identification and characterization of the novel ceacam

135
Identification and characterization of the novel CEACAM-binding adhesin of Haemophilus influenzae Dissertation Zur Erlangung des akademischen Grades des Doktors des Naturwissenschaften (Dr. rer. nat.) des Fachbereichs Biologie der Universität Konstanz vorgelegt von Arnaud Kengmo Tchoupa Konstanz 2015 Konstanzer Online-Publikations-System (KOPS) URL: http://nbn-resolving.de/urn:nbn:de:bsz:352-0-310442

Upload: others

Post on 15-Jan-2022

5 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Identification and characterization of the novel CEACAM

Identification and characterization of the

novel CEACAM-binding adhesin of

Haemophilus influenzae

Dissertation

Zur Erlangung des akademischen Grades

des Doktors des Naturwissenschaften (Dr. rer. nat.)

des Fachbereichs Biologie der Universität Konstanz

vorgelegt von

Arnaud Kengmo Tchoupa

Konstanz 2015

Konstanzer Online-Publikations-System (KOPS) URL: http://nbn-resolving.de/urn:nbn:de:bsz:352-0-310442

Page 2: Identification and characterization of the novel CEACAM
Page 3: Identification and characterization of the novel CEACAM

Date of the doctoral oral examination: 20.11.2015

Chairperson and oral examiner: Professor Dr. Bürkle

1. Oral examiner: Professor Dr. Hauck

2. Oral examiner: Professor Dr. Reidl, Karl-Franzes-University Graz

Page 4: Identification and characterization of the novel CEACAM
Page 5: Identification and characterization of the novel CEACAM

Table of Contents

I

Table of Contents

Table of Contents .............................................................................................................................. I

Acknowledgement ........................................................................................................................... III

Summary .......................................................................................................................................... IV

Zusammenfassung .......................................................................................................................... VI

General Introduction ........................................................................................................................ 1

1. Haemophilus influenzae – the bacterium ............................................................................. 1

2. Clinical significance of Haemophilus influenzae ................................................................... 2

3. Pathogenesis of Haemophilus influenzae ............................................................................. 3

4. Virulence factors of Haemophilus influenzae ........................................................................ 5

5. CEACAM-recognition by Haemophilus influenzae .............................................................. 10

6. CEACAMs – a common target for many bacteria ............................................................... 12

Aims of the study ........................................................................................................................... 16

CHAPTER I: SIGNALING BY EPITHELIAL MEMBERS OF THE CEACAM FAMILY – MUCOSAL

DOCKING SITES FOR PATHOGENIC BACTERIA ................................................ 18

I.1 Abstract ............................................................................................................................... 19

I.2 Introduction .......................................................................................................................... 19

I.3 Physiological roles of epithelial CEACAMs ......................................................................... 21

I.4 Role of CEACAM extracellular domains in mediating cis- and trans-oligomerization ......... 23

I.5 Signaling by epithelial CEACAMs ....................................................................................... 24

I.6 CEACAM1 cis-oligomerization sustained by the transmembrane domain ......................... 26

I.7 CEACAM-binding bacteria reveal the lipid raft association of their receptors .................... 27

I.8 CEACAM1 signaling initiated by the IgC2-like extracellular domains ................................... 29

I.9 CEACAM cooperation with integrins and other membrane receptors ................................ 31

I.10 Conclusions ..................................................................................................................... 34

Page 6: Identification and characterization of the novel CEACAM

Table of Contents

II

CHAPTER II: OUTER MEMBRANE PROTEIN P1 IS THE CEACAM-BINDING ADHESIN OF

HAEMOPHILUS INFLUENZAE ................................................................................ 35

II.1 Summary ............................................................................................................................. 36

II.2 Introduction .......................................................................................................................... 36

II.3 Results ................................................................................................................................. 38

II.4 Discussion ........................................................................................................................... 55

II.5 Materials and methods ........................................................................................................ 57

II.6 Acknowledgements ............................................................................................................. 60

II.7 Supporting information ........................................................................................................ 61

CHAPTER III: THE OUTER MEMBRANE PROTEIN P1 OF HAEMOPHILUS INFLUENZAE

UTILIZES ITS EXTRACELLULAR LOOPS TO GRASP HUMAN CEACAM ........ 63

III.1 Abstract ........................................................................................................................... 64

III.2 Introduction ..................................................................................................................... 64

III.3 Results ............................................................................................................................ 66

III.4 Discussion ....................................................................................................................... 77

III.5 Materials and methods .................................................................................................... 79

CHAPTER IV: THE HAEMOPHILUS INFLUENZAE ADHESIN OMP P1 IS REGULATED BY

HOST-DERIVED FATTY ACIDS ............................................................................. 83

IV.1 Abstract ........................................................................................................................... 84

IV.2 Introduction ..................................................................................................................... 84

IV.3 Results ............................................................................................................................ 87

IV.4 Discussion ....................................................................................................................... 95

IV.5 Materials and methods .................................................................................................... 98

General Discussion ...................................................................................................................... 101

Declaration of author’s contributions ........................................................................................ 107

List of publications ...................................................................................................................... 108

References .................................................................................................................................... 109

Page 7: Identification and characterization of the novel CEACAM

Acknowledgement

III

Acknowledgement

This piece of work would not have been possible without the joint efforts of

numerous people who have gained my deepest gratitude. In the following lines,

not meant to be exhaustive, I want to sincerely thank:

Prof. Dr. Christof R. Hauck, my supervisor. He offered me the unique opportunity

to conduct research in the forefront of Host-Pathogen interaction. I am amazed by

his enthusiasm, fascinated by his patience and grateful for his smart advices.

Prof Dr. Joachim Reidl for accepting to judge this work. Besides, his intensive

support was instrumental for our major findings.

Prof. Dr. Alexander Bürkle for his incessant guidance as member of my thesis

committee and graduate school, and as examiner of my thesis.

Drs. Alex Buntru, Petra Muezner-Voigt, Naja Nyffenegger, Alexandra Roth and

Maike Voges. As experienced (former) members of our lab they have introduced

me to the technics and provided me with numerous tips and tricks.

Julia Delgado-Tascón, Nina Dierdorf, Christof Paone, Alexander Timper and Yong

Shi for being irreplaceable team-players in the lab and beyond. A special mention

to Chris for proofreading my work and to Nina for the abstract in German.

Anne Keller for her guidance and inestimable help with bureaucratic issues.

Susanne Feindler-Boeckh, Claudia Hentschel and Petra Zoll-Kiewitz, for their

expert technical support on a daily basis. Susi, you are simply the best!

The AWOLI football team (Eric, Christian, Patrick, Faustin, Pericles, Daniel,

Nadège…) without which my weekends would have been monotone.

Elke Cubillos, my flat-mate and lessor. She was kind enough to host me during the

last three years of my PhD studies. “Danke für alles Elke”!

My cousin Tchoupa Marlyse for her incessant support.

My parents (Jean-Marie and Madeleine Kengmo), my brother Samsimon and my

sisters (Kévile, Suzy, Clarissa). They have been very supportive and an endless

source of motivation. Merci à vous les K.E.N.G.M.O. L’avenir nous appartient.

Page 8: Identification and characterization of the novel CEACAM

Summary

IV

Summary

Haemophilus influenzae (Hinf), a mostly commensal inhabitant of human

respiratory airways, is the causative agent of local infections such as middle ear

infections, sinusitis, bronchitis, conjunctivitis and pneumonia, but can also cause

life-threatening disseminating diseases, including meningitis and septicemia. The

success of Hinf as commensal or opportunistic pathogen relies on the ability of the

bacterium to use a combination of adhesive surface proteins, the so-called

adhesins, which intimately bind to structures on the human mucosal tissue in order

to overcome mechanical clearance and escape immune recognition. In that

respect, Hinf targets carcinoembryonic antigen-related cell adhesion molecules

(CEACAMs), which are glycoproteins of the immunoglobulin superfamily and

which are present on the apical side of nasopharyngeal epithelial cells. The first

chapter of this work presents in detail the numerous advantages for the bacteria

inherent in their interaction with epithelial CEACAMs (e.g.: strong attachment to

the mucosal surface, internalization within epithelial cells and therefore protection

against immune response).

The interaction between Hinf and CEACAMs was postulated to be mediated by the

outer membrane protein (OMP) P5, one of the major OMPs of Hinf. Therefore, we

set out to characterize the molecular requirements of CEACAM-binding by OMP

P5. However, (i) the ability of P5-deficient Hinf to strongly interact with CEACAMs;

(ii) the similarity of P5 sequences from non-CEACAM-binding and CEACAM-

binding Hinf strains, and (iii) the failure of OMP P5 to bind CEACAMs when

heterologously expressed in Escherichia coli questioned the role of OMP P5 as

the Hinf CEACAM-binding adhesin. A screen of a panel of Hinf mutants with

defined deletions in their outer membrane proteins revealed that the depletion of

OMP P1 totally abrogated the interaction of Hinf with CEACAMs. Furthermore,

non-CEACAM-binding E. coli gained CEACAM-binding properties upon ectopic

expression of Hinf OMP P1. Thus, OMP P1 is necessary and sufficient for

CEACAM-targeting.

Following the identification of OMP P1 as the bona fide CEACAM-binding adhesin

of Hinf, we sought to identify the domain(s) of OMP P1 involved in CEACAM-

recognition. Therefore, we took advantage of the high similarity between OMP P1

Page 9: Identification and characterization of the novel CEACAM

Summary

V

and its non-CEACAM-binding homolog FadL in E. coli. The solved crystal structure

of FadL helped to model OMP P1, which is predicted to build a 14-stranded β-

barrel. Interestingly, chimeras resulting from the fusion of OMP P1 with FadL

fragments combined with OMP P1 mutants deprived of specific extracellular loops

clearly underlined the involvement of the most prominent surface-exposed OMP

P1 loops (L1, L3, L4 and L7) in CEACAM recognition. The flexibility of these loops

appears important for the proper binding to CEACAM. In contrast to Hinf OMP P1,

P1 homologs of other members of the Pasteurellaceae family were unable to

recognize CEACAMs. Moreover, Hinf OMP P1 bound exclusively to human

CEACAMs, but not to other distantly related mammalian CEACAMs.

FadL, the OMP P1 homolog in E. coli, mediates the transport of long-chain fatty

acids (LCFAs) across the bacterial outer membrane. The high structural homology

shared by FadL and OMP P1 raised the question whether OMP P1 is involved in

fatty acid transport and/or is regulated by fatty acids. Though the addition of fatty

acids to the Hinf medium was able to potently stimulate bacterial growth, the role

of OMP P1 in LCFA transport requires further investigations. However, it was

obvious that LCFAs supplementations upregulated OMP P1 expression in Hinf.

Furthermore, Hinf strains treated with LCFAs were able to better interact with

recombinant CEACAMs and were more invasive in CEACAM-expressing epithelial

cells, compared to untreated bacteria.

In sum, this study not only identifies a novel CEACAM-binding adhesin in a

prominent human pathogen, but also provides insights into a new CEACAM-

binding interface built by 4 flexible loops of a bacterial outer membrane protein.

Page 10: Identification and characterization of the novel CEACAM

Zusammenfassung

VI

Zusammenfassung

Haemophilus influenzae (Hinf), ein meist kommensaler Bewohner der

menschlichen Atemwege, ist der Erreger lokaler Infektionen, wie Bronchitis,

Bindehautentzündung und Lungenentzündung. In einigen Fällen löst Hinf

lebensbedrohliche disseminierte Krankheiten wie Meningitis und Septikämie aus.

Hinf ist sehr erfolgreich als Kommensal und Pathogen, da das Bakterium eine

Vielzahl von speziellen oberflächlichen Proteinen besitzt, welche als Adhäsine

bezeichnet werden. Um eine mechanische Eliminierung und Immunerkennung zu

umgehen, binden diese Moleküle eng an Strukturen des menschlichen

Schleimhautgewebes. Hierzu bindet Hinf an CEACAMs (Glykoproteine der

Immunglobulin-Superfamilie, die auf der apikalen Seite nasopharyngealen

Epithelzellen vorhanden sind). Das erste Kapitel der vorliegenden Arbeit wird im

Einzelnen die Vorteile jener Bakterien erfassen, welche mit epithelialen CEACAMs

interagieren. Dazu gehören eine starke Bindung an der Schleimhautoberfläche,

die Internalisierung in Epithelzellen und dadurch Schutz gegen Immunantwort.

Es wurde postuliert, dass die Interaktion zwischen Hinf und CEACAMs vom

äußeren Membranprotein (OMP) P5 – ein der wichtigsten OMPs von Hinf –

vermittelt wird. Deswegen haben wir es uns zum Ziel gesetzt, die molekularen

Voraussetzungen für ein CEACAM – OMP P5 Bindung zu charakterisieren.

Jedoch (i) die Fähigkeit von P5-defizienten Hinf-Mutanten stark mit CEACAMs

zusammen zu interagieren; (ii) die Ähnlichkeit der P5-Sequenzen von nicht-

CEACAM-bindende und CEACAM-bindende Hinf Stämme, und (iii) die Unfähigkeit

der P5-Proteine CEACAMs zu erkennen, wenn sie in E. coli exprimiert wurden,

warfen ernste Zweifel an P5 als Hinf CEACAM-bindende Adhäsin auf. Ein Screen

mit verschiedenen Hinf-Mutanten, welche Deletionen in definierten

Außenmembranproteinen aufwiesen, ergab, dass die Abwesenheit von OMP P1

vollständig die Interaktion von Hinf mit CEACAMs unterdrückte. Des Weiteren

konnte gezeigt werden, dass nicht-CEACAM-bindende E. coli nach ektopischer

Expression von Hinf OMP P1 CEACAM-Bindungseigenschaften erwarben. Somit

ist OMP P1 für eine CEACAM-Bindung notwendig und ausreichend.

Nach der Identifizierung von Hinf OMP P1 als CEACAM-bindendes Adhäsin

versuchten wir, den/die Sequenzabschnitt/e von OMP P1 zu charakterisieren,

Page 11: Identification and characterization of the novel CEACAM

Zusammenfassung

VII

welcher/e für die Bindung an CEACAMs verantwortlich ist/sind. Durch Protein

Modellierung, dank dem bereits kristallisierten nicht-CEACAM-bindenden E. coli

FadL (OMP P1 Homolog), konnte eine 14-strängige β-barrel-Struktur für P1 erstellt

werden. Mit Hilfe von Chimären aus OMP P1 und FadL sowie OMP P1-Mutanten,

in denen spezifische extrazelluläre Loops deletiert wurden, konnte gezeigt werden,

dass die größten Loops (L1, L3, L4 und L7) von OMP P1 essentiell für die

CEACAM-Bindung sind. Die Flexibilität dieser Loops ist scheinbar wichtig für die

richtige Bindung an CEACAM. Im Gegensatz zu Hinf OMP P1, P1-Homologe aus

anderen Mitgliedern der Pasteurellaceae Familie konnten nicht an CEACAMs

binden. Außerdem erkennt Hinf P1 ausschließlich menschliche CEACAMs und

nicht solche entfernt verwandter Säuger.

