pharmacol Ther 22: 495-512.

140. Poquet I, Ehrlich SD, Gruss A (1998) An export-specific reporter designed for gram-

positive bacteria: application to Lactococcus lactis. J Bacteriol 180: 1904-1912.

141. Poyart C, Trieu-Cuot P (1997) A broad-host-range mobilizable shuttle vector for the

construction of transcriptional fusions to beta-galactosidase in gram-positive bacteria.

FEMS Microbiol Lett 156: 193-198.

142. Preising J, Philippe D, Gleinser M, Wei H, Blum S, Eikmanns BJ, Niess JH, Riedel

CU (2010) Selection of bifidobacteria based on adhesion and anti- inflammatory

capacity in vitro for amelioration of murine colitis. Appl Environ Microbiol 76: 3048-

3051.

143. Pugsley AP (1993) The complete general secretory pathway in gram-negative bacteria.

Microbiol Rev 57: 50-108.

144. Quan C, Zhang L, Wang Y, Ohta Y (2001) Production of phytase in a low phosphate

medium by a novel yeast Candida krusei. J Biosci Bioeng 92: 154-160.

145. Quax WJ (1997) Merits of secretion of heterologous proteins from industrial

microorganisms. Folia Microbiol (Praha) 42: 99-103.

146. Quispe-Tintaya W, Chandra D, Jahangir A, Harris M, Casadevall A, Dadachova E,

Gravekamp C (2013) Nontoxic radioactive Listeria(at) is a highly effective therapy

against metastatic pancreatic cancer. Proc Natl Acad Sci U S A 110: 8668-8673.

147. Race PR, Lovering AL, White SA, Grove JI, Searle PF, Wrighton CW, Hyde EI

(2007) Kinetic and structural characterisation of Escherichia coli nitroreductase

mutants showing improved efficacy for the prodrug substrate CB1954. J Mol Biol 368:

481-492.

148. Rainov NG (2000) A phase III clinical evaluation of herpes simplex virus type 1

thymidine kinase and ganciclovir gene therapy as an adjuvant to surgical resection and

radiation in adults with previously untreated glioblastoma multiforme. Hum Gene Ther

11: 2389-2401.

149. Recchi C, Rauzier J, Gicquel B, Reyrat JM (2002) Signal-sequence- independent

secretion of the staphylococcal nuclease in Mycobacterium smegmatis. Microbiology

148: 529-536.

78

150. Relman DA (2013) Restoration of the gut microbial habitat as a disease therapy. Nat

Biotechnol 31: 35-37.

151. Reyes Escogido ML, De Leon Rodriguez A, Barba de la Rosa AP (2007) A novel

binary expression vector for production of human IL-10 in Escherichia coli and

Bifidobacterium longum. Biotechnol Lett 29: 1249-1253.

152. Rhim SL, Park MS, Ji GE (2006) Expression and secretion of Bifidobacterium

adolescentis amylase by Bifidobacterium longum. Biotechnol Lett 28: 163-168.

153. Riedel CU, Foata F, Goldstein DR, Blum S, Eikmanns BJ (2006a) Interaction of

bifidobacteria with Caco-2 cells-adhesion and impact on expression profiles. Int J Food

Microbiol 110: 62-68.

154. Riedel CU, Foata F, Philippe D, Adolfsson O, Eikmanns BJ, Blum S (2006b) Anti-

inflammatory effects of bifidobacteria by inhibition of LPS-induced NF-kappaB

activation. World J Gastroenterol 12: 3729-3735.

155. Riedel CU, Monk IR, Casey PG, Morrissey D, O'Sullivan GC, Tangney M, Hill C,

Gahan CG (2007) Improved luciferase tagging system for Listeria monocytogenes

allows real- time monitoring in vivo and in vitro. Appl Environ Microbiol 73: 3091-

3094.

156. Ross W, Gosink KK, Salomon J, Igarashi K, Zou C, Ishihama A, Severinov K,

Gourse RL (1993) A third recognition element in bacterial promoters: DNA binding by

the alpha subunit of RNA polymerase. Science 262: 1407-1413.

157. Rossi M, Brigidi P, Rodriguez AGVY, Matteuzzi D (1996) Characterization of the

plasmid pMB1 from Bifidobacterium longum and its use for shuttle vector construction.

Res Microbiol 147: 133-143.

158. Ruiz L, Alvarez-Martin P, Mayo B, de Los Reyes-Gavilan CG, Gueimonde M,

Margolles A (2011) Controlled gene expression in bifidobacteria by use of a bile-

responsive element. Appl Environ Microbiol. 78(2):581-585.

159. Salminen S, van Loveren H (2012) Probiotics and prebiotics: health claim

substantiation. Microb Ecol Health Dis 23.

160. Sanchez B, Urdaci MC, Margolles A (2010) Extracellular proteins secreted by

probiotic bacteria as mediators of effects that promote mucosa-bacteria interactions.

Microbiology 156: 3232-3242.

79

161. Sangrador-Vegas A, Stanton C, van Sinderen D, Fitzgerald GF, Ross RP (2007)

Characterization of plasmid pASV479 from Bifidobacterium pseudolongum subsp.

globosum and its use for expression vector construction. Plasmid 58: 140-147.

162. Sasaki T, Fujimori M, Hamaji Y, Hama Y, Ito K, Amano J, Taniguchi S (2006)

Genetically engineered Bifidobacterium longum for tumor-targeting enzyme-prodrug

therapy of autochthonous mammary tumors in rats. Cancer Sci 97: 649-657.

163. Scardovi V, Sgorbati B, Zani G (1971) Starch gel electrophoresis of fructose-6-

phosphate phosphoketolase in genus Bifidobacterium. J Bacteriol 106: 1036.

164. Schell MA, Karmirantzou M, Snel B, Vilanova D, Berger B, Pessi G, Zwahlen MC,

Desiere F, Bork P, Delley M, Pridmore RD, Arigoni F (2002) The genome sequence of

Bifidobacterium longum reflects its adaptation to the human gastrointestinal tract. Proc

Natl Acad Sci U S A 99: 14422-14427.

165. Schneewind O, Missiakas DM (2012) Protein secretion and surface display in Gram-

positive bacteria. Philos Trans R Soc Lond B Biol Sci 367: 1123-1139.

166. Scotto-Lavino E, Du G, Frohman MA (2006) 5' end cDNA amplification using classic

RACE. Nat Protoc 1: 2555-2562.

