+ All Categories
Home > Documents > Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an...

Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an...

Date post: 24-Jan-2021
Category:
Upload: others
View: 3 times
Download: 0 times
Share this document with a friend
80
Infection Biology of Chlamydia pneumoniae Leslie Bailey Department of Molecular Biology Laboratory for Molecular Infection Medicine Sweden (MIMS) Department of Public Health and Clinical Medicine, Cardiology Umeå University Umeå 2008
Transcript
Page 1: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

Infection Biology of Chlamydia pneumoniae

Leslie Bailey

Department of Molecular BiologyLaboratory for Molecular Infection Medicine Sweden (MIMS)Department of Public Health and Clinical Medicine, Cardiology

Umeå UniversityUmeå 2008

Page 2: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

Department of Molecular BiologyDepartment of Public Health and Clinical Medicine, Cardiology

Department of Community Medicine and Rehabilitation, GeriatricsUmeå University

SE-901 87, Umeå, Sweden

Copyright © 2008 by Leslie BaileyISBN: 978-91-7264-532-5

Printed by Print & Media, Umeå University, Umeå, Sweden, 2008, 2004192

Page 3: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

To Lisa and Elliot

Page 4: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

•••

Page 5: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

i

ABSTRACT

There are two main human pathogens in the family of

Page 6: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

ii

TABLE OF CONTENTS

LIST OF PUBLICATIONS .............................................................................................. v Additional papers not included in thesis ................................................................. v

INTRODUCTION ............................................................................................................ 1 Chlamydiaceae ........................................................................................................ 1 Historical view of Chlamydia ................................................................................. 1 TWAR – an orienteer-related disease? ................................................................... 2 Taxonomy ............................................................................................................... 2

Structure of Chlamydia in general and C. pneumoniae in particular ........................... 4 Morphology ............................................................................................................ 4 LPS ......................................................................................................................... 4 Peptidoglycan .......................................................................................................... 4 Polymorphic membrane proteins ............................................................................ 5 Major outer membrane protein ............................................................................... 5 Heat shock proteins ................................................................................................. 6

The developmental cycle ............................................................................................. 7 The key players of Chlamydia ................................................................................ 7 Attachment .............................................................................................................. 8 Internalization ......................................................................................................... 8 Internalization: Endocytosis - mediated via activation of the actin cytoskeleton ... 8 The inclusion - a chlamydial protected site ............................................................. 9 Inclusion membrane proteins ................................................................................ 10 Primary differentiation and proliferation .............................................................. 10 Re-differentiation .................................................................................................. 11 Exit from the host cell ........................................................................................... 11 Bacteria and iron ................................................................................................... 12 Chlamydia and iron ............................................................................................... 12 Chlamydial persistence ......................................................................................... 12

Clinical manifestations .............................................................................................. 14 Acute infection ...................................................................................................... 14 Chronic infection .................................................................................................. 14

C. pneumoniae and atherosclerosis ........................................................................... 15 Atherosclerosis ...................................................................................................... 15 Atherosclerosis and animal models ....................................................................... 15 Lipoprotein (a) as a mouse model for atherosclerosis ........................................... 16 C. pneumoniae in atherosclerosis .......................................................................... 17 Antimicrobial trials have questioned the C. pneumoniae infection theory ........... 17 C. pneumoniae and inflammation of the vascular system ..................................... 18

••

Page 7: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

iii

Pro-inflammatory cytokines .................................................................................. 18 C. pneumoniae and pro-inflammatory cytokines .................................................. 19 Bacterial infection of bone demonstrates similar cytokine induction as in

atherosclerosis ....................................................................................................... 19 The type three secretion system (T3SS) .................................................................... 21

Historical observations of a possible T3SS ........................................................... 21 First evidence of a chlamydial T3SS .................................................................... 22 Composition of the chlamydial T3S injectisome .................................................. 23 Basal apparatus ..................................................................................................... 23 The Chlamydia translocon .................................................................................... 24 Chaperones of the T3SS ........................................................................................ 25 Transcriptional regulation of T3SS chaperones .................................................... 26 The needle complex .............................................................................................. 26 Gating of T3S machinery ...................................................................................... 27 Effector proteins.................................................................................................... 28 Other secreted proteins ......................................................................................... 29 Extracellular T3SS ................................................................................................ 30 Intracellular T3SS ................................................................................................. 30 Temporal transcription of T3SS genes .................................................................. 31 Stress of the T3SS ................................................................................................. 32

Antibiotics ................................................................................................................. 33 Antimicrobial resistance is a global problem ........................................................ 33 Antibiotic treatment of C. pneumoniae infection .................................................. 33

Small molecules ........................................................................................................ 34 Chemical genetics and small chemical molecules ................................................ 34 Small molecules as inhibitors of virulence ........................................................... 34 Small Molecules and inhibition of T3SS .............................................................. 34

OBJECTIVES OF THIS THESIS ............................................................................. 36 RESULTS AND DISCUSSION ..................................................................................... 37

Paper I ....................................................................................................................... 37 Screening of T3SS inhibitors using Y. pseudotuberculosis T3SS ......................... 37 INP0010 inhibits C. pneumoniae intraceullar propagation ................................... 38 INP0010 shows diverse effect in C. trachomatis .................................................. 38 INP0400 is active against both C. pneumoniae and C. trachomatis. .................... 38 Treatment with INP0010 inhibits secretion of putative T3SS effector proteins ... 39

Paper II ...................................................................................................................... 40 INP0400 inhibits proliferation but not primary differentiation ............................. 40 INP400 inhibits secretion of putative effectors ..................................................... 40 INP0400 treatment during the late cycle promotes bacterial dissociation from

the inclusion membrane ........................................................................................ 41

Page 8: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

iv

Paper III ..................................................................................................................... 42 DNA is preferable to RNA for normalization of gene expression ........................ 42 INP0010 decreases initiation of transcription ....................................................... 43 Expression of T3SS in the presence of INP0010 .................................................. 43

Paper IV .................................................................................................................... 44 INP0010 inhibits early- and mid-developmental cycle ......................................... 44 Iron initiates proliferation but not terminal re-differentiation in the presence of

INP0010 ................................................................................................................ 44 INP0010 blocks secretion of IncB into the host cell cytoplasm ............................ 45

Paper V ...................................................................................................................... 47 C. pneumoniae infection decreases bone mineral density in mice ........................ 47 C. pneumoniae growth in hFOBs increases production of IL-6 and expression

of RANKL ............................................................................................................ 47 CONCLUSIONS ............................................................................................................ 49 ACKNOWLEDGEMENTS ............................................................................................ 50 REFERENCES ............................................................................................................... 52

••

Page 9: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

v

LIST OF PUBLICATIONS This thesis is based on the following articles and manuscripts which will be referred

to by their roman numerals I-V.

I. Bailey L, Gylfe Å, Sundin C, Muschiol S, Elofsson M, Nordström P,

Henriques-Normark B, Lugert R, Waldenström A, Wolf-Watz H, and

Bergström S. Small molecule inhibitors of type III secretion in Yersinia block the

Chlamydia pneumoniae infection cycle. FEBS Lett. 2007 Feb 20;581(4):587-95.

II. Muschiol S, Bailey L, Gylfe Å, Sundin C, Hultenby K, Bergström S,

Elofsson M, Wolf-Watz H, Normark S and Henriques-Normark B. A small-

molecule inhibitor of type III secretion inhibits different stages of the infectious

cycle of Chlamydia trachomatis. Proc Natl Acad Sci U S A. 2006 Sep

26;103(39):14566-71.

III. Bailey L, Engström P, Önskog T, Bergström S and Johansson J. The T3SS-

inhibitor INP0010 decreases transcription initiation and modulates mRNA stability

during early development in Chlamydia pneumoniae. Submitted.

IV. Bailey L, Muschiol S, Engström P, Nordström P Henriques-Normark B,

Waldenström A, Gylfe Å, Elofsson M, Wolf-Watz H and Sven Bergström. The

Type three secretion: a possible mechanism for Chlamydia pneumoniae to utilize

intracellular iron acquisition - Demonstrated by using the T3SS-inhibitor INP0010.

Manuscript.

V. Bailey L, Engström P, Nordström A, Waldenström A, Bergström S and

Nordström P. Chlamydia pneumoniae Infection Results in Generalized Bone Loss

in Mice. Submitted.

Additional papers not included in thesis

(VI). Steptoe A, Shamaei-Tousi A, Gylfe A, Bailey L, Bergström S, Coates AR,

Henderson B. Protective effect of human heat shock protein 60 suggested by its

association with decreased seropositivity to pathogens.Clin Vaccine Immunol. 2007

Feb;14(2):204-7.

(VII). Ovchinnikova O, Gylfe Å, Bailey L, Nordström A, Bergström S,

Waldenström A, Hansson GK and Nordström N. Osteoprotegerin Promotes

Fibrous Cap Formation in Atherosclerotic Lesions of apoE deficient Mice.

Submitted.

Page 10: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

1

INTRODUCTION

Chlamydiaceae There are two main species (spp.) of the family Chlamydiaceae that are common

pathogens in humans. Two of the spp, Chlamydia trachomatis and C. pneumoniae, are

common pathogens in humans, whereas the other species occur mainly in animals

(summarized in Table 1). The sexually transmitted C. trachomatis has been isolated

only from humans and is comprised of two human biovars (trachoma and

lymphogranuloma venereum, LGV), including a total of 18 serovars, whereas C.

pneumoniae consists of one human biovar (TWAR) and two animal biovars, one

infecting horses (biovar equine) and the other infecting frogs1 and koalas (biovar

koala)2.

Historical view of Chlamydia Initially, these bacteria were first believed to be viruses, due to their obligate

intracellular requirements. In 1964, Schechter et al., revealed the truth about Chlamydia

when he found the presence of both RNA and DNA as well as cell wall structures,

demonstrated by electron microscopy3. However, Chlamydia was grouped with

Rickettsia until the genus Chlamydia was established and two different strains,

Chlamydia trachomatis and Chlamydia psittaci were isolated4.

Since then, several new chlamydial strains have been isolated. The first report of a

Chlamydia pneumoniae-like bacterium was described in 1965, believed to be an

atypical strain of C. psittaci. The bacterium was isolated from the eye of a child during a

trachoma vaccine trial in Taiwan, and thus given the name TW-1835. The first report of

C. pneumoniae causing clinical manifestations came in 1983, when the bacterium was

isolated in the United States from a throat swab of a university student suffering from

pharyngitis. This isolate was termed AR-39, due to its isolation as an Acute Respiratory

pathogen6. This group of organisms was eventually called TWAR, which was an

acronym for the first two isolates, i.e. TW-183 and AR-39. At that time, TWAR was

considered a human C. psittaci strain spreading from human to human without an avian

or mammalian host 6. Interestingly, TWAR was found to be a serologically unique

group among the known C. psittaci isolates. These showed differential and milder

pathogenic properties when grown in cell culture or inoculated into mice or chicken

embryos. In 1989, the strain was identified as a separate species within the genus

Chlamydia and named C. pneumoniae7. To fulfill the criteria to become a member of

the genus Chlamydia, the strain had to meet the following standards: obligate

intracellular parasitism, unique developmental cycle, share genus-specific

••

Page 11: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

2

lipopolysaccharide (LPS), and complement fixation antigen and comparable guanine-to-

cytosine content7.

TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected cardiac

death (SUD) among orienteers in Sweden8-10

. During the years 1979-92, 16 young

Swedish orienteers, of whom 14 were of the top elite level, suffered SUD, most of them

while exercising. Serological examination in all 5 cases where serum was available for

screening with a wide range of microbial antigens revealed positive serology for C.

pneumoniae10

. Accordingly, in 2001, it was concluded that Swedish elite orienteers do

not have a higher exposure rate to C. pneumoniae than blood donors11

. Therefore, C.

pneumoniae is no longer considered an orienteer-related disease, mostly due to its high

sero-prevalence in humans. Moreover, Wesslen et al. suggested that sub-acute infection

or re-activation of Bartonella played an important pathogenetic role in the Swedish

orienteers who suffered SUD12

. On the contrary, there is no association between elite

orienteers and prevalence of Bartonella antibody positivity in Denmark13

. Thus, these

results neither support nor contradict the possibility that microorganisms were involved

in some of the SUDs among young Swedish orienteers in the 90’s.

Taxonomy In 1999, a revised taxonomy of the family Chlamydiaceae was suggested based on 16S

and 23S rRNA sequence similarity clusters14

. Everett et al. recommended that the genus

Chlamydia be divided into two genera, Chlamydia and Chlamydophila. Altogether, the

revised genus family contains nine species (Table 1). However, the new reclassification

has been considered unnecessary by the Chlamydia research community15

. The authors

disagreed on the need for a new genus called Chlamydophila. Their objection to

division of the species into two separate genuses was based on the small differences

among genomes of C. trachomatis, C. psittaci and C. pneumoniae, and the report from

Tanner et al, which presents a taxonomically and phylogenetically coherent grouping

into one genus16

. Another aspect that was considered important was that the well-

recognized name Chlamydia had been accepted by the public and any change of the

name would cause confusion. In agreement with the arguments against the genus

division, the old classification has been used in the present work.

Page 12: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

3

TABLE 1. The family Chlamydiaceae as proposed by Everett et al. 199914

Species Typical host (s) Route of entry

Chlamydia

C. trachomatis Human Pharyngeal, ocular, genital, rectal

C. suis Swine Pharyngeal

C. muridarium Mouse and hamster Pharyngeal, genital

Chlamydophila*

C. psittaci Birds Pharyngeal, ocular, genital

C. pneumoniae Human, koala, horse, frog Pharyngeal, ocular

C. pecorum Domastic animals, koalas Oral

C. felis House cat Pharyngeal, ocular, genital

C. caviae Guinea pig Pharyngeal, ocular, genital, urethral

C.abortus Mammals Oral, genital

Modified from2. The Chlamydophila classification is not refered to in this thesis*

••

Page 13: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

4

Structure of Chlamydia in general and C. pneumoniae in particular

Morphology C. pneumoniae does not only differ in genomic content when compared to C.

trachomatis17,18

. Morphologically, C. pneumoniae can be distinguished from other

chlamydial species by the pear-shaped structure of its elementary bodies (EBs) as well

as a loose outer membrane19

. However, the pear-shaped appearance is not a

characteristic feature of all C. pneumoniae isolates20-23

. Thus, Wolf et al. demonstrated

by scanning electron microscopy the presence of a large periplasmic space between

the

cytoplasmic and the outer membranes of C. pneumoniae elementary bodies (EBs)24

.

LPS C. pneumoniae, like other Gram-negative bacteria, has LPS in its cell membrane.

Chlamydial LPS is a genus-specific group of antigens, which is shown to be surface-

exposed and present in both EBs and RBs25

. Therefore, it is suitable as a marker for

chlamydial infections. However, the chlamydial version of LPS seems to have lower

endotoxin activity than enterobacterial LPS and therefore is less potent as an inducer of

inflammation26,27

. Structurally and physiologically, chlamydial LPS is similar to the

rough form of LPS in enterobacteria, but is not identical among chlamydial spp.28,29

.

Peptidoglycan During all developmental stages, Chlamydia appears to be surrounded by two layers

constituting the cell membrane, a characteristic feature of Gram-negative bacteria.

However, unlike other Gram-negative bacteria, Chlamydia does not have a

peptidoglycan layer in the periplasmic space30-32

. In contrast, its cell envelope contains

penicillin-binding proteins, and the presence of peptide cross-links analogous to those

between peptidoglycan backbones has been suggested30

. It has been known for a long

time that the growth of Chlamydia is sensitive to penicillin, despite the absence of

peptidoglycan33

. This paradox is known as the chlamydial anomaly32

. Furthermore,

genomic sequence comparison of C. pneumoniae and C. trachomatis revealed the

presence of genes for peptidoglycan synthesis, membrane assembly and recycling,

respectively17,18

. Peptidoglycan has been suggested to be necessary for RB cell division

and to be produced during growth within host cells34,35

. However, many questions still

remain unanswered regarding how, when and why peptidoglycan is synthesized in

Chlamydia.

Page 14: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

5

Polymorphic membrane proteins

One of the main components of the chlamydial outer membrane is the polymorphic

membrane proteins (Pmps) localized on the surface of Chlamydia36

. This family of

proteins comprises a significant portion of the genome, approximately 4% in C.

pneumoniae18

. Overall there are 21 Pmp genes in C. pneumoniae18

. All of the 21 genes

of C. pneumoniae have been shown to be transcribed and encode large proteins that are

all phylogenetically related to one of six basic subtypes: Pmp A, B, D, E, G and H37

.

The exact function of the Pmps is not fully known, although several studies indicate that

some Pmps are potentially surface-exposed and immunogenic38,36,39

. Accordingly,

Pmp20 and Pmp21 increase the production of the pro-inflammatory cytokines

interleukin-6 (IL-6), IL-8 and monocyte chemoattractant protein-1 (MCP-1) in human

endothelial cells40

.

Major outer membrane protein The major outer membrane protein (MOMP, gene product of ompA) is the most

abundant protein in the outer membrane of Chlamydia. It was first described and

characterized in 1981 by Caldwell et al. in C. trachomatis41

. MOMP is thought to

maintain the structural integrity of EBs via disulfide bond cross-linking within the EB

outer membrane. MOMP is a multifunctional protein also considered to form pore-like

structures since it shares several biochemical properties with classical porins42

and is

believed to have a role in the infectious process43

. Initially, it was believed that C.

pneumoniae MOMP was different from the protein in C. trachomatis, in the sense of a

diverse antigenic immune determinant in C. pneumoniae MOMP44

. However, further

studies have shown that the MOMP of C. pneumoniae is indeed surface-exposed and an

immunogenic protein45

. The reason why earlier reports demonstrated opposing results

could probably be explained by conformational changes of epitopes that are easily

destroyed by detergents when the antigens are processed for immunoblotting or other

analyses45

. The antigenic properties of MOMP have been extensively studied since the

discovery that purified MOMP (the first chlamydial immunogenic molecule) was

capable of raising antibodies which could neutralize the infectivity of C. trachomatis in

vitro46

. MOMP is now one of the major candidates for development of a Chlamydia

vaccine 47

.

••

Page 15: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

6

Heat shock proteins Chlamydia contains heat shock proteins (Hsps) in the outer membrane complexes of

both EBs and RBs48,49

. The genes encoding Hsp10, Hsp60 (homologues to E. coli

GroEL) and Hsp70 (homologues to E. coli DnaK) are implicated in chlamydial

pathogenesis50-53

. These genes are constitutively expressed throughout the

developmental cycle54,55

. The Hsps are highly conserved within chlamydial species,

including C. pneumoniae48,56

. GroEL from C. pneumoniae has been detected in

atherosclerotic plaques57

and serum antibodies to GroEL correlate with the ability of C.

pneumoniae to be detected in plaques58

. Furthermore, cross-reactivity of GroEL

antibodies with human Hsp 60 has been demonstrated59,60

. Thus, this supports a role for

C. pneumoniae infection in the pathogenesis of atherosclerotic heart disease (discussed

further in section C. penumoniae and atherosclerosis).

Page 16: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

7

The developmental cycle

The key players of Chlamydia Chlamydiae are intracellular bacteria that have a unique biphasic developmental cycle

with two distinct morphological and functional forms (Fig. 1). The extracellular,

infectious form (0.3 μm) is called the elementary body (EB). The EB is metabolically

inert and the nucleoid is highly compacted in EBs due to the condensation of nuclear

material by the bacterial histone-like proteins Hc1 and Hc2, gene products of hctA and

hctB, respectively61-63

. The intracellular, replicating form (1.0 μm) is called the

reticulate body (RB) and is the metabolically active form of Chlamydia. RBs consist of

homogenous internal material and the cytoplasm appears granular with diffuse, fibrillar

nucleic acids64

. Moreover, there is an intermediate state during transition from RB to

EB, called the intermediate body (IB). The length of the complete cycle is dependent on

the infecting strain, the host cell, and the environmental conditions, varying from 48 to

96 hours in cell culture models. However, the focus here will be describing the

developmental cycle of C. pneumoniae.

FIGURE 1. Chlamydial forms. Electron micrograph of the C. pneumoniae isolate T45 in HEp-2 cell cultures

at 48 hours post infection. Elementary body (EB), reticular body (RB) and intermediate body (IB).

••

Page 17: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

8

Attachment The receptors and chlamydial ligands involved in the attachment of EBs to host cell

membranes are undefined. The initial interactions of EBs of many, but not all,

chlamydial strains and species with the host cell appear to be through reversible,

electrostatic interactions with heparan sulphate-like glycosaminoglycans (reviewed in

reference 72). Accordingly, N-glycanase treatment decreased the attachment and

infectivity of C. pneumoniae in human epithelial and endothelial cells, suggesting the

importance of chlamydial glycans containing a high-mannose oligosaccharide for entry,

in vitro and in vivo65,66

. Moreover, Puolakkainen and co-workers proposed that the

mannose 6-phosphate/insulin-like growth factor 2 receptor could serve as a receptor

specifically for C. pneumoniae attachment67,68

. In addition, infectivity was inhibited

when the mannose 6- phosphate receptor was blocked by retinoic acid68

. Chlamydial

surface-exposed proteins are also anticipated to be involved in attachment, for example

of MOMP43

, Omp2, Hsp7069

and PmpD/Pmp270

.

Internalization After attachment, the EB is rapidly internalized into the host cell cytoplasm. There are

several models proposing how EBs enters the host. For example, in 1978, Byrne and

Moulder described the entry of C. psittaci and C. trachomatis into non-professional

phagocytic cells as a process termed “parasite-mediated endocytosis”71

. Lipid-raft

mediated uptake has been implicated as another alternative entry mechanism. However,

because of conflicting results further work is needed to clarify the relevance of lipid-raft

mediated uptake72

. Thus, the internalization step is a crucial step for chlamydial survival

and development of pathogenesis. The mechanism however is still unknown.

Internalization: Endocytosis - mediated via activation of the

actin cytoskeleton Chlamydia is highly dependent on reorganizing the actin cytoskeleton to facilitate their

entry into host cells in vitro73

. C. pneumoniae induces transient microvillar hypertrophy

upon binding to epithelial cells74

. Chlamydia is known to induce tyrosine

phosphorylation of several proteins regulating eukaryotic signaling pathways75-77

.

Several different signaling pathways regulating actin dynamics and organization, such

as PI3-kinase (PI3K) and MEK-ERK kinases, as well as the Rho family of GTPases,

have been shown to be targeted by Chlamydia74,77

. Although C. pneumoniae and C.

trachomatis induce similar morphological changes in microvillar rearrangements, they

target distinct signaling pathways mediating cell alterations. C. pneumoniae uptake has

been shown to be PI3K-dependent involving MAPK kinase ERK1/2 activity74

. In

contrast, C. trachomatis entry involves TARP phosphorylation78

(described in T3SS

section) and subsequently host signal transduction pathways recruiting Rac and

activation of WAVE2/Arp2/379

.