FadL, das OMP P1 Homolog in E. coli, vermittelt den Transport von langkettigen

Fettsäuren (LCFAs) über die äußere Membran. Die hohe strukturelle Homologie

von FadL und OMP P1 warf die Frage auf, ob OMP P1 am Fettsäuretransport

beteiligt ist und / oder von Fettsäuren reguliert wird. Durch die Zugabe von

Fettsäure in das Kulturmedium von Hinf konnte das Wachstum dieser Bakterien

effektiv stimuliert werden. Es sind jedoch weitere Untersuchungen notwendig, um

die Rolle von OMP P1 am LCFA-Transport zu evaluieren. Es war offensichtlich,

dass die LCFA-Zugabe die OMP P1 Expression in Hinf erhöhte. Darüber hinaus

waren Hinf Stämme nach LCFA-Behandlung in der Lage besser mit

rekombinanten CEACAMs zu interagieren und schneller in CEACAM-

exprimierenden Epithelzellen einzudringen.

Zusammengefasst, identifiziert diese Studie nicht nur ein neuartiges CEACAM-

bindendes Adhäsin, sondern liefert auch Einblicke in ein neues CEACAM-

bindendes Interface, bestehend aus 4 flexiblen Loops eines bakteriellen

Außenmembranproteins.

Page 12: Identification and characterization of the novel CEACAM
Page 13: Identification and characterization of the novel CEACAM

General Introduction

1

General Introduction

1. Haemophilus influenzae – the bacterium

Haemophilus influenzae (Hinf) is a non-motile, pleomorphic, Gram-negative

coccobacillus. Its genus’ name, Haemophilus (from Greek: blood loving),

describes the requirement by this bacterium of blood-derived nutrients, namely

hemin and nicotinamide-adenine dinucleotide (NAD). Haemophilus influenzae,

formerly Pfeiffer’s bacillus, was discovered during an influenza pandemic and was

wrongly thought, during more than 3 decades, to be the causative agent of the flu

(Norskov-Lauritsen, 2014).

Haemophilus influenzae species is phenotypically heterogeneous. For instance,

some strains are shielded by a polysaccharide capsule that is absent in other

strains. The encapsulated Hinf strains (typeable) can be categorized, depending

on the composition of their capsular polysaccharides, into six serotypes,

designated a through f. The unencapsulated strains consist of encapsulated

bacteria that have switched off their capsule expression or bacteria that completely

lack the genetic information for capsule synthesis (non-typeable) (Agrawal and

Murphy, 2011).

At a genomic level, by multilocus sequence analysis, Hinf strains are divided into

phylogenetic groups I and II. The group I represents the core of the Hinf species

and includes all strains of serotypes c and d, most of strains of serotypes a and b,

and the majority of non-typeable isolates. Conversely, the phylogenetic group II is

small and encompasses all strains of serotypes e and f, few strains of a and b, and

some non-typeable isolates. However, there is an increasing number of

unencapsulated isolates that can be assigned to either group I or II (Norskov-

Lauritsen, 2014). DNA-based analyses remain nevertheless the method of choice

when it comes to subtyping Hinf. This is exemplified by the former species

Haemophilus aegyptius that was shown in DNA hybridization studies to belong to

Haemophilus influenzae species and therefore renamed H. influenzae biogroup

aegyptius (Harrison et al., 2008).

Page 14: Identification and characterization of the novel CEACAM

General Introduction

2

2. Clinical significance of Haemophilus influenzae

Hinf is a commensal colonizer of the human respiratory tract. The carriage rates

vary with the type of Hinf strains and the age of the healthy carrier. For instance,

the colonization rate is close to zero for Haemophilus influenzae type b (Hib)

where the Hib vaccination is widespread (Agrawal and Murphy, 2011). Since the

Hib vaccine is ineffective against non-typeable Hinf (NTHi) strains, these

organisms can colonize up to 80% of both children and adults (Rao et al., 1999).

By contiguous spread (in the case of NTHi) or invasion of the bloodstream

(encapsulated strains), Hinf reaches privileged anatomical sites where it, under

predisposing conditions (e.g.: age, viral infections, immunodeficiency), can cause

diverse affections. Some examples are listed below:

(i) Meningitis. In the pre-Hib-vaccine era, Hinf meningitis were mostly due to type

b strains. Nowadays (in the post-Hib-vaccine era), non-typeable strains are

responsible for most invasive infections, followed by serotypes f and b (Norskov-

Lauritsen, 2014).

(ii) Otitis media. It is the childhood disease for which medical assistance is mostly

sought. NTHi accounts for 27–37% of acute otitis media and is therefore the

second most common etiological agent of this disease. Moreover, NTHi represents

the most common bacterial cause of chronic otitis media with effusion (Rao et al.,

1999).

(iii) Conjunctivitis. Children are more affected by bacterial conjunctivitis than

adults and NTHi causes 44–68% of cases in children, compared with about 25% in

adults (Van Eldere et al., 2014). Importantly, 20–70% of children with bacterial

conjunctivitis also suffer from acute otitis media. Given the high prevalence of

NTHi in bacterial conjunctivitis, it is likely that NTHi is the causative agent of the

concomitant otitis media.

(iv) Sinusitis. Hinf is accountable for approximately one-third of episodes of acute

and chronic sinusitis. Besides, the usage of pneumococcal conjugate vaccines

seems to have relatively increased the contribution of Hinf as causative agent of

sinusitis (Agrawal and Murphy, 2011).

(v) Exacerbations in adults with COPD (chronic obstructive pulmonary

disease). COPD is a leading cause of death worldwide. NTHi is a major cause of

Page 15: Identification and characterization of the novel CEACAM

General Introduction

3

acute exacerbations of COPD by inducing airway inflammation and tissue damage

(Van Eldere et al., 2014).

As numerous studies report the widespread resistance of Hinf to ampicillin, the

main drug of choice in proven Hinf infections, the clinicians still have a lot of

alternative antibiotics. Antibiotic therapy is even not necessary in case of a mild

infection, as the symptomatic therapy is, in that case, enough to accelerate the

healing of the patient. For moderate or severe infections, where the involvement of

Hinf is suspected, empirical therapy (with for example, advance macrolides,

cephalosporin and fluoroquinolone) can be successful. However, bacterial culture

and antimicrobial susceptibility testing enable the clinician to optimize the antibiotic

treatment according to the identified bacterial strain.

3. Pathogenesis of Haemophilus influenzae

As a common commensal inhabitant of the human nasopharyngeal mucosa, Hinf

has to face and withstand host clearance mechanisms, including mucociliary

escalator and local immunity (St. Geme III, 2002). Undeniably, the mucus layer of

the respiratory mucosa efficiently traps most of the incoming bacteria and viruses

(Rao et al., 1999). Although able to bind the mucin, the main glycoprotein of the

mucus (Reddy et al., 1996), Hinf preferentially adheres to non-ciliated cells and to

areas of damaged epithelium via numerous adhesins (St. Geme III, 2002).

Conditions impairing mucociliary clearance or damaging the epithelial barrier

therefore favor Hinf colonization (Fig. 1). However, the bacterium on its own is

able to reduce the ciliary beat frequency, to promote the loss of cilia by epithelial

cells, or to directly damage the mucociliary escalator (Foxwell et al., 1998).

To circumvent host humoral response, Hinf secretes IgA1 proteases which cleave

secretory IgA1, the most abundant mucosal immunoglobulin. A strong support for

extreme importance of IgA1 proteases for Hinf pathogenesis was provided by a

recent study with human volunteers colonized with Hinf. There, the phase variable

igaB gene of IgA1 protease was significantly more switched on after 3–6 days of

human nasopharyngeal colonization, compared to the initial inoculum (Poole et al.,

2013).

Page 16: Identification and characterization of the novel CEACAM

General Introduction

4

The strong adherence of Hinf to epithelial cells, following the interaction of Hinf

adhesins with plasma membrane receptors, results in some cases in the invasion

of the human cells by the bacteria (Clementi and Murphy, 2011). Internalized Hinf,

escaping antibiotic therapy and immune response, are thought to constitute a

protective reservoir for persistence and recurrent infections (Clementi and Murphy,

2011). Besides, good proportion of intracellular Hinf is likely released beneath the

epithelium by the so-called transcytosis. Alternatively, Hinf can reach the

basement membrane by paracytosis, which is the passage of bacterium between

cells. It is elusive how Hinf degrades the basement membrane in order to get

access to the capillary enterocytes and thereby invades the bloodstream. Once in

blood, the rigid polysaccharide capsule is an enormous advantage for survival.

This explains why the unencapsulated NTHi isolates cause mainly local infections.

Fig. 1. Pathogenesis of Haemophilus influenzae. (1) Viral infections, exposure to environmentaltoxicants and other factors impairing the mucociliary function increase the likelihood of bacterialcolonization. Bacteria (red ovals), which are not trapped in the mucus and cleared, are able toreduce the ciliary beat frequency or to promote the loss of cilia by epithelial cells. (2) Upon damageof the mucociliary escalator, bacteria can attach to non-ciliated epithelial cells. (3) Adherentbacteria can form aggregates or be internalized. (4) Under certain conditions, bacterialmicrocolonies break up to disperse organisms, spreading within the respiratory tract (modified from(Rao et al., 1999)).

Page 17: Identification and characterization of the novel CEACAM

General Introduction

5

The presence of Hinf on the mucosal surface of its host does not remain

undetected. There is a robust production of proinflammatory cytokines in response

to Hinf. Fascinatingly, the resulting influx of neutrophils, monocytes, eosinophils,

and macrophages, accentuates the epithelial cell damage, stimulates mucus

production and therefore fosters the spreading of the bacteria (Rao et al., 1999).

Eventually, the outcome of acquisition of a Hinf strain will depend on both host

fitness and pathogen virulence.

4. Virulence factors of Haemophilus influenzae

In an apparently redundant manner, Hinf utilizes numerous factors to accomplish

the same function, which may be attachment to and invasion into epithelial cells or

immune response evasion. These virulence factors are summarized in the Fig. 2.

Fig. 2. Overview of Hinf major virulence factors. In typeable Hinf the capsule is acknowledgedto mask other virulence factors, such as lipooligosaccharides (LOS) or integral outer membraneadhesins. However, huge surface appendages, including pili and autotransporters, traverse thecapsule.

Page 18: Identification and characterization of the novel CEACAM

General Introduction

6

i. The capsule

Encapsulated Hinf can readily lose their capsule. When present, the

polysaccharide capsule of Hinf, by protecting the bacterium from desiccation,

improves the survival of the bacterium in the environment and therefore increases

the propagation to new host (St. Geme III and Cutter, 1996). In the host, Hinf

capsule induces resistance to complement-mediated killing and phagocytosis,

both of which promote bacterial survival in human blood stream. However, the

capsule masks bacterial adhesins that are instrumental in the initial steps of the

bacterial colonization (e.g: adhesion to and invasion into epithelial cells).

Therefore, it has been proposed that, depending on the stage of the infection and

in response to environmental stimuli, encapsulated Hinf strains are able to

modulate the expression of their capsule (St. Geme III and Cutter, 1996).

ii. The pili

Selected typeable and non-typeable Hinf express pili, which are hairlike surface

appendages reaching up to 450 nm in length. Pili are used by Hinf to establish

long-range contact with epithelial cells, mucin or even extracellular matrix proteins

(St. Geme III, 2002). The adhesive pilus is encoded by a gene cluster made of 5

genes (hifA–hifE), with hifE coding for the pilus adhesin. Of note, these huge pili

play a role in the bacteria-induced agglutination of red blood cells and are

therefore called hemagglutinating pili.

Besides, the hemagglutinating pili not involved in bacterial motility, the relatively

small (5–7nm) type IV pilus (TfP) was initially characterized in the NTHi isolate

086-026NP and shown to be indispensable for the twitching motility of this strain

(Bakaletz et al., 2005). Since, the implication of TfP has been extended to:

adherence to epithelial cells, biofilm formation (in vitro and in vivo) and efficient

colonization of animal models (Carruthers et al., 2012).

iii. Fibrils

Fibrils are adhesive structures significantly thinner and smaller than the

hemagglutinating pilus and coded by hsf or hia gene in encapsulated or

nonencapsulated Hinf, respectively.

Hsf (Haemophilus surface fibril) is a huge trimeric autotransporter (100 nm) that

was proven, in Hinf type b, to promote adherence to respiratory epithelial cells

Page 19: Identification and characterization of the novel CEACAM

General Introduction

7

and, via binding to vitronectin, to contribute to serum resistance (Singh et al.,

2014).

Hia (Haemophilus influenzae adhesin), smaller than Hsf, is a high-molecular-

weight autotransporter (115 kDa) sharing significant homology with Hsf. Hia is also

an adhesin, whose adhesive properties are conserved when ectopically expressed

in Escherichia coli (Rao et al., 1999).

iv. HMW1 and HMW2 proteins

High-molecular-weight (HMW) adhesins HMW1 and HMW2 are autotransporters

present in up to 75% of all NTHi isolates and absent in typeable Hinf. The huge

majority of NTHi strains deficient in HMW1 and HMW2 expressed alternatively

Hia. HMW1 and HMW2 mediate binding to epithelial cells. Although very similar

(80% similarity), HMW1 and HMW2 differ in their binding specificity. For instance,

when expressed in E. coli, HMW1-mediated adherence to laryngeal Hep-2 cells

was 3-fold higher compared to HMW2 (Hultgren et al., 1993).

v. Hap

Hap (Haemophilus adhesion and penetration protein) is also an autotransporter

present in NTHi strains. The mature protein encompasses two domains: the

autotransporter β-barrel embedded in the outer membrane (designated Hapβ) and

the extracellular passenger domain (designated HapS). Once Hap is translocated

on the bacterial surface, an autoproteolytic cleavage event releases HapS from

Hapβ (St. Geme III, 2002). Interestingly, HapS mediates attachment to epithelial

cells and interaction with extracellular matrix proteins (fibronectin, laminin and

collagen IV). HapS also promotes bacterial aggregation and microcolony formation.

Most importantly, secretory leukocyte protease inhibitor, abundant in human upper

respiratory tract, inhibits Hap autoproteolytic cleavage and therefore potentiates its

adhesive functions (Rao et al., 1999). Moreover, when expressed in the otherwise

Hap-deficient Hinf Rd strain, Hap consistently increases the attachment of Rd to

epithelial cells (Euba et al., 2015).

vi. LOS

Lipooligosaccharide (LOS) represents a major component of the Hinf cell wall. The

role of LOS in Hinf virulence has been better characterized in NTHi isolates where

it is not masked by a capsule. LOS is involved in the early stages of Hinf

Page 20: Identification and characterization of the novel CEACAM

General Introduction

8

pathogenesis where it contributes to the adherence of the bacterium to epithelial

cells by interacting with the PAF (platelet-activating factor) receptor (Swords et al.,

2000). Importantly, LOS needs to incorporate phosphorylcholine (ChoP) in order to

bind the PAF receptor. The relevance of ChoP+ LOS is supported further by the

finding in NTHi that phase variable licA gene coding for the phosphorylcholine

kinase incorporating ChoP into LOS, was turned on 7 times more after 6 days of

human colonization compared to the initial inoculum (Poole et al., 2013).