167. Serafini F, Turroni F, Guglielmetti S, Gioiosa L, Foroni E, Sanghez V, Bartolomucci

A, Motherway MO, Palanza P, van Sinderen D, Ventura M (2012) An efficient and

reproducible method for transformation of genetically recalcitrant bifidobacteria.

FEMS Microbiol Lett 333: 146-152.

168. Sgorbati B, Scardovi V, Leblanc DJ (1982) Plasmids in the Genus Bifidobacterium. J

Gen Microbiol 128: 2121-2131.

169. Shimamura S, Abe F, Ishibashi N, Miyakawa H, Yaeshima T, Araya T, Tomita M

(1992) Relationship between oxygen sensitivity and oxygen-metabolism of

Bifidobacterium species. J Dairy Sci 75: 3296-3306.

170. Shkoporov AN, Efimov BA, Khokhlova EV, Kafarskaia LI, Smeianov VV (2008)

Production of human basic fibroblast growth factor (FGF-2) in Bifidobacterium breve

using a series of novel expression/secretion vectors. Biotechnol Lett 30: 1983-1988.

171. Shortle D (1983) A genetic system for analysis of staphylococcal nuclease. Gene 22:

181-189.

80

172. Shouldice SR, Cho SH, Boyd D, Heras B, Eser M, Beckwith J, Riggs P, Martin JL,

Berkmen M (2010) In vivo oxidative protein folding can be facilitated by oxidation-

reduction cycling. Mol Microbiol 75: 13-28.

173. Stritzker J, Pilgrim S, Szalay AA, Goebel W (2008) Prodrug converting enzyme gene

delivery by L. monocytogenes. Bmc Cancer 8: 94.

174. Sun Z, Baur A, Zhurina D, Yuan J, Riedel CU (2012) Accessing the inaccessible:

molecular tools for bifidobacteria. Appl Environ Microbiol 78: 5035-5042.

175. Sun Z, He X, Brancaccio VF, Yuan J, Riedel CU (2014) Bifidobacteria exhibit LuxS-

dependent autoinducer 2 activity and biofilm formation. PLoS One 9: e88260.

176. Tabor S, Richardson CC (1985) A bacteriophage T7 RNA polymerase/promoter

system for controlled exclusive expression of specific genes. Proc Natl Acad Sci U S A

82: 1074-1078.

177. Takata T, Shirakawa T, Kawasaki Y, Kinoshita S, Gotoh A, Kano Y, Kawabata M

(2006) Genetically engineered Bifidobacterium animalis expressing the Salmonella

flagellin gene for the mucosat immunization in a mouse model. J Gene Med 8: 1341-

1346.

178. Takeuchi A, Matsumura H, Kano Y (2002) Cloning and expression in Escherichia

coli of a gene, hup, encoding the histone- like protein HU of Bifidobacterium longum.

Biosci Biotechnol Biochem 66: 598-603.

179. Tang W, He Y, Zhou S, Ma Y, Liu G (2009) A novel Bifidobacterium infantis-

mediated TK/GCV suicide gene therapy system exhibits antitumor activity in a rat

model of bladder cancer. J Exp Clin Cancer Res 28: 155.

180. Tangney M (2010) Gene therapy for cancer: dairy bacteria as delivery vectors. Discov

Med 10: 195-200.

181. Thorne SH, Barak Y, Liang W, Bachmann MH, Rao J, Contag CH, Matin A (2009)

CNOB/ChrR6, a new prodrug enzyme cancer chemotherapy. Mol Cancer Ther 8: 333-

341.

182. Toso JF, Gill VJ, Hwu P, Marincola FM, Restifo NP, Schwartzentruber DJ, Sherry

RM, Topalian SL, Yang JC, Stock F, Freezer LJ, Morton KE, Seipp C, Haworth L,

Mavroukakis S, White D, MacDonald S, Mao J, Sznol M, Rosenberg SA (2002) Phase

I study of the intravenous administration of attenuated Salmonella typhimurium to

81

patients with metastatic melanoma. J Clin Oncol 20: 142-152.

183. Tseng TT, Tyler BM, Setubal JC (2009) Protein secretion systems in bacterial-host

associations, and their description in the Gene Ontology. BMC Microbiol 9 Suppl 1: S2.

184. Turroni F, Foroni E, Pizzetti P, Giubellini V, Ribbera A, Merusi P, Cagnasso P,

Bizzarri B, de'Angelis GL, Shanahan F, van Sinderen D, Ventura M (2009) Exploring

the diversity of the bifidobacterial population in the human intestinal tract. Appl

Environ Microbiol 75: 1534-1545.

185. Turroni F, Serafini F, Foroni E, Duranti S, O'Connell Motherway M, Taverniti V,

Mangifesta M, Milani C, Viappiani A, Roversi T, Sanchez B, Santoni A, Gioiosa L,

Ferrarini A, Delledonne M, Margolles A, Piazza L, Palanza P, Bolchi A, Guglielmetti S,

van Sinderen D, Ventura M (2013) Role of sortase-dependent pili of Bifidobacterium

bifidum PRL2010 in modulating bacterium-host interactions. Proc Natl Acad Sci U S

A 110: 11151-11156.

186. van Kuilenburg AB (2004) Dihydropyrimidine dehydrogenase and the efficacy and

toxicity of 5-fluorouracil. Eur J Cancer 40: 939-950.

187. van Roosmalen ML, Geukens N, Jongbloed JD, Tjalsma H, Dubois JY, Bron S, van

Dijl JM, Anne J (2004) Type I signal peptidases of Gram-positive bacteria. Biochim

Biophys Acta 1694: 279-297.

188. Vass SO, Jarrom D, Wilson WR, Hyde EI, Searle PF (2009) E. coli NfsA: an

alternative nitroreductase for prodrug activation gene therapy in combination with

CB1954. Br J Cancer 100: 1903-1911.

189. Waidmann MS, Bleichrodt FS, Laslo T, Riedel CU (2011) Bacterial luciferase

reporters: the Swiss army knife of molecular biology. Bioeng Bugs 2: 8-16.

190. Warmann SW, Armeanu S, Heigoldt H, Ruck P, Vonthein R, Heitmann H, Seitz G,

Lemken ML, Bitzer M, Fuchs J, Lauer UM (2009) Adenovirus-mediated cytosine

deaminase/5-fluorocytosine suicide gene therapy of human hepatoblastoma in vitro.