Page 18: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

9

The inclusion - a chlamydial protected site When the EB have been endocytosed and localized inside the host cell, EBs are

protected within a modified vacuole, called the inclusion, throughout the developmental

cycle (Fig. 2). In this compartment the bacteria can avoid fusion with vesicular

trafficking pathways of the host cell and internalization of lyzosomes and endosomes

(reviewed in reference 77). Trafficking of the EB to the peri-nuclear region and the

Golgi area is dependent on early chlamydial gene expression80

. Morphologically,

C. pneumoniae inclusions are smaller in size when compared to inclusions of

C. trachomatis. Moreover, multiple inclusions are seen during C. pneumoniae infection,

whereas C. trachomatis inclusions normally fuse.

FIGURE 2. Developmental cycle of C. pneumoniae. See text for abbreviations.

••

Page 19: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

10

Inclusion membrane proteins In the inclusion membrane, there exists a group of Chlamydia-specific proteins called

inclusion membrane proteins (Incs). The first report of an Inc protein was demonstrated

in C. psittaci and named IncA81

. Since then, six other Incs, from IncB to IncG, have

been characterized82,83

. Inc proteins are not well conserved between Chlamydia spp.18

.

However, Incs display similarities in their hydropathy profiles indicating a structural

homology, comprised of a large bi-lobed hydrophobic region of approximately 50–80

amino acids84

. It is hypothesized that the major hydrophobic domain in each Inc protein

is embedded in the inclusion membrane. Toh et al. have identified 90 putative Inc

proteins in C. pneumoniae and 36 in C. trachomatis based on the general structure of

the predicted proteins85

. The potential to export such a large number of Incs to the

inclusion membrane suggests that the inclusion membrane may have several functions

in vesicle trafficking, inclusion development, avoidance of lysosomal fusion, nutrient

acquisition and signaling associated with EB-RB-EB reorganization77,86

. Since Incs in

general lack a signal sequence, the T3SS has been postulated, and to some extent

demonstrated, to translocate Incs (more in the T3SS section). IncA has been shown to

have a function in the homotypic fusion of inclusions during C. trachomatis infection87

.

IncA and G of C. trachomatis have been shown to be phosporylated by undefined host

kinases88,89

. IncG has further been shown to interact with the eukaryotic protein 14-3-

3β, implicated in a number of signal transduction pathways by taking advantage of their

ability to alter the intracellular distribution of bound ligands89

. However, the function of

the interaction remains unknown.

Primary differentiation and proliferation Around 8-12 hours post-infection (p.i), C. pneumoniae EBs start to differentiate into

RBs. This occurs before intracellular replication/proliferation/growth begins by binary

fission24

(own observations). It has been suggested that de novo protein expression is

required to initiate intracellular growth, as differentiation can be blocked by the addition

of antibiotic inhibitors of transcription or translation, e.g. antibiotics belonging to the

class of macrolides 90

. But the underlying signal(s) are yet undefined. Between 12 to 48

hours p.i., RBs multiply and are highly dependent on host cells supplying iron, amino

acids and energy80,91

. However, there are differences in the specific needs for nutrients

and energy between species and strains. For example, C. psittaci and C. trachomatis

have a complete set of genes required for tryptophan biosynthesis, whereas these genes

are absent in the C. pneumoniae genome17,18

. Moreover, naturally occurring amino acids

differentially influence the development of C. pneumoniae and C. trachomatis92

. There

are morphological differences in the localization of RBs within the inclusion between C.

pneumoniae and C. trachomatis during equivalent stages of development. RBs of C.

pneumoniae are located throughout the entire lumen of the inclusion, whereas

C. trachomatis RBs are typically tightly juxtaposed to the inclusion membrane24

.

Page 20: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

11

Certain circumstances, for example nutrient deficiency, may result in morphological

alterations of RBs and the emergence of enlarged, atypical chlamydial forms termed

“persistent forms.”

Re-differentiation After multiple rounds of division, C. pneumoniae RBs start to re-differentiate back to

infectious EBs around 36-48 hours p.i24

(own observations). During re-differentiation of

RBs to EBs, IBs can be detected. Expression of a number of late-cycle genes occurs

during the re-differentiation process and is described in the review by Abdelrahman and

Belland 64

. However, the signal(s) regulating this event is unknown and one speculative

mechanism has been proposed involving the T3SS (discussed in the T3SS section).

Exit from the host cell When the intracellular growth of C. pneumoniae is completed, infectious EBs are

released into the cytoplasm to initiate new cycles in new host cells by exocytosis or host

cell lysis (72-92 hours p.i, Fig. 3). Chlamydial exit has not been extensively studied,

until recently. Hybiske et al., describes lysis of the host cell membrane as a sequential

process in which the inclusion ruptures before the cell membrane93

. The same authors

described a second mechanism for host cell entry of C. trachomatis and C. caviae as a

packaged-release mechanism, called extrusion93

. The extrusion of Chlamydia involves

the large Chlamydia-containing vacuole pinching off and extruding out of the cell. This

leaves the host cell intact, with a residual bacteria-containing inclusion. The

implications of this exit strategy have not been fully explored or demonstrated for C.

pneumoniae.

FIGURE 3. Growth of C. pneumoniae isolate T45 in HEp-2 cells.

••

Page 21: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

12

Bacteria and iron Iron is an important growth factor for pathogenic bacteria, with the exception of the

Lyme disease agent, Borrelia burgdorferi94

. Intracellular bacteria, such as Listeria and

Mycobacteria, depend on intracellular iron sources that are host-derived95-97

. Highly

sophisticated iron-uptake systems have been identified in most bacterial species with

iron-binding siderophores as the major mechanism for bacteria to respond to iron

limitation. For instance, Mycobacteria express at least three known iron uptake systems

and Listeria can access cytoplasmic iron from ferritin98

. However, no iron uptake

mechanism has so far been identified in Chlamydia.

Chlamydia and iron One of the best understood pathways of iron delivery into the eukaryotic host cell is via

the iron-saturated form of transferrin, holo-transferrin, and its high affinity binding to

the transmembrane transferrin receptor99

. Intracellular iron is transferred to the

endosomal pathway (early endosomes) by acidification and an NADH-dependent

reductase activity which converts ferric iron to ferrous iron, which is next exported into

the cytoplasm 99

. Chlamydia escapes this pathway when neither the nascent nor the

mature inclusion fuses with the early endosome90,96,97,100-102

. However, the early

endosome, containing transferrin, is recruited early during the infection and stays

closely associated with the chlamydial inclusion through the entire developmental cycle.

This might suggest a possible chlamydial source of iron, since Chlamydia has been

shown to utilize holo-transferrin as an iron-source95,97

. Moreover, YtgA, a predicted

periplasmic metal-binding protein, has recently been proposed to be involved in iron

acquisition in Chlamydia103

. Furthermore, the divalent cation-dependent regulator A

(DcrA) homologue of ferric uptake regulator (Fur) in E. coli has also been suggested to

be involved in yet unexplained molecular regulatory mechanisms104,105

.

Chlamydial persistence Physicians and the medical community have long thought that long-term chronic or

repeated chlamydial infection might make Chlamydia-related diseases worse.

Accordingly, it has been proposed that C. pneumoniae and C. trachomatis are capable

of causing long-term infections continuing for months or even years in the absence of

treatment. Moreover, when Chlamydia enters a persistent state associated with an

incomplete developmental cycle, the infection is difficult to eradicate with antibiotics or

could, even worse, enter persistence due to antibiotic treatment106

. It has been suggested

that persistence is induced as a result of Chlamydia lacking some essential amino acids,

such as tryptophan, which is essential for complete chlamydial development. How well

these in vitro models really support the in vivo situation is unclear.

Page 22: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

13

Exposing chlamydial infections in vitro, with interferon-gamma (IFN-) has become a

conventional procedure to promote persistence (reviewed in reference 91). IFN-

induces tryptophan depletion through activation of the host tryptophan-degrading

enzyme indoleamine 2, 3-dioxygenase (IDO) resulting in the inability of RBs to

differentiate into EBs. Interestingly, C. trachomatis uses a chlamydial tryptophan

synthase to counteract the absence of tryptophan when the amino acid is removed from

the cell culture medium, suggesting the ability to synthesize tryptophan in vivo107

.

However, C. pneumoniae lacks the trpBA operon encoding the tryptophan synthase and

uses another mechanism for tryptophan synthesis108

. Iron depletion, using the iron

chelator deferoxamine-mesylate (DAM), has also been shown to induce persistence of

Chlamydia, in vitro95,97,109-111

. The key clinical question is still open: Does long-term

chronic or repeated infection lead to the intermittent appearance of disease? And how

relevant are the in vitro models described above? Can the models truly mimic the

presence of a rigorous host immune response battling the pathogen to persist in vivo?

••

Page 23: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

14

Clinical manifestations

Acute infection C. pneumoniae is a respiratory pathogen that causes upper and lower respiratory tract

diseases and invades the epithelial cells of the respiratory tract and lung tissue. In the

acute infection, C. pneumoniae is transmitted from person to person via respiratory

secretions and droplets112

. Transmission is relatively inefficient, and the incubation

period may be several weeks, which slows down the spread of outbreaks113

. The

majority of C. pneumoniae infections are asymptomatic or mild upper respiratory tract

infections, most of which are mild and self-restricted and therefore often go

undiagnosed114

. For example, in a collaborative study we analyzed 500 asymptomatic

subjects for prevalence of C. pneumoniae infection. In total, 63% were IgG-positive,

indicative of a previous C. pneumoniae infection115

. Accordingly, other studies have

demonstrated a sero-prevalence around 40% to 70% in the adult population globally116

(reviewed in reference 116). Thus, the majority of humans are infected with C.

pneumoniae at some stage of their lives. However, the incubation period of infection

due to C. pneumoniae can be several weeks, which is longer than that of many other

respiratory pathogens117

. Involvement of C. pneumoniae infection has been described in

pneumonia, acute bronchitis, common cold, persistent cough, pharyngitis, sinusitis and

otitis media118(reviewed in reference 118).

Chronic infection In addition to acute respiratory infections, several chronic respiratory tract inflammatory

diseases have been associated with C. pneumoniae infection. These include chronic

bronchitis and chronic obstructive pulmonary disease (COPD) as well as sarcoidosis

(reviewed in reference 118). Several studies also address an association between C.

pneumoniae and asthma119(reviewed in reference 119). Furthermore, C. pneumoniae

may play a role in neurological disorders such as multiple sclerosis120

. C. pneumoniae

has also been associated with reactive arthritis, where C. pneumoniae DNA and mRNA

were found in synovial material from patients suffering from inflammatory joint

disease121,122

. In addition to respiratory tract infections, C. pneumoniae has been

associated with cardiovascular diseases, in particular in the development of

atherosclerosis.

Page 24: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

15

C. pneumoniae and atherosclerosis

Atherosclerosis Atherosclerosis develops as a reaction to damage of the arterial wall

123. It is now

considered an inflammatory response to retain and modify lipids in the vessel wall. Both

innate and adaptive immune defense mechanisms are believed to be important in

pathogenesis124,125

. Classical risk factors for developing atherosclerosis are

hypercholesterolemia, smoking, diabetes mellitus and hypertension as well as

environmental and genetic factors123

. However, exactly how the different established

risk factors contribute to the initiation and/or progression of the atherosclerotic lesions

is unclear. This has led to studies of other possible factors that could be involved in the

etiology and pathogenesis of atherosclerosis and its complications. An alternative

explanation that has raised a lot of controversy is the atherosclerotic infection theory.

This hypothesis was formulated at the beginning of the last century when Frothingham

suggested that, “The sclerosis of old age may simply be a summation of lesions arising

from infectious or metabolic toxins”126

. However, this idea did not gain much interest

until the end of the seventies when Fabricant and co-workers127

showed that chickens

experimentally infected with a herpes virus developed vascular lesions that resembled

atherosclerosis in man. Later studies have especially pointed out C. pneumoniae, but

also Helicobacter pylori, Herpes simplex virus and cytomegalovirus as possible primary

etiological factors or co-factors in the pathogenesis of atherosclerotic diseases like

ischemic heart disease and cerebrovascular disease116

.

Atherosclerosis and animal models Mice are the most frequently used animals in experimental atherosclerosis studies.

These rodents are easy to breed, and importantly, it is possible to knock out and replace

endogenous genes in mice as well as create transgenic models128

. Wild-type mice are

normocholesterolemic, and most lipids in the blood are carried by High Density

Lipoprotein (HDL), which is known to be protective against atherosclerosis in

humans123

. Atherosclerotic lesions do not develop spontaneously in mice, and

hypercholesterolemia has to be induced either by genetic manipulation or special diets

containing high levels of fat and cholesterol. Lesion formation in mice is restricted to

the aortic root or aortic sinus area, and the lesions are mainly early stage fatty lesions

with invading smooth muscle cells (SMC) as a characteristic feature128

. Apolipoprotein

E (ApoE) deficient mice are the most commonly used genetically modified mouse

model since they develop atherosclerosis spontaneously when fed rodent chow129

.

Moreover, these mice have total cholesterol levels about five times higher than wild-

type mice and at 10 weeks of age develop atherosclerotic lesions in the aorta and

coronary and pulmonary arteries128,129

. In addition, ApoE-deficient mouse models of C.

••

Page 25: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

16

pneumoniae infection have produced contradictory results. Studies demonstrate either

increased atherosclerosis or no effect on progression130-133

.

Lipoprotein (a) as a mouse model for atherosclerosis Dahlén and co-workers have shown an exceptionally strong statistical association

between plasma levels of lipoprotein (a) (Lp(a)) and atherosclerosis134

. Lp(a) has so far

only been found in humans, some monkeys and in hedgehogs135

. Lp(a) is very similar to

Low Density Lipoprotein (LDL) in its constitution. Lp(a) contains, just like LDL, apo-B

100, but Lp (a) differs from LDL in that it contains an extra large protein,

apolipoprotein (a) (apo(a)) that is linked to apoB by a disulphide bridge. Accordingly,

mice cannot produce Lp(a). Therefore, a double transgenic mouse expressing human

apo(a)/apoBH produces Lp(a) and develops atherosclerosis136

. Thus, these mice can be

used in an atherosclerotic model to investigate whether C. pneumoniae infection in

combination with Lp(a) accelerates the development of atherosclerosis.

In addition, 20-week-old Lp(a) mice sacrificed at 10 weeks p.i. with C. pneumoniae

following three repeated inoculations, did not show any macroscopic plaque formations

in the aortic cusps of the aortic root, after staining with oil red. However, infiltration of

macrophages and possibly foam cells was observed (Fig. 4A-B). Moreover, when

compared to ApoE-deficient mice at same age, the Lp(a) mice display less pronounced

lesions of atherosclerotic plaques (Fig. 4C). Accordingly, Berg et al. reported that Lp(a)

mice develop atherosclerotic lesions after 46 weeks of age when fed a normal diet137

.

Thus, these mice were considered to be time-consuming and difficult to work with as a

model for spontaneous development of atherosclerosis.

FIGURE 4. Photomicrographs of cross-sections stained for lipids in the aortic root of 20-week-old mice.

(A) Accumulation of lipoid or foam cells expanding the subendothelial space in aortic cusp of a Lp(a) double

transgenic mouse. The wall of the aorta is thickened by this accumulation of material (magnification 100).

(B) A closer view of the area marked in (A) (magnification 400). (C) Oil red staining shows accumulation of

lipids within atherosclerotic plaques in the three aortic cusps.

Page 26: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

17

C. pneumoniae in atherosclerosis Among the chronic inflammatory conditions associated with C. pneumoniae,

atherosclerosis is the most widely studied. The first report of an association of C.

pneumoniae with atherosclerosis was published in 1988. Saikku and co-workers

demonstrated elevated levels of C. pneumoniae antibodies in patients suffering from

myocardial infarctions and coronary heart disease compared to controls138

. Since then,

more than 50 seroepidemiological reports have strengthened the early findings of

Saikku. However, large scale serological prospective studies investigating the

association with C. pneumoniae and the risk of coronary events did not find any

connection (reviewed in reference 116). Moreover, other strong associations exist

between C. pneumoniae infection and atherosclerosis as demonstrated by detection of

the organism within atherosclerotic lesions, but not in adjacent normal tissue by

immunohistochemistry, polymerase chain reaction and electron microscopy139-141

.

Viable C. pneumoniae have also been cultured from atheromatous plaques, suggesting a

more causal relationship142-145

. Animal models demonstrate that C. pneumoniae can

either initiate lesion development or cause exacerbation of lesions in rabbit and mouse

animal models, respectively146 (reviewed in reference 146).

Antimicrobial trials have questioned the C. pneumoniae

infection theory To better understand the pathogenic significance of C. pneumoniae in the development

of atherosclerosis, several pharmacological intervention trails with antibiotics have been

performed. Initially, clinical antibiotic interventions were promising, with some

protective effects reported147 (reviewed in reference 147). However, most of these studies

were considered to suffer from major weaknesses such as small patient groups and

treatment courses too short to be effective against chronic C. pneumoniae infection.

Therefore, two larger prospective clinical trials were performed148,149

. Despite long-term

treatment with a bactericidal antibiotic effective against C. pneumoniae, no reduction in

the rate of cardiovascular events was observed148

. Accordingly, a one-year course of

weekly azithromycin did not alter the risk of cardiac events among patients with stable

coronary artery disease149

. However, these studies have been questioned as antibiotic

treatment is not effective against chronic and persistent C. pneumoniae infections106,150-

152. Another consideration is whether the atherosclerotic process already has been

initiated prior to antibiotic treatment. Thus, taking antibiotics too late in the

inflammatory process is unlikely to have an effect and factors unaffected by antibiotics

are likely to obscure any beneficial anti-chlamydial effect153

. Therefore, C. pneumoniae

infections remain a reasonable risk factor for development of atherosclerosis.

••

Page 27: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

18

C. pneumoniae and inflammation of the vascular system The ability of C. pneumoniae to infect several cell types and to disseminate into various

tissues after respiratory infection is fundamental for the various pathogenic

consequences. Endothelial cells become activated upon infection with C. pneumoniae

and increase production of inflammatory mediators and express adhesion molecules

such as intercellular adhesion molecule (ICAM-1) and vascular cell adhesion molecule-

1 (VCAM-1) which subsequently leads to the transendothelial migration of leukocytes

to the inflammatory site154

. Additionally, C. pneumoniae-infected endothelial cells

increase secretion of monocyte chemoattractant protein-1 (MCP-1) and IL-8 which

stimulate the transendothelial migration of monocytes and neutrophils as well as

monocyte differentiation into macrophages (reviewed in reference 154). Moreover C.

pneumoniae has been found in foam cells and is believed to mediate oxidation of

LDL155

. In addition, Rödel and collaborators have published several reports on the

ability of C. pneumoniae to infect and proliferate in SMC156-158

. Taken together, the

collective activation of several cell types in the vascular system would create a

sustained inflammatory environment. Contributing to the initiated inflammatory

response following the infection is the production of a cascade of pro-inflammatory

cytokines e.g. IL-1β, IL-6, IL-8 and tumor necrosis factor (TNF-α) that helps sustain the

state of the inflammatory site by acting as cell signaling molecules.

Pro-inflammatory cytokines The first cytokine to be discovered was IL-1 in the early 1950’s

159. In general, cytokines

are regulatory proteins presented by leukocytes and other cell types such as fibroblasts,

epithelial cells, osteoblasts, e.g. used for cell-cell communication. Cytokines do not only

act in a paracrine manner but can also signal in an autocrine fashion and activate the cell

that is producing them. Cytokines produced during a bacterial infection are e.g. the pro-

inflammatory cytokines IL-1β, IL-6, IL-8. These cytokines exhibit pleiotropic actions

that include the regulation of host innate and adaptive immune responses and the

modulation of inflammatory responses160

. Moreover, these pro-inflammatory cytokines

reveal biological activity including osteoclastgenesis stimulating bone resorption161

(reviewed in 161). On the other hand, cytokine responses by epithelial or phagocytic

cells can be disrupted by some pathogenic bacteria, such as Yersinia enterocolitica and

Pseudomonas aeruginosa, eventually enabling their escape from the host's immune

system160,162

. A number of studies have shown that cytokines can affect the growth of a

variety of bacteria inside host cells. For example, TNF- stimulates the growth of

Mycobacterium tuberculosis in human monocytes163

. IL-6 has been shown to increase

the growth of Mycobacterium in macrophages, thereby inhibiting T cell activation164

.

However, not much is described regarding whether or not a bacterium benefits from the

production of pro-inflammatory cytokines. It is speculative, but it could be a good

strategy for recruitment of immune cells to the site of infection to subsequently infect

Page 28: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

19

these cells. In this manner, the infection could enter the circulation and spread to distant

tissues.

C. pneumoniae and pro-inflammatory cytokines As mentioned earlier, C. pneumoniae is believed to be associated with manifestations of

inflammatory chronic diseases such as atherosclerosis, asthma, neurological disorders

and arthritis. C. pneumoniae has been shown to infect and proliferate in vascular cells

e.g. endothelial cells, smooth muscle cells, macrophages and in atherosclerotic

lesions140,165

. Several in vitro experiments reveal that the infection induces expression

and release of pro-inflammatory cytokines such as TNF- , IL-1 and IL-6 in vascular

cells166-170

. Moreover, C. pneumoniae infection of respiratory cells is shown to induce

IL-8 (CXCL8) production171,172

. Recently, it was reported that C. pneumoniae infection

of microglial cells and astrocyte cells increased production of MCP-1, IL-1, IL-6 and

TNF-173

.

Bacterial infection of bone demonstrates similar cytokine

induction as in atherosclerosis Clinical studies have revealed that bacterial infection of bone tissue is a possible

explanation for imbalanced bone remodeling, but the mechanisms responsible for these

observations are not defined161

. Interestingly, the inflammatory response of bone

destruction resembles the pathogenesis of atherosclerosis. Several studies have

demonstrated that Staphylococcus aureus and Salmonella can invade and persist within

cultured murine and human osteoblasts and the infection promotes the ability to produce

IL-1, IL-6 and TNF-, known to initiate the inflammation that results in progressive

bone destruction174-179

.