When established in the mucosa, the bacteria use secreted form of LOS to induce

paralysis and loss of cilia by the epithelial cells (Rao et al., 1999). Importantly,

LOS plays a major role in the persistence of Hinf. In details, LOS contributes to the

bacterial resistance to complement killing. Also, by integrating host molecules,

such as ChoP or sialic acid, in its structure, LOS participates to bacterial host

mimicry and immune evasion (Bouchet et al., 2003).

vii. The outer membrane proteins

Outer membrane protein (OMP) P2. The OMP P2 is the most abundant protein

in the Hinf outer membrane. It is a trimeric, β-barreled protein with porin activity.

P2 binds to human nasopharyngeal mucin (Reddy et al., 1996). OMP P2 may also

play a role during systemic disease caused by Hinf type b as an otherwise

isogenic P2 mutant of a virulent type b Hinf strain was unable to provoke

bacteremia in infant rat (Cope et al., 1990). Besides, OMP P2 was shown to be

upregulated on bacteria within biofilm and also in the biofilm extracellular matrix

(Wu et al., 2014). It is however unclear, if P2 is necessary for biofilm formation in

Hinf.

OMP P4. The outer membrane protein P4 (or lipoprotein e) is primordial for the

transport hemin and nicotinamide nucleotides, across the outer membrane of Hinf

(Reidl and Mekalanos, 1996; Reidl et al., 2000). Interestingly a P4-deficient mutant

was proven to be less virulent than its isogenic wild-type Hib strain in rat model of

invasive disease (Morton et al., 2007). Lately, OMP P4 was shown to: (i) bind

extracellular matrix (ECM) proteins; (ii) promote bacterial attachment to epithelial

cells; (iii) improve serum resistance and (iv) be necessary for bacterial colonization

of mouse middle ear (Su et al., 2015).

OMP P5. The outer membrane P5 is a β-barrel transmembrane protein whose

porin activity has been not yet characterized. In clear contrast, its role as an

Page 21: Identification and characterization of the novel CEACAM

General Introduction

9

adhesin has been extensively investigated. For instance, OMP P5 associates with

human nasopharyngeal mucin (Reddy et al., 1996). P5 depletion decreases both

the adherence of Hib to human oropharyngeal cells and the severity of otitis media

in chinchilla model (Sirakova et al., 1994). The receptors of P5 on human epithelial

cells were suggested to be CEACAM1 (carcinoembryonic antigen-related cell

adhesion molecule 1) and ICAM1 (intercellular adhesion molecule 1) (Avadhanula

et al., 2006; Hill et al., 2001). OMP P5 is also implicated in the resistance of NTHi

to both classical and alternative complement pathways in typeable and non-

typeable Hinf strains (Rosadini et al., 2014). More recently, OMP P5 importance

for human epithelial cell invasion and bacterial persistence in mice lungs was

nicely demonstrated (Euba et al., 2015).

TbpA and TbpB. The transferrin binding protein (Tbp) A and B are outer

membrane proteins whose expression is iron-repressible. TbpA and TbpB interact

together with human transferrin, which is an iron transporter. Upon binding to

transferrin, TbpA releases the iron in the bacterial periplasm (Rao et al., 1999).

Tbps and iron uptake systems as a whole are essential for bacterial persistence as

the human host maintains the free iron at levels too low to sustain bacterial

survival and growth.

The other outer membrane proteins. As our understanding of Hinf pathogenesis

increases, so does the number of outer membrane proteins that are somehow

involved in Hinf virulence. For example, during the last decade two novel adhesins

were discovered in the Riesbeck lab, namely Protein E (PE) and Protein F (PE).

PE and PF are able to adhere to extracellular matrix proteins as well as to host

cells. Furthermore, PE and PF play in an important role in the subversion of host

innate immunity (Jalalvand and Riesbeck, 2014). In sum, many other outer

membrane proteins exploited by Hinf to colonize its human host may remain

unknown and the relevance of many other known OMPs in Hinf pathogenesis

certainly require further investigations.

viii. The genetic plasticity of Haemophilus influenzae

Hinf is naturally competent. In other words, Hinf is able to take up free DNA from

its environment. The bacterium is transformed, when this DNA uptake leads to

genotype change by either recombination or establishment of a plasmid. Given the

competence of many Hinf strains, it is not a surprise that the number of genes

Page 22: Identification and characterization of the novel CEACAM

General Introduction

10

within the Hinf species varies between 1765 and 2355, with the core genome

containing only 1485 genes (Jalalvand and Riesbeck, 2014).

Beside this genetic heterogeneity, all the genes possessed by Hinf are not evenly

expressed, even within a clonal population. Interestingly, some genes are phase

variable. These genes possess short nucleotide repeats in either their coding or

upstream promoter regions. Spontaneous gain or loss of these repeats results in

translational frame shifts, with genes being kept on or turned off. For a phase

variable gene coding for a simple molecule, this will result in expression or not of

the molecule. When the phase variable gene product is involved in the synthesis

of a complex macromolecule, such as LOS, phase variation can result in a vast

array of structures. As an example, the NTHi strain 9274 has as many as 25

different LOS forms (Rahman et al., 1999).

Once expressed, numerous Hinf surface molecules are different from one strain to

another within the immunogenic regions. These differences result from genetic

polymorphisms, including gene point mutations, insertions or deletions. The

ensuing antigenic variations prevent the antibody cross-reaction on heterologous

strains. The OMP P2, for instance, is able to modify its sequence within a clonal

population during the course of persisting infection (St. Geme III, 2002).

Virtually all the virulence factors that were discussed above (in the Subheading 4.)

and other surface-exposed Hinf are highly immunogenic. However, the genetic

and phenotypic variabilities of the bacterium maintain a highly heterogeneous

bacterial population that is able to evade host immune response and persist within

its preferred human niche.

5. CEACAM-recognition by Haemophilus influenzae

The carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) are a

subgroup of heavily glycosylated proteins belonging to immunoglobulin

superfamily. CEACAMs are expressed by a wide range of cells, including

granulocytes, lymphocytes, endothelial and epithelial cells. In addition to their

physiological roles (extensively discussed in the Chapter I), CEACAMs are

targeted by selected bacteria in order to get a grip on the human mucosal surface.

Since CEACAMs were known to be expressed by epithelial cells of the respiratory

Page 23: Identification and characterization of the novel CEACAM

General Introduction

11

tract (Virji et al., 1996a) and Hinf attachment to epithelial cells acknowledged as an

irreplaceable step of the bacterial pathogenesis (Rao et al., 1999), Virji and

colleagues had the good idea to screen typeable and non-typeable Hinf isolates

for CEACAM-binding. Rewardingly, they showed that most of the strains tested

were able to interact with CEACAM1 (Virji et al., 2000). Additionally,

unencapsulated Hinf of the biogroup aegyptius was interacting also with CEA (the

protein product of the CEACAM5 gene). The Hinf binding site on the CEACAM1

side was defined to be the non-glycosylated CFG face of its amino-terminal Ig

variable-like domain (Virji et al., 2000). Additional experiments provided hints that

the Hinf CEACAM-binding adhesin was proteinaceous in nature. After deletion of

its outer membrane protein P5, the otherwise CEACAM-binding Hinf Rd strain

failed to recognize recombinant, soluble CEACAM1 but still interacted with

CEACAM1-expressing cells. Therefore, the authors suggested P5 as the

CEACAM1-binding adhesin of Hinf and speculated that an additional ligand may

intervene in cellular context (Hill et al., 2001).

Aiming to test the relevance of Hinf – CEACAM interaction in vivo, Bookwalker and

colleagues used the chinchilla model of Hinf nasopharyngeal colonization. Of note,

chinchilla express an orthologue of human CEACAM1, chinchilla CEACAM1

(cCEACAM1). cCEACAM1 cross-reacted with anti-human CEACAM1 antibody.

Strikingly, pretreatment of the animals with anti-human CEACAM1 antibody

severely impaired the colonization of chinchilla nasopharynx with Hinf, suggesting

the importance of cCEACAM1 for successful colonization (Bookwalter et al.,

2007). Since these authors had previously shown that the Hinf OMP P5 is required

for nasopharyngeal colonization of chinchilla (Sirakova et al., 1994), they deduced

that P5 was interacting with cCEACAM during chinchilla colonization. Therefore,

evidence of direct interaction between Hinf P5 and cCEACAM1 are missing.

Nevertheless, a recent study has demonstrated that CEACAMs are expressed in

non-cancer, human, lung tissues (Klaile et al., 2013). Most importantly, normal

human brochoepithelial cells upregulated their CEACAM1 expression after 24 h of

infection with NTHi isolates. (Klaile et al., 2013). It is unclear whether or not this

effect is restricted to bacteria exclusively colonizing human respiratory airways, as

no negative controls, such as commensal, gut bacteria were included by Klaile and

coworkers (Klaile et al., 2013).

Page 24: Identification and characterization of the novel CEACAM

General Introduction

12

6. CEACAMs – a common target for many bacteria

A quarter-century has passed since Leusch and colleagues first reported that

human E. coli isolates were able to bind members of the CEACAM family, namely

CEA and CEACAM6 (Leusch et al., 1990). At present, almost a dozen of bacterial

species have been characterized to target CEACAMs and it is very likely that more

CEACAM-binding bacteria will be uncovered in the future. For better or for worse,

much of the current research on the bacteria interacting with CEACAMs has

focused on the most pathogenic amongst them. Few of them will be briefly

presented in the following lines.

Neisseria gonorrhoeae. Probably the most studied of the CEACAM-binding

bacteria, Neisseria gonorrhoeae (Ngo), the causative agent of the sexually

transmitted disease gonorrhea, binds many members of the CEACAM family via

its “opacity-associated” (Opa) proteins (Virji et al., 1996a; Virji et al., 1996b). The

opa gene is phase variable owing to several pentameric repeats present in its 5’

coding region and each gonococcal strain expresses up to 12 distinct opa genes

(Stern et al., 1986). Noteworthy, each opa gene is independently turned off or on,

and each expressed Opa protein exhibits a unique CEACAM-binding profile

(ranging from no binding to CEACAMs, to multiple interactions with CEACAM1,

CEACAM3, CEA and CEACAM6) (Bos et al., 1997). Thus, the CEACAM-binding

capabilities of a clonal population integrate these genetic and phenotypic

variations. The outcome of the Ngo-CEACAM interaction depends on the

CEACAMs that are engaged. Targeting of epithelial CEACAMs (CEACAM1, CEA

and CEACAM6) provide the bacteria with a solid foothold on the mucosal surface.

Signaling cascades induced upon engagement of epithelial CEACAMs increase

integrin-mediated host cell adhesion to extracellular matrix and block exfoliation of

infected epithelial cells (Muenzner et al., 2010). Whereas the engagement of the

epithelial CEACAMs is beneficial to the bacteria, their recognition by granulocyte

receptor CEACAM3 leads to their opsonin-independent phagocytosis and rapid

elimination (Schmitter et al., 2004). Amazingly, Ngo isolates causing disseminating

diseases seem to have evolved to avoid interaction with CEACAM3 (Roth et al.,

2013). However, it remains puzzling which subtle structural features dictate the

binding of the β-barreled Opa protein to CEACAM1 and not to CEACAM3 (both

Page 25: Identification and characterization of the novel CEACAM

General Introduction

13

molecules sharing ~90% amino acid identity in their Opa-binding, N-terminal

domain).

Neisseria meningitidis. Living in the human nasopharynx, as a commensal,

Neisseria meningitidis (Nme) is able to cause life-threatening and fulminant

meningitis or septicaemia (Apicella, 2010). The interaction of Nme with CEACAMs

is also mediated by Opa proteins (Virji et al., 1996b). Similar to gonococcal Opa

proteins, menningococal Opa proteins utilize the surface-exposed hypervariable

(HV) regions 1 and 2 of their loops 2 and 3, respectively, to associate with

CEACAMs. Although highly variable, HV-2 regions of various Nme Opa proteins

have a conserved GxI/V/LxQ motif that is critical for CEACAM engagement (de

Jonge et al., 2003). In clear contrast, this motif is absent in Ngo Opa proteins.

Another remarkable difference between Ngo and Nme is the presence in the latter

of a polysaccharide capsule. The capsule of Nme, masking the CEACAM binding

OMP, has long cast doubt on the relevance of Opa – CEACAM interaction in vivo.

A recent study nicely demonstrated that capsulated Nme strain MC58 colonizes

better and persists longer in the nasopharynx of mice transgenic for human

CEACAM1 compared to wild-type mice (Johswich et al., 2013). Besides, the Nme

strain MC58 was also shown to keep binding to CEACAM1 even in absence of

Opa proteins, suggesting an additional CEACAM-targeting ligand in Nme

(Kuespert et al., 2011). Still, the identity of this adhesin has to be determined.

Moraxella catarrhalis. Another inhabitant of human respiratory tract, Moraxella

catarrhalis (Mca) is one the major causes of otitis media in children and respiratory

diseases in adults (Hill and Virji, 2003). Mca associates to CEACAMs via its

ubiquitous surface protein (Usp) A1, which is a “lollipop”-like autotransporter (Hill

and Virji, 2003). Of the ~90 kDa UspA1 protein, a ~17 kDa polypeptide named rD-

7 and representing 16% of UspA1 is able to interact with CEACAMs (Hill et al.,

2005). Multiple sequence alignment of this CEACAM-targeting portion of UspA1

from CEACAM-binding and non-CEACAM-binding strains of Mca, revealed a

putative CEACAM-binding motif (Brooks et al., 2008) that was refined to a 20-

amino acid stretch by structural studies (Conners et al., 2008). Thus, UspA1

exhibits a conserved CEACAM-binding motif within its coiled-coil stalk (Conners et

al., 2008). Of note, an additional non-CEACAM binding ubiquitous surface protein,

namely UspA2, is expressed by Mca. Upon recombination, UspA1/UspA2 chimeric

Page 26: Identification and characterization of the novel CEACAM

General Introduction

14

molecules can be generated. Intriguingly, Hill and colleagues demonstrated that

14% of the CEACAM-binding Mca clinical isolates use an UspA2 variant that has

incorporated the CEACAM-targeting motif of UspA1 (Hill et al., 2012). These

findings underline the flexibility of the bacteria interacting with CEACAMs, gaining

or losing the binding capabilities depending on their specific needs. An in vivo

study snapshotting the transfer of the CEACAM-binding motif from a Mca strain to

another in order to promote bacterial persistence on the mucosal surface is

warmly awaited.