Pediatr Blood Cancer 53: 145-151.

191. Weaver JD, Ullah AH, Sethumadhavan K, Mullaney EJ, Lei XG (2009) Impact of

assay conditions on activity estimate and kinetics comparison of Aspergillus niger

PhyA and Escherichia coli AppA2 phytases. J Agric Food Chem 57: 5315-5320.

192. Wegmann U, Horn N, Carding SR (2013) Defining the bacteroides ribosomal binding

82

site. Appl Environ Microbiol 79: 1980-1989.

193. Whelan K, Quigley EM (2013) Probiotics in the management of irritable bowel

syndrome and inflammatory bowel disease. Curr Opin Gastroenterol 29: 184-189.

194. Wille T, Blank K, Schmidt C, Vogt V, Gerlach RG (2012) Gaussia princeps luciferase

as a reporter for transcriptional activity, protein secretion, and protein-protein

interactions in Salmonella enterica serovar typhimurium. Appl Environ Microbiol 78:

250-257.

195. Wu MH, Pan TM, Wu YJ, Chang SJ, Chang MS, Hu CY (2010) Exopolysaccharide

activities from probiotic bifidobacterium: Immunomodulatory effects (on J774A.1

macrophages) and antimicrobial properties. Int J Food Microbiol 144: 104-110.

196. Xiong AS, Peng RH, Li X, Fan HQ, Yao QH, Guo MJ, Zhang SL (2003) Influence of

signal peptide sequences on the expression of heterogeneous proteins in Pichia pastoris.

Sheng Wu Hua Xue Yu Sheng Wu Wu Li Xue Bao (Shanghai) 35: 154-160.

197. Xu YF, Zhu LP, Hu B, Fu GF, Zhang HY, Wang JJ, Xu GX (2007) A new expression

plasmid in Bifidobacterium longum as a delivery system of endostatin for cancer gene

therapy. Cancer Gene Ther 14: 151-157.

198. Yamamoto S, Wada J, Katayama T, Jikimoto T, Nakamura M, Kinoshita S, Lee KM,

Kawabata M, Shirakawa T (2010) Genetically modified Bifidobacterium displaying

Salmonella-antigen protects mice from lethal challenge of Salmonella Typhimurium in

a murine typhoid fever model. Vaccine 28: 6684-6691.

199. Yao J, Wang JY, Lai MG, Li YX, Zhu HM, Shi RY, Mo J, Xun AY, Jia CH, Feng JL,

Wang LS, Zeng WS, Liu L (2011) Treatment of Mice with Dextran Sulfate Sodium-

Induced Colitis with Human Interleukin 10 Secreted by Transformed Bifidobacterium

longum. Mol Pharmaceut 8: 488-497.

200. Yazawa K, Fujimori M, Amano J, Kano Y, Taniguchi S (2000) Bifidobacterium

longum as a delivery system for cancer gene therapy: Selective localization and growth

in hypoxic tumors. Cancer Gene Therapy 7: 269-274.

201. Yi C, Huang Y, Guo ZY, Wang SR (2005) Antitumor effect of cytosine deaminase/5-

fluorocytosine suicide gene therapy system mediated by Bifidobacterium infantis on

melanoma. Acta Pharmacol Sin 26: 629-634.

202. Yildirim Z, Winters DK, Johnson MG (1999) Purification, amino acid sequence and

83

mode of action of bifidocin B produced by Bifidobacterium bifidum NCFB 1454. J

Appl Microbiol 86: 45-54.

203. Yin X, Yu B, Tang Z, He B, Ren J, Xiao X, Tang W (2013) Bifidobacterium infantis-

mediated HSV-TK/GCV suicide gene therapy induces both extrinsic and intrinsic

apoptosis in a rat model of bladder cancer. Cancer Gene Ther 20: 77-81.

204. Yin Y, Kou L, Wang JJ, Xu GX (2012) Therapeutic efficacy of Bifidobacterium

longum-mediated human interleukin-2 with endostatin or TRAIL in transplanted

tumors in mice. Exp Ther Med 3: 481-486.

205. Yu Z, Huang Z, Sao C, Huang Y, Zhang F, Yang J, Lian J, Zeng Z, Luo W, Zeng W,

Deng Q (2011) Bifidobacterium as an oral delivery carrier o f interleukin-12 for the

treatment of Coxsackie virus B3- induced myocarditis in the Balb/c mice. Int

Immunopharmacol.12(1):125-30.

206. Yuan J, Wang B, Sun Z, Bo X, Yuan X, He X, Zhao H, Du X, Wang F, Jiang Z,

Zhang L, Jia L, Wang Y, Wei K, Wang J, Zhang X, Sun Y, Huang L, Zeng M (2008)

Analysis of host- inducing proteome changes in bifidobacterium longum NCC2705

grown in Vivo. J Proteome Res 7: 375-385.

207. Zhang LL, Chen X, Zheng PY, Luo Y, Lu GF, Liu ZQ, Huang H, Yang PC (2010)

Oral Bifidobacterium modulates intestinal immune inflammation in mice with food

allergy. J Gastroen Hepatol 25: 928-934.

208. Zhu LP, Yin Y, Xing J, Li C, Kou L, Hu B, Wu ZW, Wang JJ, Xu GX (2009)

Therapeutic efficacy of Bifidobacterium longum-mediated human granulocyte colony-

stimulating factor and/or endostatin combined with cyclophosphamide in mouse-

transplanted tumors. Cancer Science 100: 1986-1990.

209. Zhurina D, Dudnik A, Waidmann MS, Grimm V, Westermann C, Breitinger KJ, Yuan

J, van Sinderen D, Riedel CU (2013) High-Quality Draft Genome Sequence of

Bifidobacterium longum E18, Isolated from a Healthy Adult. Genome Announc 1.

210. Zhurina D, Zomer A, Gleinser M, Brancaccio VF, Auchter M, Waidmann MS,

Westermann C, van Sinderen D, Riedel CU (2011) Complete Genome Sequence o f

Bifidobacterium bifidum S17. J Bacteriol 193: 301-302.

211. Zimmermann P, Zardi G, Lehmann M, Zeder C, Amrhein N, Frossard E, Bucher M

(2003) Engineering the root-soil interface via targeted expression of a synthetic phytase

84

gene in trichoblasts. Plant Biotechnol J 1: 353-360.