The bone resorption and remodelling system is regulated through increased expression

of the receptor activator of NF-B ligand (RANKL180

), its receptor (RANK) and the

synchronized production of a decoy receptor named osteoprotegerin (OPG181

). Under

normal circumstances, the maintenance of bone tissue depends on the dynamic interplay

between RANKL/OPG regulated by osteoblasts and the activation of bone resorption

mediated via RANK expressed on pre-osteoclasts. In bone disorders, bone resorption

exceeds bone formation and the result will be bone destruction. Interestingly,

microorganisms like S. aureus, Salmonella and the peroidontitis-associated bacterium

Actinobacillus actinomycetemcomitans have been previously shown to induce the

expression of RANKL in osteoblast-like cells and synovial fibroblasts mediating

osteoclast activation, therefore they are suggested to be mediators of

osteoclastgenesis177,182,183

.

••

Page 29: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

20

Accordingly, a case study reported two patients suffering from unexplained chronic

anaemia with detected C. pneumoniae in their bone marrow184

. This could be of interest,

as osteoblast progenitor cells are generated from multi-potent cells that differentiate e.g.

into mature osteoblasts and are thereafter recruited to the bone-forming sites.

Moreover, accumulating evidence indicates a pathopyhsiological link between

atherosclerosis and osteoporosis185-192

. We have performed a study demonstrating that

OPG promotes fibrous cap formation in atherosclerotic lesions of ApoE-deficient mice

(manuscript VII in this thesis). Could infections, in particular C. pneumoniae infection,

be implicated in the pathogenesis of both diseases?

Page 30: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

21

The type three secretion system (T3SS)

Bacterial infection is dependent on pathogenic traits that benefit survival and

proliferation of the bacterium in its host. Bacterial proteins involved in pathogenesis

need to be anchored to the bacterial cell wall, or delivered across the cell wall. Such

actions demand special bacterial protein secretion systems. One such example is the

type three secretion system (T3SS). This system allows Gram-negative bacteria in a

close contact with a host cell, to transport proteins from the bacterium into the

cytoplasm of the eukaryotic host cell. This transport system was first studied and

characterized in pathogenic Yersina spp.193

. However, T3SS was first visualized in

Salmonella194

followed by enteropathogenic E. coli195

and Shigella196

. Thereafter,

T3SSs have been characterized in many additional bacteria 197

. All human pathogenic

bacteria possessing a T3SS use it to translocate effector proteins into host cells. These

host cell-localized effectors subsequently modulate host cellular functions198

. The T3SS

is a complex apparatus, consisting of more than 25 proteins spanning the entire bacterial

envelope. The exterior component of all T3SSs is the syringe-like structure protruding

out from the bacterium, known as the needle complex or injectisome. The needle

complex projects from the bacterium and mediates delivery of the bacterial effector

proteins through its channel. Secretion by this mechanism is contact-dependent199

.

I will focus further on the composition and regulation of the T3SS in the section

describing chlamydial T3SS. However, many assumptions of the chlamydial T3SS are

based on information on assembly and regulation of the T3SS in Yersinia.

Historical observations of a possible T3SS In 1973, 15 years before the first visualization of the T3SS in Salmonella

194, Matsumoto

published electron micrographs showing rosette-like structures and projections at the

surface of C. psittaci200

. Each of these rosettes had a diameter of about 20 nm and

appeared to be arranged in a radial formation of nine subunits. However, the research

community was doubtful of these observations and speculated that the projections were

artifacts formed during purification of EBs rather than natural structures. From the years

1975 to 1981, Matsumoto continued studying and analyzing his observations 201,202

.

Hence, in 1982, he published a new freeze-deep-etching technique that rendered

examination in more detail. In his new electron micrographs he noticed that each

projection on the RB surface emerged from the center of a flower-like structure, about

30 nm in diameter. These flower-like structures appeared to be a radial arrangement of

nine leaves with a projection reaching out from the bacterial surface (Fig. 5). Further,

the morphology of the RB projections was identical to the projections on EBs. Each

projection was anchored in to the cytoplasmic membrane of the Chlamydia, from where

it protruded out through the inclusion membrane of the cell. All RBs had these

••

Page 31: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

22

projections and the number of projections peaked 10 h after infection after which they

gradually decreased to approximately the same number of projections as on EBs203,204

.

Comparable structures have thereafter been observed on the surface of all other human-

pathogenic Chlamydia (C. trachomatis205

and C. pneumoniae23

). Today, many other

researchers apply these early observations from Matsumoto to support their own

hypotheses and results on T3SS in Chlamydia.

FIGURE 5. Early observations of Matsumoto – the chlamydial projections. A: T3S projections (arrows)

are viewed from the cytosolic side of the infected cell extending across the inclusion membrane from

underlying RBs. B: Projections on the RB surface emerged from the center of a flower-like structure, in an

arrangement of nine leaves (insert). C. caviae were examined by scanning and transmission electron

microscopy (TEM). Micrographs are modified and reprinted with permission from Trends in Microbiology

(Picture A: Copyrigh 2007) and ASM (Picture B: Copyright 1982), respectively.

First evidence of a chlamydial T3SS The first report of a chlamydial T3SS came in 1997

206. By sequencing a fragment of the

C. psittaci genome the authors detected four open reading frames that were predicted to

encode proteins with similarity to T3S apparatus components CdsU and CdsV (contact-

dependent secretion), CopN (Chlamydia outer protein homologue of YopN) and Scc1

(specific Chlamydia chaperone). Analysis of the C. trachomatis serovar D genome later

confirmed the results from Hsia et al., that Chlamydia possesses a complete set of genes

that could encode a T3S apparatus17

. Later, the same authors analyzed the genome

sequence of C. pneumoniae CWL029, revealing same repertoire of T3S-related genes as

seen in C. trachomatis18

. Interestingly, the T3S-associated genes were scattered

throughout the genome. This contrasted to other Gram-negative bacteria whose T3S-

Page 32: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

23

genes are typically located at one distinct locus of the chromosome, termed the

pathogenicity island, or on a virulence plasmid17,207

. A common feature of T3SS genes

is their low G/C content relative to the rest of the genome. This is not the case for

Chlamydia, whose entire genome is low in G/C206,208,209

. Taken together, these early

reports have been the starting-point for intensive research on the chlamydial T3SS.

Composition of the chlamydial T3S injectisome Several C. pneumoniae genes are homologous to genes coding for units resembling the

T3S apparatus of Yersinia and other known T3SS (summarized in Fig 6). Function has

been predicted based upon studies of their homologues in Yersinia, for which the T3SS

is the best characterized. As mentioned earlier, the T3S apparatus has a complex

architecture with the majority of its key players believed to be associated with the

bacterial membrane. Many of the apparatus components, in Yersinia referred to as the

Yersinia secretion-Yersinia outer proteins system (Ysc-Yops), are well conserved

among bacteria207

. Accordingly, T3S-associated genes of Chlamydia were originally

labeled with references to the Ysc-Yop nomenclature. However, this nomenclature has

been revised by Fields et al.210

based on the convention of contact-dependent secretion

(Cds) in Chlamydia, proposed by Hsia et al206

. Therefore, the apparatus components

will be denoted as Cds in this work.

Basal apparatus In Chlamydia, the predicted basal body of the T3SS is anchored to the inner membrane

of the bacterium. The integral membrane protein CdsV (also known as LcrD and

previously termed Cds2) is predicted to form a central protein-conducting channel

across the cytoplasmic membrane based upon work done in Yersinia pestis207,211

. Other

proteins suggested to be involved in the formation of such a channel include CdsD,

CdsR, CdsS, CdsT and CdsU (previously termed Cds1). Recently, CdsD was reported

to be phosphorylated by PknD, an integral membrane protein with a kinase activity

domain located in the cytoplasm212

. The CdsU homologue, YscU, was recently

described as an essential component of the export apparatus of the Yersinia injectisome.

YscU cleavage is needed to acquire a conformation allowing recognition of a subset of

secreted substrates, termed the translocators that form a translocon213

. However, cdsU

transcripts have not been detected at any time-point throughout the developmental cycle

in C. pneumoniae54

, whereas they have been detected in C. trachomatis 210

. These

findings suggest variable importance of CdsU in Chlamydia. CdsL and CdsN are

predicted to cooperate in regulating translocation according to their function in Yersinia

and other T3SS 214

. CdsN mediates ATP hydrolysis providing the energy for secretion

and CdsL might function as its regulator, as in Yersinia215

. CdsJ is an outer membrane

lipoprotein spanning the periplasmic space and therefore possibly bridges the inner and

outer membranes. The final component of the basal apparatus is the distal outer

••

Page 33: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

24

membrane protein CdsC. CdsC, which is the homologue of YscC in Yersinia, belongs to

the secretin family of proteins and forms a stable ring-like structure in the outer

membrane216

. In other systems, these outer-ring structures are thought to function as

transport channels for macromolecules. Interestingly, this structure shares similarities to

the rosette-like structures observed by Matsumoto217

.

FIGURE 6. Schematic illustration of the chlamydial T3S-injectisome. The putative structure of the

chlamydial injectisome is derived by comparison with homologus proteins of the Yersinia T3S apparatus. The

figure is modified from (www.genome.jp – KEGG). See text for abbreviations.

The Chlamydia translocon To be able to transport effector proteins through the eukaryotic cell membrane,

microorganisms with a T3SS apparatus have to be in close contact with the host cell.

Upon cell contact the bacteria are believed to insert a channel-complex into the cell

plasma membrane that allows translocation of proteins into the cell218 (reviewed in

reference 218). This complex is commonly refered to as the translocon. In Yersinia,

YopB and YopD interact at the interface between bacteria and host cell, and have been

suggested to form a translocon pore. This pore complex allows the direct Yop effector

translocation into the target cell219

. A third translocator in Yersinia, LcrV (low calcium

response protein V) is localized at the distal tip of the needle where it forms a bridge

that connects the needle to the translocon220

. So far, no LcrV homologue has been

reported in Chlamydia.

Page 34: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

25

YopB and YopD in Yersinia have homologues in C. pneumoniae and C. trachomatis,

termed Chlamydia outer proteins CopB and CopD208

. This assertion is not based upon

sequence homology between these proteins but rather similar size and hydrophobicity

profiles. Moreover, both CopB and CopD lack signal sequences and lie in close

proximity to the postulated T3SS chaperones, Scc2 (also termed LcrH-1) and Scc3 (also

termed LcrH-2, both discussed in more detail in the next section). Thus, their predicted

function is to act as a gating mechanism facilitating translocation of secreted effectors

across the inclusion membrane. CopB has been detected in the C. trachomatis inclusion

membrane, consistent with its presumed function as a T3S translocator221

. Additionally,

CopD has been demonstrated to be translocated in the heterologous system of the SPI-2

T3SS of S. enterica (serovar Typhimurium)222

. Interestingly, a paralogue of CopB has

been described, named CopB2223

. CopB2 can be detected in the host cell cytosol,

probably demonstrating distinct functions when compared to other known translocators.

However further studies are needed to evaluate its function. Taken together, this

suggests that Chlamydia most certainly acquires a translocon in the inclusion membrane

to complete delivery of effectors.

Chaperones of the T3SS The T3S chaperones can be grouped into different classes according to their binding

affinities to their corresponding cognate substrates (reviewed in reference 220). As the

name implies, effector-class chaperones bind to one or at most a few translocated

effectors. In contrast, translocator-class T3S chaperones bind to the proteins that form

the translocon. Common to these two classes is their ability to facilitate secretion by

guiding proteins to the T3S-apparatus and keeping them in an unfolded state prior to

secretion. They also prevent premature interactions between different T3S components.

Some of the best-studied T3S-associated chaperones are those grouped in the

translocator class of chaperones224

. In Y. pseudotuberculosis, LcrH (low calcium

response protein H) belonging to the chaperones termed Sycs, binds and stabilizes

YopB and YopD in the bacterial cytoplasm prior to secretion225

. In Chlamydia,

orthologs to syc-gene products have been found and are believed to be specific

Chlamydia chaperones according to their structural similarities to known chaperones in

other T3SSs206,210,221

. In the first report of a possible T3SS of Chlamydia, Scc1

(homologous to SycE in Yersinia) was discovered206,210,221

. SycE is thought to act as a

stabilizing chaperone for YopE, a anti-phagocytic factor produced by Yersinia226

.

Recently, it has been demonstrated in C. trachomatis serovar L2 that the two

chaperones Scc2 (LcrH-1) and Scc3 (LcrH-2), homologous to LcrH, were associated

with the same translocator protein, YopD, in a heterologous Yersinia T3SS model

223.

Moreover, LcrH in Yersinia interacts with YopB and YopD at a unique binding site

established by the folding of its three tandem tetratricopeptide repeats (TPRs)

••

Page 35: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

26

facilitating protein-protein interactions227

. Both Scc2 and Scc3 of Chlamydia possess a

TRP domain223

. Additionally, it was shown that Scc2 interacts with CopN, and is

suggested to serve as a chaperone for this protein228

. Recently, two new chaperones

were discovered and shown to be involved in regulation of CdsF in C. trachomatis.

These were denoted as CdsE and CdsG according to their similarities to the

Pseudomonas spp. needle chaperones PscE and PscG, respectively229

. However, these

chaperones only share 66% and 67% similarity to the predicted protein sequences in C.

pneumoniae, respectively, as determined by the multiple sequence alignment program

ClustalW2 (author contribution).

Transcriptional regulation of T3SS chaperones In 2000, Subtil et al. speculated that scc2 and scc3 could be expressed at different time-

points during the infection, for example in EBs at the entry phase and during

proliferation in RBs, and enable the sequential translocation of structurally related

proteins208

. Five years later, Byrne et al. proposed that protein products of the scc2

operon are likely to function during attachment since these genes are

expressed

following EB arrangement and the protein products of the scc3 operon likely function

during intracellular survival230

. Temporal expression studies with C. pneumoniae

revealed that Scc3 was one of the few T3S genes expressed as early as 1.5 h following

infection and expressed continuously during the developmental cycle. In contrast, Scc2

was apparently not to be expressed until late in the developmental cycle, suggesting a

different function for each of the gene products228

. Expression of copB, and copB2 are

both mid- to late-expressed T3S genes. SicA, the chaperone of the translocators SipB

and SipC of Salmonella, activates the expression of late SPI-1 T3S genes encoding only

late effector proteins. This occurs by SicA forming a regulatory complex with a

transcriptional activator InvF231

. Interestingly, the Scc homolog LcrH in Yersinia also

acts as a negative regulator of Yop expression, in disagreement with its chaperone

function as a stabilizator of the translocon232

. Further, LcrH has been shown to interact

with YscY in Yersinia233

. Thus, this implicates the multi-functional activity of T3S

chaperones as stabilizers, activator pilots and as regulator(s) of the T3SS. It is tempting

to speculate that Sccs can function as negative regulators of the Chlamydia translocon

or even possess more distinct purposes.

The needle complex Bacteria possessing a T3S apparatus can simply be described as bacteria using a needle

to become virulent. Early on in the field of Chlamydia research, the most firmly

established hypothesis concerning this needle structure was that the projections function

as channels that facilitate the uptake of nutrients from the host cytosol by Chlamydia, an

idea designated as the “soup-through-a-straw” hypothesis234

. The needle-like structure,

referred to as the needle complex, has been shown by scanning transmission electron

Page 36: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

27

microscopy to consist of approximately 150 YscF subunits and the tip complex is

formed by three to five LcrV monomers in Yersinia enterocolitica235

. Moreover,

purified needles from Pseudomonas aeruginosa detached from the bacterial surface

were 60-80 nm in length and 7 nm in width, resembling purified YscF needles from

Yersinia236

. Recently, using a bioinformatics approach, gene CT666 in C. trachomatis

was revealed to be homologous to yscF in Yersinia, termed cdsF229

. The open reading

frame for cdsF is located within an operon encoding other putative components of the

apparatus (Fig. 7). It was further shown by immunoblotting and electron microscopy

that CdsF was concentrated in the outer membrane of EBs and surface-exposed as a

component of an extracellular needle-like projection. During infection, CdsF was

detectible in the inclusion membrane with a punctate distribution adjacent to membrane-

associated RBs229

. CdsF of C. trachomatis shares 89% similarity to the predicted

protein sequence in C. pneumoniae, suggesting the existence of CdsF in this species as

well (author contribution).

Gating of T3S machinery CopN is the protein corresponding to YopN of Yersinia. It is proposed to be the lid of

the T3SS apparatus in the inclusion membrane regulating the gating function of

T3SS221

. CopN of C. trachomatis was the first protein shown to be translocated in a

T3S-dependent manner in a heterologus system using Y. enterocolitica221

and later S.

typhimurium SPI-1222

. CopN has been associated with the inclusion membrane during

intracellular growth, supporting the idea of a gating mechanism. In Yersinia, YopN can

form a complex with TyeA and its chaperones SycN and YscB237

. Subsequently, this

complex, under non-permissive conditions, is presumed to block secretion, as YopN

cannot be secreted while interacting with TyeA. When dissociated from TyeA, YopN is

released opening the gate for translocation of Yops. However, the underlying signal(s)

regulating the release of YopN from TyeA is undefined237

. This plug-like mechanism is

extremely interesting from a chlamydial point of view. Messenger RNA of copN is

known to be expressed late in the developmental cycle of both C. pneumoniae and C.

trachomatis54,217,238

. J. Peters et al., suggests a theory based on unpublished data, where

CopN was not expressed in EBs, only late in normal RBs. Therefore it, is hypothesized

that CopN can provoke RB detachment (concerning C. trachomatis, author’s

annotation) and shutting off the T3SS as a consequence54,217

. This is in agreement with

their mathematical theory, later described in the intracellular T3SS section. Their

assumption is that there is a possible function for the regulation of CopN involved as a

T3SS on/off switch important for the re-differentiation of infectious EBs. Whether or

not detachment from the inclusion membrane is sufficient enough to regulate the switch

is speculative. However, more functional studies on CopN and a possible complex

interaction with a TyeA-like protein are needed. Scc3 is a possible chaperone

regulating such a complex since it has been demonstrated to interact with CopN

228.

••

Page 37: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

28

Effector proteins Often, effector proteins of Chlamydia are referred to as secreted substrates in the

literature. However, proteins secreted by the T3SS in other species are divided into two

classes of substrates/proteins according to their function and time of action. First, there

are those proteins secreted when the apparatus assembled and ready to secrete the

translocators, which generates the pore formed in the cell membrane. After this, the

effectors are then delivered through the T3S-apparatus, either to the inclusion

membrane or into the cell cytoplasm. Therefore, in this section, I have separated the

predicted Chlamydia effectors into a single class of secreted proteins (summarized in

Table 2).

There is not a common function for all known T3S effector proteins. For instance, Yops

secreted by Yersinia, have one mission, targeting intracellular host cell signaling

pathways to benefit their survival outside the cell and escape from the innate immune

system. Additionally, Salmonella Sips secreted by the SP-1 T3SS induce cell invasion,

while Sses secreted by the SP-2 T3SS support their proliferation in macrophages

(reviewed in reference 207). The diversity among T3SS provides, in the context of

Chlamydia, both pros and cons. It would have been much simpler if effectors were

homologues between different bacteria or if chlamydial T3SS genes were located at a

single location, as in other pathogens constituting a T3SS. However, surrogate T3SS

have been successful in revealing secretion of potential chlamydial effectors (Table 2).

Using heterologous T3SS the inclusion membrane proteins IncA, IncB and IncC were

predicted to be T3S-mediated effector proteins of C. pneumoniae. This was possible by

constructing hybrid genes by fusing the 5' part of the chlamydial inc-gene of interest in

frame with the calmodulin-dependent adenylate cyclase (Cya) gene of Bordetella

pertussis and subsequently measuring the production and secretion of hybrid proteins239

.

Interestingly, the transcription of incA, incB and incC has been shown to occur earlier

than transcription of genes encoding components of the T3SS itself 240

. Additionally,

IncC of C. trachomatis, has been shown to be secreted through heterologous T3SS210

. In

a randomized search of proteins with unknown function conserved among C.

pneumoniae, C. trachomatis and C. caviae, 24 new candidate proteins were found.

None of the predicted proteins belonged to the Inc family of proteins. Out of these 24

candidates, four were shown to be secreted as full-length proteins by a T3S mechanism

in S. flexneri. One of the protein homologs of Cpn705 was found to be secreted in the

cytoplasm of infected cells241

. Pkn5 was originally identified and postulated to be T3SS-

related when the genome of C. trachomatis was sequenced17

. In one of the known T3S

sub-clusters (Fig. 7) the pkn5 gene is located upstream of cdsC and encodes a putative

serine/threonine kinase208

. Pkn5 has been shown to be translocated via the SPI-2 T3SS

of S. enterica222

. Recently a new member was included in the family of Chlamydia

effectors242

. The protein CT847 was revealed to be secreted in a T3SS-mediated model

Page 38: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

29

of Y. pseudotuberculosis. Moreover, the protein interacted in the cell cytoplasm with a

eukaryotic protein termed GPIC (Grap2 cyclin D-interacting protein). CT847 shares

62% sequence similarity to C. pneumoniae gene product Cpn1004 (author contribution).

TABLE 2. Chlamydial effectors

C. pneumoniae C. trachomatis

Effector Location Secreted Gene-

number Secreted

Gene-

number Possible function

IncAa Inclusion Yes

Cpn0186

Cpn0595 Yes CT119

Membrane protein

IncBa Inclusion Yes Cpn0291 No CT232

Membrane protein/host cell

interactions*

IncCa/b

Inclusion Yes Cpn0292 Yes CT233 Membrane protein

Pkn5c S/T kinase Yes Cpn0703 No CT673 S/T kinase activity

TARPd Surface of EBs No Cpn0572 Yes CT456 Invasion

Hypothetical proteind Unknown No Cpn1004 Yes CT847

Interaction with

eukaryotic GPIC

Hypothetical proteina

Secreted to

cytoplasm Yes Cpn0705 No CT671 Host cell interaction(s)

Hypothetical proteina Unknown Yes Cpn0725 No CT652.1 Unknown

Hypothetical proteina Unknown Yes Cpn0859 No CT718 Unknown

Hypothetical proteina Unknown Yes Cpn1005 No CT848 Unknown

Effector proteins secreted in heterologues system: S. flexneria, Y. enterocoliticab, S. typhimuriumc, Y.

pseudotuberculosisd. For references; see text.

Other secreted proteins There are other proteins proposed to be secreted into the cytoplasm of host cells. For

instance, Cap1 (homologous to Cpn0648) is a protein of unknown function recognized

by cytotoxic T-cells243

and CADD (homologous to Cpn0761) has been shown to be

involved in modulation of host cell apoptosis and to be located in close contact with the

inclusion244

. Another protein identified as secreted is the chlamydial protease-like

activity factor (CPAF). CPAF has the ability to cleave a eukaryotic transcription

factor245

. Translocation of CPAF does not seem to occur via the T3SS (referred to246

and

unpublished data by A. Subtil). There are two genes coding for products with unknown

function in C. pneumoniae, Cpn0796 and Cpn0797 and have no homologues in C.

trachomatis. Both are found to be secreted in the host cell cytoplasm247,248

. However,

••

Page 39: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

30

whether or not any of these proteins are T3SS-related is unknown. With ongoing efforts

to develop novel tools to better study and characterize virulence mechanisms in

Chlamydia, it is likely that the predicted function of many effector proteins will be

updated in the future.