Diffusely adhering Escherichia coli (DAEC). One group of the pathogenic

strains of E.coli interacting with CEACAMs is represented by the diffusely adhering

E. coli (DAEC). The DAEC are associated with both enteric and urinary tract

infections in humans and animals (Servin, 2005). The CEACAM-binding strains of

this group use their Afa/Dr adhesins to target CEACAMs. The Afa/Dr family of

adhesins includes both fimbrial and afimbrial adhesins. Afa adhesins, for instance,

have afimbrial adhesive sheaths encoded by an afa operon containing up to 6

genes (Servin, 2005). The products of these genes are transcriptional regulators,

periplasmic chaperon proteins, anchoring proteins or adhesin subunits. In most of

the cases the subunit AfaE and AfaD build up the adhesive sheath of the Afa

adhesins. However, AfaE adhesin was shown to be necessary and sufficient for

CEACAM-binding (Guignot et al., 2009). On the structural level, AfaE-III, AfaE

subunit of the uropathogenic E. coli strain A30, had its crystal structure solved a

decade ago. AfaE-III consists of an immunoglobulin-like fold missing a central

antiparallel β-strand and possessing an N-terminal extension. During the Afa

adhesin assembly, a periplasmic chaperone (AfaB) provides the missing strand in

a process that aids folding and targets the adhesin subunits to the outer

membrane usher protein for export (Anderson et al., 2004). The subsequent

incorporation of the adhesin subunit into the adhesive sheath on the bacterial

surface requires the N-terminal strand of AfaE-III to attach another adhesin subunit

by taking over the role previously performed by the chaperone in an anti-parallel

arrangement (Anderson et al., 2004). The CEACAM-binding interface, present on

side of AfaE-III, is a 1,446 Å-area encompassing portions of the strands A, B, E

and D (Korotkova et al., 2006). Even if some mutagenesis analyses have been

carried out in an attempt to pinpoint which AfaE-III amino residues are important

for CEACAM-targeting (Korotkova et al., 2006), molecular and structural features

Page 27: Identification and characterization of the novel CEACAM

General Introduction

15

explaining the failure of some Afa adhesins (e.g.: AfaE-VIII) to bind CEACAMs are

still a mystery.

In sum, the exploitation by distinct bacteria of structurally unrelated proteins to

target the common human CEACAM receptor is a clear indication of the pivotal

role of CEACAM-targeting in bacterial pathogenesis. The numerous unsolved

questions in this fascinating scientific field warrant further investigations.

Page 28: Identification and characterization of the novel CEACAM

Aims of the study

16

Aims of the study

Haemophilus influenzae (Hinf) exhibits an impressing genetic and phenotypic

amenability. However, most of its strains safeguard their interaction with the

human CEACAMs. Therefore, we embarked in this study with the enthusiastic

wish of contributing to the molecular understanding of Hinf pathogenesis,

especially with respect to the exquisite interaction between the postulated adhesin,

the antigenic variable OMP P5, and its human receptor CEACAM. We speculated

that the identification of a conserved epitope sustaining CEACAM-binding may

serve directly as a vaccine candidate or indirectly as rationale for the development

of adhesion-disrupting therapeutics. The fruit of our investigations is organized

here in 4 chapters, as follows:

1) Epithelial CEACAMs highly abundant on the mucosal surface are target of

choice for the incoming bacterial pathogens, including Haemophilus

influenzae. In the first chapter, we review the physiological roles of CEACAMs

before providing an overview on how the bacteria can hijack signaling induced

upon epithelial CEACAM engagement for their own profit. We postulate that

bacterial ligands of CEACAM may be used in the future in order to decrypt

physiological and pathologic processes involving epithelial CEACAMs.

2) Hinf was reported to use the outer membrane protein P5 to target CEACAM1.

The initial aim of this chapter was to determine the CEACAM-binding motif of

OMP P5. However, our initials results cast serious doubt on P5 as the Hinf

CEACAM-binding adhesin. Consequently, we screened a panel of wild-type

and mutants Hinf strains for CEACAM-recognition and identified the outer

membrane protein P1 as the Hinf ligand targeting human CEACAMs. One of

the remaining opened questions of this section was to provide an explanation,

as to why OMP P5 was wrongly considered as CEACAM-ligand. Further, by

expressing OMP P1 of Hinf strains with distinct CEACAM aptitudes in E. coli, it

should be tested whether the E. coli transformants recapitulate the profile of

the respective parent Hinf strain. Lastly, it should be analyzed if the OMP P1

targets exclusively human CEACAMs and if this species-specific recognition of

the receptor is also preserved when OMP 1 is ectopically expressed in E. coli.

Page 29: Identification and characterization of the novel CEACAM

Aims of the study

17

3) With the exception of its surface-exposed loops, OMP P1 is highly conserved

within the Pasteurellaceae family. Here, we sought to investigate if the

conserved structure corresponds to preserved CEACAM binding capability for

all the Pasteurellaceae family members. Mutagenesis analyses and molecular

modeling were used in order to pinpoint the molecular determinants of

CEACAM-targeting by OMP P1. Also, extracellular loops restriction

approaches were established in order to rigorously test the contribution of

loops flexibility in efficient interaction of OMP P1 with CEACAMs. Finally,

three-dimensional modeling of the bimolecular complex OMP P1 – CEACAM

should give visual appreciation of the extent to which extracellular loops of

OMP P1 are implicated in CEACAM-targeting.

4) The predicted structure of Hinf OMP P1 is mesmerizingly similar to the solved

structure of the E. coli fatty acid transporter FadL. In this chapter, we aimed at

analyzing the modulation of OMP P1 levels by fatty acids. The modified OMP

P1 levels were tested for any functional relevance with regard to bacterial

binding to recombinant CEACAMs, CEACAM-induced invasion of epithelial

cells and phagocytosis by human granulocytes.

Page 30: Identification and characterization of the novel CEACAM

18

CHAPTER I

Signaling by epithelial members of the

CEACAM family – mucosal docking sites

for pathogenic bacteria

Arnaud Kengmo Tchoupa1, Tamara Schumacher1 and Christof R. Hauck1,2

1Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany and 2Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz,

Germany

Cell Communication and Signaling 2014: 12:27

Page 31: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

19

I.1 Abstract

Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) comprise

a group of immunoglobulin-related vertebrate glycoproteins. Several family

members, including CEACAM1, CEA, and CEACAM6, are found on epithelial

tissues throughout the human body. As they modulate diverse cellular functions,

their signaling capacity is in the focus of current research. In this review we will

summarize the knowledge about common signaling processes initiated by

epithelial CEACAMs and suggest a model of signal transduction by CEACAM

family members lacking significant cytoplasmic domains. As pathogenic and non-

pathogenic bacteria exploit these receptors during mucosal colonization, we try to

highlight the connection between CEACAMs, microbes, and cellular responses.

Special emphasis in this context is placed on the functional interplay between

CEACAMs and integrins that influences matrix adhesion of epithelial cells. The

cooperation between these two receptor families provides an intriguing example of

the fine tuning of cellular responses and their manipulation by specialized

microorganisms.

I.2 Introduction

The carcinoembryonic antigen-related cell adhesion molecules (CEACAMs), a

subgroup of the CEA family of immunoglobulin-related proteins, are encoded in

the human genome by 12 genes (Kuespert et al., 2006; Zebhauser et al., 2005)

(Figure I-1). All 12 expressed CEACAM genes and a number of derived

pseudogenes cluster on chromosome 19q13 (Kammerer et al., 2007; Teglund et

al., 1994). CEACAMs show distinct expression patterns on different cell types

(Hammarstrom, 1999; Zebhauser et al., 2005). Whereas particular CEACAMs are

only expressed in certain epithelial or myeloid cells, others are found in various

tissues (Gray-Owen and Blumberg, 2006). Some family members play a precise

functional role in particular events such as hearing in the inner ear (CEACAM16)

or phagocytosis of specific bacterial pathogens (CEACAM3) (Schmitter et al.,

2004; Zheng et al., 2011). However, most CEACAMs can be seen as modulators

of general cellular processes such as cell adhesion, differentiation, proliferation,

and survival. To fulfill such diverse functions, CEACAMs have to intersect with

Page 32: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

20

other cellular receptors and to transmit signals into the cell. Indeed, signal

transduction mediated by distinct CEACAM family members, which encompass a

cytoplasmic domain, such as CEACAM3 and a splice variant of CEACAM1 with

long cytoplasmic domain, has been studied in great detail (Buntru et al., 2012;

Gray-Owen and Blumberg, 2006). Given the fact that several CEACAMs are GPI-

anchored proteins or that they sustain functionality in the absence of a cytoplasmic

domain, the mechanistic details of signal transduction processes initiated by these

CEACAM family members are still widely unresolved. Interestingly, CEACAMs are

utilized by bacterial pathogens as host receptors on epithelial cells. Similar to

physiological stimulation of CEACAMs, bacteria-initiated clustering of CEACAMs

can induce robust cellular responses including activation of certain kinases,

stimulation of small G proteins, cytoskeletal rearrangements, induction of novel

gene expression events, enhanced cell adhesion, and receptor endocytosis. It has

become clear that CEACAM-binding bacterial pathogens exploit the signaling

capacity of these immunoglobulin superfamily receptors to enhance their chances

of colonizing the mucosal surface. As CEACAM family members without significant

cytoplasmic domains dominate on several epithelial surfaces such as breast, liver,

or prostate (Gaur et al., 2008), we will use this review, to summarize the current

knowledge about the signaling function of these epithelial CEACAMs. By

highlighting recent advances in the understanding of bacteria-induced CEACAM-

mediated processes, we provide a framework for further dissecting the molecular

signaling connections emanating from epithelial members of this family.

Page 33: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

21

I.3 Physiological roles of epithelial CEACAMs

Since the discovery of carcinoembryonic antigen (CEA) some 50 years ago (Gold

and Freedman, 1965), and the subsequent appreciation of a family of CEA-related

cell adhesion molecules (Beauchemin et al., 1999) (Figure I-1), numerous

physiological and pathological processes have been associated with these

mammalian membrane glycoproteins. Historically, cancer is one of the disease

states linked to aberrant CEACAM function and the role of epithelial CEACAMs in

tumour progression and metastasis has been summarized in an excellent review

recently (Beauchemin and Arabzadeh, 2013). In particular, human CEACAM1,

CEA, and CEACAM6, which can be found on various epithelial cell types and

derived carcinomas, are thought to shape the interaction between tumour cells

and their stromal counterparts as well as immune cells. Apart from their potential

utilization as clinical biomarkers and promising therapeutic targets in melanoma,

lung, colorectal, and pancreatic cancers, these epithelial CEACAMs are also

implicated in morphogenesis (Kirshner et al., 2003; Yokoyama et al., 2007),

Figure I-1: The human CEACAM family. Schematic depiction of the twelve members of the human carcinoembryonic antigen-related cell adhesion molecules. The red spheres indicate IgV-like domains, the blue spheres indicate IgC2-like domains, which are stabilized by disulfide bonds (S-S). The green spirals indicate transmembrane helices. GPI-anchors are depicted in the form of a green arrow ending in the lipid bilayer. CEACAM20 encodes only a partial IgV-like domain (N*). Graph modified from http://www.carcinoembryonic-antigen.de/.

Page 34: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

22

angiogenesis (Ergun et al., 2000; Horst et al., 2006), cell proliferation (Singer et

al., 2010), cell motility (Klaile et al., 2005; Muller et al., 2005), apoptosis (Singer et

al., 2005), regulation of cell matrix attachment (Muenzner et al., 2005; Wong et al.,

2009), as well as epithelial cell-cell interaction and cell polarisation (Benchimol et

al., 1989; Huang et al., 1999). Clearly, forward and reverse genetic approaches in

animal models have suggested that CEACAMs are not essential for all these

processes. For example, mice lacking CEACAM1 are viable and fertile and do not

show gross morphological alterations (Leung et al., 2006). Furthermore,

heterologous expression of human CEACAM1 in the mouse or expression of

additional human epithelial CEACAMs, which are not encoded in the murine

genome (such as CEA and CEACAM6), does not result in perturbation of tissue

architecture or normal tissue homeostasis (Chan and Stanners, 2004; Eades-

Perner et al., 1994; Gu et al., 2010). Therefore, epithelial CEACAMs seem to

contribute to the fine-tuning of cellular behaviour and their contribution might

become critical during stressful conditions, such as tissue damage and repair,

which are not readily obvious in laboratory kept animals.

Most studies of CEACAM-initiated signal transduction have focused on CEACAM1

in immune cells and transformed epithelial cells (nicely summarized in

(Beauchemin and Arabzadeh, 2013; Gray-Owen and Blumberg, 2006)).

Investigations into CEACAM1 structure and function have also profited from the

fact that this family member is expressed in different cell types and that CEACAM1

orthologs exist in other mammalian species (Kammerer and Zimmermann, 2010).

Due to differential splicing, human CEACAM1 occurs in 11 isoforms with the

number of extracellular Ig domains ranging from one to four (see the CEA

homepage at http://www.carcinoembryonic-antigen.de/index.html; (Barnett et al.,

1993)). The major isoforms in human cells are CEACAM1-4 and CEACAM1-3,

which possess an extracellular amino-terminal IgV-like domain, followed by three

(A1, B, A2) or two (A1, B) IgC2-like domains, respectively. Similarly, in other

epithelial CEACAMs, such as CEA or CEACAM6, up to six extracellular IgC2-like

domains follow the amino-terminal IgV-like domain (Figure I-1). Accordingly,

engagement of the extracellular domains of epithelial CEACAMs serves as the

primary stimulus for CEACAM-mediated transmembrane signaling. Under

physiologic conditions, homophilic interactions between CEACAMs on opposing

cells are thought to be the major trigger of CEACAM-initiated signaling processes,

Page 35: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

23

although CEACAMs can also engage in heterophilic interactions, e.g. with

selectins (Obrink, 1997).

I.4 Role of CEACAM extracellular domains in mediating cis- and

trans-oligomerization

Trans-oligomerization resulting from homophilic interactions between the amino-

terminal IgV-like domains of CEACAMs on neighbouring epithelial cells is the basis

of CEACAM-mediated cell-cell adhesion (Teixeira et al., 1994; Watt et al., 2001;

Wikstrom et al., 1996; Zhou et al., 1993a). However, it has become clear that this

homophilic type of trans-oligomerization is further supported by the presence of

IgC2-like domains (Cheung et al., 1993; Zhou et al., 1993a). In a tissue context,

these additional extracellular Ig domains might allow these receptors to extend

farther from the membrane surface to facilitate binding, but they might also be

directly involved in homophilic trans-interactions (Zhou et al., 1993a; Zhou et al.,

1993b). Moreover, recent electron tomography studies of soluble and membrane-

attached CEACAM1 ectodomains have not only confirmed the critical role of the

IgV-like amino-terminal domain for trans-oligomerisation, but also pointed to

additional cis-interactions in the extracellular part of CEACAM1 (Klaile et al.,

2009). Indeed, the extracelluar chain of Ig domains in CEACAM1 appears to be

rather flexible, but can be stabilized by cis-interactions between either IgV-like

domains or IgC2-like domains of parallel CEACAM1 molecules in the same

membrane plane (Klaile et al., 2009). As a consequence, CEACAMs might occur

in different oligomerization states, partially dictated by the occurrence of trans- or

cis-interactions between their extracellular domains. At least in the case of

CEACAM1, these different oligomerization states clearly have an influence on its

signaling function (Muller et al., 2009). In one of the following sections, it will

become clear that the issue of CEACAM1 oligomerization is even more complex,

as the transmembrane domain of this receptor also sustains cis-interactions,

presumably depending on the lipid context.