212. Zu C, Wang J (2014) Tumor-colonizing bacteria: a potential tumor targeting therapy.

Crit Rev Microbiol 40: 225-235.

213. Zuo F, Yu R, Feng X, Khaskheli GB, Chen L, Ma H, Chen S (2014) Combination of

heterogeneous catalase and superoxide dismutase protects Bifidobacterium longum

strain NCC2705 from oxidative stress. Appl Microbiol Biotechnol. [Epub ahead of print]

PMID: 24903816

85

7 APPENDICES

7.1 Bacteria strains

All bacteria strains, with short description and their sources were listed in table 7-1.

Table 7-1: Bacteria strains used in this study

Strain Relevant genotype or description Source

E. coli DH5a Type strain Invitrogen

E. coli DH10B Type strain Invitrogen

E. coli DH10B/pMDY23 E. coli DH10B harboring plasmid pMDY23 This study

E. coli DH10B/pMDY23-PluxS E. coli DH10B harboring plasmid pMDY23-PluxS This study

E. coli DH10B/pMDY23-P16S

rRNA

E. coli DH10B harboring plasmid pMDY23-P16S rRNA This study

E. coli DH10B/pMDY23-Phup E. coli DH10B harboring plasmid pMDY23-Phup This study

E. coli DH10B/pMDY23-Pgap E. coli DH10B harboring plasmid pMDY23-Pgap This study

E. coli DH10B/pMgapP E. coli DH10B harboring plasmid pMgapP This study

E. coli DH10B/pMgapS0P E. coli DH10B harboring plasmid pMgapS0P This study

E. coli DH10B/pMgapS1P E. coli DH10B harboring plasmid pMgapS1P This study

E. coli DH10B/pMgapS2P E. coli DH10B harboring plasmid pMgapS2P This study

E. coli DH10B/pMgapS3P E. coli DH10B harboring plasmid pMgapS3P This study

E. coli DH10B/pMgapS4P E. coli DH10B harboring plasmid pMgapS4P This study

E. coli DH10B/pMgapS5P E. coli DH10B harboring plasmid pMgapS5P This study

E. coli DH10B/pMgapS6P E. coli DH10B harboring plasmid pMgapS6P This study

E. coli DH10B/pMgapS7P E. coli DH10B harboring plasmid pMgapS7P This study

E. coli DH10B/pMgapS6C E. coli DH10B harboring plasmid pMgapS6C This study

B. longum NCC2705 Sequenced type strain (Schell et al.,

2002)

86

Table 7-1 (continued): Bacteria strains used in this study

Strain Relevant genotype or description Source

B. bifidum S17 Sequenced type strain (Zhurina et al.,

2011)

B. longum E18 Sequenced infants isolate (Zhurina et al.,

2013)

B. breve S27 Infants isolate Our lab

B. bifidum S17/pMDY23 B. bifidum S17 harboring plasmid pMDY23 This study

B. bifidum S17/pMDY23-PluxS B. bifidumS17 harboring plas mid pMDY23-PluxS This study

B. bifidum S17/pMDY23-P16S

rRNA

B. bifidumS17 harboring plas mid pMDY23-P16S

rRNA

This study

B. bifidum S17 /pMDY23-Phup B. bifidum S17 harboring plasmid pMDY23-Phup This study

B. bifidum S17/pMDY23-Pgap B. bifidum S17 harboring plasmid pMDY23-Pgap This study

B. bifidum S17/pMgapP B. bifidum S17 harboring plasmid pMgapP This study

B. bifidum S17/pMgapS0P B. bifidum S17 harboring plasmid pMgapS0P This study

B. bifidum S17/pMgapS1P B. bifidum S17 harboring plasmid pMgapS1P This study

B. bifidum S17/pMgapS2P B. bifidum S17 harboring plasmid pMgapS2P This study

B. bifidum S17/pMgapS3P B. bifidum S17 harboring plasmid pMgapS3P This study

B. bifidum S17/pMgapS4P B. bifidum S17 harboring plasmid pMgapS4P This study

B. bifidum S17/pMgapS5P B. bifidum S17 harboring plasmid pMgapS5P This study

B. bifidum S17/pMgapS6P B. bifidum S17 harboring plasmid pMgapS6P This study

B. bifidum S17/pMgapS7P B. bifidum S17 harboring plasmid pMgapS7P This study

B. bifidum S17/pMgapS6C B. bifidum S17 harboring plasmid pMgapS6C This study

87

7.2 Plasmids

All plasmids with short description and their sources were listed in table 7-2.

Table 7-2: Plasmids used in this study

Plasmid Description Source

pJET1.2 Linear b lunt end cloning vector Life Science

pMDY23 E. coli – Bifidobacterium sp. shuttle vector, promoter-less, gusA

reporter

Nestle

pMDY23-PluxS pMDY23 inserted 208 bp potential LuxS promoter region This study

pMDY23-P16S rRNA pMDY23 inserted 569 bp 16S rRNA promoter This study

pMDY23-Phup pMDY23 inserted 241 bp hup promoter region This study

pMDY23-Pgap pMDY23 inserted 209 bp gap promoter region This study

pMgapP gusA was replaced by appA in pMDY23-Pgap, for phytase

overexpression

This study

pMgapS0P S0 was fused in pMgapP, for secretion of phytase This study

pMgapS1P S1 was fused in pMgapP, for secretion of phytase This study

pMgapS2P S2 was fused in pMgapP, for secretion of phytase This study

pMgapS3P S3 was fused in pMgapP, for secretion of phytase This study

pMgapS4P S4 was fused in pMgapP, for secretion of phytase This study

pMgapS5P S5 was fused in pMgapP, for secretion of phytase This study

pMgapS6P S6 was fused in pMgapP, for secretion of phytase This study

pMgapS7P S7 was fused in pMgapP, for secretion of phytase This study

pZA-SchrR cloning vector containing synthetic codon adapted ChrR6 This study

pMgapS6C gusA was replaced by S6 fused ChrR6 in pMgap, for secretion of

ChrR6

This study

S0: pred icted signal peptide (SP) of sialidase from B.bifidum S17 encoded by gene bbif_1734

S1: pred icted SP of Tat-secreted glycosidase from B. longum E18 encoded by gene blong_0223

S2: pred icted SP of putative Tat-secreted pectin lyase-like protein from B. longum E18 encoded by gene

blong_1620

S3: pred icted SP of hypothetical secreted protein with NlpC/P60 domain from B. longum E18 encoded by

gene blong_1728

88

S4: pred icted SP of conserved hypothetical secreted protein with CHAP domain from B. longum E18

encoded by gene blong_0476

S5: pred icted SP of subtilisin family peptidase (lactocepin) from B. bifidum S17 encoded by gene bbif_1681

S6: pred icted SP of surface protein with Gram-positive anchor and Cna protein B-type domains from B.

bifidum S17 encoded by gene bbif_1761

S7: pred icted SP of conserved hypothetical secreted protein with CHAP domain from B. breve S27 encoded

by gene S27_0508

7.3 Oligonucleotides

Oligonucleotides were designed based on the DNA sequences available from the National

Center for Biotechnology Information (NCBI) and listed in table 7-3 with short description.