Extracellular T3SS Most T3SSs characterized in other spp. are functional upon host cell contact e.g. when

internalized or phagocytosed. This has also been postulated as a function for the

chlamydial T3SS. The first report of a T3SS-translocated protein that was tyrosine-

phosphorylated upon cell contact was a protein named Tir (translocated intimin

receptor) described in enteropathogenic E. coli (EPEC249

). A tyrosine-phosphorylated

effector protein has also been found in C. trachomatis, named TARP (translocated

actin-recruiting phosphoprotein78

). TARP is translocated through T3S by Y.

pseudotuberculosis and is demonstrated to be involved in the recruitment of actin,

leading to internalization of C. trachomatis. Moreover, it has been reported that C.

muridarum, C. caviae, and C. pneumoniae TARP fails to become phosphorylated at

tyrosine residues at the site of entry initiating internalization, therefore indicating the

action of TARP is species-specific for C. trachomatis250

. However, all chlamydial

species examined to date show recruitment of actin to the site of entry. Thus, Chlamydia

likely shares at least some common mechanism(s) for entry.

Intracellular T3SS Chlamydia are strictly intracellular bacteria that rely on mechanisms facilitating their

required host environmental needs. Therefore, it is not remarkable that many predictions

and assumptions suggest employment of a T3SS to maintain intracellular survival. It

may be essential to understand the involvement of T3S in the development of

Chlamydia. In addition, the function of T3SS in other pathogens is most often crucial

during invasion of cells and escaping the innate immune system, and usually not for

intracellular proliferation207

.

The genome has been completely sequenced for the Parachlamydia-related

Acanthamoeba symbiont UWE25251

. UWE25 is thought to have diverged from the

family of chlamydiaceae about 700 million years ago and also encodes a complete

T3SS251

. Thus, it is possible that the common ancestor of the T3SS evolved from

Parachlamydia. The T3SS is described as a contact-dependent secretion system. It

follows that Bavoil and colleagues have formed an hypothesis using a mathematical

model that proposes that RBs grow strictly in contact with the inclusion membrane and

that this contact is mediated by the T3S apparatus217,252

. As the inclusion compartment

grows, the T3S activity decreases for every RB until the RB detaches from the inclusion

membrane, and this detachment from the chlamydial inclusion membrane constitutes

Page 40: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

31

the signal for late RB-to-EB differentiation. Moreover, this could explain why

chlamydial development, initially almost synchronous, becomes asynchronous in the

mature inclusion as EBs differentiate from RBs253

.

Temporal transcription of T3SS genes T3SS genes in Chlamydia exhibit temporal expression throughout the developmental

cycle54,210,221,240

. These genes have been clustered into three major classes of genes,

termed early-, mid- and late-cycle genes54,254

. The early-cycle genes expressed in C.

pneumoniae include cdsC, cdsS, cdsL, cdsJ and scc3. The genes expressed during mid-

developmental cycle are cdsD, cdsN and cdsR, and finally the late-cycle genes include

copN, scc1, scc2, and cdsT54

(Fig. 7). Recently, a fourth classes of genes temporally

expressed in the developmental cycle was described and defined as tardy genes110

. The

tardy gene class was established and defined based on a transcriptional profile that

differed from the late genes110

. In the study by Mayer and co-workers, there is a

difference in the classification of some genes, as for example scc3 was moved to the

mid-cycle class of genes and scc2 was placed in the tardy class. Lugert et al., also

demonstrated a different temporal expression profile, where transcription of cdsN, copN

and scc1 was detected 8 h p.i in comparison to Slepenkin et al. who were able to detect

cdsN at the time of infection, whereas copN and scc1 were not transcribed before 12 h

p.i.54,240

. These differences in transcriptional kinetics can probably be explained by the

sensitive methodology/technique of RT-PCR and different inoculation doses. The fact

that scc2 is expressed in the tardy class of genes suggests a regulatory function in the

early phase of the following infectious cycle, coding for early EB proteins. This

speculation is supported by the study where Mayer et al. compared their expression data

with data from the proteomic study of EBs of Vandahl et al.255

. Their study

demonstrate that genes coding for EB proteins were primarily connected with products

in late gene clusters, whereas transcripts coding for EB mRNAs were mainly connected

with tardy clusters110,255

. Proteomic analyses of C. pneumoniae revealed that all

components of T3S-apparatus were present in EBs255

, whereas only cdsC was found in

the outer membrane complex proteome and proposed to be surface-exposed256

.

••

Page 41: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

32

FIGURE 7. Organization of the T3SS gene cluster in the C. pneumoniae TW183 genome. Gene size is

not displayed to scale and location shown on the genomic illustration is not exact (modified from54,257).

Stress of the T3SS When Chlamydia are stressed by the cell-mediated immune system, T3SS-modulated

processes may become of importance. For instance, IFN--treated C. pneumoniae-

infected cells decrease transcription of scc1, scc2 and cdsV54

. In contrast, scc3

expressed throughout the normal developmental cycle, did not appear to be affected by

IFN- treatment54,55

. In C. psittaci, transcriptional levels of incA were analyzed during

early-mid developmental cycle (12 h p.i.) and incA was demonstrated to be up-regulated

when the infection was treated with IFN- or when iron was depleted using desferal258

.

In contrast, we have demonstrated that INP0010 treatment reduced the expression of

incA and incB transcripts in C. pneumoniae 12 h p.i. (paper III in this thesis). Moreover,

an extensive study combining the microarray-technique with qRT-PCR demonstrated a

down-regulation of the T3SS-associated genes scc1, scc2 and cdsC in an iron-induced

persistence model110

. In addition, Ouellette et al. reported that transcription of scc2 and

additional genes involved in bacterial growth were up-regulated during IFN-γ

persistence259

in contrast to previous works demonstrating an inhibition of transcription

following IFN-γ stimulation238,258,260

. Thus, transcriptional profiles of T3SS-associated

genes change under different stress-induced conditions and remain to be further

investigated.

Page 42: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

33

Antibiotics

Antimicrobial resistance is a global problem

Traditional antibiotics are used for their capacity to kill bacteria or inhibit their growth.

However, antimicrobial resistance has become a global problem and is probably one of

the most essential assignments for microbiologists and physicians in combating the

bacterial infections. Therefore, increasing efforts are present challenging new

antimicrobial strategies compensating the use of traditional antibiotics and the inability

of microorganisms to circumvent its action. Consequently, in 2002, the European Union

council of health ministers presented a strategy for battling antibiotic resistance and

proposed a policy recommendation for use of antibiotics and for better surveillance of

antibiotic resistance (according to the homepage of The Swedish Institute for Infectious

Disease Control, online) In 2005, the World Health Assembly (WHA) of the World

Health Organization (WHO) presented the resolution “Improving the containment of

antimicrobial resistance” to counteract antibiotic resistance (according to their

homepage, online). A frightening example of the consequences of antimicrobial

resistance, have been reported from the Centers for Disease Control and Prevention

(CDC) in the United States. According to their statistics, almost 2 million patients in US

that get an infection following hospitalized each year and more than 70% of the bacteria

that cause hospital-acquired infections are resistant to at least one of the antibiotics most

commonly used for treatment (according to homepage of CDC, online).

Antibiotic treatment of C. pneumoniae infection C. pneumoniae is susceptible to antibiotics belonging to the classes of macrolides,

tetracyclines and rifamycines in vivo261

. However, Chlamydia can develop antibiotic

resistance in vitro and therefore antibiotic resistance may become a future clinical

problem262-265

.

Clinical experience has shown that C. pneumoniae infections can recur after short

course therapy, why high dose treatment for 14 days is recommended117

. In vitro,

inappropriate treatment of C. pneumoniae infection may induce an aberrant persistent

infection state, unresponsive to antimicrobial treatment 91

. The nature of C. pneumoniae

infection makes prevention difficult, and since the recommended first choice treatment

includes tetracyclins and macrolides the development of new anti-chlamydial strategies

remains an important aim to reduce the use of these valuable antibiotics.

••

Page 43: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

34

Small molecules

Chemical genetics and small chemical molecules Genetic approaches have been used for decades to identify genes that regulate a

biological process of interest. Knocking out a gene of interest is possible in many

organisms today although Chlamydia and several other bacterial species lack tools for

genetic manipulations. Instead, a chemical genetic approach can be instrumental for

studying bacterial functions (reviewed in reference 266). This strategy uses small

organic molecules to reveal the specific macromolecules responsible for regulating

biological systems. First, an assay for the biological process of interest is developed,

secondly, the process is systematically inhibited with small molecules and finally the

component affected by each molecule is determined to reveal the proteins or genes

regulating the process266

. The small molecules directly alter protein function by binding

to their targets and either enhancing or inhibiting the function of the targets.

Small molecules as inhibitors of virulence When developing new antimicrobial drugs, reducing the risk of resistance is important.

While antibiotics target bacterial growth, an alternative approach would be to inhibit

bacterial virulence mechanisms. Antimicrobial resistance rarely develops in virulent

bacteria during treatment. Instead, resistance develops in the large number of non-

virulent bacteria of the resident commensal flora and spreads through horizontal gene

transfer to neighboring bacteria. Small molecule inhibitors have a high molecular

affinity, targeting a specific virulence-related factor and therefore are potentially less

likely to interact with non-virulent bacteria. Thus, resistance against virulence-specific

inhibitors is unlikely to develop and spread among the resident commensal flora.

Examples of potential applications targeting virulence include two-component signal

transduction systems, quorum sensing, biofilm formation and T3SS267-270

. For example,

virstatin blocks the transcriptional regulator ToxT in Vibrio cholerae, thereby inhibiting

the expression of virulence factors regulating toxin and pili assembly271

. Moreover, a

hydroxymate, termed LFI, inhibits the anthrax toxin in Bacillus anthracis272

. Another

example of small molecules as inhibitors of virulence is a class of compounds termed

pilicides that targets and inhibits the formation of virulence-associated pili of

uropathogenic E. coli270

.

Small Molecules and inhibition of T3SS

In an effort to identify compounds that target and inhibit T3S-induced virulence,

Elofsson and Wolf-Watz designed a high-throughput screen and examined a

commercial library of more than 9,000 compounds273

. Their approach was based on the

ability to monitor the expression of yopE of Y. pseudotuberculosis in a luciferase

Page 44: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

35

reporter assay. The system identified 12 potential T3SS inhibitors. One of the hits,

compound INP0007 belonging to a class of acylated hydrazones of salicylaldehydes,

was later shown to specifically block YopH secretion in infected HeLa cells274

.

Recently, INP0010 and INP0403, belonging to the same class of compounds as

INP0007, have been shown to inhibit T3SS of S. enterica, in two independent

studies275,276

. In the work by Hudson et al., the authors demonstrated an inhibition of

secretion via the SPI-1 T3SS-related effectors as well as an inhibition of invasion into

HeLa cells. However, INP0007 and INP0403 had to be pre-incubated with the bacteria

to suppress T3SS-dependent responses in a bovine intestinal loop model. In other work

done by Rehn et al., the authors confirmed similar inhibitory effects on invasion of

epithelial MDCK cells and secretion of SPI-1 T3SS-related effector proteins.

Interestingly, one of the tested compounds, INP0010 affected intracellular S. enterica

proliferation in macrophage-like cells at concentrations of 40 µM. Another example of

small molecules as inhibitors of T3SS has been described in EPEC277

. The most

efficient compound, composed of a halogenated salicylaldehyde derivative condensed

with a 3-aminoacetophenone, inhibited expression of T3SS-associated genes named

esps (for E. coli-secreted proteins). Notably, this compound does not belong to the same

class of compounds as INP0007.

••

Page 45: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

36

OBJECTIVES OF THIS THESIS

The general aim of this thesis was to investigate Chlamydia infection biology,

especially the T3SS.

The more specific objectives were as follows:

To investigate the virulence properties of Chlamydia spp. using chemical

genetics.

To investigate the effect of different T3S-inhibtors in the developmental cycle

of C. pneumoniae and C. trachomatis.

To obtain a valid method for monitoring initiation of transcription in C.

pneumoniae in the presence of a T3S-inhibtor.

To investigate the effect of C. pneumoniae infection on bone biology in mice.

To investigate whether C. pneumoniae can infect osteoblasts and characterize

the cytokine response.

Page 46: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

37

RESULTS AND DISCUSSION

Paper I

There are two major reasons the employment of small molecules could have important

implications in Chlamydia biology:

1. There are no molecular biological approaches to accomplish necessary genetic

manipulation in the spp. of Chlamydia. Thus, blocking a function by addition of

chemicals enables studies of its function.

2. Small molecules may present a novel opportunity for therapeutic interventions in

Chlamydia. Small molecules have previously been shown to inhibit the T3SS in

Yersinia. Given the close homology between T3SSs in Yersinia and Chlamydia we

hypothesized that small molecules could inhibit the T3SS also in Chlamydia.

Accordingly, this might evaluate the essentiality of the T3SS in pathogenesis of

Chlamydia, e.g. molecular mechanisms involved in attachment, entry, proliferation and

differentiation.

Screening of T3SS inhibitors using Y. pseudotuberculosis T3SS In a previous study, INP0007, belonging to the class of acylated hydrazones of

salicylaldehydes, prevented Y. Pseudotuberculosis YopH translocation into HeLa

cells274

. However, INP0007 displays limited solubility in water and exhibited cytotoxic

properties when used at higher concentrations in our HEp-2 infection model (data not

shown). Therefore, our interest was shifted towards the more soluble analogue

INP0010. Thus, the inhibitory as well as cytotoxic effects of INP0010 had to be

determined first. For this purpose we applied the rapid screening assay of T3S-inhibitors

that measures both virulence inhibition of Yersinia infection in cell culture, as well as

compound cytotoxicity. INP0010 was not cytotoxic in the assay at concentrations as

high as 50 µM. Moreover, microscopic examinations revealed that cell proliferation and

morphology were unaltered in HEp-2 cells grown at 10 µM of INP0010 for up to 72 h

(data not shown). Thus, based on these data INP0010 was chosen as a suitable T3S-

inhibitor in our infection model. Moreover, in order to ensure that any observed effects

on Chlamydia are the result of T3SS blockage, we also included the compound

INP0406 which belongs to the same chemical class as INP0010, but is unable to inhibit

the T3SS of Yersinia.

••

Page 47: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

38

INP0010 inhibits C. pneumoniae intraceullar propagation When INP0010 at 10 µM was added to C. pneumoniae infected cells, no chlamydial

inclusions could be observed. These results suggest an altered developmental cycle. To

further investigate the effect of INP0010 on bacterial proliferation, we performed a set

of control experiments:

1. Pre-treatment of the host cells with INP0010 prior to cell infection had no

effect on C. pneumoniae propagation.

2. Independent of when INP0010 was added during the early developmental cycle

(0-12 h), C. pneumoniae proliferation was inhibited.

3. A dose-dependent effect of INP0010 was demonstrated both with immuno-

fluorescence and quantitative real-time PCR at 48 h p.i., respectively.

4. Heat-inactivated C. pneumoniae did not proliferate and was present at similar

bacterial numbers as when INP0010 at 10 µM was added to the cell culture.

INP0010 shows diverse effect in C. trachomatis To our surprise, when HEp-2 cells were infected with C. trachomatis serovar L2 and

treated with INP0010 at concentrations ranging from 10 – 30 µM, no inhibitory effect

could be observed. Importantly, no inhibitory effect could be observed on either C.

pneumoniae- or C. trachomatis-infected cells treated with INP0406. Together, these

results suggest important differences in the T3SS of different species. However, the

underlying reason for this is not clear since the T3SSs in the two Chlamydia species are

likely to be more closely related to each other than to Y. pseudotuberculosis.

Interestingly, C. pneumoniae have a broader repertoire of postulated effector proteins

than C. trachomatis, suggesting differential biology, tropism and pathogenesis between

the two different spp. This is supported by the ability of C. pneumoniae to be more

invasive and survive in a broader range of host cell types than C. trachomatis 18

. This

suggests the possibility of using small molecules to inhibit different targets and

functions in Chlamydia.

INP0400 is active against both C. pneumoniae and C.

trachomatis. Intracellular replication of C. trachomatis was not affected by 30 µM of INP0010.

Therefore we expanded the search for putative T3S-inhibitors that could block

proliferation for both C. pneumoniae and C. trachomatis. Compound INP0400 inhibited

propagation of C. pneumoniae and C. trachomatis at 10 µM and demonstrated similar

inhibitory effect in Y. pseudotuberculosis, without cytotoxic effects. In conclusion, by

modifying the chemical structure(s) and screening each compound in our infection

Page 48: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

39

model, it was possible to identify a novel compound which was active against both

chlamydial spp. This is in contrast to INP0010, which was selectively active against C.

pneumoniae. This indicates that the target for this class of compound shows minor

differences in the two Chlamydia spp. examined herein. Thus, different small molecules

made it possible to further characterise the differences in the T3SS in the different

Chlamydia spp. This strategy constitutes a good substitute for the lack of genetic

methods in these pathogens.

Treatment with INP0010 inhibits secretion of putative T3SS

effector proteins Inclusion membrane proteins IncA, IncB and IncC are predicted to be T3S-mediated

effector proteins of C. pneumoniae as they are secreted in heterlogous T3SS 239

. We

demonstrated by using immunofluorescence microscopy, that IncB and IncC was

localized to the chlamydial inclusion in close association with the bacteria. However,

no specific staining of IncB and IncC could be observed in the presence of INP0010 at

10µM. These results suggest of an inhibition in translocation of the predicted effector

proteins when INP0010 is present. This is later supported in paper IV.

••

Page 49: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

40

Paper II

In paper I we identified INP0400 as an inhibitor of Chlamydia propagation. In paper II

we have further evaluated the action of INP0400 on C. trachomatis growth-mediated

processes, revealing the T3SS as essential for development and pathogenesis of C.

trachomatis.

INP0400 inhibits proliferation but not primary differentiation C. trachomatis-infected McCoy cells treated with INP0400 at 20 µM showed similar

inhibitory effects as that observed in HEp-2-infected cells (paper I). This was later

corroborated by another group, whereas INP400 at 20 µM inhibited growth of C.

trachomatis serotype D in McCoy and HeLa cells278

.

However, the process of re-differentiation from RBs-to-EBs is poorly investigated.

During the re-diffrentiation process, the expression of a number of late-cycle T3SS-

associated genes occurs. We demonstrated a dose-dependent reduction in inclusion size

and a concomitant reduction in the number of intracellular bacteria in infected cells

following INP0400-treatment, 30 h p.i. This is indicative of an inhibition of

proliferation, as RBs have already started to re-differentiate to infectious EBs, making

the visual inclusion larger in size at 30 h p.i. By electron microscopy we confirmed that

internalized EBs in the presence of INP400 converted to RBs. However, the

proliferation of RBs were inhibited in a dose-dependent manner, resulting in smaller

inclusion bodies containing just one or a few RBs. Accordingly, this implicates that the

T3SS might regulate the terminal differentiation of Chlamydia This is supported by the

work of Fields and co-workers279

.

INP400 inhibits secretion of putative effectors We further investigated whether INP0400 could interfere with secretion of T3S-effector

proteins. We could demonstrate that homotypic inclusion fusions were inhibited when

INP0400 was given at 8 h p.i, a time-point in the developmental cycle that precedes the

onset of IncA production87

. Accordingly, no detection of IncA-antibody signals was

seen in the non-fused inclusions. This proposes an inhibition of effector translocation

when INP0400 is present. This result is supported by the work by Wolf et al., who also

could not detect IncA in the inclusion membrane following INP0007-treatment of C.

trachomatis infected cells279

.

Moreover, IncG of C. trachomatis has been shown to interact with the eukaryotic

protein 14-3-3β89

. Accordingly, we demonstrated that treatment with INP0400, early in

establishment of infection in McCoy cells, prevented the host protein 14-3-3 from

localizing to the inclusion membrane. Therefore, it can be proposed that the T3S-

Page 50: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

41

inhibition prevents translocation of IncG into the inclusion membrane. Interestingly, C.

pneumoniae do not have homologues to IncG nor undergo homotypic fusions resulting

in a single inclusion in cells that are multiply infected. This might support the idea of

two differentially regulated T3SS between C. pneumoniae and C. trachomatis, as

proposed in paper II.

INP0400 treatment during the late cycle promotes bacterial

dissociation from the inclusion membrane C. trachomatis RBs are typically found juxtaposed to the inner surface of the inclusion

membrane during mid-cycle development. However, it is unknown whether this

attachment requires effectors delivered by the chlamydial TTS system. Likewise, it is

not known whether RB association with the inclusion membrane is required for

bacterial multiplication and/or RB-to-EB transition. Peters et al., have predicted a model

for T3S contact-dependent development using a mathematical model, which predicts

that RBs detach from the inclusion membrane after secretion of T3S effectors, thus

T3SS become inactivated and late differentiation begins217,252

. It can be described as a

“switch button” turning the T3SS on-and-off. T3SS is on (active) when the bacteria are

in contact with the inclusion membrane and off (inactive) when terminal RB-to-EB

transition occurs. We showed that INP0400-treatment during mid-developmental cycle

induced a failure of the bacteria to localize to the inclusion membrane, instead leaving

free space between the inclusion membrane and the bacteria. Surprisingly, when

INP0400 was removed at the end of the developmental cycle and infected cells further

cultured for a short period (4 hours) in fresh culture media, the bacteria re-associated

with the inclusion membrane. This might support the “switch button” theory whereby

the presence of the T3S-inhibitor mimics the naturally occurring stage when RBs detach

from the inclusion membrane and begin differentiation.

••

Page 51: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

42

Paper III

In paper III, we describe the inconvenience of using different internal gene-expression

controls for normalizing the expression of a target gene of interest. Instead, DNA should

be considered, since it is always present, stable and a varied DNA level directly reflects

proliferation of the bacteria. Moreover, INP0010 decreases initiation of transcription in

C. pneumoniae during the early developmental cycle as demonstrated by a novel

mathematical model.

DNA is preferable to RNA for normalization of gene expression In general, stable transcripts are used to normalize the expression of a target gene. This

can, however, be deceptive given:

1. Variation in transcript stability between control and target mRNAs.

2. Levels of mRNA can vary dependent of when it is essential in the

developmental cycle.