Page 36: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

24

I.5 Signaling by epithelial CEACAMs

As transmembrane signaling requires a connection to the cytosol, the

transmembrane domain containing CEACAM1 has been the focus of a multitude

of studies (Beauchemin and Arabzadeh, 2013; Gray-Owen and Blumberg, 2006).

Indeed, CEACAM1 harbors a cytoplasmic domain, which can either be long (L; 71

amino acids in humans) or short (S; 10 amino acids). The “L” isoforms encompass

a functional immunoreceptor tyrosine-based inhibitory motif (ITIM) and both

CEACAM1-L and CEACAM1-S isoforms are often co-expressed in the same cell,

with expression ratios varying between different cell types and between different

cellular states (Singer et al., 2010; Singer et al., 2000). In many cases, expression

of the short isoform interferes with CEACAM1-L generated signals (Muller et al.,

2009; Turbide et al., 1997). Therefore, the signal transduction role of CEACAM1

has been mostly attributed to the CEACAM1-L isoform and its cytoplasmic

domain. Indeed, CEACAM1-L can interact with cytoplasmic protein tyrosine

kinases and protein tyrosine phosphatases, as well as with calmodulin, β-catenin,

actin, filamin, shc, and tropomyosin (for review see (Beauchemin and Arabzadeh,

2013)). Only few of these interactions are sustained by the short cytoplasmic

domain of CEACAM1-4S. However, investigations of transformed mammary

epithelial cells (MCF7 cells) grown in a 3D-matrigel environment have suggested

that CEACAM1-4S can induce lumen formation in these carcinoma cells resulting

in acinar-like structures (Kirshner et al., 2003). In follow up studies, the effect of

CEACAM1-4S was pinpointed to binding interactions of the short cytoplasmic

domain. In particular, in CEACAM1-4S the membrane-proximal phenylalanine

F454 or lysine K456 residues (-HFGKTGSSGPLQ), respectively, interact with

cytoskeletal components and T457 (-HFGKTGSSGPLQ) is phosphorylated (Chen

et al., 2007). Furthermore, MCF7 cells injected together with human fibroblasts in

the fat pad of mice show a more normal phenotype (with lumen formation), when

CEACAM1 is stably expressed in these cells (Li et al., 2009). In this situation, both

CEACAM1-4S and CEACAM1-4L are able to induce lumen formation and gland

development in the xenograft (Samineni et al., 2011). Therefore, despite major

differences in their cytoplasmic sequences and their distinct profiles of protein-

protein interactions, both CEACAM1-4L as well as CEACAM1-4S appear to

modulate the growth behaviour of epithelial cells in a similar manner. These

findings imply that they can transmit at least some overlapping signals into the

Page 37: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

25

cells. Indeed, phosphorylation of the membrane proximal threonine residue

(T457), present in the cytoplasmic domains of CEACAM1-4S and CEACAM1-4L,

by calmodulin kinase IID (CaMKIID) is the critical event required for CEACAM1-

driven lumen formation in transformed breast epithelial cells (Nguyen et al., 2014).

A similar contribution of CEACAM1 to morphogenesis has now been reported in

3D cultures of prostate cells (Zhang et al., 2013). The primary human prostate

cells formed organoids with a lumen and small tubular outgrowth, which was

inhibited, when anti-CEACAM1 antibodies were added to the cultures or when

CEACAM1 expression was reduced by about 50% with antisense oligonucleotides

(Zhang et al., 2013). As these cells express both CEACAM1 isoforms, with either

short or long cytoplasmic domain, it is unclear if one or both proteins are

responsible for the phenotype. Prostate epithelial cells express an additional

member of the CEACAM family, CEACAM20, which is found together with

CEACAM1 on the luminal surface of normal prostate glands. Again, antisense

oligonucleotides against CEACAM20 reduced tubule outgrowth (Zhang et al.,

2013). Clearly, CEACAM20 has a cytoplasmic domain sequence distinct from

CEACAM1. Even more striking, CEACAM20 lacks a complete IgV-like amino-

terminal domain, which is instrumental in CEACAM1 for homophilic interactions

between CEACAM1 on neighbouring cells. Together, these recent insights point to

functional commonalities between epithelial CEACAM family members, which

show striking sequence divergence in their amino-terminal IgV-like domain or their

cytoplasmic sequences.

One important implication arising from these results is the realization that signaling

by epithelial CEACAMs could involve parts of these receptors other than the

cytoplasmic domain or the amino-terminal IgV-like domain, such as the

transmembrane or additional extracellular domains. Indeed, recent experiments

employing either carcinoma cell lines or using bacterial pathogens as CEACAM

ligands have pointed into this unexpected direction.

Page 38: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

26

I.6 CEACAM1 cis-oligomerization sustained by the

transmembrane domain

A long standing observation in the field is the reduced expression of CEACAM1

that accompanies transformation of epithelial cells from different tissues

(Beauchemin and Arabzadeh, 2013), including the transition from hepatocytes to

hepatoma cells. It is therefore not surprising, that re-expression of CEACAM1-4L

in rat hepatocellular carcinoma cells results in growth suppression in vitro and

reduced tumour formation in vivo (Laurie et al., 2005). In contrast, expression of

CEACAM1-4S in an anchorage-dependent hepatocellular carcinoma cell line

promoted robust growth of the cells in soft-agar, suggesting that CEACAM1-4S-

initiated signaling rendered the cells anchorage-independent (Lawson et al.,

2012). Strikingly, this effect could be abolished by mutations in the transmembrane

domain. In particular, point mutations disrupting a membrane-integral GxxxG motif

resulted in the loss of the anchorage-independent growth promoting properties of

CEACAM1-4S. As GxxxG motifs in α-helical domains are known to support helix-

helix interactions, it was proposed that such mutations might disrupt cis-dimer

formation of CEACAM1. Recent biochemical approaches based on chemical

crosslinking support the idea that CEACAM1 oligomerizes laterally via the

transmembrane domain to sustain downstream function (Patel et al., 2013).

Together, these results indicate that the transmembrane domain of CEACAM1

promotes clustering and oligomerization of the receptor as a pre-requisite for

signaling into the cell (Figure I-2).

Page 39: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

27

I.7 CEACAM-binding bacteria reveal the lipid raft association of

their receptors

Further insight into CEACAM signaling connections has been gained by the use of

bacterial pathogens as selective and multivalent stimuli of these receptors. Over

the last two decades, diverse CEACAM-binding pathogens including pathogenic

Escherichia coli strains, Neisseria gonorrhoeae, Neisseria meningitidis,

Haemophilus influenzae, and Moraxella catarrhalis, have been found to bind to

CEACAM1 or other epithelial CEACAMs such as CEA and CEACAM6 (Chen and

Gotschlich, 1996; Chen et al., 1997; Hill and Virji, 2003; Leusch et al., 1991; Virji et

al., 2000; Virji et al., 1996a). In an intriguing example of convergent evolution,

these bacteria employ structurally distinct adhesive surface proteins (adhesins) to

Figure I-2: Signaling initiated by epithelial CEACAMs. Schematic summary of recent findingswith regard to CEACAM-initiated signaling events in epithelial cells. Upon ligand binding,CEACAM1 forms oligomers supported by cis-interactions between the extracellular and thetransmembrane domains (1) and is recruited to membrane microdomains (2). GPI-anchoredepithelial CEACAMs, such as CEA or CEACAM6, constitutively localize to membranemicrodomains (3). In membrane microdomains, epithelial CEACAMs connect to putative co-receptor(s) (black) via extracellular IgC2-like domains (4). Intracellular signaling triggered byepithelial CEACAMs either directly or indirectly via co-receptor(s) leads to phosphatidylinositol-3’-kinase dependent signaling processes connected to receptor-mediated endocytosis (5).Furthermore, stimulation of epithelial CEACAMs triggers novel gene expression events, e.g. denovo expression of CD105, which extracts zyxin from basal integrin-rich focal adhesion sites (6),resulting in increased integrin activity and enhanced binding to the basal extracellular matrix (ECM)(7).

Page 40: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

28

connect to the same group of human receptors (Table 1). As CEACAM1, CEA,

and CEACAM6 are exposed on the apical membrane of mucosal cells, they

provide an accessible handle for incoming bacteria (for review see (Kuespert et

al., 2006)). Indeed, all CEACAM-binding pathogenic bacteria characterized so far

exploit the human mucosa as a platform for colonization, multiplication and further

spread (Virji, 2009) . In addition to mere binding to host cells, CEACAM

engagement triggers endocytosis of the bacteria into epithelial cells and

transcytosis of microorganisms through intact epithelial layers (Gray-Owen et al.,

1997; Virji et al., 1996a; Wang et al., 1998). In this respect, it has been reported

before that GPI-anchored CEA and CEACAM6 as well as CEACAM1 initiate a

characteristic uptake pathway that is distinct from phagocytosis mediated by the

granulocyte receptor CEACAM3 (McCaw et al., 2004; Schmitter et al., 2007). Due

to its exceptional phagocytosis promoting properties, CEACAM3-initiated signaling

has been studied in great detail (for review see (Buntru et al., 2012)). In contrast to

epithelial CEACAMs, CEACAM3-initiated uptake of bacteria critically relies on a

cytoplasmic sequence motif and involves extensive actin cytoskeleton

rearrangements orchestrated by the small GTPase Rac and its effector protein

WAVE2 (Pils et al., 2012; Schmitter et al., 2004). Importantly, CEACAM3-

mediated phagocytosis is independent of sphingolipid- and cholesterol-rich

membrane microdomains, as cholesterol chelators do not interfere with this

process (Guignot et al., 2009; Schmitter et al., 2007). This is strikingly different for

epithelial CEACAMs, where internalization of bacteria is sensitive against

cholesterol depletion (Korotkova et al., 2008; Schmitter et al., 2007). Therefore, in

addition to receptor dimerization and oligomerization, it appears that signaling

initiated by epithelial CEACAMs also requires the proper lipid environment in the

membrane. For GPI-linked CEA and CEACAM6 it is known for some time that

these glycoproteins localize to detergent-resistant membrane fractions (Screaton

et al., 2000). In this regard, the GPI anchor of CEA is sufficient to localize proteins

to membrane microdomains (Nicholson and Stanners, 2006). Transmembrane

CEACAM1 has also been found in detergent-resistant membrane microdomains in

epithelial and endothelial cells (Muenzner et al., 2008; Muller et al., 2005). In

contrast to GPI-anchored CEACAMs, which constitutively localize to the detergent-

resistant membrane fraction, CEACAM1 is only found in membrane microdomains

upon receptor clustering (Muenzner et al., 2008). This suggests an additional layer

Page 41: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

29

of regulation, which drives this receptor into specific membrane regions upon

receptor engagement. As mutations in the transmembrane, but not the

cytoplasmic domain of CEACAM1 affect localization in detergent-resistant

membrane fractions (Muenzner et al., 2008), it is tempting to speculate that the

receptor oligomerization function of the CEACAM1 transmembrane domain directs

this receptor into membrane microdomains (Figure I-2). Together, epithelial

CEACAMs require a specific lipid environment in the plasma membrane for proper

function, where GPI-anchored CEACAMs constitutively localize and where

CEACAM1 can be recruited to upon receptor oligomerization.

Bacterial species Primary target tissue

Adhesin CEACAM specificity References

1 3 5 6

Adherent-invasive Escherichia coli (AIEC)

Digestive tract FimH - NC - + (Barnich et al., 2007)

Diffusely adhering Escherichia coli (DAEC)

Digestive tract Urogenital tract

Afa/Dr-I + - + + (Berger et al., 2004)

Haemophilus influenzae Nasopharynx OmpP5 + NC + NC (Hill et al., 2001; Virji et al., 2000)

Moraxella catarrhalis Nasopharynx UspA1 + + + NC (Brooks et al., 2008; Hill and Virji, 2003)

Neisseria gonorrhoeae Urogenital tract OpaCEA + + + + (Chen et al., 1997; Virji et al., 1996b)

Neisseria meningitidis Nasopharynx OpaCEA + + + + (Virji et al., 1996a)

commensal Neisseria Nasopharynx OpaCEA + NC NC NC (Toleman et al., 2001)

Salmonella sp Digestive tract Uncharacterized fimbrial adhesin

+ NC + + (Leusch et al., 1991)

NC: not yet characterized

I.8 CEACAM1 signaling initiated by the IgC2-like extracellular

domains

Though the localization in membrane microdomains is shared by epithelial

CEACAMs this does not provide a direct explanation for their signaling capacity.

Again, CEACAM-mediated internalization of bacterial pathogens provided novel

insight into how epithelial CEACAMs might be mechanistically connected to

intracellular signaling pathways. In numerous endocytic processes,

phosphatidylinositol phosphates (PIPs) play an important role (Araki et al., 1996;

Table 1: CEACAM-binding bacteria and their adhesive proteins

Page 42: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

30

Indik et al., 1995). Therefore, the observation that CEACAM3–mediated

internalization is not blocked by wortmannin, an inhibitor of phosphatidylinositol-3’

kinase (PI3K), was particularly striking (Buntru et al., 2011). This surprising finding

with regard to CEACAM3 prompted an investigation of PI3K and PIPs in bacterial

internalization via epithelial CEACAMs. Interestingly, in CEACAM1-expressing

cells, a strong accumulation of phosphatidylinositol 3’,4’,5’-trisphosphate

(PI3,4,5P) was observed around bacterial uptake sites (Voges et al., 2012).

Furthermore, overexpression of class I PI3K increased bacterial uptake, whereas

wortmannin blocked CEACAM1-, CEA-, and CEACAM6-mediated internalisation.

Expression of the 5’-phosphate-directed PIP phosphatase SHIP (SH2 domain-

containing inositol phosphatase), which dephosphorylates PI3,4,5P, reduces

CEACAM1-mediated internalization. Intriguingly, PI3K-dependent endocytosis via

CEACAM1 was not linked to cytoplasmic determinants of the receptor, but rather

required the extracellular IgC2-like domains of CEACAM1 (Voges et al., 2012).

Accordingly, expression of CEACAM1 mutants lacking either one or all IgC2-like

domains resulted in lower numbers of endocytosed bacteria in comparison to

wildtype CEACAM1 despite a similar binding of the microorganisms to the

truncated receptor. It is interesting to note, that inhibition of PI3K by wortmannin

did not interfere with the re-location of CEACAM1 to membrane microdomains,

suggesting that PI3K signaling is downstream of receptor oligomerization and

membrane microdomain association of the receptor. A plausible explanation would

be that the IgC2 domains of CEACAM1 connect bacteria-bound CEACAM1,

presumably via the extracellular part of a membrane-microdomain located

receptor, with PI3K signaling inside cells (Figure I-2).

It is interesting to note that the IgC2 domains of CEACAM1 orthologs from human,

cattle, mouse and rat show a higher degree of sequence conservation than the

amino-terminal IgV-like domain (Kammerer et al., 2004; Kammerer and

Zimmermann, 2010). The lower sequence conservation in the amino-terminal IgV-

like domain compared to the IgC2-like domains has always been interpreted as a

sign of positive selection for sequence variation in the amino-terminal domain.