Table 7-3: Primers used in this study

Name Sequence Description

GSPR1 GGAGTGAGGGATGGTGTTGA Reverse transcription

GSPR3 GTTCTTGACCCACGGGATGT First round PCR

GSPR4 CATCCTTCTCGGCGTAGACC Second round PCR

PMF GCATATGACTAGTGAGCTC Colonies screen

PMR AGGACGTAAGTCGACATG Colonies screen

PgapF GAAGATCTGCGGAATGCCTCGCATCGAATC For. primer of Pgap

PgapR GGCCTCGAGCTCCCTTTGTAGGGTAGA Rev. primer of Pgap

PhupF GAAGATCTACGAGGAGTACCGGTGGATCG For. primer of Phup

PhupR GGCCTCGAGCTCCTGTACTCCTGCACAG Rev. primer of Phup

PluxF GAAGATCTTCGCTACCAATGCGTTCGG For. primer of PluxS

PluxR GGCCTCGAGCGCTCGTTGTCGTTGTCCAT Rev. primer of PluxS

P16SF GAAGATCTCGGTGTACTGGATCAATGATGATTCCG For. primer o f P16S rRNA

P16SR GGCCTCGAGA CCTCCTTACGCCGCCAGCGTTCATCC Rev. primer of P16S rRNA

PhyF GGCCTCGAGATG CAGAGTGAGCCGGAGCTGA For. primer of appA

PhyR CCCAAGCTTAGCCTCAGAGCATTCAGGTAAC Rev. primer of appA

S0F GGCCTCGAGATGGTTCGTTCGACCAAGCCATCGCT For. primer of S0

S1F GGCCTCGAGATGCATCAATCAACACGAAAGCGGTG For. primer of S1

S2F GGCCTCGAGATGACATCCCGTCAGGGCAGA For. primer of S2

S3F GGCCTCGAGATGAAGACCAAAACTGTAGCTTCT For. primer of S3

S4F GGCCTCGAGATGACGAACGTACGTGTGATCAA For. primer of S4

S5F GGCCTCGAGATGGCCAATAAGCAATGGCCTCGCTG For. primer of S5

89

Table 7-3 (continued): Primers used in this study

Name Sequence Description

S6F GGCCTCGAGATGAAATCACTGATGAAAAAGGTTTTCGC For. primer of S6

S7F GGCCTCGAGATGACTAAGGCGAAAACGATGAGGCCAG For. primer of S7

S0R TCAGCTCCGGCTCACTCTGGCTGGCCGCACTCGCGGTGGATA Rev. primer of S0

S1R TCAGCTCCGGCTCACTCTGATCGGCTGCCTGCGCGGT Rev. primer of S1

S2R TCAGCTCCGGCTCACTCTGTGACTGCGCGAACGCAGC Rev. primer of S2

S3R TCAGCTCCGGCTCACTCTGCTCAGCTGCAGTTGCTGTAG Rev. primer of S3

S4R TCAGCTCCGGCTCACTCTGGGTGTCTGCCTGGGCAGG Rev. primer of S4

S5R TCAGCTCCGGCTCACTCTGCGGAGCCGCGAGCGCGGTA Rev. primer of S5

S6R TCAGCTCCGGCTCACTCTGATCCGCTGCGTTGGCCGT Rev. primer of S6

S7R TCAGCTCCGGCTCACTCTGGGTGTTTGCCCGCGCCGG Rev. primer of S7

P0F TATCCACCGCGAGTGCGGCCAGCCAGAGTGAGCCGGAGCTGA For. primer of P0

P1F ACCGCGCAGGCAGCCGATCAGAGTGAGCCGGAGCTGA For. primer of P1

P2F GCTGCGTTCGCGCAGTCACAGAGTGAGCCGGAGCTGA For. primer of P2

P3F CTACAGCAACTGCAGCTGAGCAGAGTGAGCCGGAGCTGA For. primer of P3

P4F CCTGCCCAGGCAGACACCCAGAGTGAGCCGGAGCTGA For. primer of P4

P5F TACCGCGCTCGCGGCTCCGCAGAGTGAGCCGGAGCTGA For. primer of P5

P6F ACGGCCAACGCAGCGGATCAGAGTGAGCCGGAGCTGA For. primer of P6

P7F CCGGCGCGGGCAAACACCCAGAGTGAGCCGGAGCTGA For. primer of P7

Restriction sites are underlined. For., forward; Rev., reverse

7.4 Promoter sequences

DNA sequences for potential promoters of gene gap, luxS in B. bifidum S17, hup and 16S

rRNA in B. longum NCC2705 were cloned and sequenced as following.