In this study, we monitored expression of T3SS-associated genes and genes encoding

proteins involved in RNA-polymerisation, heat shock responses and DNA replication.

Accordingly, some of the genes were presumed to be efficient control genes for

normalization, at 12 hours p.i. This window of time was suitable when the primary

differentiation takes place and replication is initiated. To test transcript stability, we

used rifampicin which inhibits de novo protein synthesis by binding to the RNA-

polymerase. Rifampicin was added to infected cells with or without INP0010 at two

distinct phases of the developmental cycle:

1. During early infection (0 hours p.i.) when represented as infectious EBs.

2. During early mid-developmental growth (12 hours p.i.) and when RBs are

present.

In general, transcripts of EBs were more stable, when compared to RB transcripts. In

contrary to the situation at 0 hours p.i., transcripts of rpoD, cdsS and incA could not be

detected after one hour of rifampicin treatment, at 12 h p.i. This suggests that these

transcripts are quickly turned-over, during the transition from EBs to RBs. Taken

together, target and control RNAs display a varied stability at different developmental

phases, both in the presence or absence of INP0010.

Relative gene expression is not necessarily correlated to proliferation since the

expression might fluctuate significantly throughout the developmental cycle. This

would give a relative target mRNA expression that follows the expression-pattern of the

control mRNA, rather than reflecting the bacterial proliferation. Accordingly, several

control and target mRNAs were induced at the early-mid developmental cycle (12 hours

Page 52: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

43

p.i.) when compared to early infection (2 h p.i.). Consequently, using 16S, rpoA and

rpoD as controls for measuring the relative target gene-expression throughout the

developmental cycle, would result in a markedly reduced relative gene-expression at 12

hours p.i., compared to early infection. Although the numbers of bacteria remains

unaltered. Relative gene expression should be correlated both to the expression of the

target and the control mRNA as well as the degradation, when using RNA as control.

Moreover, the diverse stability between different/or the same RNA species could

depend on the growth phase and presence/absence of INP0010. Consequently, RNA

should be avoided as control for normalization against your target gene expression of

interest, instead, DNA should be used as a control.

INP0010 decreases initiation of transcription Determination of mRNA half-life is important to fully understand mechanisms involved

in gene regulation in response to INP0010 or by developmental signals. In addition, the

stability of mRNA may indicate how rapidly the translation of the encoded protein can

be shut down. Therefore, we have determined the relative transcription initiation

constant (k) using expression of C. pneumoniae genes and by introducing the relative

transcript amount and decay into a mathematical model. Using this model, we could

demonstrate that when INP0010 is present, initiation of transcription decreases. This

suggests that stabilized mRNA levels induced with INP0010, in theory, could increase

protein amount. This proposal is consistent with the work by Wolf et al., who

demonstrated accumulation of T3SS-associated substrates within INP0007-treated

RBs279

. However, further work is required to determine whether there is a correlation

between an increased chemical half-life and a more functional mRNA.

The developmental cycle is initiated as the metabolically inactive EBs are endocytosed

by the host cell, where they are tightly bound within a membranous vesicle. Within this

vesicle, unknown environmental signals trigger an initial round of chlamydial

transcription. Our results suggest that INP0010 in general decreases initiation of

transcription.

Expression of T3SS in the presence of INP0010 In paper I, we presented results indicating that expression of most T3SS-associated

genes were down-regulated after INP0010-treatment. However, these results were based

on normalisation against the ribosomal 16S RNA. We therefore refined previous results

and normalised the presence of mRNA against the native DNA. When normalised

against DNA, a reduction was detected for all RNAs tested in the presence of INP0010.

Thus, the inhibition of expression was not specific for T3SS-associated genes during

early developmental events. This indicates that INP0010 functions generally during

early developmental cycle and reduces expression of all, at least those tested herein,

genes by a hitherto unknown mechanism.

••

Page 53: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

44

Paper IV

In paper IV, we suggest that INP0010 target the T3SS in C. pneumoniae infected cells

and thereby arrest the RB, either after primary differentiation or during terminal re-

differentiation. Moreover, INP0010 inhibited expression of T3S-genes necessary for the

exterior part of the T3S injectisome during the late developmental cycle. Consequently,

the effector protein IncB was not detected in the cytoplasm following INP0010

treatment. The effect of INP0010 on iron-responsive genes indicates a role for T3S in

iron acquisition. Accordingly, our results suggest a possibility for C. pneumoniae to

acquire iron via the intracellular trafficking pathway of endocytosed transferrin.

INP0010 inhibits early- and mid-developmental cycle

C. pneumoniae modulates its entry by rearranging the host cells cytoskeleton upon

binding. Other pathogens are known to use their T3SS to inject effector proteins upon

host cell contact. However, our uptake experiments demonstrated that INP0010 did not

interfere with C. pneumoniae internalisation, suggesting either a non-active T3S

apparatus or an independent T3S-mechanism during entry. Accordingly, INP0010

analogue in infection models of C. trachomatis L2 and serovar D also failed to interfere

with invasion278,279

. We have previously suggested (paper II) that INP400 inhibits

progression of C. trachomatis in either the mid-developmental phase when replication

of the RBs occurs or during the re-differentiation of RBs back into infectious EBs. In

addition, we could show that the metabolically inactive EB converts to an RB, but in

contrast to when C. trachomatis is INP400-treated, RB multiplication is totally inhibited

when INP0010 is present. Thus, INP0010 interferes with signals required for early RB

proliferation processes which are in agreement with the results presented in paper III,

demonstrating a general reduction of transcription.

Iron initiates proliferation but not terminal re-differentiation in

the presence of INP0010 Dual action of T3S-inhibitors has been suggested, whereby INPs are also capable of

chelating iron278

. Thus, this enabled us to use INP0010 to examine the T3SS and its

ability to regulate iron required for Chlamydia propagation. To be able to monitor

potential iron modulated process governed by the T3S, INP0010-effects were compared

with the effects achieved by the classical iron-chelator desferal (DAM). We showed that

Fe2+

could reverse the RB proliferation inhibitory effect of INP0010 when additional

Fe2+

was available during infection. However, reorganization of C. pneumoniae RBs

into EBs was inhibited as demonstrated by re-infection experiments and TEM. This

suggests an essential role of the T3S and iron in transducing an undefined signal for

terminal differentiation in C. pneumoniae. Moreover, expression of the putative iron-

Page 54: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

45

binding proteins, YtgA and DcrA, increased when compared to wild-type infection,

supporting a function for the T3SS in iron acquisition. Furthermore, addition of iron-

saturated holo-transferrin to INP0010-treated C. pneumoniae-infected cells resulted in

100% inhibition of EB production. In contrast, Slepenkin et al. demonstrated 100%

recovery of C. trachomatis when holo-transferrin was added in the presence of

INP0341278

. This further suggests differences in T3SSs between the two Chlamydia spp.

It has previously been shown that the early endosome, containing transferrin is a likely

Chlamydia-source of iron95,97

. In addition, we could demonstrate when using DAM that

decreasing the host cellular pool of iron re-directed transferrin receptors to the inclusion

sites, presumably due to lack of iron-accessibility for Chlamydia. However, when

INP0010 was present, no accumulation of transferrin receptors was shown in close

contact to the inclusions, even though INPs has iron-chelating properties. Thus,

INP0010 could have blocked the T3S-secretion of effectors essential for host trafficking

pathway e.g. endosomal trafficking. Presumably, the Chlamydia could use the T3S-

injectisome to interfere with the early endosome to sequester iron deposited by

transferrin and this is might have been initiated by secretion of effector proteins for e.g.

redirection of the endosome.

INP0010 blocks secretion of IncB into the host cell cytoplasm We propose that IncB can be secreted into the host cell cytosol via a T3S iron-

dependent mechanism. Supplementation of ferrous iron was sufficient to re-establish the

expression of IncB in the bacteria, both at the transcriptional and translational levels.

However, it appears that secretion was affected when INP0010 was added in the

presence Fe2+

. Taken together, our results suggest that C. pneumoniae might use the

T3SS needle structure to interfere with the endosome to sequester iron deposited by

transferrin (summarised in Fig. 8).

••

Page 55: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

46

FIGURE 8. Schematic illustration of C. pneumoniae development in the presence of the T3S-inhibtior

INP0010 at 10 µM. C. pneumoniae is endocytosed independently of INP0010 1. INP0010 arrests the RB after

primary differentiation 2. IncB secretion is inhibited in the presence of INP0010 3. Effector protein(s)

presumably interacts with the transferrin receptor trafficking pathway and/or early endosomes for iron

acquisition. RBs prolifierate in the presence of INP0010 and ferrous iron (insert). 4. Re-differentiation is

inhibited in the presence of INP0010 and ferrous iron.

Page 56: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

47

Paper V

In paper 5, we have for the first time presented data showing generalized bone loss in C.

pneumoniae infection in mice. The infection was associated with increased levels of the

bone resorptive cytokines IL-6 and IL-1, as well as demonstrating an increased sub-

population of T-cells expressing RANKL on their cell surface. In addition, C.

pneumoniae established an infection in a human osteoblast cell line in vitro with similar

cytokine profiles as seen in vivo, supporting a causal linkage. Collectively, these data

may indicate a previously unknown pathologic role of C. pneumoniae in generalized

bone loss.

C. pneumoniae infection decreases bone mineral density in mice Mice were inoculated intranasal once a week for three consecutive weeks and bone

mineral density was measured before and 16 days p.i. Infected mice demonstrated a

significant reduction in bone mineral density of the distal femur and proximal tibia

when compared with sham infected mice. Importantly, there was no significant

difference in weight gain during the infection period when comparing sham infected and

infected mice. The infection was associated with increased levels of the bone resorptive

cytokines IL-6 and IL-1, as well as demonstrating an increased sub-population of T-

cells expressing RANKL. Taken together, these results suggest that C. pneumoniae

mediated induction of the innate immune system might modulate bone biology

processes in mice. This was further evaluated in vitro.

C. pneumoniae growth in hFOBs increases production of IL-6

and expression of RANKL In this paper we used the hFOB cell line that expresses the normal osteoblastic

phenotype as well as RANKL280,281

to study the effect of C. pneumoniae infection. The

infected hFOBs demonstrated significantly higher amounts of IL-6, both on mRNA and

protein level, when compared to non-infected cells. Moreover, the secretion increased in

a dose-dependent manner when the cells were infected. Interestingly, other studies have

demonstrated similar production of IL-6 at sites of bacterial infection in human S.

aureus-associated osteomyelitis175

. In addition, the increased production of IL-6, in our

study, was dependent of chlamydial protein synthesis as demonstrated by a set of

control experiments:

1. Supernatants collected from previously infected hFOBs could not induce

expression of IL-6.

••

Page 57: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

48

2. Heat-inactivated C. pneumoniae did not induce secretion of IL-6. Thus, neither

LPS nor surface exposed glycoproteins mediates the IL-6 production.

3. The presence of non-proliferating C. trachomatis could not induce significant

cytokine secretion when compared to non-infected cells.

Additionally, IL-1 expression was up-regulated in hFOB cells in response to infection.

However, the levels of secreted IL-1 were not increased in the supernatant of infected

cells when compared to non-infected cells. This was further investigated and we found

that hFOBs do not secret high levels of IL-1 even when stimulated with pure LPS from

E. coli (data not shown). Finally, RANKL expression was 3-fold higher in infected

cells.

Interestingly, the inflammatory response of bone destruction resembles the pathogenesis

of atherosclerosis, postulated to be accelerated by C. pneumoniae infection. Therefore,

C. pneumoniae could be involved in processes of the vascular- and bone remodelling-

system (summarized in Fig. 9). Our novel results indicate that certain bacteria can

contribute to general bone loss, and this needs to be further investigated in humans.

FIGURE 9. Influence of C. pneumoniae on bone and vascular tissue.

Page 58: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

49

CONCLUSIONS

The T3S-inhibtors INP0010 and INP0400 block the developmental cycle of C.

pneumoniae and C. trachomatis, without any cytotoxic effect, respectively.

INP0400 inhibits secretion of the T3S effector-protein IncA in C. trachomatis, which is

needed for inclusion fusion.

INP0400 targets the T3SS and provokes a bacterial dissociation from the inclusion

membrane presumed to mimic the natural occurrence of terminal differentiation

INP0010 decreases initiation of transcription in C. pneumoniae during the early mid-

developmental cycle as demonstrated by a novel calculation, useful for measurement of

transcription initiation in any intracellular pathogen.

INP0010 targets the T3SS of C. pneumoniae and thereby arrests RB proliferation as

well as RB to EB re-differentiation.

C. pneumoniae uses the transferrin receptor pathway as an iron-uptake system, and may

be regulated via the T3SS.

C. pneumoniae infection decreases bone mineral density in mice, and is associated with

increased production of IL-6, IL-1 and RANKL.

C. pneumoniae infection of human osteoblast-like cells in vitro increases production of

IL-6 and expression of IL-1 and RANKL.

••

Page 59: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

50

ACKNOWLEDGEMENTS

Reflections of writing a thesis: Stayed without my family for weeks, crashed my car,

lived at IKSU without using any of their facilities, ate pasta daily, tried not to be

distracted even though my hockey team had their worse season ever… Suddenly I heard

an echo of Sven Bergström’s voice in my head; you are always so positive Lelle! I

almost forgot…

I like to acknowledge all the people at MolBiol and UmU who have helped me through

the years, without any ranking score. Sven, my co-supervisor (sounds strange to call co-

) who gave me the opportunity to be in his lab and for taking care of me. I have never

told you this before, but you have always remained me of one the greatest leaders in the

history of football– Peter Antonine! Peter, my supervisor, who has been a great

inspiration with lots of exciting ideas through the years. Anders, co-supervisor, for

always being so positive and open-minded in our projects. Åsa co-worker and co-

supervisor, you have been great to work with and helpful in all aspects. Johnny

(Hegerfors), co-worker, co-trainer, co-driver, co-babysitter but most of all, my beloved

friend. Hasse, for being an endless source of ideas and knowledge. J.J, a great

researcher and an even greater hockey-fan, Forza LHF! Mikael E., for being so helpful

and interested in the T3S project. Matt, for with a very short notice, reading my thesis –

You did a great job! Thanks!! Our collaborators at MTC, Sandra, Birgitta and

Staffan. Thanks for a great work with the PNAS paper. Göran K Hansson and co-

workers, for sharing knowledge concerning atherosclerosis. Ulf and co-worker Anita,

for sharing your enthusiasm in infections and bone biology. Patte, the one I have shared

most ideas and lab-time with through the years. You have become like a younger

brother to me. It has been a privilege to work with you and “stafettpinnen” is yours now.

Just remember to always have a plan B. Lenore for being incredible with the electron

microscope.

The Borrelia clan: Betty, or simply B, for being the best corrector and co-worker. I

don’t know how to pay you back but I will come up with something! Iggy for being a

great room-mate on tour and all your assistance in the office – Iggyyy!! Christer, is-it-

good-or-is-it-bad, Larsson, for being a joy of life. Coma, full with unknown talents, at

least for me You really kicked ass with the Finns! (Anna: TACK!). Marie A, a.k.a Ms

Bindefeldt, for always being so helpful, with everything. You are probably the most

pedagogic person I ever met – have seen you in action at course! Ingela for being the

coordinator per se. Your contribution is eternal. Elin, the best belly-dancer in groupSB.

You are a sunshine! Lisette, for spreading some Norrbottens-spirit in the group. You

are a true lab-inspiration! Marie B, for sharing your energy every morning, always a

smile! Former members of group SB; Yngve, Pinnen och Palle, you were all inspiring.

Page 60: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

51

All my former students, who have been great colleagues and interested in our projects:

Nisse, Wolfie, Tobbe, Marco, Lisa, Tina and Susanne.

Marita, Ethel, Berith, and the rest of the coordinating-staff for always being so caring!

Kerstin at Medicine, you have been amazing helping me handling all papers. Lab

service, Agnetha and co-workers for doing the best job, for us and our students! Britt-

Inger and Jenny (51135) at Djuris, for taking care of our mice.

Ulle, for all nice conversions, Smut, Eddie, Jenny, Babsan – the funniest person ever.

Keep it up Babsan! Stina, Anna, Annika and Connie – thanks for the breakfasts!

Jempa for being DJ Roccos biggest fan and for having the nicest mum – love her food

and bakeries. Janne, Maria N, Sofia, Petra, Sara G, Lissandro, Jeanette, David,

Jocke, Linda J, Sara C, Sara R, Gosia, Monica, Katrin, Olena and Stefan for

supporting DJ Rocco. Krisse, Pelle and Mike for your kindness and support. Micke W,

still go strong! Näsorna, Mari, Vickan, Johan, Maria, Mats and the latin-lover

Geovanni for all laughs in tha Bunkern. Rutan, for your patience in the FACS room.

Till alla mina fotbollsvänner: GCK P84-86, MSK P87-90 . Många av er har blivit mina

vänner, men kom ihåg att jag fortfarande är CoachB! Fredde, Fuad, Andy, Babbe,

Zacke, Adam, Lasse, Nisse, Said, Solle, the Dunkels, Are, Nille, Jacke, Ragge,

Jeppe, Robban, Calle, Tobbe, Peter och alla ni andra! Håkan Arestav – keep up the

good work. Ettan väntar! Till fotbollsfamiljerna Stenman’s och Chennoufi’s. Ni är

väldigt betydelsefulla för mig och min familj, med den kärlek ni visat Elliot.

Till alla mina Rosviks-bröder med respektive familj: Pierre (still…), Sotar´n, Sniff,

Andreas, Micke, Fredde, Pelle L och Janne. Mina Pite-bröder med respektive familj:

Eng (TACK mannen!), Ros, Pelle B, Macce, Ibbe och OK/Q8-Kalle. Nitte, I´m your

biggest fan (TACK mannen!). Jag är evigt tacksam er vänskap. Fastän vi är utspridda i

landet så finns det ingen gårdag när vi träffas. Jag älskar er alla, och kommer alltid finns

där för er, det vet ni!

Elliots bästis Emma, du är en pärla!

Till Lisas föräldrar Birgitta och Tomas. Ni har varit fantastiska under denna tid. Jag

lovar att aldrig mer skriva en avhandling! Resterande ”Lundbergare” Sollan, Curthen,

Sofi, Anders och storkusinerna Theo och Vilmer – Tack för all kärlek ni ger oss!!

Till min far och bäste vän, Leslie Sr. Du har alltid uppmuntrat mig till att göra det jag

tror på. Mannen, jag tror Barcelona fortfarande har en chans! Lilian, min syster, mitt

stöd och min ögonsten. Du är bäst!!! Jag älskar er!

Dragamama, ovom knjigom zelim ti pokloniti svu moju ljubav i postovanje. Ti si mi

podarila zivot, tvoja ljubav ispunjava svaki trenutak u mom zivotu. Volim te i

obozavam najvise na svijetu. Tvoj sin Lelle.

Till sist vill jag tacka Lisa och Elliot som skrivit denna bok tillsammans med mig. Ni är

de käraste jag har och jag ser fram emot att leva resten av mitt liv med er. Jag älskar er!

••

Page 61: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

52

REFERENCES 1. Berger, L., Volp, K., Mathews, S., Speare, R. & Timms, P. Chlamydia

pneumoniae in a free-ranging giant barred frog (Mixophyes iteratus) from

Australia. J Clin Microbiol 37, 2378-80 (1999).

2. Everett, K.D. Chlamydia and Chlamydiales: more than meets the eye. Vet

Microbiol 75, 109-26 (2000).

3. Schechter, E.M., Tribby, II & Moulder, J.W. Nucleic Acid Metabolism in L Cells

Infected with a Member of the Psittacosis Group. Science 145, 819-21 (1964).

4. Page, L. Proposal for the recognition of two species in the genus Chlamydia.

International Journal of Systematic Bacteriology 18, 51–66 (1968).

5. Grayston, J.T. Immunisation against trachoma. Pan American Health Organization

Scientific Publication 147, 549 (1965).

6. Grayston, J.T., Kuo, C.C., Wang, S.P. & Altman, J. A new Chlamydia psittaci

strain, TWAR, isolated in acute respiratory tract infections. N Engl J Med 315,

161-8 (1986).

7. Grayston, J.T., Wang, S.P., Kuo, C.C. & Campbell, L.A. Current knowledge on

Chlamydia pneumoniae, strain TWAR, an important cause of pneumonia and other

acute respiratory diseases. Eur J Clin Microbiol Infect Dis 8, 191-202 (1989).

8. Gnarpe, H., Gnarpe, J., Sundelof, B., Gustafson, R. & Gardulf, A. Prevalence of

specific antibodies to Chlamydia pneumoniae (TWAR) in Swedish orienteers.

Lancet 340, 1047-8 (1992).

9. Wesslen, L., Pahlson, C., Friman, G., Fohlman, J., Lindquist, O. & Johansson, C.

Myocarditis caused by Chlamydia pneumoniae (TWAR) and sudden unexpected

death in a Swedish elite orienteer. Lancet 340, 427-8 (1992).

10. Wesslen, L., Pahlson, C., Lindquist, O., Hjelm, E., Gnarpe, J., Larsson, E.,

Baandrup, U., Eriksson, L., Fohlman, J., Engstrand, L., Linglof, T., Nystrom-

Rosander, C., Gnarpe, H., Magnius, L., Rolf, C. & Friman, G. An increase in

sudden unexpected cardiac deaths among young Swedish orienteers during 1979-

1992. Eur Heart J 17, 902-10 (1996).

11. Hjelm, E., Wesslen, L., Gnarpe, H., Gnarpe, J., Nystrom-Rosander, C., Rolf, C. &

Friman, G. Antibodies to Chlamydia pneumoniae in young Swedish orienteers.

Scand J Infect Dis 33, 589-92 (2001).

12. Wesslen, L., Ehrenborg, C., Holmberg, M., McGill, S., Hjelm, E., Lindquist, O.,

Henriksen, E., Rolf, C., Larsson, E. & Friman, G. Subacute bartonella infection in

Swedish orienteers succumbing to sudden unexpected cardiac death or having

malignant arrhythmias. Scand J Infect Dis 33, 429-38 (2001).

13. Schiellerup, P., Dyhr, T., Rolain, J.M., Christensen, M., Damsgaard, R., Ethelberg,

S., Fisker, N., Frost Andersen, N., Raoult, D. & Krogfelt, K.A. Low

seroprevalence of bartonella species in danish elite orienteers. Scand J Infect Dis

36, 604-6 (2004).

14. Everett, K.D., Bush, R.M. & Andersen, A.A. Emended description of the order

Chlamydiales, proposal of Parachlamydiaceae fam. nov. and Simkaniaceae fam.

nov., each containing one monotypic genus, revised taxonomy of the family

Page 62: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

53

Chlamydiaceae, including a new genus and five new species, and standards for the

identification of organisms. Int J Syst Bacteriol 49 Pt 2, 415-40 (1999).