However, together with the loss of function upon deletion of the IgC2 domains, the

relative conservation of the IgC2 domains of epithelial CEACAMs may reflect

conserved functions and therefore evolutionary constraints on this region.

Importantly, whereas all isoforms of CEACAM1, CEA, and CEACAM6 encompass

Page 43: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

31

at least one IgC2-like extracellular domain, CEACAM3 lacks such an extracellular

domain. The absence of an IgC2-like extracellular domain in CEACAM3 correlates

well with the mechanistically distinct endocytosis mediated by CEACAM3 in

comparison to epithelial CEACAMs. Altogether, it is very tempting to speculate

that engagement of epithelial CEACAMs will promote the association of their

extracelluar IgC2 domain(s) with not yet identified co-receptor(s), which in turn

transmit the PI3K activating signal into the cell (Figure I-2). This model would also

explain why CEACAMs with differences in the amino-terminal and the cytoplasmic

domain (such as CEACAM1 and CEACAM20) can promote similar cellular

responses as discussed above for prostate morphogenesis. Such a common co-

receptor for multiple CEACAMs might also be located in membrane microdomains,

where CEACAM1 re-locates upon oligomerization and where GPI-anchored

CEACAMs constitutively localize. The identification of this putative co-receptor

might be the turning point in the quest to completely understand the fascinating

physiology of epithelial CEACAMs.

I.9 CEACAM cooperation with integrins and other membrane

receptors

Several cellular receptors have already been proposed to act as co-receptors for

CEA or to co-operate with epithelial CEACAMs (Li et al., 2010; Samara et al.,

2007; Slevogt et al., 2008). For example, in lung epithelial cells, CEACAM1 has

been shown to co-immunoprecipitate with Toll-like receptor 2 (TLR2) and bacterial

engagement of CEACAM1 has been suggested to interfere with TLR2-induced

pro-inflammatory responses (Slevogt et al., 2008). However, as the Moraxella

catarrhalis strain O35E employed in these studies does not bind to any CEACAM

(Brooks et al., 2008), it is unclear, how CEACAM-initiated responses are triggered

in this context.

In several studies, it has been observed that CEACAM stimulation has a positive

effect on cell-matrix adhesion of epithelial cells as well as on integrin-mediated

cell-cell adhesion in leukocytes (Lobo et al., 2009; Muenzner et al., 2005; Muller et

al., 2005). In the case of CEACAM1, a phosphorylation-dependent interaction with

integrin β3 has been reported (Brummer et al., 2001) and CEACAM1 colocalizes

with integrin β1 in MCF7 cells grown in Matrigel (Kirshner et al., 2004) suggesting

Page 44: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

32

that CEACAMs functionally interact with integrins. Since ligand-bound integrins

locally organize membrane microdomains, they could constitute a co-receptor for

epithelial CEACAMs (del Pozo et al., 2004; Hoffmann et al., 2010). Indeed, the

observed functional co-operation was suggested to result from co-clustering of

GPI-linked CEACAMs together with integrins in these membrane areas (Ordonez

et al., 2007). A co-operation between CEACAMs and integrins would nicely

explain the modulation of cellular functions such as cell adhesion and cell survival

in the absence of matrix-attachment (Camacho-Leal and Stanners, 2008;

Camacho-Leal et al., 2007). However, biochemical evidence for a close physical

interaction between CEA or CEACAM6 and integrins is lacking. Furthermore,

CEACAMs localize to lateral cell-cell contacts or the apical membrane

compartment in polarized cells, whereas ligand-bound integrins cluster at basal

cell-matrix adhesion sites. The seeming contradiction between functional

cooperation and distinct subcellular localization of epithelial CEACAMs and

integrins has been nicely resolved. Using CEACAM-binding bacteria as a naturally

occurring, highly selective and multivalent ligand for CEACAM1, CEA, and

CEACAM6, an unbiased gene-expression analysis revealed a number of genes,

which are specifically induced following CEACAM stimulation in epithelial cells

(Muenzner et al., 2005). Further analysis showed that upregulation of a member of

the TGF-β1 receptor family, termed endoglin or CD105, is observed upon

stimulation of GPI-anchored CEACAMs or stimulation of a CEACAM1 mutant

lacking the complete cytoplasmic domain (Muenzner et al., 2005). In all these

cases, CEACAM engagement by bacteria results in an elevated CD105 mRNA

level, which is observed within 1–3 hours after bacterial infection (Muenzner et al.,

2005). In a similar time frame, the infected epithelial cells display enhanced

integrin-mediated adhesion to the extracellular matrix and CD105 expression is

necessary and sufficient for this phenotype (Muenzner et al., 2005). CD105

expression in turn does not alter the amount of integrins on the cells, but initiates

the redistribution of the focal adhesion protein zyxin. Indeed, zyxin binds with high

affinity to the cytoplasmic domain of CD105, and disappears from integrin-rich

focal adhesion sites as soon as CD105 is expressed in epithelial cells (Muenzner

et al., 2010) (Figure I-2). Due to the lack of zyxin at focal adhesions, integrin

activity, and therefore extracellular matrix (ECM) binding of the infected cells,

increases over the course of several hours following contact with CEACAM-

Page 45: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

33

binding bacteria. Increased integrin activity and strengthened ECM-binding is also

observed in zyxin-deficient or CD105-overexpressing cells, suggesting that

CEACAM-binding bacteria exploit physiological regulators of cell adhesion to

indirectly manipulate integrin activity (Muenzner et al., 2010) (Figure I-2). This

functional interplay between CEACAM stimulation, CD105 expression and its

effect on focal adhesion site composition provide a plausible scenario, how

CEACAMs can modulate integrin-mediated cell adhesion even without directly

associating with integrins. However, it should be noted that several CEACAM-

binding bacteria also possess surface adhesins, which associate with extracellular

matrix (ECM) proteins of their host, such as fibronectin or vitronectin (Brooks et al.,

2008; Hill and Virji, 2003; Virji, 2009). In this manner, ECM protein binding could

allow such bacteria to simultaneously engage integrins and CEACAMs once the

integrity of the epithelial barrier and the spatial separation of CEACAMs and

integrins might be compromised. If such a potential co-stimulation of integrins and

CEACAMs by pathogenic microbes has consequences for the outcome of

bacteria-host interaction has not been explored so far.

Nevertheless, already the indirect connection between CEACAMs and integrins

must be advantageous for bacteria trying to get a foothold on the mucosal surface,

given the fact that so many unrelated microbes target CEACAMs (Table 1).

Indeed, this functional connection allows bacteria to engage receptors on the

apical side of the epithelium, while ultimately impacting on the activity of integrins,

which are located on the basolateral side of polarized epithelial cells. In the case

of CEACAM-binding Neisseria gonorrhoeae, which infects the urogenital tract, it

has been observed that the enhanced matrix binding of the infected epithelial cells

strongly reduces the exfoliation of the superficial mucosal cell layer (Muenzner et

al., 2010). Suppressing CD105 upregulation or inhibiting the zyxin-CD105

interaction in the urogenital tract of CEA-transgenic mice allows exfoliation to

proceed despite the presence of CEACAM-binding bacteria, providing

experimental proof that CEACAM engagement is instrumental for successful

colonization of the mucosal surface (Muenzner et al., 2010). Further examples

have emerged, which demonstrate that colonization of the nasopharyngeal

mucosa by Neisseria meningitidis or Moraxella catarrhalis profits from the

presence of epithelial CEACAMs (Bookwalter et al., 2007; Johswich et al., 2013).

In the case of N. meningitidis, bacteria are not detected in wildtype mice three

Page 46: Identification and characterization of the novel CEACAM

Pathogen interaction and signaling by epithelial CEACAMs

34

days after inoculation, whereas the same bacterial strain is present for up to a

week in the nasopharynx of CEACAM1-transgenic mice (Johswich et al., 2013). It

is currently unclear, if suppression of epithelial exfoliation, CEACAM-integrin

cooperation, or other forms of CEACAM-initiated cellular responses are involved in

nasopharyngeal colonization. However, these examples again demonstrate that

epithelial CEACAMs, either with or without cytoplasmic domain, can orchestrate

signaling events in epithelial cells. Furthermore, they also showcase that much of

CEACAM functionality can be learned by using CEACAM-binding bacteria, such

as N. gonorrhoeae, as selective and potent stimuli.

I.10 Conclusions

Over the last decade, CEACAMs have emerged as important modulators of

signaling events in leukocytes, endothelial, and epithelial cells. The simultaneous

expression of multiple CEACAM family members by most human epithelial tissues,

including GPI-anchored and transmembrane forms in different splice variants, has

hampered progress in deciphering the molecular signaling connections initiated by

CEACAM-mediated cell-cell interactions. To understand the contribution of

CEACAMs in these processes, well characterized antibodies have been employed

to interfere with CEACAM-CEACAM interactions, but due to sterical hindrance

such approaches might also block a number of other cell-cell interactions. The use

of CEACAM-binding bacteria as multivalent, high affinity ligands for a number of

epithelial CEACAMs has provided an additional opportunity to selectively trigger

CEACAM signaling in vitro and in vivo. These natural probes allow the

visualization of local CEACAM-initiated signaling complexes as well as signaling

intermediates and, therefore, have provided novel insight. Combining these

different approaches will further help to refine our understanding of epithelial

CEACAM physiology and of the involved molecular and cellular processes.

Page 47: Identification and characterization of the novel CEACAM

35

CHAPTER II

Outer membrane protein P1 is the

CEACAM-binding adhesin of

Haemophilus influenzae

Arnaud Kengmo Tchoupa1, Sabine Lichtenegger2, Joachim Reidl2 and Christof

R. Hauck1,3

1Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany

2Institute of Molecular Biosciences, University of Graz, Graz, Austria

3Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz,

Germany

Mol Microbiol. 2015 Jul 16. doi: 10.1111/mmi.13134. [Epub ahead of print]

Page 48: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

36

II.1 Summary

Haemophilus influenzae is a Gram-negative pathogen colonizing the upper

respiratory tract mucosa. H. influenzae is one of several human-restricted bacteria,

which bind to carcinoembryonic antigen related cell adhesion molecules

(CEACAMs) on the epithelium leading to bacterial uptake by the eukaryotic cells.

Adhesion to CEACAMs is thought to be mediated by the H. influenzae outer

membrane protein (OMP) P5. However, CEACAMs still strongly bound to H.

influenzae lacking OMP P5 expression, and soluble CEACAM receptor

ectodomains failed to bind to OMP P5, when heterologously expressed in E. coli.

Screening of a panel of H. influenzae OMP mutants revealed that lack of OMP P1

completely abrogated CEACAM binding and supressed CEACAM-mediated

engulfment of H. influenzae by epithelial cells. Moreover, ectopic expression of

OMP P1 in E. coli was sufficient to induce CEACAM-binding and to promote

attachment to and internalization into CEACAM-expressing cells. Interestingly,

OMP P1 selectively recognizes human CEACAMs, but not homologs from other

mammals and this binding preference is preserved upon expression in E. coli.

Together, our data identify OMP P1 as the bona-fide CEACAM-binding invasin of

H. influenzae. This is the first report providing evidence for an involvement of the

major outer membrane protein P1 of H. influenzae in pathogenesis.

II.2 Introduction

Haemophilus influenzae is a human-restricted Gram-negative coccobacillus that

colonizes asymptomatically the human respiratory tract of healthy individuals. The

shift from commensal to pathogen happens when the bacterium reaches privileged

anatomical sites under various predisposing conditions, such as age, viral

infections, or a constant exposure to pollutants. Haemophilus infection can result

in otitis media, sinusitis, conjunctivitis, respiratory infections, septicaemia, or

meningitis (Foxwell et al., 1998; Rao et al., 1999). The capsulate members of this

species (typeable Hinf) are grouped into six serotypes (a-f) depending on the

composition of their capsular polysaccharides, with serotype b strains causing

invasive infections in infants and children (Agrawal and Murphy, 2011). These

strains may switch off capsule expression in vivo and in vitro (Hoiseth and

Page 49: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

37

Gilsdorf, 1988), whereas the truly acapsulate strains lack the genetic information

for capsulation (non-typeable H. influenzae, NTHi). During the last 2 decades, the

widespread use of H. influenzae type b (Hib) conjugate vaccines has nearly

eradicated invasive disease by Hib in countries where the vaccines are used

widely (Agrawal and Murphy, 2011). In contrast, NTHi remains one of the most

common causes of otitis media, while also playing major roles in sinusitis and

conjunctivitis (Murphy et al., 2009). Furthermore, chronic obstructive pulmonary

disease (COPD), the fourth leading cause of death worldwide, can be exacerbated

by bacterial pathogens with NTHi being the most common (Roier et al., 2012).

Besides, the non-capsulate H. influenzae biogroup aegyptius (H. influenzae-aeg)

is an important cause of self-limiting conjunctivitis and Brazilian purpuric fever, a

fulminant systemic disease observed in children (Harrison et al., 2008).

The first step of in H. influenzae pathogenesis is the colonization of the

nasopharynx, which requires that the microbe overcomes host clearance

mechanisms, including the mucociliary escalator and local immunity (St. Geme III,

2002). Similar to other pathogenic bacteria, H. influenzae can express multiple

adhesins, which mediate attachment to host target tissues (Abraham et al., 1998;

Finlay and Falkow, 1997). One group of host receptors recognized by both typable

H. influenzae and NTHi belongs to the carcinoembryonic antigen-related cell

adhesion molecules (CEACAMs), a subgroup of the immunoglobulin superfamily

glycoproteins present on the apical side of nasopharyngeal epithelial cells (Virji et

al., 2000). It has been reported earlier, that the outer membrane protein (OMP) P5

of H. influenzae is the bacterial adhesin responsible for CEACAM binding (Hill et

al., 2001). However, P5 shows large antigenic variation between different strains

and it is unclear, which structural feature of this OMP mediates the interaction with

host CEACAMs.

In this study, we report that the OMP P5 of typeable H. influenzae or NTHi fails to

promote CEACAM binding. Instead, we identify H. influenzae OMP P1, a

homologue of E. coli FadL, as the CEACAM-binding adhesin. Deletion of P1 in H.

influenzae as well as heterologous expression in E. coli demonstrates that P1 is

necessary and sufficient to bind several human CEACAMs. Concordantly, when

expressed on the surface of E. coli, P1 mediates adhesion to and invasion into

epithelial cells. By identifying the bona fide CEACAM-binding adhesin of typeable

Page 50: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

38

and non-typeable H. influenzae our study opens up novel avenues in designing

preventive strategies against these human pathogens.