Pgap (bbif_0612)

AGATCTGCGGAATGCCTCGCATCGAATCGCCGCAGGCTGTACAGACATATTTGTTAGCGTTAAC

GAAATATGGCCGTTTTATGCTCAAAGCAAGCGCGACACCGTTGCTCTAGTACAGACGGCGCATT

ACAGTAGACACTGTTGGTAAACAAAGGCCATAGCGCATCCATGCGCAAACGGTCTACCCTACA

AAGGGAG CTCGAG

PluxS (bbif_1299)

AGATCTGCAGTTCCTCGCCGTCAACGGCGTGGGTGACAGCAAGCTGGCCAAATACGGCAAGCG

ATTCATGGAGGCCATCGCCGGCTTCGACGATTGACAGCGCCGTTACAGCGCACCGGACTCGCCT

CGTGCCGTATATGCCGTATATGAGGAACGCCTATAGCCAAGAATGCGGCGAGACGCGCGCGAT

90

AGTGAGACAATGGTCGATGGAATGCCGGCTAACGGTCGTACGCGCCGGCGTCATATCGAGCAA

AAGGAGTCTGCG CTCGAG

Phup (bl_1798)

AGATCTCCGCCACTTTGCTGCACCGCGTGGTGTTCTACTGGCTGCGCATTCCGCTGGGCGCGGCG

GCCATGAAGTGGCTTGACAAGCATAATCTTGTCTGATTCGTCTATTTTCATACCCCCTTCGGGGA

AATAGATGTGAAAACCCTTATAAAACGCGGGTTTTCGCAGAAACATGCGCTAGTATCATTGATG

ACAACATGGACTAAGCAAAAGTGCTTGTCCCCTGACCCAAGAAGGATGCTTT CTCGAG

P16S rRNA (blr_01)

AGATCTACACGCCGAGCGAAGCCTGTCGTTTCAACGATTTGGGCTTATCTTCGTGCGCCCGATTT

GCGCGAGTCCGTGAATGCGTGTAAGTTATTCCCTTGCTGCCGCTCAGCGAGTGGTTCTCCTCCGG

GGAGCTGCGAGGTGGGTGAGTGGTGGTGGTTTGAGAACTCAAGAGCGTGTTTGTACTACTTCTT

TATAGTCAATGATTGCCAGTTCATTCCTCGCCTGATTGCCTGTCGTGGTGGTTGGGTACCCGGGA

GGGTTTGATGAGGGGTGAGGTTTTTTGAGGGCGTCCTTCCTTAAGGACGTGCTCGTCAATTTTTG

TTTTGAGAGTCATCTATTCGGATGCTTTTCATGAAGTTTTTTTGTGGAGGGTTCGATTCTGGCTCA

GGATGAACGCTGGCGGCGtaaggaggCTCGAG

7.5 Putative gap promoters of representative bifidobacteria

Sequences of putative gap promoters were extracted from the genomes of corresponding

bifidobacteria strains publicly in NCBI database.

B. bifidum S17, promoter of bbif_0612

>gi|310286520:c752203-751998

GGCCGCGGTGCGGAATGCCTCGCATCGAATCGCCGCAGGCTGTACAGACATA TTTGTTAGCGTT

AACGAAATATGGCCGTTTTATGCTCAAAGCAAGCGCGACACCGTTGCTCTAGTACAGACGGCGC

ATTACAGTAGACACTGTTGGTAAACAAAGGCCATAGCGCATCCATGCGCAAACGGTCTACCCTA

CAAAGGGAGAACAC

B. dentium Bd1, promoter of bdp_1514

>gi|283454959:1693570-1693730

AAACTCATTGTGAGCGTTCACGGAAACGACGTCGTTTTCTGAAACACACGACCGCCATACGTTG

CCCTAAGTACAGGTGACGCACTAAAGTGGGCGGTGTTGGTAAACAATGGCTTCATTGCGCCTCG

CGCATGTGTGGCCTACCCTAAAGGGAGAATTAC

91

B. adolescentis ATCC 15703, promoter of bad_1079

>gi|119025018:1338696-1338866

ACGCATGCGGGACATGCATGTGAGCGTTCACTGAAAATCGCACTGCATTTTGGAACACGCGCCT

CGGAACGTTGCCCTAAGTACAGGTGACGCACTACAGTGGACGGTGTTGGTAAACAATGGCTTCA

GTGTGCCTGGCACACGCGTGGCCTACCCTAAAGGGAGAATTAC

B. breve UCC2003, promoter of bbr_1233

>gi|476417161:1536384-1536616

CCGGACGTGAGCCCCAACTTCCGTGGATTCACGCCGTCGGTCCAATACGGGAAACCGCATATCG

GTTGCCGGCGATCCACAATGTGAGCGCTCACAAAAAAACGACGTTTTTGCTCAAAACGCACGCT

CAGACATTGCCATGTGTACAGAGTCGGCATTACAGTAGCAACTGTTGGTAAACAATGGCCCGGT

GTGCCAGAGCGCACCACGGGCACCCTACAAGGGAGAATTAC

B. longum E18, promoter of blong_1334

>gi|58036264:c266275-266034

CTTGCATGCCGCGCGCTTGCTCATGATGATTCGAGACATTCCTCCAAAGGAGAGGAAAAATCAC

ATGTCGGTTGCGGGCAATCCGAATTGTGAGCGCTCACAGAAAATACGGCATTTTTGCCCAAAAC

GCACGCTGAAACGTTGCCATGTGTACAGAGTCGGCATTACAGTAGCAACTGTTGGTAAACAATG

GCCCGGTGTGCCAAAGCGCGCCAAGGCCACCCTACAAGGGAGAATTACAT

7.6 Reporter gene appA

DNA sequence of mature form appA in Escherichia coli DH10B (GenBank: CP000948.1)

was used as a reference for cloning of reporter.