15. Schachter, J., Stephens, R.S., Timms, P., Kuo, C., Bavoil, P.M., Birkelund, S.,

Boman, J., Caldwell, H., Campbell, L.A., Chernesky, M., Christiansen, G., Clarke,

I.N., Gaydos, C., Grayston, J.T., Hackstadt, T., Hsia, R., Kaltenboeck, B.,

Leinonnen, M., Ojcius, D., McClarty, G., Orfila, J., Peeling, R., Puolakkainen, M.,

Quinn, T.C., Rank, R.G., Raulston, J., Ridgeway, G.L., Saikku, P., Stamm, W.E.,

Taylor-Robinson, D.T., Wang, S.P. & Wyrick, P.B. Radical changes to chlamydial

taxonomy are not necessary just yet. Int J Syst Evol Microbiol 51, 249; author

reply 251-3 (2001).

16. Tanner, M.A., Harris, J.K. & Pace, N.R. Molecular phylogeny of Chlamydia and

relatives. In Chlamydia: Intracellular Biology, Pathogenesis, and Immunity. Edited

by R. S. Stephens. Washington, DC: American Society for Microbiology., 1-8

(1999).

17. Stephens, R.S., Kalman, S., Lammel, C., Fan, J., Marathe, R., Aravind, L.,

Mitchell, W., Olinger, L., Tatusov, R.L., Zhao, Q., Koonin, E.V. & Davis, R.W.

Genome sequence of an obligate intracellular pathogen of humans: Chlamydia

trachomatis. Science 282, 754-9 (1998).

18. Kalman, S., Mitchell, W., Marathe, R., Lammel, C., Fan, J., Hyman, R.W.,

Olinger, L., Grimwood, J., Davis, R.W. & Stephens, R.S. Comparative genomes of

Chlamydia pneumoniae and C. trachomatis. Nat Genet 21, 385-9 (1999).

19. Chi, E.Y., Kuo, C.C. & Grayston, J.T. Unique ultrastructure in the elementary

body of Chlamydia sp. strain TWAR. J Bacteriol 169, 3757-63 (1987).

20. Carter, M.W., al-Mahdawi, S.A., Giles, I.G., Treharne, J.D., Ward, M.E. & Clark,

I.N. Nucleotide sequence and taxonomic value of the major outer membrane

protein gene of Chlamydia pneumoniae IOL-207. J Gen Microbiol 137, 465-75

(1991).

21. Popov, V.L., Shatkin, A.A., Pankratova, V.N., Smirnova, N.S., von Bonsdorff,

C.H., Ekman, M.R., Morttinen, A. & Saikku, P. Ultrastructure of Chlamydia

pneumoniae in cell culture. FEMS Microbiol Lett 68, 129-34 (1991).

22. Popov, V.L., Pankratova, V.N., Prozorovskii, S.V., Saikku, P., Smirnova, N.S. &

Shatkin, A.A. [The ultrastructure of a new species of Chlamydia--Chlamydia

pneumoniae]. Zh Mikrobiol Epidemiol Immunobiol, 2-8 (1992).

23. Miyashita, N., Kanamoto, Y. & Matsumoto, A. The morphology of Chlamydia

pneumoniae. J Med Microbiol 38, 418-25 (1993).

24. Wolf, K., Fischer, E. & Hackstadt, T. Ultrastructural analysis of developmental

events in Chlamydia pneumoniae-infected cells. Infect Immun 68, 2379-85 (2000).

25. Brade, L., Zych, K., Rozalski, A., Kosma, P., Bock, K. & Brade, H. Structural

requirements of synthetic oligosaccharides to bind monoclonal antibodies against

Chlamydia lipopolysaccharide. Glycobiology 7, 819-27 (1997).

26. Birkelund, S., Lundemose, A.G. & Christiansen, G. Characterization of native and

recombinant 75-kilodalton immunogens from Chlamydia trachomatis serovar L2.

Infect Immun 57, 2683-90 (1989).

27. Ingalls, R.R., Rice, P.A., Qureshi, N., Takayama, K., Lin, J.S. & Golenbock, D.T.

The inflammatory cytokine response to Chlamydia trachomatis infection is

endotoxin mediated. Infect Immun 63, 3125-30 (1995).

••

Page 63: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

54

28. Brade, H., Brade, L. & Nano, F.E. Chemical and serological investigations on the

genus-specific lipopolysaccharide epitope of Chlamydia. Proc Natl Acad Sci U S A

84, 2508-12 (1987).

29. Holst, O., Brade, L., Kosma, P. & Brade, H. Structure, serological specificity, and

synthesis of artificial glycoconjugates representing the genus-specific

lipopolysaccharide epitope of Chlamydia spp. J Bacteriol 173, 1862-6 (1991).

30. Barbour, A.G., Amano, K., Hackstadt, T., Perry, L. & Caldwell, H.D. Chlamydia

trachomatis has penicillin-binding proteins but not detectable muramic acid. J

Bacteriol 151, 420-8 (1982).

31. Fox, A., Rogers, J.C., Gilbart, J., Morgan, S., Davis, C.H., Knight, S. & Wyrick,

P.B. Muramic acid is not detectable in Chlamydia psittaci or Chlamydia

trachomatis by gas chromatography-mass spectrometry. Infect Immun 58, 835-7

(1990).

32. Moulder, J.W. Why is Chlamydia sensitive to penicillin in the absence of

peptidoglycan? Infect Agents Dis 2, 87-99 (1993).

33. Matsumoto, A. & Manire, G.P. Electron microscopic observations on the effects of

penicillin on the morphology of Chlamydia psittaci. J Bacteriol 101, 278-85

(1970).

34. Brown, W.J. & Rockey, D.D. Identification of an antigen localized to an apparent

septum within dividing chlamydiae. Infect Immun 68, 708-15 (2000).

35. Skipp, P., Robinson, J., O'Connor, C.D. & Clarke, I.N. Shotgun proteomic analysis

of Chlamydia trachomatis. Proteomics 5, 1558-73 (2005).

36. Knudsen, K., Madsen, A.S., Mygind, P., Christiansen, G. & Birkelund, S.

Identification of two novel genes encoding 97- to 99-kilodalton outer membrane

proteins of Chlamydia pneumoniae. Infect Immun 67, 375-83 (1999).

37. Grimwood, J., Olinger, L. & Stephens, R.S. Expression of Chlamydia pneumoniae

polymorphic membrane protein family genes. Infect Immun 69, 2383-9 (2001).

38. Longbottom, D., Findlay, J., Vretou, E. & Dunbar, S.M. Immunoelectron

microscopic localisation of the OMP90 family on the outer membrane surface of

Chlamydia psittaci. FEMS Microbiol Lett 164, 111-7 (1998).

39. Tanzer, R.J., Longbottom, D. & Hatch, T.P. Identification of polymorphic outer

membrane proteins of Chlamydia psittaci 6BC. Infect Immun 69, 2428-34 (2001).

40. Niessner, A., Kaun, C., Zorn, G., Speidl, W., Turel, Z., Christiansen, G., Pedersen,

A.S., Birkelund, S., Simon, S., Georgopoulos, A., Graninger, W., de Martin, R.,

Lipp, J., Binder, B.R., Maurer, G., Huber, K. & Wojta, J. Polymorphic membrane

protein (PMP) 20 and PMP 21 of Chlamydia pneumoniae induce proinflammatory

mediators in human endothelial cells in vitro by activation of the nuclear factor-

kappaB pathway. J Infect Dis 188, 108-13 (2003).

41. Caldwell, H.D., Kromhout, J. & Schachter, J. Purification and partial

characterization of the major outer membrane protein of Chlamydia trachomatis.

Infect Immun 31, 1161-76 (1981).

42. Wyllie, S., Ashley, R.H., Longbottom, D. & Herring, A.J. The major outer

membrane protein of Chlamydia psittaci functions as a porin-like ion channel.

Infect Immun 66, 5202-7 (1998).

43. Su, H., Raymond, L., Rockey, D.D., Fischer, E., Hackstadt, T. & Caldwell, H.D. A

recombinant Chlamydia trachomatis major outer membrane protein binds to

Page 64: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

55

heparan sulfate receptors on epithelial cells. Proc Natl Acad Sci U S A 93, 11143-8

(1996).

44. Campbell, L.A., Kuo, C.C., Wang, S.P. & Grayston, J.T. Serological response to

Chlamydia pneumoniae infection. J Clin Microbiol 28, 1261-4 (1990).

45. Wolf, K., Fischer, E., Mead, D., Zhong, G., Peeling, R., Whitmire, B. & Caldwell,

H.D. Chlamydia pneumoniae major outer membrane protein is a surface-exposed

antigen that elicits antibodies primarily directed against conformation-dependent

determinants. Infect Immun 69, 3082-91 (2001).

46. Caldwell, H.D. & Perry, L.J. Neutralization of Chlamydia trachomatis infectivity

with antibodies to the major outer membrane protein. Infect Immun 38, 745-54

(1982).

47. Christiansen, G. & Birkelund, S. Is a Chlamydia vaccine a reality? Best Pract Res

Clin Obstet Gynaecol 16, 889-900 (2002).

48. Kornak, J.M., Kuo, C.C. & Campbell, L.A. Sequence analysis of the gene

encoding the Chlamydia pneumoniae DnaK protein homolog. Infect Immun 59,

721-5 (1991).

49. Brunham, R.C. & Peeling, R.W. Chlamydia trachomatis antigens: role in immunity

and pathogenesis. Infect Agents Dis 3, 218-33 (1994).

50. Zhong, G. & Brunham, R.C. Antibody responses to the chlamydial heat shock

proteins hsp60 and hsp70 are H-2 linked. Infect Immun 60, 3143-9 (1992).

51. Mayr, M., Metzler, B., Kiechl, S., Willeit, J., Schett, G., Xu, Q. & Wick, G.

Endothelial cytotoxicity mediated by serum antibodies to heat shock proteins of

Escherichia coli and Chlamydia pneumoniae: immune reactions to heat shock

proteins as a possible link between infection and atherosclerosis. Circulation 99,

1560-6 (1999).

52. Sasu, S., LaVerda, D., Qureshi, N., Golenbock, D.T. & Beasley, D. Chlamydia

pneumoniae and chlamydial heat shock protein 60 stimulate proliferation of human

vascular smooth muscle cells via toll-like receptor 4 and p44/p42 mitogen-

activated protein kinase activation. Circ Res 89, 244-50 (2001).

53. Wilson, A.C., Wu, C.C., Yates, J.R., 3rd & Tan, M. Chlamydial GroEL

autoregulates its own expression through direct interactions with the HrcA

repressor protein. J Bacteriol 187, 7535-42 (2005).

54. Slepenkin, A., Motin, V., de la Maza, L.M. & Peterson, E.M. Temporal expression

of type III secretion genes of Chlamydia pneumoniae. Infect Immun 71, 2555-62

(2003).

55. Mukhopadhyay, S., Miller, R.D., Sullivan, E.D., Theodoropoulos, C., Mathews,

S.A., Timms, P. & Summersgill, J.T. Protein expression profiles of Chlamydia

pneumoniae in models of persistence versus those of heat shock stress response.

Infect Immun 74, 3853-63 (2006).

56. Kikuta, L.C., Puolakkainen, M., Kuo, C.C. & Campbell, L.A. Isolation and

sequence analysis of the Chlamydia pneumoniae GroE operon. Infect Immun 59,

4665-9 (1991).

57. Kol, A., Sukhova, G.K., Lichtman, A.H. & Libby, P. Chlamydial heat shock

protein 60 localizes in human atheroma and regulates macrophage tumor necrosis

factor-alpha and matrix metalloproteinase expression. Circulation 98, 300-7

(1998).

••

Page 65: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

56

58. Fong, I., Chiu, B., Viira, E., Tucker, W., Wood, H. & Peeling, R. Chlamydial heat-

shock protein-60 antibody and correlation with Chlamydia pneumoniae in

atherosclerotic plaques. J Infect Dis. 15;186(2002).

59. Ismail, A., Khosravi, H. & Olson, H. The role of infection in atherosclerosis and

coronary artery disease: a new therapeutic target. Heart Dis 1, 233-40 (1999).

60. Ford, P.J., Gemmell, E., Hamlet, S.M., Hasan, A., Walker, P.J., West, M.J.,

Cullinan, M.P. & Seymour, G.J. Cross-reactivity of GroEL antibodies with human

heat shock protein 60 and quantification of pathogens in atherosclerosis. Oral

Microbiol Immunol 20, 296-302 (2005).

61. Barry, C.E., 3rd, Hayes, S.F. & Hackstadt, T. Nucleoid condensation in

Escherichia coli that express a chlamydial histone homolog. Science 256, 377-9

(1992).

62. Barry, C.E., 3rd, Brickman, T.J. & Hackstadt, T. Hc1-mediated effects on DNA

structure: a potential regulator of chlamydial development. Mol Microbiol 9, 273-

83 (1993).

63. Hackstadt, T., Brickman, T.J., Barry, C.E., 3rd & Sager, J. Diversity in the

Chlamydia trachomatis histone homologue Hc2. Gene 132, 137-41 (1993).

64. Abdelrahman, Y.M. & Belland, R.J. The chlamydial developmental cycle. FEMS

Microbiol Rev 29, 949-59 (2005).

65. Kuo, C.C., Lee, A. & Campbell, L.A. Cleavage of the N-linked oligosaccharide

from the surfaces of Chlamydia species affects attachment and infectivity of the

organisms in human epithelial and endothelial cells. Infect Immun 72, 6699-701

(2004).

66. Kuo, C.C., Lee, A., Jiang, S.J., Yaraei, K. & Campbell, L.A. Inoculation of

Chlamydia pneumoniae or Chlamydia trachomatis with ligands that inhibit

attachment to host cells reduces infectivity in the mouse model of lung infection:

implication for anti-adhesive therapy. Microbes Infect 9, 1139-41 (2007).

67. Puolakkainen, M., Kuo, C.C. & Campbell, L.A. Chlamydia pneumoniae uses the

mannose 6-phosphate/insulin-like growth factor 2 receptor for infection of

endothelial cells. Infect Immun 73, 4620-5 (2005).

68. Puolakkainen, M., Lee, A., Nosaka, T., Fukushi, H., Kuo, C.C. & Campbell, L.A.

Retinoic acid inhibits the infectivity and growth of Chlamydia pneumoniae in

epithelial and endothelial cells through different receptors. Microb Pathog (2007).

69. Raulston, J.E., Davis, C.H., Paul, T.R., Hobbs, J.D. & Wyrick, P.B. Surface

accessibility of the 70-kilodalton Chlamydia trachomatis heat shock protein

following reduction of outer membrane protein disulfide bonds. Infect Immun 70,

535-43 (2002).

70. Wehrl, W., Brinkmann, V., Jungblut, P.R., Meyer, T.F. & Szczepek, A.J. From the

inside out--processing of the Chlamydial autotransporter PmpD and its role in

bacterial adhesion and activation of human host cells. Mol Microbiol 51, 319-34

(2004).

71. Byrne, G.I. & Moulder, J.W. Parasite-specified phagocytosis of Chlamydia psittaci

and Chlamydia trachomatis by L and HeLa cells. Infect Immun 19, 598-606

(1978).

72. Dautry-Varsat, A., Subtil, A. & Hackstadt, T. Recent insights into the mechanisms

of Chlamydia entry. Cell Microbiol 7, 1714-22 (2005).

Page 66: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

57

73. Majeed, M. & Kihlstrom, E. Mobilization of F-actin and clathrin during

redistribution of Chlamydia trachomatis to an intracellular site in eucaryotic cells.

Infect Immun 59, 4465-72 (1991).

74. Coombes, B.K. & Mahony, J.B. Identification of MEK- and phosphoinositide 3-

kinase-dependent signalling as essential events during Chlamydia pneumoniae

invasion of HEp2 cells. Cell Microbiol 4, 447-60 (2002).

75. Birkelund, S., Johnsen, H. & Christiansen, G. Chlamydia trachomatis serovar L2

induces protein tyrosine phosphorylation during uptake by HeLa cells. Infect

Immun 62, 4900-8 (1994).

76. Fawaz, F.S., van Ooij, C., Homola, E., Mutka, S.C. & Engel, J.N. Infection with

Chlamydia trachomatis alters the tyrosine phosphorylation and/or localization of

several host cell proteins including cortactin. Infect Immun 65, 5301-8 (1997).

77. Fields, K.A. & Hackstadt, T. The chlamydial inclusion: escape from the endocytic

pathway. Annu Rev Cell Dev Biol 18, 221-45 (2002).

78. Clifton, D.R., Fields, K.A., Grieshaber, S.S., Dooley, C.A., Fischer, E.R., Mead,

D.J., Carabeo, R.A. & Hackstadt, T. A chlamydial type III translocated protein is

tyrosine-phosphorylated at the site of entry and associated with recruitment of

actin. Proc Natl Acad Sci U S A 101, 10166-71 (2004).

79. Scidmore, M.A. Chlamydia - Genomics and Pathogenesis. Edited by P.M Bavoil

and P. B. Wyrick. Norfolk, Horizone Bioscienc, U.K., 255-295 (2006).

80. Wyrick, P.B. Intracellular survival by Chlamydia. Cell Microbiol 2, 275-82

(2000).

81. Rockey, D.D., Heinzen, R.A. & Hackstadt, T. Cloning and characterization of a

Chlamydia psittaci gene coding for a protein localized in the inclusion membrane

of infected cells. Mol Microbiol 15, 617-26 (1995).

82. Bannantine, J.P., Rockey, D.D. & Hackstadt, T. Tandem genes of Chlamydia

psittaci that encode proteins localized to the inclusion membrane. Mol Microbiol

28, 1017-26 (1998).

83. Scidmore-Carlson, M.A., Shaw, E.I., Dooley, C.A., Fischer, E.R. & Hackstadt, T.

Identification and characterization of a Chlamydia trachomatis early operon

encoding four novel inclusion membrane proteins. Mol Microbiol 33, 753-65

(1999).

84. Bannantine, J.P., Griffiths, R.S., Viratyosin, W., Brown, W.J. & Rockey, D.D. A

secondary structure motif predictive of protein localization to the chlamydial

inclusion membrane. Cell Microbiol 2, 35-47 (2000).

85. Toh, H., Miura, K., Shirai, M. & Hattori, M. In silico inference of inclusion

membrane protein family in obligate intracellular parasites chlamydiae. DNA Res

10, 9-17 (2003).

86. Rockey, D.D., Scidmore, M.A., Bannantine, J.P. & Brown, W.J. Proteins in the

chlamydial inclusion membrane. Microbes Infect 4, 333-40 (2002).

87. Hackstadt, T., Scidmore-Carlson, M.A., Shaw, E.I. & Fischer, E.R. The

Chlamydia trachomatis IncA protein is required for homotypic vesicle fusion. Cell

Microbiol 1, 119-30 (1999).

88. Rockey, D.D., Grosenbach, D., Hruby, D.E., Peacock, M.G., Heinzen, R.A. &

Hackstadt, T. Chlamydia psittaci IncA is phosphorylated by the host cell and is

••

Page 67: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

58

exposed on the cytoplasmic face of the developing inclusion. Mol Microbiol 24,

217-28 (1997).

89. Scidmore, M.A. & Hackstadt, T. Mammalian 14-3-3beta associates with the

Chlamydia trachomatis inclusion membrane via its interaction with IncG. Mol

Microbiol 39, 1638-50 (2001).

90. Scidmore, M.A., Rockey, D.D., Fischer, E.R., Heinzen, R.A. & Hackstadt, T.

Vesicular interactions of the Chlamydia trachomatis inclusion are determined by

chlamydial early protein synthesis rather than route of entry. Infect Immun 64,

5366-72 (1996).

91. Hogan, R.J., Mathews, S.A., Mukhopadhyay, S., Summersgill, J.T. & Timms, P.

Chlamydial persistence: beyond the biphasic paradigm. Infect Immun 72, 1843-55

(2004).

92. Al-Younes, H.M., Gussmann, J., Braun, P.R., Brinkmann, V. & Meyer, T.F.

Naturally occurring amino acids differentially influence the development of

Chlamydia trachomatis and Chlamydia (Chlamydophila) pneumoniae. J Med

Microbiol 55, 879-86 (2006).

93. Hybiske, K. & Stephens, R.S. Mechanisms of host cell exit by the intracellular

bacterium Chlamydia. Proc Natl Acad Sci U S A 104, 11430-5 (2007).

94. Posey, J.E. & Gherardini, F.C. Lack of a role for iron in the Lyme disease

pathogen. Science 288, 1651-3 (2000).

95. Raulston, J.E. Response of Chlamydia trachomatis serovar E to iron restriction in

vitro and evidence for iron-regulated chlamydial proteins. Infect Immun 65, 4539-

47 (1997).

96. Al-Younes, H.M., Rudel, T. & Meyer, T.F. Characterization and intracellular

trafficking pattern of vacuoles containing Chlamydia pneumoniae in human

epithelial cells. Cell Microbiol 1, 237-47 (1999).

97. Al-Younes, H.M., Rudel, T., Brinkmann, V., Szczepek, A.J. & Meyer, T.F. Low

iron availability modulates the course of Chlamydia pneumoniae infection. Cell

Microbiol 3, 427-37 (2001).

98. Schaible, U.E. & Kaufmann, S.H. Iron and microbial infection. Nat Rev Microbiol

2, 946-53 (2004).

99. Ratledge, C. & Dover, L.G. Iron metabolism in pathogenic bacteria. Annu Rev

Microbiol 54, 881-941 (2000).

100. Taraska, T., Ward, D.M., Ajioka, R.S., Wyrick, P.B., Davis-Kaplan, S.R., Davis,

C.H. & Kaplan, J. The late chlamydial inclusion membrane is not derived from the

endocytic pathway and is relatively deficient in host proteins. Infect Immun 64,

3713-27 (1996).

101. van Ooij, C., Apodaca, G. & Engel, J. Characterization of the Chlamydia

trachomatis vacuole and its interaction with the host endocytic pathway in HeLa

cells. Infect Immun 65, 758-66 (1997).

102. Scidmore, M.A., Fischer, E.R. & Hackstadt, T. Restricted fusion of Chlamydia

trachomatis vesicles with endocytic compartments during the initial stages of

infection. Infect Immun 71, 973-84 (2003).

103. Raulston, J.E., Miller, J.D., Davis, C.H., Schell, M., Baldwin, A., Ferguson, K. &

Lane, H. Identification of an iron-responsive protein that is antigenic in patients

Page 68: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

59

with Chlamydia trachomatis genital infections. FEMS Immunol Med Microbiol 51,

569-76 (2007).