II.3 Results

Haemophilus influenzae strains display distinct CEACAM binding profiles

More than a decade after the initial discovery, that typeable H. influenzae and

NTHi strains bind to the amino-terminal immunoglobulin variable (Igv)-like domain

of human CEACAM1 (Virji et al., 2000), the structural details of this interaction are

still unknown. Given the fact that the proposed CEACAM-binding adhesin, the

OMP P5, is highly variable, we wondered, whether this variation dictates binding to

selected human CEACAM family members. Accordingly, fifteen different H.

influenzae strains were employed in bacterial pull-down assays with soluble Igv-

like amino-terminal domains of human CEACAM1, CEACAM3, CEACAM4,

CEACAM5, and CEACAM6 expressed as GFP-fusion proteins (Figure II-1A and

B). Four different CEACAM-binding profiles were observed for the various strains:

14 out of 15 H. influenzae strains recognized CEACAM1, about a quarter of the

strains recognized one additional CEACAM family member, namely CEA, and one

strain of H. influenzae biogroup aegyptius (ATCC 11116) was able to bind

CEACAM1, CEA, and CEACAM3 (Figure II-1B and C). None of the H. influenzae

strains bound to CEACAM4, CEACAM6, or CEACAM8, which share a sequence

identity of 45, 90, or 72%, respectively, with human CEACAM1 in their N-terminal

domains (Figure II-1B and C).

Importantly, one NTHi isolate (strain 7502) completely lacked CEACAM binding

and even prolonged exposure did not reveal any positive signals (Figure II-1B).

Our recombinant CEACAM proteins comprised the Igv-like amino-terminal

domains only. However, the IgC2-like domains of CEACAM1 (A1, B or A2) were

suggested to contribute to the interaction of H. influenzae with CEACAM1 (Virji et

al., 2000). Therefore, we tested whether NTHi 7502 would be able to bind a

recombinant CEACAM1 protein comprising all extracellular domains (hCEA1-

NA1BA2; Figure II-1A).

Page 51: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

39

Figure II-1. Differential recognition of human CEACAMs by various H. influenzae strains. A. Schematic drawing of human CEACAM1, the recombinant CEACAM1 amino-terminal Igv-like domain fused to GFP (hCEA1-N), the soluble CEACAM1 containing 4 extracellular domains (hCEA1-NA1BA2) and the recombinant chimera consisting of N-terminal Igv-like domain of CEACAM8 followed by 3 IgC2-like domains of CEACAM1 (hCEA8/1-NA1BA2). Soluble, GFP-fused amino-terminal Igv-like domains of different human and mammalian CEACAMs were used in this study. B. GFP-tagged N-terminal domains of human CEACAM1 (hCEA1-N), CEACAM3 (hCEA3-N), CEA (hCEA-N), CEACAM6 (hCEA6-N) and CEACAM8 (hCEA8-N) were generated as secreted proteins in 293 cells. Equivalent levels of the different soluble N-terminal domains were verified by probing the supernatants with anti-GFP antibodies (lowest panel). Supernatants containing the indicated GFP-fusion proteins were incubated with Hinf-aeg or NTHi strains 7502, KN4 or H86, respectively, and CEACAM-binding was determined upon bacterial pull-down and anti-GFP

Page 52: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

40

Western Blot (upper panels). Pull-downs using NTHi strain 7502 (uppermost panel) were exposed 3-times longer than the other panels. C. Receptor binding assays as described in (B) were performed with eleven additional H. influenzae strains. The binding to CEACAMs is symbolized by “+” in case of binding or “─” in absence of binding. D. Cell culture supernatants (supe, left panel), enriched in equivalent amounts of GFP-tagged soluble proteins containing hCEA1-NA1BA2 or hCEA8/1-NA1BA2, respectively, were incubated with NTHi 7502 (middle panel) or H. influenzae Rd (right panel). The binding to CEACAMs was analyzed by flow cytometry. Bacteria incubated in plain medium (gray line) were included as negative controls. The number (6x) shows the -fold change in mean fluorescence intensity (MFI) of bacteria incubated with hCEA1-NA1BA2 in comparison to bacteria incubated with hCEA8/1-NA1BA2.

Similar to its inability to bind the CEACAM1 N-terminal domain (Figure II-1B), NTHi

7502 failed to recognize hCEA1-NA1BA2 (Figure II-1D). To investigate the

contribution of the Igv-like amino-terminal domain versus the IgC2-like domains of

CEACAM1 in H. influenzae binding we used hCEA1-NA1BA2 as well as a

chimeric protein, where the CEACAM1 N-terminal domain was exchanged for the

CEACAM8 N-terminal domain (hCEA8/1-NA1BA2; Figure II-1A). Clearly, H.

influenzae Rd associated with hCEA1-NA1BA2, but not with hCEA8/1-NA1BA2

highlighting the essential role of the Igv-like amino-terminal domain for the H.

influenzae-CEACAM1-interaction (Figure II-1D). Together, these data demonstrate

that the Igv-like domain of CEACAM1 is necessary and sufficient to mediate H.

influenzae binding, but that there is considerable heterogeneity in CEACAM-

recognition by H. influenzae strains that may be linked to variations in the

CEACAM-binding adhesin.

The OMP P5 protein of H. influenzae does not confer CEACAM binding

properties

The -barrel outer membrane protein (OMP) P5 was proposed to constitute the

CEACAM-binding adhesin of H. influenzae (Hill et al., 2001). To better define the

structure-activity relationship of this bacterial adhesin, we set out to compare P5

sequence variation to the observed CEACAM binding profiles. Therefore, we

cloned and sequenced the ompP5 gene from the 15 H. influenzae strains used in

the CEACAM-binding assay and aligned the protein sequences to delineate a

CEACAM-binding motif (Figure II-2A). By concentrating on surface exposed loops

3 and 4 of OMP P5, which form the putative binding interface (Bookwalter et al.,

2008), the amino acid sequence variation in these regions became obvious

(Figure II-2A). Interestingly, homologies in loop sequences did not co-segregate

with the observed pattern of CEACAM-binding. In particular, the OMP P5

Page 53: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

41

sequence of strain 7502, which showed no CEACAM binding in our assays, was

not grossly different from the other strains (e.g. compare strain 7502 with strains

2019 or H51; Figure II-1C and Figure II-2A.). To directly link OMP P5 with the

ability of bacteria to bind CEACAM1, we expressed OMP P5 derived from one of

the CEACAM-binding H. influenzae strains, strain Rd, heterologously in E. coli

(Eco-P5-Rd). OMP P5 was not only expressed, but also integrated into the outer

membrane of E. coli as demonstrated by its accessibility to anti-P5 antibodies

(Figure II-2B and C) and its heat modifiable migration in gel electrophoresis

(Figure. S2). However, similar to the control E. coli strain harboring the empty

plasmid (Eco pET28), Eco-P5-Rd was not able to bind to the GFP-fused

CEACAM1 Igv-like amino-terminal domain, whereas Hinf Rd showed the expected

strong binding to CEACAM1-GFP in this assay (Fig. 2D). In addition, we

heterologously expressed two additional OmpP5 genes derived from strains H51

and H86, respectively, in E. coli (Eco-P5-H51; Eco-P5-H86). Similar to Eco-P5-Rd

we did not observe any binding to CEACAMs (data not shown). Together, these

results suggested that either OMP P5 alone is not sufficient to promote CEACAM1

binding or that this membrane protein is not the CEACAM-binding adhesin of H.

influenzae.

Page 54: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

42

Figure II-2. The OMP P5 of H. influenzae is not the CEACAM-binding adhesin. A. 15 H.influenzae OMP P5 sequences spanning the predicted surface-exposed loops 3 and 4 werealigned with Clustal Omega. Spaces (-) are introduced to maximize homology. The dashed linesseparate H. influenzae strains exhibiting the same CEACAM-binding profile. B. Presence of H.influenzae OMP P5 in the outer membrane of recombinant E. coli (Eco-P5-Rd) was detected byWestern blotting with polyclonal anti-P5 mouse serum (-P5 blot; upper panel). E. coli harboringthe empty vector (Eco pET28) and outer membrane protein preparation of H. influenzae Rd servedas negative and positive controls, respectively. The arrow head points to Hinf OMP P5, whereasthe asterisk indicates a non-specific cross-reactive band from E. coli, presumably OMP A.Equivalent protein loading was demonstrated by Coomassie staining of the membrane (bottompanel). C. Surface location of OMP P5 in Eco-P5-Rd was confirmed by flow cytometry followingstaining with -P5 serum. Grey area indicates staining of the Eco-pET28 control strain. D. IPTG-induced Eco-pET28 and Eco-P5-Rd were incubated with cell culture supernatants containinghuman CEACAM1-N-GFP (hCEA1-N), human CEACAM8-N-GFP (hCEA8-N), or plain culturemedium. After washing, samples were analysed by flow cytometry to determine bacteria-associated GFP fluorescence. H. influenzae Rd was included as positive control for humanCEACAM1-binding. Grey area indicates staining of bacteria incubated in plain culture medium. Thenumber (5x) shows the -fold change in mean fluorescence intensity (MFI) of H. influenzae Rdincubated with hCEA1-N in comparison to hCEA8-N.

Page 55: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

43

Genetic deletion of OMP P1 abrogates Hinf interaction with CEACAM1

The failure of heterologously expressed OMP P5 to engage CEACAMs prompted

us to search for alternative CEACAM-binding outer membrane proteins in H.

influenzae. Accordingly, we tested a panel of isogenic H. influenzae mutants with

deletions of defined outer membrane proteins for their ability to associate with the

recombinant CEACAM1-GFP fusion protein (see Table S1 for a list of strains and

mutants). Importantly, whereas the P5-deficient mutant of H. influenzae strain Rd

showed reduced CEACAM1 binding, the deletion of the OMP P1 of this strain

completely abrogated CEACAM1 binding (Figure II-3A). Deletion of genes

encoding OMP P2, OMP P4, or transferrin-binding protein did not alter CEACAM

recognition by Hinf strain Rd (Figure II-3A). To corroborate these findings in a

more physiological context, we analyzed adhesion of H. influenzae strain Rd to

human lung adenocarcinoma A549 cells. To this end, cells were infected with

either the wildtype bacteria (Hinf Rd), the OMP P5 mutant (Hinf Rd ∆P5), or the

OMP P1 mutant (Hinf Rd ∆P1), respectively. Two hours after infection, samples

were washed and processed for scanning electron microscopy. Clearly, whereas

numerous wildtype bacteria and similar numbers of the isogenic Hinf Rd ∆P5

mutant were found attached to the epithelial cells, cell adhesion by Hinf Rd ∆P1

was almost completely absent (Figure II-3B). Viability and growth of all three

strains in cell culture medium was not affected by the presence or absence of

OMP P1 or OMP P5, respectively (Suppl. Figure S1A). Together, these data

highlight the necessity of H. influenzae OMP P1 for CEACAM-targeting.

Page 56: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

44

Figure II-3. Genetic deletion of OMP P1 abrogates the interaction of H. influenzae withhuman CEACAMs. A. H. influenzae Rd and its respective P5- (Hinf Rd ∆P5), P1- (Hinf Rd ∆P1),P2- (Hinf Rd ∆P2), P4- (Hinf Rd ∆P4) or TbpX- (Hinf Rd ∆TbpX) deficient mutants were incubatedwith the indicated human CEACAM-GFP fusion proteins or cell culture medium. Flow cytometrywas used to detect CEACAM binding. The fold change in MFI upon incubation with hCEA1-Ncompared to hCEA8-N is indicated for each sample. B. A549 cells were infected for 2 hours withwild-type H. influenzae Rd strain, its isogenic P5 or P1 mutant and processed for scanning electronmicroscopy upon fixation. Shown are pseudocolored bacteria (blue) attached to A549 cells (grey).

Page 57: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

45

CEACAM1-mediated host cell invasion of H. influenzae is OMP P1-dependent

Although being considered a classical extracellular pathogen, H. influenzae has

been consistently reported within host cells, including epithelial cells (for a

comprehensive review, please refer to (Clementi and Murphy, 2011)). In order to

unambiguously identify the contribution of CEACAM1 in epithelial cell invasion by

H. influenzae, we used HeLa cells, which do not express CEACAMs

endogenously. To allow CEACAM-mediated interactions, HeLa cells were stably

transfected with GFP-tagged human CEACAM1 or with GFP alone (Suppl. Fig.

S1B). These stable cell lines were infected for 2 h with Hinf Rd, Hinf Rd ∆P1, or

Hinf Rd ∆P5, respectively, and internalized bacteria were evaluated by gentamicin

protection assays. Importantly, Hinf Rd invaded HeLa cells in a CEACAM1-

dependent manner and also Hinf Rd ∆P5 showed a CEACAM1-dependent

invasion, which was reduced to about 50% of the wildtype strain (Figure II-4A).

Consistent with its inability to bind CEACAM1, Hinf Rd ∆P1 completely failed to

invade HeLa CEACAM1 cells (Figure II-4A). Analysis of the outer membrane

protein profile of the three strains indicated that OMP P5 expression in Hinf Rd

∆P1 was comparable to the wildtype Rd strain (Figure II-4B). However, OMP P1

expression was clearly reduced in the Hinf Rd ∆P5 mutant indicating that deletion

of OMP P5 could affect surface expression of OMP P1 (Figure II-4B and C).

Alterations in outer membrane content of OMP P1 in Hinf Rd ∆P5 were not

correlated with gross morphological changes of the microorganisms as observed

by scanning electron microscopy (Figure II-4D). Together, our observation of

compromised OMP P1 levels in a Hinf Rd ∆P5 mutant might explain the reduction

in CEACAM binding and CEACAM-mediated invasion by P5-deficient H.

influenzae strains (Figure II-3A and Figure II-4A).

Page 58: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

46

Figure II-4. H. influenzae invades CEACAM-expressing cells in an OMP P1-dependentmanner. A. HeLa cells stably expressing GFP alone or GFP-tagged human CEACAM1 wereinfected with H. influenzae Rd or the indicated mutants (Rd ∆P5 or Rd ∆P1). Two hours post-infection, extracellular bacteria were killed with gentamicin and the amount of viable, intracellularbacteria was determined via colony counts. Results represent means ± SD of three independentexperiments done in triplicate (n=3) *P < 0.05, ***P < 0.001). B. Wild-type H. influenzae Rd strain(WT), the OMP P5 mutant (∆P5) and the OMP P1 mutant (∆P1) outer membrane proteins wereisolated and immunoblotted with anti-P5 (upper panel) or anti-P1 (middle panel) mouse sera.Similar amounts of protein in the different samples were verified by Coomassie staining of themembrane (lower panel). C. Densitometry of intensities of OMP P1 band in ∆P5 relative to that ofthe wild-type Rd strain. Values are mean ± SD of the amount of OMP P1 as observed in threeindependent experiments. (*P < 0.05). D. Scanning electron microscopy pictures of H. influenzaeRd wildtype and Rd ∆P5, respectively. Scale bars 200 nm.