CAGAGTGAGCCGGAGCTGAAGCTGGAAAGTGTGGTGATTGTCAGTCGTCATGGTGTGCGTGCTC

CAACCAAGGCCACGCAACTGATGCAGGATGTCACCCCAGACGCATGGCCAACCTGGCCGGTAA

AACTGGGTTGGCTGACACCGCGNGGTGGTGAGCTAATCGCCTATCTCGGACATTACCAACGCCA

GCGTCTGGTAGCCGACGGATTGCTGGCGAAAAAGGGCTGCCCGCAGTCTGGTCAGGTCGCGATT

ATTGCTGATGTCGACGAGCGTACCCGTAAAACAGGCGAAGCCTTCGCCGCCGGGCTGGCACCTG

ACTGTGCAATAACCGTACATACCCAGGCAGATACGTCCAGTCCCGATCCGTTATTTAATCCTCTA

AAAACTGGCGTTTGCCAACTGGATAACGCGAACGTGACTGACGCGATCCTCAGCAGGGCAGGA

GGGTCAATTGCTGACTTTACCGGGCATCGGCAAACGGCGTTTCGCGAACTGGAACGGGTGCTTA

ATTTTCCGCAATCAAACTTGTGCCTTAAACGTGAGAAACAGGACGAAAGCTGTTCATTAACGCA

GGCATTACCATCGGAACTCAAGGTGAGCGCCGACAATGTCTCATTAACCGGTGCGGTAAGCCTC

GCATCAATGCTGACGGAGATATTTCTCCTGCAACAAGCACAGGGAATGCCGGAGCCGGGGTGG

GGAAGGATCACCGATTCACACCAGTGGAACACCTTGCTAAGTTTGCATAACGCGCAATTTTATT

92

TGCTACAACGCACGCCAGAGGTTGCCCGCAGCCGCGCCACCCCGTTATTAGATTTGATCAAGAC

AGCGTTGACGCCCCATCCACCGCAAAAACAGGCGTATGGTGTGACATTACCCACTTCAGTGCTG

TTTATCGCCGGACACGATACTAATCTGGCAAATCTCGGCGGCGCACTGGAGCTCAACTGGACGC

TTCCCGGTCAGCCGGATAACACGCCGCCAGGTGGTGAACTGGTGTTTGAACGCTGGCGTCGGCT

AAGCGATAACAGCCAGTGGATTCAGGTTTCGCTGGTCTTCCAGACTTTACAGCAGATGCGTGAT

AAAACGCCGCTGTCATTAAATACGCCGCCCGGAGAGGTGAAACTGACCCTGGCAGGATGTGAA

GAGCGAAATGCGCAGGGCATGTGTTCGTTGGCAGGTTTTACGCAAATCGTGAATGAAGCACGC

ATACCGGCGTGCAGTTTGTAA

7.7 SP sequences

SPs were predicted by SignalP and corresponding DNA sequences were listed as following

including 6 nucleotides after predicted cleavage sites.

S0 (bbif_1734)

ATGGTTCGTTCGACCAAGCCATCGCTGCTGCGCCGTCTTGGCGCGCTGGTGGCGGCCGCTGCCA

TGCTGGTGGTACTGCCCGCAGGGGTATCCACCGCGAGTGCGGCCAGC

S1 (blong_0223)

ATGCATCAATCAACACGAAAGCGGTGGCTTGCGTCAATCGGCGCGGTTGCAGCGGTCGCCACAC

TGGCCACCGGCGGTGCAGTCACCGCGCAGGCAGCCGAT

S2 (blong_1620)

ATGACATCCCGTCAGGGCAGACAAGCCATAGCCGCGACGGCCGCAATGGGCGTGGCCGTCGCG

CTGGCGTTGCCGACCGCTGCGTTCGCGCAGTCA

S3 (blong_1728)

ATGAAGACCAAAACTGTAGCTTCTTCCATTGTTGCCGCGATTGTCTCCTGTGCCTGCTTGTTTAT

GTTTGTTCCTACAGCAACTGCAGCTGAG

S4 (blong_0476)

ATGACGAACGTACGTGTGATCAAGCCCGCACTGGCGGCACTGGTGGCCGCCGCAGCCTGTGTGG

GAGGTCTGGCGTTCAGCTCGGCACAGCCTGCCCAGGCAGACACC

S5 (bbif_1681)

ATGGCCAATAAGCAATGGCCTCGCTGGGTTGCCGCTGTAGCGGCCCTGGCGATGGCAGGCAGCC

TGCCCGGTACCGCGCTCGCGGCTCCG

93

S6 (bbif_1761)

ATGAAATCACTGATGAAAAAGGTTTTCGCTGCCGCCGCGGCGATTGCCACCGTATTTGGATTGG

CTGCGACGACAGTCGCCACGGCCAACGCAGCGGAT

S7 (S27_0508)

ATGACTAAGGCGAAAACGATGAGGCCAGTGTTGGCCGCATGCGCGGCCAGCGCCATATGCGTG

GCCGGGCTGCTGGCCAGCCCTGTTCAGCCGGCGCGGGCAAACACC

7.8 chrR6 sequences

DNA sequences of E. coli and B. bifidum S17 codon adapted chrR6 were supplied as

following.