104. Wyllie, S. & Raulston, J.E. Identifying regulators of transcription in an obligate

intracellular pathogen: a metal-dependent repressor in Chlamydia trachomatis. Mol

Microbiol 40, 1027-36 (2001).

105. Rau, A., Wyllie, S., Whittimore, J. & Raulston, J.E. Identification of Chlamydia

trachomatis genomic sequences recognized by chlamydial divalent cation-

dependent regulator A (DcrA). J Bacteriol 187, 443-8 (2005).

106. Gieffers, J., Rupp, J., Gebert, A., Solbach, W. & Klinger, M. First-choice

antibiotics at subinhibitory concentrations induce persistence of Chlamydia

pneumoniae. Antimicrob Agents Chemother 48, 1402-5 (2004).

107. Wood, H., Fehlner-Gardner, C., Berry, J., Fischer, E., Graham, B., Hackstadt, T.,

Roshick, C. & McClarty, G. Regulation of tryptophan synthase gene expression in

Chlamydia trachomatis. Mol Microbiol 49, 1347-59 (2003).

108. Sakash, J.B., Byrne, G.I., Lichtman, A. & Libby, P. Cytokines induce indoleamine

2,3-dioxygenase expression in human atheroma-asociated cells: implications for

persistent Chlamydophila pneumoniae infection. Infect Immun 70, 3959-61 (2002).

109. Wehrl, W., Meyer, T.F., Jungblut, P.R., Muller, E.C. & Szczepek, A.J. Action and

reaction: Chlamydophila pneumoniae proteome alteration in a persistent infection

induced by iron deficiency. Proteomics 4, 2969-81 (2004).

110. Maurer, A.P., Mehlitz, A., Mollenkopf, H.J. & Meyer, T.F. Gene expression

profiles of Chlamydophila pneumoniae during the developmental cycle and iron

depletion-mediated persistence. PLoS Pathog 3, e83 (2007).

111. Dill, B.D. & Raulston, J.E. Examination of an inducible expression system for

limiting iron availability during Chlamydia trachomatis infection. Microbes Infect

9, 947-53 (2007).

112. Falsey, A.R. & Walsh, E.E. Transmission of Chlamydia pneumoniae. J Infect Dis

168, 493-6 (1993).

113. Kleemola, M., Saikku, P., Visakorpi, R., Wang, S.P. & Grayston, J.T. Epidemics

of pneumonia caused by TWAR, a new Chlamydia organism, in military trainees

in Finland. J Infect Dis 157, 230-6 (1988).

114. Miyashita, N., Niki, Y., Nakajima, M., Fukano, H. & Matsushima, T. Prevalence

of asymptomatic infection with Chlamydia pneumoniae in subjectively healthy

adults. Chest 119, 1416-9 (2001).

115. Steptoe, A., Shamaei-Tousi, A., Gylfe, A., Bailey, L., Bergström, S., Coates, A.R.

& Henderson, B. Protective effect of human heat shock protein 60 suggested by its

association with decreased seropositivity to pathogens. Clin Vaccine Immunol 14,

204-7 (2007).

116. Campbell, L.A. & Kuo, C.C. Chlamydia pneumoniae--an infectious risk factor for

atherosclerosis? Nat Rev Microbiol 2, 23-32 (2004).

117. Kuo, C.C., Jackson, L.A., Campbell, L.A. & Grayston, J.T. Chlamydia

pneumoniae (TWAR). Clin Microbiol Rev 8, 451-61 (1995).

118. Blasi, F., Cosentini, R. & Tarsia, P. Chlamydia pneumoniae respiratory infections.

Curr Opin Infect Dis 13, 161-164 (2000).

119. Sutherland, E.R. & Martin, R.J. Asthma and atypical bacterial infection. Chest

132, 1962-6 (2007).

••

Page 69: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

60

120. Stratton, C.W. & Wheldon, D.B. Multiple sclerosis: an infectious syndrome

involving Chlamydophila pneumoniae. Trends Microbiol 14, 474-9 (2006).

121. Hannu, T., Puolakkainen, M. & Leirisalo-Repo, M. Chlamydia pneumoniae as a

triggering infection in reactive arthritis. Rheumatology (Oxford) 38, 411-4 (1999).

122. Gerard, H.C., Schumacher, H.R., El-Gabalawy, H., Goldbach-Mansky, R. &

Hudson, A.P. Chlamydia pneumoniae present in the human synovium are viable

and metabolically active. Microb Pathog 29, 17-24 (2000).

123. Ross, R. The pathogenesis of atherosclerosis: a perspective for the 1990s. Nature

362, 801-9 (1993).

124. Hansson, G.K. Inflammation, atherosclerosis, and coronary artery disease. N Engl

J Med 352, 1685-95 (2005).

125. Hansson, G.K. & Libby, P. The immune response in atherosclerosis: a double-

edged sword. Nat Rev Immunol 6, 508-19 (2006).

126. C, F. The relationship between acute infectious diseases and arterial lesions. Arch

Intern Med. 8, 153–162 (1911).

127. Fabricant, C.G., Fabricant, J., Litrenta, M.M. & Minick, C.R. Virus-induced

atherosclerosis. J Exp Med 148, 335-40 (1978).

128. Smith, J.D. Mouse models of atherosclerosis. Lab Anim Sci 48, 573-9 (1998).

129. Plump, A.S., Smith, J.D., Hayek, T., Aalto-Setala, K., Walsh, A., Verstuyft, J.G.,

Rubin, E.M. & Breslow, J.L. Severe hypercholesterolemia and atherosclerosis in

apolipoprotein E-deficient mice created by homologous recombination in ES cells.

Cell 71, 343-53 (1992).

130. Moazed, T.C., Campbell, L.A., Rosenfeld, M.E., Grayston, J.T. & Kuo, C.C.

Chlamydia pneumoniae infection accelerates the progression of atherosclerosis in

apolipoprotein E-deficient mice. J Infect Dis 180, 238-41 (1999).

131. Burnett, M.S., Gaydos, C.A., Madico, G.E., Glad, S.M., Paigen, B., Quinn, T.C. &

Epstein, S.E. Atherosclerosis in apoE knockout mice infected with multiple

pathogens. J Infect Dis 183, 226-231 (2001).

132. Caligiuri, G., Rottenberg, M., Nicoletti, A., Wigzell, H. & Hansson, G.K.

Chlamydia pneumoniae infection does not induce or modify atherosclerosis in

mice. Circulation 103, 2834-8 (2001).

133. Aalto-Setala, K., Laitinen, K., Erkkila, L., Leinonen, M., Jauhiainen, M., Ehnholm,

C., Tamminen, M., Puolakkainen, M., Penttila, I. & Saikku, P. Chlamydia

pneumoniae does not increase atherosclerosis in the aortic root of apolipoprotein

E-deficient mice. Arterioscler Thromb Vasc Biol 21, 578-84 (2001).

134. Dahlen, G.H. Lp(a) lipoprotein in cardiovascular disease. Atherosclerosis 108,

111-26 (1994).

135. Laplaud, P.M., Beaubatie, L., Rall, S.C., Jr., Luc, G. & Saboureau, M.

Lipoprotein[a] is the major apoB-containing lipoprotein in the plasma of a

hibernator, the hedgehog (Erinaceus europaeus). J Lipid Res 29, 1157-70 (1988).

136. Callow, M.J., Verstuyft, J., Tangirala, R., Palinski, W. & Rubin, E.M.

Atherogenesis in transgenic mice with human apolipoprotein B and lipoprotein (a).

J Clin Invest 96, 1639-46 (1995).

137. Teivainen, P.A., Eliassen, K.A., Berg, K., Torsdalen, K. & Svindland, A.

Atherogenesis and vascular calcification in mice expressing the human LPA gene.

Pathophysiology 11, 113-120 (2004).

Page 70: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

61

138. Saikku, P., Leinonen, M., Mattila, K., Ekman, M.R., Nieminen, M.S., Makela,

P.H., Huttunen, J.K. & Valtonen, V. Serological evidence of an association of a

novel Chlamydia, TWAR, with chronic coronary heart disease and acute

myocardial infarction. Lancet 2, 983-6 (1988).

139. Shor, A., Kuo, C.C. & Patton, D.L. Detection of Chlamydia pneumoniae in

coronary arterial fatty streaks and atheromatous plaques. S Afr Med J 82, 158-61

(1992).

140. Kuo, C.C., Gown, A.M., Benditt, E.P. & Grayston, J.T. Detection of Chlamydia

pneumoniae in aortic lesions of atherosclerosis by immunocytochemical stain.

Arterioscler Thromb 13, 1501-4 (1993).

141. Kuo, C.C., Shor, A., Campbell, L.A., Fukushi, H., Patton, D.L. & Grayston, J.T.

Demonstration of Chlamydia pneumoniae in atherosclerotic lesions of coronary

arteries. J Infect Dis 167, 841-9 (1993).

142. Ramirez, J.A. Isolation of Chlamydia pneumoniae from the coronary artery of a

patient with coronary atherosclerosis. The Chlamydia pneumoniae/Atherosclerosis

Study Group. Ann Intern Med 125, 979-82 (1996).

143. Maass, M., Bartels, C., Engel, P.M., Mamat, U. & Sievers, H.H. Endovascular

presence of viable Chlamydia pneumoniae is a common phenomenon in coronary

artery disease. J Am Coll Cardiol 31, 827-32 (1998).

144. Jackson, L.A., Campbell, L.A., Kuo, C.C., Rodriguez, D.I., Lee, A. & Grayston,

J.T. Isolation of Chlamydia pneumoniae from a carotid endarterectomy specimen.

J Infect Dis 176, 292-5 (1997).

145. Esposito, G., Blasi, F., Allegra, L., Chiesa, R., Melissano, G., Cosentini, R.,

Tarsia, P., Dordoni, L., Cantoni, C., Arosio, C. & Fagetti, L. Demonstration of

viable Chlamydia pneumoniae in atherosclerotic plaques of carotid arteries by

reverse transcriptase polymerase chain reaction. Ann Vasc Surg 13, 421-5 (1999).

146. de Kruif, M.D., van Gorp, E.C., Keller, T.T., Ossewaarde, J.M. & ten Cate, H.

Chlamydia pneumoniae infections in mouse models: relevance for atherosclerosis

research. Cardiovasc Res 65, 317-27 (2005).

147. Grayston, J.T. Antibiotic treatment of atherosclerotic cardiovascular disease.

Circulation 107, 1228-30 (2003).

148. Cannon, C.P., Braunwald, E., McCabe, C.H., Grayston, J.T., Muhlestein, B.,

Giugliano, R.P., Cairns, R. & Skene, A.M. Antibiotic treatment of Chlamydia

pneumoniae after acute coronary syndrome. N Engl J Med 352, 1646-54 (2005).

149. Grayston, J.T., Kronmal, R.A., Jackson, L.A., Parisi, A.F., Muhlestein, J.B.,

Cohen, J.D., Rogers, W.J., Crouse, J.R., Borrowdale, S.L., Schron, E. & Knirsch,

C. Azithromycin for the secondary prevention of coronary events. N Engl J Med

352, 1637-45 (2005).

150. Kutlin, A., Roblin, P.M. & Hammerschlag, M.R. In vitro activities of azithromycin

and ofloxacin against Chlamydia pneumoniae in a continuous-infection model.

Antimicrob Agents Chemother 43, 2268-72 (1999).

151. Kutlin, A., Roblin, P.M. & Hammerschlag, M.R. Effect of gemifloxacin on

viability of Chlamydia pneumoniae (Chlamydophila pneumoniae) in an in vitro

continuous infection model. J Antimicrob Chemother 49, 763-7 (2002).

••

Page 71: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

62

152. Gieffers, J., Fullgraf, H., Jahn, J., Klinger, M., Dalhoff, K., Katus, H.A., Solbach,

W. & Maass, M. Chlamydia pneumoniae infection in circulating human

monocytes is refractory to antibiotic treatment. Circulation 103, 351-6 (2001).

153. Taylor-Robinson, D. & Boman, J. The failure of antibiotics to prevent heart

attacks. Bmj 331, 361-2 (2005).

154. Mahony, J. & Coombes, B. Chlamydia pneumoniae and atherosclerosis: does the

evidence support a causal or contributory role? FEMS Microbiol Lett. Apr

1;197(1), 1-9. Review (2001).

155. Kalayoglu, M. & Byrne, G. A Chlamydia pneumoniae component that induces

macrophage foam cell formation is chlamydial lipopolysaccharide. Infect Immun

(11), 5067-72 (1998).

156. Rödel, J., Woytas, M., Groh, A., Schmidt, K., Hartmann, M., Lehmann, M. &

Straube, E. Production of basic fibroblast growth factor and interleukin 6 by

human smooth muscle cells following infection with Chlamydia pneumoniae.

Infect Immun 68, 3635-41. (2000).

157. Rödel, J., Prochnau, D., Prager, K., Baumert, J., Schmidt, K. & Straube, E.

Chlamydia pneumoniae decreases smooth muscle cell proliferation through

induction of prostaglandin E2 synthesis. Infect Immun 72, 4900-4. (2004).

158. Rödel, J., Lehmann, M., Vogelsang, H. & Straube, E. Chlamydia pneumoniae

infection of aortic smooth muscle cells reduces platelet-derived growth factor

receptor-beta expression. FEMS Immunol Med Microbiol 51, 363-71 (2007).

159. Bennett, I.L., Jr. & Beeson, P.B. Studies on the pathogenesis of fever. II.

Characterization of fever-producing substances from polymorphonuclear

leukocytes and from the fluid of sterile exudates. J Exp Med 98, 493-508 (1953).

160. Hornef, M.W., Wick, M.J., Rhen, M. & Normark, S. Bacterial strategies for

overcoming host innate and adaptive immune responses. Nat Immunol 3, 1033-40

(2002).

161. Henderson, B. & Nair, S.P. Hard labour: bacterial infection of the skeleton. Trends

Microbiol 11, 570-7 (2003).

162. Wilson, M., Seymour, R. & Henderson, B. Bacterial perturbation of cytokine

networks. Infect Immun 66, 2401-9 (1998).

163. Byrd, T.F. Tumor necrosis factor alpha (TNFalpha) promotes growth of virulent

Mycobacterium tuberculosis in human monocytes iron-mediated growth

suppression is correlated with decreased release of TNFalpha from iron-treated

infected monocytes. J Clin Invest 99, 2518-29 (1997).

164. Denis, M. & Gregg, E.O. Recombinant interleukin-6 increases the intracellular and

extracellular growth of Mycobacterium avium. Can J Microbiol 37, 479-83

(1991).

165. Ouchi, K., Fujii, B., Kudo, S., Shirai, M., Yamashita, K., Gondo, T., Ishihara, T.,

Ito, H. & Nakazawa, T. Chlamydia pneumoniae in atherosclerotic and

nonatherosclerotic tissue. J Infect Dis 181 Suppl 3, S441-3 (2000).

166. Coombes, B.K. & Mahony, J.B. cDNA array analysis of altered gene expression in

human endothelial cells in response to Chlamydia pneumoniae infection. Infect

Immun 69, 1420-7 (2001).

167. Kaukoranta-Tolvanen, S.S., Teppo, A.M., Laitinen, K., Saikku, P., Linnavuori, K.

& Leinonen, M. Growth of Chlamydia pneumoniae in cultured human peripheral

Page 72: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

63

blood mononuclear cells and induction of a cytokine response. Microb Pathog 21,

215-21 (1996).

168. Summersgill, J.T., Molestina, R.E., Miller, R.D. & Ramirez, J.A. Interactions of

Chlamydia pneumoniae with human endothelial cells. J Infect Dis 181 Suppl 3,

S479-82 (2000).

169. Kol, A., Bourcier, T., Lichtman, A.H. & Libby, P. Chlamydial and human heat

shock protein 60s activate human vascular endothelium, smooth muscle cells, and

macrophages. J Clin Invest 103, 571-7 (1999).

170. Netea, M.G., Selzman, C.H., Kullberg, B.J., Galama, J.M., Weinberg, A.,

Stalenhoef, A.F., Van der Meer, J.W. & Dinarello, C.A. Acellular components of

Chlamydia pneumoniae stimulate cytokine production in human blood

mononuclear cells. Eur J Immunol 30, 541-9 (2000).

171. Jahn, H.U., Krull, M., Wuppermann, F.N., Klucken, A.C., Rosseau, S., Seybold, J.,

Hegemann, J.H., Jantos, C.A. & Suttorp, N. Infection and activation of airway

epithelial cells by Chlamydia pneumoniae. J Infect Dis 182, 1678-87 (2000).

172. Yang, J., Hooper, W.C., Phillips, D.J., Tondella, M.L. & Talkington, D.F.

Induction of proinflammatory cytokines in human lung epithelial cells during

Chlamydia pneumoniae infection. Infect Immun 71, 614-20 (2003).

173. Boelen, E., Steinbusch, H.W., Pronk, I., Grauls, G., Rennert, P., Bailly, V.,

Bruggeman, C.A. & Stassen, F.R. Inflammatory responses following Chlamydia

pneumoniae infection of glial cells. Eur J Neurosci 25, 753-60 (2007).

174. Bost, K.L., Ramp, W.K., Nicholson, N.C., Bento, J.L., Marriott, I. & Hudson,

M.C. Staphylococcus aureus infection of mouse or human osteoblasts induces high

levels of interleukin-6 and interleukin-12 production. J Infect Dis 180, 1912-20

(1999).

175. Marriott, I. Osteoblast responses to bacterial pathogens: a previously

unappreciated role for bone-forming cells in host defense and disease progression.

Immunol Res 30, 291-308 (2004).

176. Marriott, I., Gray, D.L., Tranguch, S.L., Fowler, V.G., Jr., Stryjewski, M., Scott

Levin, L., Hudson, M.C. & Bost, K.L. Osteoblasts express the inflammatory

cytokine interleukin-6 in a murine model of Staphylococcus aureus osteomyelitis

and infected human bone tissue. Am J Pathol 164, 1399-406 (2004).

177. Ishida, I., Kohda, C., Yanagawa, Y., Miyaoka, H. & Shimamura, T.

Epigallocatechin gallate suppresses expression of receptor activator of NF-kappaB

ligand (RANKL) in Staphylococcus aureus infection in osteoblast-like NRG cells.

J Med Microbiol 56, 1042-6 (2007).

178. Yoshii, T., Magara, S., Miyai, D., Nishimura, H., Kuroki, E., Furudoi, S., Komori,

T. & Ohbayashi, C. Local levels of interleukin-1beta, -4, -6 and tumor necrosis

factor alpha in an experimental model of murine osteomyelitis due to

staphylococcus aureus. Cytokine 19, 59-65 (2002).

179. Fullilove, S., Jellis, J., Hughes, S.P., Remick, D.G. & Friedland, J.S. Local and

systemic concentrations of tumour necrosis factor-alpha, interleukin-6 and

interleukin-8 in bacterial osteomyelitis. Trans R Soc Trop Med Hyg 94, 221-4

(2000).

180. Lacey, D.L., Timms, E., Tan, H.L., Kelley, M.J., Dunstan, C.R., Burgess, T.,

Elliott, R., Colombero, A., Elliott, G., Scully, S., Hsu, H., Sullivan, J., Hawkins,

••

Page 73: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

64

N., Davy, E., Capparelli, C., Eli, A., Qian, Y.X., Kaufman, S., Sarosi, I., Shalhoub,

V., Senaldi, G., Guo, J., Delaney, J. & Boyle, W.J. Osteoprotegerin ligand is a

cytokine that regulates osteoclast differentiation and activation. Cell 93, 165-76

(1998).

181. Simonet, W.S., Lacey, D.L., Dunstan, C.R., Kelley, M., Chang, M.S., Luthy, R.,

Nguyen, H.Q., Wooden, S., Bennett, L., Boone, T., Shimamoto, G., DeRose, M.,

Elliott, R., Colombero, A., Tan, H.L., Trail, G., Sullivan, J., Davy, E., Bucay, N.,

Renshaw-Gegg, L., Hughes, T.M., Hill, D., Pattison, W., Campbell, P., Sander, S.,

Van, G., Tarpley, J., Derby, P., Lee, R. & Boyle, W.J. Osteoprotegerin: a novel

secreted protein involved in the regulation of bone density. Cell 89, 309-19 (1997).

182. Zhang, X., Aubin, J.E., Kim, T.H., Payne, U., Chiu, B. & Inman, R.D. Synovial

fibroblasts infected with Salmonella enterica serovar Typhimurium mediate

osteoclast differentiation and activation. Infect Immun 72, 7183-9 (2004).

183. Belibasakis, G.N., Johansson, A., Wang, Y., Chen, C., Kalfas, S. & Lerner, U.H.

The cytolethal distending toxin induces receptor activator of NF-kappaB ligand

expression in human gingival fibroblasts and periodontal ligament cells. Infect

Immun 73, 342-51 (2005).

184. Nebe, C.T., Rother, M., Brechtel, I., Costina, V., Neumaier, M., Zentgraf, H.,

Bocker, U., Meyer, T.F. & Szczepek, A.J. Detection of Chlamydophila

pneumoniae in the bone marrow of two patients with unexplained chronic

anaemia. Eur J Haematol 74, 77-83 (2005).

185. Sioka, C., Goudevenos, J., Pappas, K., Bougias, C., Papadopoulos, A.,

Grammatikopoulos, K. & Fotopoulos, A. Bone mineral density and coronary

atherosclerosis. Calcif Tissue Int 81, 333 (2007).

186. Kiechl, S., Schett, G., Wenning, G., Redlich, K., Oberhollenzer, M., Mayr, A.,

Santer, P., Smolen, J., Poewe, W. & Willeit, J. Osteoprotegerin is a risk factor for

progressive atherosclerosis and cardiovascular disease. Circulation 109, 2175-80

(2004).

187. Kiechl, S., Werner, P., Knoflach, M., Furtner, M., Willeit, J. & Schett, G. The

osteoprotegerin/RANK/RANKL system: a bone key to vascular disease. Expert

Rev Cardiovasc Ther 4, 801-11 (2006).

188. Schoppet, M., Preissner, K.T. & Hofbauer, L.C. RANK ligand and

osteoprotegerin: paracrine regulators of bone metabolism and vascular function.

Arterioscler Thromb Vasc Biol 22, 549-53 (2002).

189. Schoppet, M., Sattler, A.M., Schaefer, J.R., Herzum, M., Maisch, B. & Hofbauer,

L.C. Increased osteoprotegerin serum levels in men with coronary artery disease. J

Clin Endocrinol Metab 88, 1024-8 (2003).