E. coli chrR6, GenBank: DQ987901.1

ATGTCTGAAAAATTGCAGGTGGTTACGTTACTGGGGAGCCTGCGCAAAGGCTCATTTAATGGCA

TGGTTGCACGTACCCTGCCGAAAATTGCTCCGGCGAGCATGGAAGTCAATGCGTTACCATCCAT

TGCCGACATTCCCTTGTATGACGCTGACGTACAGCAGGAAGAAGGTTTTCCAGCAACGGTTGAA

GCTCTGGCGGAACAGATCCGTCAGGCTGACGGTGTGGTGATCGTCACGCCGGAATATAACTACT

CGGTACCGGGTGGGCTGAAAAATGCCATCGACTGGCTTTCCCGCCTGCCGGATCAACCGCTGGC

AGGTAAACCGGTGTTGATTCAGACCAGCTCAATGGGCGCGATTGGCGGCGCGCGCTGTCAGAAT

CACCTGCGCCAGATTCTGGTCTTCCTCGATGCAATGGTGATGAACAAGCCGGAATTTATGGGCG

GCGTGATTCAGAACAAAGTTGATCCGCAAAACGGAGAAGTGATTGATCAGGGTACGCTGGACC

ACCTGACCGGGCTATTGACCGCATTTGGTGAGTTTATTCAGCGAGTTAAGATCTAA

B. bifidum S17 codon adapted chrR6

ATGTCGGAAAAGCTCCAGGTGGTCACGTTGCTCGGCAGTCTGCGCAAAGGGAGCTTCAACGGC

ATGGTCGCCCGTACCCTTCCGAAGATTGCACCGGCGAGCATGGAGGTGAATGCCCTGCCTTCGA

TAGCGGACATACCCCTGTATGACGCGGATGTGCAGCA GGAAGAAGGCTTTCCGGCTACCGTCGA

AGCCTTGGCCGAACAGATACGCCAAGCCGATGGAGTGGTCATCGTTACCCCGGAGTACAACTAC

TCCGTACCAGGCGGACTGAAGAACGCGATCGACTGGCTCTCCAGACTGCCGGATCAACCCCTTG

CTGGTAAGCCGGTCCTGATCCAGACCTCCTCCATGGGCGCGATTGGCGGTGCAAGGTGCCAGAA

CCACTTGCGGCAGATCCTGGTGTTCCTGGACGCCATGGTCATGAACAAACCCGAGTTCATGGGT

GGCGTGATCCAGAACAAGGTAGACCCGCAGAATGGCGAGGTTATCGACCAGGGAACGCTGGAC

CATCTCACTGGGCTGCTGACAGCCTTTGGCGAGTTCATCCAACGCGTGAAAATTTAA

94

ABBREVIATIONS

% percent

Ω Ohm

σ sigma

μ micro

℃ degree centigrade

4-HX 4-hydroxylamine

4-MU 4-methylumbelliferone

4-MUG 4-methylumbelliferxl-β-D-glucuronide

5-FC 5-fluorocytosine

5-FU 5-fluorouracil

5’-RACE Rapid amplification of cDNA 5’-end

AT adenine and thymine

ATP adenosine triphosphate

B. Bifidobacterium

BCA bicinchoninic acid

BLAST basic local alignment search tool

bp base pair

BSA bovine serum albumin

CB1954 5-aziridin- l-yl-2,4-dinitrobenzamide

CD cytosine deaminase

cDNA complementary deoxyribonucleic acid

CSB citrate sucrose buffer

95

dATP deoxyadenosine triphosphate

ddH2O double distilled water

dH2O distilled water

DMSO dimethyl sulfoxide

DNA deoxyribonucleic acid

DNase deoxyribonuclease

DTT dithiothreitol

E. Escherichia

EDTA Ethylenediaminetetraacetatic acid

EFSA theEuropean Food Safety Authority

EP Eppendorf

FGF-2 basic fibroblast growth factor

Fig. figure

g gram

GAPDH glyceraldehyde 3-phosphate dehydrogenase

GC guanine and cytosine

GCV ganciclovir

GDEPT gene directed enzyme prodrug therapy

GFP green fluorescent protein

GIT gastrointestinal tract

GRAS generally recognized as safe

GUS β-glucuronidase

h hour

HEPES N-2-hydroxyethylpiperazine-N-2-ethane sulfonic acid

HPSF high purity salt free

96

HSV-TK herpes simplex virus thymidine kinase

IBD inflammatory bowel disease

ICAM-1 intercellular adhesion molecule 1

IECs intestinal epithelial cells

IFN Interferon

l liter

IL interleukin

LB Luria Bertani

LDF linear discrimination function

LPS lipopolysaccharides

m mili

min minute

mRNA messenger ribonucleic acid

MRS de Man-Rogosa-Sharpe medium

NADH nicotinamide adenine dinucleotide (reduced)

NCBI National Center for Biotechnology Information

NF-κB nuclear factor kappa-light-chain-enhancer of activated B

cells

NNPP neural network promoter prediction

NTR nitroreductase

OD600 optical density at wavelength 600nm

OE-PCR overlap extension polymerase chain reaction

ORF open reading frame

PCR polymerase chain reaction

PSM phytase screening medium

97

RBS ribosome binding site

rcf relative centrifuge force

RCM reinforced clostridial medium

rpm resolutions per minute

RNA ribonucleic acid

RNAP RNA polymerase

s second

SDS sodium dodecyl sulphate

SOD superoxide dismutase

SP signal peptide

SPase signal peptidase

Spc spectinomycin

TAE tris acetate EDTA buffer

Tat two arginine translocation

TCA trichloroacetic acid

TdT terminal deoxynucleotidyl transferase

TNF tumor necrotic factor

tRNA transfer RNA

TSS transcription start site

U unit

UV ultraviolet

V volt

v/v volume per volume

w/v weight per volume

98

PUBLICATIONS

1. Sun Z, Baur A, Riedel CU. A secretion reporter system and heterologous secretion of

nitroreductase in Bifidobacterium bifidum S17 for tumor therapy. Manuscript in

preparation

2. Sun Z, Westermann C, Yuan J, Riedel CU. Experimental determination and

characterization of the gap promoter of Bifidobacterium bifidum S17. Bioengineered.

2014. 5(6):1-7.

3. Sun Z, He X, Brancaccio VF, Yuan J, Riedel CU. Bifidobacteria exhibit LuxS-

dependent autoinducer 2 activity and biofilm formation. PLoS ONE. 2014. 9(2):

e88260.

4. Sun Z, Baur A, Zhurina D, Yuan J, Riedel CU. Accessing the inaccessible: molecular

tools for bifidobacteria. Appl Environ Microbiol. 2012. 78(15):5035-42.

99

ACKNOWLEDGEMENT

During the last four years, many of you contributed in a certain way to the completion of

this thesis. Without those help and support from you, this thesis cannot be done timely. I

would like to express my sincere gratitude to all of you listed below.

Prof. Dr. Bernhard Eikmanns, thank you for giving me the opportunity to be a PhD student

under your official supervision. Your kind help and supportive comments in the institute

seminars are much appreciated. Prof. Dr. Peter Dürre also supplied valuable suggestions.

Dr. Christian Riedel, you supervised my study from writing proposa l to finalizing results.

Your contribution on the completion of the thesis is highly acknowledged. Thank you very

much for your patience on my work, and spending your limited time on correct every

pieces of my paper carefully. I cannot manage to conduct the project smoothly without

your firm support and constructive suggestions.

Much appreciation goes to Prof. Dr. Stefan Binder. Thank you for spending time to review

my thesis. Gratitude to Prof. Dr. Steven Jansen, Prof. Dr. Marion Kazda, and Dr. Frank

Rosenau as well, for you support as examiners in my defense.

I would also like to thank all former and present members of AG Riedel, especially Dr.

Daria, Dr. Marita, Dr. Mark, Verena, Marion, Annika, and Christina for their support in lab

and tolerance to share a crowded office.

This project would not have been possible without the financial support provided by the

German Academic Exchange Service (DAAD). I am very grateful to DAAD former staff

Mr. David Hildebrand. Thank you very much for your kind support in the early phase

when I reached Germany.

At last, I want to express deep gratitude to my wife and my father. Without their support

and sacrifice, I will be impossible to pursue higher education abroad for years. I dedicate

this thesis to them.

100

DECLARATION

I hereby declare that this thesis presented herein was composed by me independently, and

its content was originated entirely from my own work during the PhD study in Ulm

University. Information from the published and unpublished work of others has been fully

acknowledged in the text and cited in references.

I certify that this thesis has not been submitted in any form for acquiring another degree at

any university or institute. I also declare that the work presented in the thesis is original

and I was the person primarily involved in those related publications.

Ulm, July 2014

Zhongke Sun