190. Schoppet, M., Schaefer, J.R. & Hofbauer, L.C. Low serum levels of soluble

RANK ligand are associated with the presence of coronary artery disease in men.

Circulation 107, e76; author reply e76 (2003).

191. Sattler, A.M., Schoppet, M., Schaefer, J.R. & Hofbauer, L.C. Novel aspects on

RANK ligand and osteoprotegerin in osteoporosis and vascular disease. Calcif

Tissue Int 74, 103-6 (2004).

192. Bennett, B.J., Scatena, M., Kirk, E.A., Rattazzi, M., Varon, R.M., Averill, M.,

Schwartz, S.M., Giachelli, C.M. & Rosenfeld, M.E. Osteoprotegerin inactivation

Page 74: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

65

accelerates advanced atherosclerotic lesion progression and calcification in older

ApoE-/- mice. Arterioscler Thromb Vasc Biol 26, 2117-24 (2006).

193. Gemski, P., Lazere, J. & Casey, T. Plasmid associated with pathogenicity and

calcium dependency of Yersinia enterocolitica. Infect Immun. 27, 682-5 (1980).

194. Kubori, T., Matsushima, Y., Nakamura, D., Uralil, J., Lara-Tejero, M., Sukhan, A.,

Galan, J.E. & Aizawa, S.I. Supramolecular structure of the Salmonella

typhimurium type III protein secretion system. Science 280, 602-5 (1998).

195. Knutton, S., Rosenshine, I., Pallen, M.J., Nisan, I., Neves, B.C., Bain, C., Wolff,

C., Dougan, G. & Frankel, G. A novel EspA-associated surface organelle of

enteropathogenic Escherichia coli involved in protein translocation into epithelial

cells. Embo J 17, 2166-76 (1998).

196. Blocker, A., Gounon, P., Larquet, E., Niebuhr, K., Cabiaux, V., Parsot, C. &

Sansonetti, P. The tripartite type III secreton of Shigella flexneri inserts IpaB and

IpaC into host membranes. J Cell Biol 147, 683-93 (1999).

197. Troisfontaines, P. & Cornelis, G.R. Type III secretion: more systems than you

think. Physiology (Bethesda) 20, 326-39 (2005).

198. Galan, J.E. & Collmer, A. Type III secretion machines: bacterial devices for

protein delivery into host cells. Science 284, 1322-8 (1999).

199. Forsberg A, V.A.M., Skurnik M., Wolf-Watz H. The surface-located YopN protein

is involved in calcium signal transduction in Yersinia pseudotuberculosis. Mol

Microbiol. Apr;5, 977-86. (1991).

200. Matsumoto, A. Fine structures of cell envelopes of Chlamydia organisms as

revealed by freeze-etching and negative staining techniques. J Bacteriol 116,

1355-63 (1973).

201. Matsumoto, A., Fujiwara, E. & Higashi, N. Observations of the surface projections

of infectious small cell of Chlamydia psittaci in thin sections. J Electron Microsc

(Tokyo) 25, 169-70 (1976).

202. Matsumoto, A. Electron microscopic observations of surface projections and

related intracellular structures of Chlamydia organisms. J Electron Microsc

(Tokyo) 30, 315-20 (1981).

203. Matsumoto, A. Surface projections of Chlamydia psittaci elementary bodies as

revealed by freeze-deep-etching. J Bacteriol 151, 1040-2 (1982).

204. Matsumoto, A. Electron microscopic observations of surface projections on

Chlamydia psittaci reticulate bodies. J Bacteriol 150, 358-64 (1982).

205. Gregory, W.W., Gardner, M., Byrne, G.I. & Moulder, J.W. Arrays of hemispheric

surface projections on Chlamydia psittaci and Chlamydia trachomatis observed by

scanning electron microscopy. J Bacteriol 138, 241-4 (1979).

206. Hsia, R.C., Pannekoek, Y., Ingerowski, E. & Bavoil, P.M. Type III secretion genes

identify a putative virulence locus of Chlamydia. Mol Microbiol 25, 351-9 (1997).

207. Hueck, C.J. Type III protein secretion systems in bacterial pathogens of animals

and plants. Microbiol Mol Biol Rev 62, 379-433 (1998).

208. Subtil, A., Blocker, A. & Dautry-Varsat, A. Type III secretion system in

Chlamydia species: identified members and candidates. Microbes Infect 2, 367-9

(2000).

209. Kim, J.F. Revisiting the chlamydial type III protein secretion system: clues to the

origin of type III protein secretion. Trends Genet 17, 65-9 (2001).

••

Page 75: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

66

210. Fields, K.A., Mead, D.J., Dooley, C.A. & Hackstadt, T. Chlamydia trachomatis

type III secretion: evidence for a functional apparatus during early-cycle

development. Mol Microbiol 48, 671-83 (2003).

211. Plano, G.V., Barve, S.S. & Straley, S.C. LcrD, a membrane-bound regulator of the

Yersinia pestis low-calcium response. J Bacteriol 173, 7293-303 (1991).

212. Johnson, D.L. & Mahony, J.B. Chlamydophila pneumoniae PknD exhibits dual

amino acid specificity and phosphorylates Cpn0712, a putative type III secretion

YscD homolog. J Bacteriol 189, 7549-55 (2007).

213. Sorg, I., Wagner, S., Amstutz, M., Muller, S.A., Broz, P., Lussi, Y., Engel, A. &

Cornelis, G.R. YscU recognizes translocators as export substrates of the Yersinia

injectisome. Embo J 26, 3015-24 (2007).

214. Woestyn, S., Allaoui, A., Wattiau, P. & Cornelis, G.R. YscN, the putative

energizer of the Yersinia Yop secretion machinery. J Bacteriol 176, 1561-9

(1994).

215. Blaylock, B., Riordan, K.E., Missiakas, D.M. & Schneewind, O. Characterization

of the Yersinia enterocolitica type III secretion ATPase YscN and its regulator,

YscL. J Bacteriol 188, 3525-34 (2006).

216. Koster, M., Bitter, W., de Cock, H., Allaoui, A., Cornelis, G.R. & Tommassen, J.

The outer membrane component, YscC, of the Yop secretion machinery of

Yersinia enterocolitica forms a ring-shaped multimeric complex. Mol Microbiol

26, 789-97 (1997).

217. Peters, J., Wilson, D.P., Myers, G., Timms, P. & Bavoil, P.M. Type III secretion a

la Chlamydia. Trends Microbiol 15, 241-51 (2007).

218. Coombes, B.K. & Finlay, B.B. Insertion of the bacterial type III translocon: not

your average needle stick. Trends Microbiol 13, 92-5 (2005).

219. Francis, M.S. & Wolf-Watz, H. YopD of Yersinia pseudotuberculosis is

translocated into the cytosol of HeLa epithelial cells: evidence of a structural

domain necessary for translocation. Mol Microbiol 29, 799-813 (1998).

220. Mueller, C.A., Broz, P., Muller, S.A., Ringler, P., Erne-Brand, F., Sorg, I., Kuhn,

M., Engel, A. & Cornelis, G.R. The V-antigen of Yersinia forms a distinct

structure at the tip of injectisome needles. Science 310, 674-6 (2005).

221. Fields, K.A. & Hackstadt, T. Evidence for the secretion of Chlamydia trachomatis

CopN by a type III secretion mechanism. Mol Microbiol 38, 1048-60 (2000).

222. Ho, T.D. & Starnbach, M.N. The Salmonella enterica serovar typhimurium-

encoded type III secretion systems can translocate Chlamydia trachomatis proteins

into the cytosol of host cells. Infect Immun 73, 905-11 (2005).

223. Fields, K.A., Fischer, E.R., Mead, D.J. & Hackstadt, T. Analysis of putative

Chlamydia trachomatis chaperones Scc2 and Scc3 and their use in the

identification of type III secretion substrates. J Bacteriol 187, 6466-78 (2005).

224. Parsot, C., Hamiaux, C. & Page, A.L. The various and varying roles of specific

chaperones in type III secretion systems. Curr Opin Microbiol 6, 7-14 (2003).

225. Neyt, C. & Cornelis, G.R. Role of SycD, the chaperone of the Yersinia Yop

translocators YopB and YopD. Mol Microbiol 31, 143-56 (1999).

226. Frithz-Lindsten, E., Rosqvist, R., Johansson, L. & Forsberg, A. The chaperone-like

protein YerA of Yersinia pseudotuberculosis stabilizes YopE in the cytoplasm but

is dispensible for targeting to the secretion loci. Mol Microbiol 16, 635-47 (1995).

Page 76: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

67

227. Edqvist, P.J., Broms, J.E., Betts, H.J., Forsberg, A., Pallen, M.J. & Francis, M.S.

Tetratricopeptide repeats in the type III secretion chaperone, LcrH: their role in

substrate binding and secretion. Mol Microbiol 59, 31-44 (2006).

228. Slepenkin, A., de la Maza, L.M. & Peterson, E.M. Interaction between components

of the type III secretion system of Chlamydiaceae. J Bacteriol 187, 473-9 (2005).

229. Betts, H.J., Twiggs, L.E., Sal, M.S., Wyrick, P.B. & Fields, K.A. Bioinformatic

and biochemical evidence for the identification of the type III secretion system

needle protein of Chlamydia trachomatis. J Bacteriol 190, 1680-90 (2008).

230. Ouellette, S.P., Abdelrahman, Y.M., Belland, R.J. & Byrne, G.I. The Chlamydia

pneumoniae type III secretion-related lcrH gene clusters are developmentally

expressed operons. J Bacteriol 187, 7853-6 (2005).

231. Darwin, K.H. & Miller, V.L. The putative invasion protein chaperone SicA acts

together with InvF to activate the expression of Salmonella typhimurium virulence

genes. Mol Microbiol 35, 949-60 (2000).

232. Olsson, J., Edqvist, P.J., Broms, J.E., Forsberg, A., Wolf-Watz, H. & Francis, M.S.

The YopD translocator of Yersinia pseudotuberculosis is a multifunctional protein

comprised of discrete domains. J Bacteriol 186, 4110-23 (2004).

233. Francis, M.S., Lloyd, S.A. & Wolf-Watz, H. The type III secretion chaperone

LcrH co-operates with YopD to establish a negative, regulatory loop for control of

Yop synthesis in Yersinia pseudotuberculosis. Mol Microbiol 42, 1075-93 (2001).

234. Stephens, R. Challenge of Chlamydia research. Infect Agents Dis Dec;1(6), 279–

293 (1993).

235. Broz, P., Mueller, C.A., Muller, S.A., Philippsen, A., Sorg, I., Engel, A. &

Cornelis, G.R. Function and molecular architecture of the Yersinia injectisome tip

complex. Mol Microbiol 65, 1311-20 (2007).

236. Pastor, A., Chabert, J., Louwagie, M., Garin, J. & Attree, I. PscF is a major

component of the Pseudomonas aeruginosa type III secretion needle. FEMS

Microbiol Lett 253, 95-101 (2005).

237. Schubot, F.D., Jackson, M.W., Penrose, K.J., Cherry, S., Tropea, J.E., Plano, G.V.

& Waugh, D.S. Three-dimensional structure of a macromolecular assembly that

regulates type III secretion in Yersinia pestis. J Mol Biol 346, 1147-61 (2005).

238. Belland, R.J., Nelson, D.E., Virok, D., Crane, D.D., Hogan, D., Sturdevant, D.,

Beatty, W.L. & Caldwell, H.D. Transcriptome analysis of chlamydial growth

during IFN-gamma-mediated persistence and reactivation. Proc Natl Acad Sci U S

A 100, 15971-6 (2003).

239. Subtil, A., Parsot, C. & Dautry-Varsat, A. Secretion of predicted Inc proteins of

Chlamydia pneumoniae by a heterologous type III machinery. Mol Microbiol 39,

792-800 (2001).

240. Lugert, R., Kuhns, M., Polch, T. & Gross, U. Expression and localization of type

III secretion-related proteins of Chlamydia pneumoniae. Med Microbiol Immunol

(Berl) 193, 163-71 (2004).

241. Subtil, A., Delevoye, C., Balana, M.E., Tastevin, L., Perrinet, S. & Dautry-Varsat,

A. A directed screen for chlamydial proteins secreted by a type III mechanism

identifies a translocated protein and numerous other new candidates. Mol

Microbiol 56, 1636-47 (2005).

••

Page 77: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

68

242. Chellas-Gery, B., Linton, C.N. & Fields, K.A. Human GCIP interacts with CT847,

a novel Chlamydia trachomatis type III secretion substrate, and is degraded in a

tissue-culture infection model. Cell Microbiol 9, 2417-30 (2007).

243. Fling, S.P., Sutherland, R.A., Steele, L.N., Hess, B., D'Orazio, S.E., Maisonneuve,

J., Lampe, M.F., Probst, P. & Starnbach, M.N. CD8+ T cells recognize an

inclusion membrane-associated protein from the vacuolar pathogen Chlamydia

trachomatis. Proc Natl Acad Sci U S A 98, 1160-5 (2001).

244. Stenner-Liewen, F., Liewen, H., Zapata, J.M., Pawlowski, K., Godzik, A. & Reed,

J.C. CADD, a Chlamydia protein that interacts with death receptors. J Biol Chem

277, 9633-6 (2002).

245. Zhong, G., Fan, P., Ji, H., Dong, F. & Huang, Y. Identification of a chlamydial

protease-like activity factor responsible for the degradation of host transcription

factors. J Exp Med 193, 935-42 (2001).

246. Dautry-Varsat, A., Balana, M.E. & Wyplosz, B. Chlamydia--host cell interactions:

recent advances on bacterial entry and intracellular development. Traffic 5, 561-70

(2004).

247. Vandahl, B.B., Stensballe, A., Roepstorff, P., Christiansen, G. & Birkelund, S.

Secretion of Cpn0796 from Chlamydia pneumoniae into the host cell cytoplasm by

an autotransporter mechanism. Cell Microbiol 7, 825-36 (2005).

248. Dong, F., Flores, R., Chen, D., Luo, J., Zhong, Y., Wu, Z. & Zhong, G.

Localization of the hypothetical protein Cpn0797 in the cytoplasm of Chlamydia

pneumoniae-infected host cells. Infect Immun 74, 6479-86 (2006).

249. Kenny, B., DeVinney, R., Stein, M., Reinscheid, D.J., Frey, E.A. & Finlay, B.B.

Enteropathogenic E. coli (EPEC) transfers its receptor for intimate adherence into

mammalian cells. Cell 91, 511-20 (1997).

250. Clifton, D.R., Dooley, C.A., Grieshaber, S.S., Carabeo, R.A., Fields, K.A. &

Hackstadt, T. Tyrosine phosphorylation of the chlamydial effector protein Tarp is

species specific and not required for recruitment of actin. Infect Immun 73, 3860-8

(2005).

251. Horn, M., Collingro, A., Schmitz-Esser, S., Beier, C.L., Purkhold, U., Fartmann,

B., Brandt, P., Nyakatura, G.J., Droege, M., Frishman, D., Rattei, T., Mewes,

H.W. & Wagner, M. Illuminating the evolutionary history of chlamydiae. Science

304, 728-30 (2004).

252. Hoare, A., Timms, P., Bavoil, P.M. & Wilson, D.P. Spatial constraints within the

chlamydial host cell inclusion predict interrupted development and persistence.

BMC Microbiol 8, 5 (2008).

253. Bavoil, P.M., Hsia, R. & Ojcius, D.M. Closing in on Chlamydia and its

intracellular bag of tricks. Microbiology 146 ( Pt 11), 2723-31 (2000).

254. Shaw, E.I., Dooley, C.A., Fischer, E.R., Scidmore, M.A., Fields, K.A. &

Hackstadt, T. Three temporal classes of gene expression during the Chlamydia

trachomatis developmental cycle. Mol Microbiol 37, 913-25 (2000).

255. Vandahl, B.B., Birkelund, S., Demol, H., Hoorelbeke, B., Christiansen, G.,

Vandekerckhove, J. & Gevaert, K. Proteome analysis of the Chlamydia

pneumoniae elementary body. Electrophoresis 22, 1204-23 (2001).

256. Vandahl, B.B., Birkelund, S. & Christiansen, G. Genome and proteome analysis of

Chlamydia. Proteomics 4, 2831-42 (2004).

Page 78: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

69

257. Hefty, P.S. & Stephens, R.S. Chlamydial type III secretion system is encoded on

ten operons preceded by sigma 70-like promoter elements. J Bacteriol 189, 198-

206 (2007).

258. Goellner, S., Schubert, E., Liebler-Tenorio, E., Hotzel, H., Saluz, H.P. & Sachse,

K. Transcriptional response patterns of Chlamydophila psittaci in different in vitro

models of persistent infection. Infect Immun 74, 4801-8 (2006).

259. Ouellette, S.P., Hatch, T.P., AbdelRahman, Y.M., Rose, L.A., Belland, R.J. &

Byrne, G.I. Global transcriptional upregulation in the absence of increased

translation in Chlamydia during IFNgamma-mediated host cell tryptophan

starvation. Mol Microbiol 62, 1387-401 (2006).

260. Mathews, S., George, C., Flegg, C., Stenzel, D. & Timms, P. Differential

expression of ompA, ompB, pyk, nlpD and Cpn0585 genes between normal and

interferon-gamma treated cultures of Chlamydia pneumoniae. Microb Pathog 30,

337-45 (2001).

261. Hammerschlag, M.R. Advances in the management of Chlamydia pneumoniae

infections. Expert Rev Anti Infect Ther 1, 493-503 (2003).

262. Rupp, J., Solbach, W. & Gieffers, J. Variation in the mutation frequency

determining quinolone resistance in Chlamydia trachomatis serovars L2 and D. J

Antimicrob Chemother 61, 91-4 (2008).

263. Binet, R. & Maurelli, A.T. Frequency of development and associated physiological

cost of azithromycin resistance in Chlamydia psittaci 6BC and C. trachomatis L2.

Antimicrob Agents Chemother 51, 4267-75 (2007).

264. Morrissey, I., Salman, H., Bakker, S., Farrell, D., Bebear, C.M. & Ridgway, G.

Serial passage of Chlamydia spp. in sub-inhibitory fluoroquinolone concentrations.

J Antimicrob Chemother 49, 757-61 (2002).

265. Dreses-Werringloer, U., Padubrin, I., Kohler, L. & Hudson, A.P. Detection of

nucleotide variability in rpoB in both rifampin-sensitive and rifampin-resistant

strains of Chlamydia trachomatis. Antimicrob Agents Chemother 47, 2316-8

(2003).

266. Smukste, I. & Stockwell, B.R. Advances in chemical genetics. Annu Rev

Genomics Hum Genet 6, 261-86 (2005).

267. Marra, A. Can virulence factors be viable antibacterial targets? Expert Rev Anti

Infect Ther 2, 61-72 (2004).

268. Marra, A. Targeting virulence for antibacterial chemotherapy: identifying and

characterising virulence factors for lead discovery. Drugs R D 7, 1-16 (2006).

269. Clatworthy, A.E., Pierson, E. & Hung, D.T. Targeting virulence: a new paradigm

for antimicrobial therapy. Nat Chem Biol 3, 541-8 (2007).

270. Aberg, V. & Almqvist, F. Pilicides-small molecules targeting bacterial virulence.

Org Biomol Chem 5, 1827-34 (2007).

271. Hung, D.T., Shakhnovich, E.A., Pierson, E. & Mekalanos, J.J. Small-molecule

inhibitor of Vibrio cholerae virulence and intestinal colonization. Science 310,

670-4 (2005).

272. Panchal, R.G., Hermone, A.R., Nguyen, T.L., Wong, T.Y., Schwarzenbacher, R.,

Schmidt, J., Lane, D., McGrath, C., Turk, B.E., Burnett, J., Aman, M.J., Little, S.,

Sausville, E.A., Zaharevitz, D.W., Cantley, L.C., Liddington, R.C., Gussio, R. &

••

Page 79: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

70

Bavari, S. Identification of small molecule inhibitors of anthrax lethal factor. Nat

Struct Mol Biol 11, 67-72 (2004).

273. Kauppi, A.M., Nordfelth, R., Uvell, H., Wolf-Watz, H. & Elofsson, M. Targeting

bacterial virulence: inhibitors of type III secretion in Yersinia. Chem Biol 10, 241-

9 (2003).

274. Nordfelth, R., Kauppi, A.M., Norberg, H.A., Wolf-Watz, H. & Elofsson, M.

Small-molecule inhibitors specifically targeting type III secretion. Infect Immun

73, 3104-14 (2005).

275. Hudson, D.L., Layton, A.N., Field, T.R., Bowen, A.J., Wolf-Watz, H., Elofsson,

M., Stevens, M.P. & Galyov, E.E. Inhibition of type III secretion in Salmonella

enterica serovar Typhimurium by small-molecule inhibitors. Antimicrob Agents

Chemother 51, 2631-5 (2007).

276. Negrea, A., Bjur, E., Ygberg, S.E., Elofsson, M., Wolf-Watz, H. & Rhen, M.

Salicylidene acylhydrazides that affect type III protein secretion in Salmonella

enterica serovar typhimurium. Antimicrob Agents Chemother 51, 2867-76 (2007).

277. Gauthier, A., Robertson, M.L., Lowden, M., Ibarra, J.A., Puente, J.L. & Finlay,

B.B. Transcriptional inhibitor of virulence factors in enteropathogenic Escherichia

coli. Antimicrob Agents Chemother 49, 4101-9 (2005).

278. Slepenkin, A., Enquist, P.A., Hagglund, U., de la Maza, L.M., Elofsson, M. &

Peterson, E.M. Reversal of the antichlamydial activity of putative type III secretion

inhibitors by iron. Infect Immun 75, 3478-89 (2007).

279. Wolf, K., Betts, H.J., Chellas-Gery, B., Hower, S., Linton, C.N. & Fields, K.A.

Treatment of Chlamydia trachomatis with a small molecule inhibitor of the

Yersinia type III secretion system disrupts progression of the chlamydial

developmental cycle. Mol Microbiol 61, 1543-55 (2006).

280. Humphrey, E.L., Williams, J.H., Davie, M.W. & Marshall, M.J. Effects of

dissociated glucocorticoids on OPG and RANKL in osteoblastic cells. Bone 38 38,

652-661 (2006).

281. Harris, S., Tau, K., Enger, R., Toft, D., Riggs, B. & Spelsberg, T. Estrogen

response in the hFOB 1.19 human fetal osteoblastic cell line stably transfected

with the human estrogen receptor gene. J Cell Biochem 59, 193-201 (1995).

Page 80: Infection Biology of Chlamydia pneumoniae141442/... · 2008. 3. 17. · TWAR – an orienteer-related disease? In the 90’s, TWAR was believed to be the common cause of sudden unexpected

•••


Recommended