Page 59: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

47

Complementation of OMP P1 in Hinf Rd ∆P1 restores CEACAM-binding

To firmly establish OMP P1 as a CEACAM-binding adhesin, we complemented the

P1-deficient Hinf Rd mutant (Hinf Rd ∆P1) by restoration of the chromosomal OMP

P1 locus. The resulting strain (Hinf Rd ∆P1 P1+) expressed P1 at levels similar to

the H. influenzae Rd parent strain (Figure II-5A) and bound to the soluble

CEACAM1-N-domain (Figure II-5B). To determine if re-expression of OMP P1 in

Hinf Rd ∆P1 leads to cellular uptake of the bacteria, flow cytometry-based invasion

assays were conducted with 293 cells and FITC-labeled bacteria. Cells were

transfected with a full length CEACAM1-4L-encoding expression plasmid or the

empty control plasmid. Cells were then infected with Hinf Rd, the OMP P5 mutant

(Hinf Rd ∆P5), the OMP P1 mutant (Hinf Rd ∆P1), or the P1 complemented strain

(Hinf Rd ∆P1 P+). After 2h of infection, the internalization of FITC-labeled microbes

was quantified by flow cytometry in the presence of trypan blue, which quenches

the fluorescence of extracellular bacteria. Consistent with the gentamicin

protection assays (Figure II-4A), Hinf Rd and, to a slightly lesser extent, Hinf Rd

∆P5 were internalized by CEACAM1-expressing 293 cells (Figure II-5C). However,

Hinf Rd ∆P1 showed no invasion and this non-invasive phenotype of Hinf Rd ∆P1

was completely reversed upon complementation with OMP P1 (Figure II-5C).

Together, these results confirmed the critical role of OMP P1 in CEACAM-

mediated interactions with host cells and suggested that OMP P1 can function as

a H. influenzae invasin for CEACAM-expressing human cells.

Page 60: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

48

Figure II-5. Restoration of ompP1 in H. influenzae OMP P1 mutant reverses its CEACAM-binding phenotype. A. Outer membrane proteins of H. influenzae Rd (WT), its P1 mutant (∆P1)and the P1-restored strain (∆P1 P1+) were analyzed for P1 or P5 expression using anti-P1 or anti-P5 mouse sera, respectively. B. CEACAM-binding of the P1-complemented ∆P1 mutant Hinf Rd(Hinf Rd ∆P1 P1+) was determined by flow cytometry after incubation with the indicated GFP-fusedCEACAM soluble proteins. Grey area indicates staining of bacteria incubated in plain culturemedium. The fold change in MFI upon incubation with hCEA1-N compared to hCEA8-N isindicated. C. 293 cells were transiently transfected with the empty vector (pcDNA) or with a vectorencoding CEACAM1-4L-HA. Expression of CEACAM1-4L-HA in the transfected 293 cells wasdetected by Western Blotting of whole cell lysates (WCL) with monoclonal CEACAM-antibody(clone D14HD11; upper panel) and similar amounts of loaded WCL was verified by anti-tubulinantibodies (lower panel). Transfected cells were infected for 2 hours with FITC-labelled H.influenzae Rd wildtype, Rd ∆P5, Rd ∆P1, or the P1 complemented strain (Rd ∆P1 P1+),respectively. The infected cells were suspended and the fluorescence of extracellular bacteria wasquenched by trypan blue. Cell-associated fluorescence, which was proportional to the amount ofintracellular bacteria, was analyzed by flow cytometry. Uptake of H. influenzae Rd wildtype byCEACAM1-expressing cells was set to 100%. Bars show means ± SD of the intracellular bacteriaas observed in three independent experiments (*P < 0.05, ***P < 0.001).

Page 61: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

49

Modelling of H. influenzae OMP P1 identifies surface-exposed structures

In an attempt to delineate CEACAM-binding structural features of OMP P1, we

combined three-dimensional modeling of OMP P1 with multiple protein sequence

alignment of OMP P1 from H. influenzae strains exhibiting distinct CEACAM-

recognition profiles. Interestingly, H. influenzae OMP P1 is a homologue of the E.

coli long-chain fatty acid transporter (FadL), which share around 39% amino acid

sequence identity. Using the atomic structure of FadL (van den Berg et al., 2004),

Hinf OMP P1 was modeled using SWISS-MODEL (http://swissmodel.expasy.org

/interactive). The resulting three-dimensional structure suggested that OMP P1

consists of a 14-stranded β barrel embedded in the Haemophilus outer membrane

Figure II-6A). The lumen of the barrel is occluded by the amino-terminal domain

and there are seven extracellular loops (L1 to L7), of which four, L1, L3, L4, and

L7, are prominent and easily accessible (Figure II-6A). Next, we extracted the

OMP P1 coding sequences of strains Hinf Rd KW20 (access. No. NC_000907

REGION: 422144..423523) and NTHi 86-028 NP (access. No. NC_007146

REGION: 491079..492455) from NCBI (http://www.ncbi.nlm.nih.gov). Furthermore,

ompP1 genes from thirteen additional isolates (see Table S1) were cloned and

sequenced. The derived protein sequences were aligned with Clustal Omega

(Sievers et al., 2011). The alignment highlighted conserved as well as the highly

variable regions of OMP P1, which coincide with the surface exposed loops 1, 3,

4, and 7 (Figure II-6B). Strikingly, the only strain in our collection, which had been

negative for CEACAM-binding, strain NTHi 7502, turned out to be a natural OMP

P1 mutant. Indeed, the ompP1 coding sequence of this strain contained a non-

sense mutation at position 875 creating a premature stop codon (Figure II-6B).

The disruption of expression was confirmed at the protein level, as no OMP P1

was detected in outer membrane preparations of strain NTHi 7502, while OMP P5

was expressed at normal levels (Figure II-6C). This result provides further genetic

support for the idea that OMP P1 is the CEACAM-binding adhesin. Further

inspection of the surface exposed loops of the CEACAM-binding OMP P1 variants

did not reveal a consistent sequence motif, which would explain binding to one or

more CEACAMs (Figure II-6B). Therefore, these findings suggest that a three-

dimensional structure rather than a conserved linear stretch of amino acids

determines CEACAM-targeting by H. influenzae OMP P1.

Page 62: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

50

Figure II-6. H. influenzae OMP P1 is a β-barrel porin with highly variable extracellular loops.A. Putative Hinf-aeg OMP P1 three-dimensional structure generated by SWISS-Model. OMP P1 ispredicted to consist of a 14-stranded transmembrane β-barrel obstructed by an N-terminal hatchdomain. B. The deduced amino acid sequences of OMP P1 from 15 H. influenzae strains werealigned with Clustal Omega. Spaces [.] are introduced to allow maximal alignment. The colorshighlight amino acids, which are conserved in >90% of strains (dark grey), 60 – 90 % of strains(grey), or 45 – 60% (light grey) of analysed strains. The black arrows above the sequencesdelineate the surface-exposed loops (loop 1 — 7). C. Outer membrane proteins were isolated fromH. influenzae 7502 or H. influenzae Rd, respectively, and immunoblotted with anti-P5 (upper panel)or anti-P1 (lower panel) mouse sera.

Page 63: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

51

Heterologous expression of H. influenzae OMP P1 transfers CEACAM-

binding capability to E. coli

To investigate if OMP P1 is sufficient to recognize CEACAMs, we cloned and

expressed the ompP1 gene of H. influenzae Rd in E. coli (Eco-P1-Rd).

Immunoblot analysis of outer membrane preparations as well as antibody staining

of intact bacteria indicated that OMP P1 is expressed and located on the surface

of Eco-P1-Rd (Figure II-7A and B). Importantly, expression of H. influenzae OMP

P1 converted E. coli into a CEACAM1-binding bacterium (Figure II-7C). To verify

OMP P1–CEACAM interaction in a cellular context, we infected HeLa cells

expressing GFP or GFP-tagged human CEACAM1 with Eco-P1-Rd, Eco-P5-Rd,

or E. coli harboring the empty vector (Eco-pET28), respectively. Neither Eco-

pET28, nor Hinf OMP P5-expressing E. coli (Eco-P5-Rd) was taken up by GFP- or

CEACAM1-GFP HeLa cells (Figure II-7D). In sharp contrast, Eco-P1-Rd was

readily internalized by CEACAM1-expressing cells (Figure II-7D). The results

obtained with antibiotic protection assays were corroborated with microscopic

analysis of infected samples. Differential staining of intracellular vs. extracellular

bacteria revealed that HeLa-CEACAM1-GFP cells contained numerous

intracellular Eco-P1-Rd (Figure II-7E). However, intracellular bacteria were not

observed in the case of HeLa-CEACAM1-GFP cells infected with Eco-P5-Rd or

Eco-pET28 (Figure II-7E). Taken together, these data demonstrate that OMP P1

of H. influenzae is not only necessary, but also sufficient to target CEACAMs

present on human cells and to trigger bacterial invasion in non-professional

phagocytic cells.

Page 64: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

52

Figure II-7. . OMP P1 expression in E. coli promotes binding to recombinant CEACAMs and invasion into CEACAM-expressing cells. A. Outer membrane proteins (OMPs) were isolated from Eco pET28 or E. coli expressing the ompP1 gene of H. influenzae Rd (Eco P1-Rd) and immunoblotted with anti-P1 antibodies (upper panel). Similar loading of OMPs was verified by Coomassie staining. B. Surface location of P1 in Eco-P1-Rd was confirmed by flow cytometry following staining with -P1 serum. Black line indicates staining of the Eco pET28 control strain. C. GFP-tagged amino-terminal domains of CEACAM1 (hCEA1-N) or CEACAM8 (hCEA8-N) as well as plain cell culture medium were incubated with either Eco P1-Rd or Eco pET28. Flow cytometry analysis was used to determine bacteria-associated GFP fluorescence as a measure of binding to human CEACAMs. The fold change in MFI upon incubation with hCEA1-N compared to hCEA8-N is indicated. D. HeLa GFP and HeLa CEACAM1-GFP were infected with Eco pET28, Eco P5-Rd,

Page 65: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

53

or Eco P1-Rd for 2 hours at MOI 100. The number of viable, internalized bacteria was subsequently determined by gentamicin protection assays. The data represent means ± standard deviation of three independent experiments done in triplicate (**P < 0.01). E. HeLa CEACAM1-GFP cells were infected as in (D). Samples were fixed and extracellular bacteria (red) were stained with polyclonal anti- E. coli LPS antibody in combination with a Cy3-conjugated secondary antibody. After cell permeabilization, both intra- and extracellular bacteria were stained again using polyclonal anti-E. coli lipopolysaccharide antibody in combination with a Cy5-conjugated secondary antibody (cyan). Arrowheads point to extracellular bacteria labelled with Cy3 and Cy5, while arrows indicate internalized bacteria selectively stained with Cy5. Scale bars 10 µM.

Heterologous expression of OMP P1 recapitulates the species-specific

CEACAM binding profile of H. influenzae strains

CEACAM-binding adhesins of several Gram-negative bacteria, including Neisseria

gonorrhoeae, N. meningitidis as well as Moraxella catarrhalis, selectively

associate with human CEACAM1, but not with CEACAM1 orthologues from other

mammals (Voges et al., 2010). The species–specificity of the H. influenzae

CEACAM-binding adhesin has not been evaluated. To first test, if H. influenzae

OMP P1 expressed in E. coli faithfully recapitulates the binding specificity of the

adhesin, we cloned and expressed in E. coli two additional OMP P1 genes derived

from NTHi KN4 or H. influenzae biogroup aegyptius (Eco-P1-KN4 and Eco-P1-

Hae), respectively. Similar to the CEACAM binding spectrum observed for the

parent H. influenzae strains, Eco-P1-KN4 recognized exclusively CEACAM1, Eco-

P1-Rd bound to CEACAM1 or CEA, whereas Eco-P1-Hae associated with human

CEACAM1, CEACAM3, and CEA (Figure II-8A). Most importantly, recombinant

OMP P1 levels in E. coli were similar to endogenous OMP P1 levels in the

respective H. influenzae parent strains (Figure II-8). Combined, these results

demonstrate that heterologous expression of OMP P1 in E. coli preserves the

binding specificity of the adhesin. Next, we used soluble GFP fusion proteins of

the IgV-like amino-terminal domains derived from human, bovine, murine, or

canine CEACAM1 orthologues (Figure II-8C) and employed them in binding

assays with H. influenzae strain Rd. Clearly, H. influenzae selectively bound to

human CEACAM1, but not to any other mammalian CEACAM1 orthologue (Figure

II-8C).

Page 66: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

54

Furthermore, upon expression of OMP P1 in E. coli the identical binding specificity

was observed (Figure II-8D). Together, these results demonstrate that H.

influenzae OMP P1 has exquisite selectivity for human CEACAMs and indicate

that this outer membrane protein co-evolved with the human receptor(s). From

these detailed binding studies and their evolutionary implications, we speculate

that this molecular interaction plays a vital role in the microbe–host relationship.

Figure II-8. E. coli heterologously expressing H. influenzae OMP P1 mimic the species-specific CEACAM-recognition profile of parent H. influenzae strains. A. E. coli expressingOMP P1 of NTHi KN4 (Eco-P1-KN4), Hinf Rd (Eco-P1-Rd), or Hinf-aeg (Eco-P1-Hae) as well as E.coli pET28 (Eco-pET28) were incubated with equivalent levels of the indicated hCEA-N-GFPproteins. Presence of hCEA-N domains was verified by probing the supernatants with anti-GFPantibodies (supe; top panel). After incubation, bacteria-associated CEACAM-N domains weredetected by Western blotting with GFP antibodies (pull-down; bottom panels). B. The E. coli strainsexpressing different OMP P1 proteins from A) were compared side-by-side for OMP P1 expressionwith equal amount (10 µg) of outer membrane proteins derived from the parental H. influenzaestrains. C. The amino-terminal Igv-like domains (N) of human CEACAM8 (hCEA8), humanCEACAM1 (hCEA1), canine CEACAM1 (cCEA1), bovine CEACAM1b (bCEA1b), bovineCEACAM1a (bCEA1a) and murine CEACAM1 (mCEA1), were expressed as secreted GFP-fusionproteins in 293 cells. Cell culture supernatants containing the indicated GFP-fusion proteins wereincubated with H. influenzae Rd. After washing, bacteria-associated CEACAMs were detected byWestern blotting with anti-GFP antibodies (top panel). Equivalent levels of soluble N-terminaldomains in the cell culture supernatants were verified by probing the supernatants with anti-GFPantibodies (supe; bottom panel). D. Binding of Eco-P1-Rd to GFP-tagged amino-terminal domain ofvarious mammalian CEACAM1 of bovine, canine, human or mouse origin was analyzed as in C).

Page 67: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

55

II.4 Discussion

Haemophilus influenzae is a clinically relevant pathogen inhabiting the human

nasopharynx. It has been previously reported that H. influenzae attaches to host

receptors of the CEACAM family via its antigenic variable outer membrane protein

P5 (Hill et al., 2001). In vivo, CEACAM-binding seems to be instrumental for H.

influenzae to successfully colonize model organisms (Bookwalter et al., 2007).

Here, we report that OMP P5 does not support receptor binding and OMP P5-

deficient H. influenzae have only a slightly reduced ability to engage CEACAMs.

Instead, a distinct membrane protein, the so-called OMP P1, is the critical

CEACAM-binding adhesin of H. influenzae. OMP P1 is one of the major surface-

exposed proteins and accounts for ~10% of H. influenzae OMP content (Bolduc et

al., 2000). Different binding assays and functional investigations with typeable and

non-typeable H. influenzae strains, deletion and complementation of OMP P1 as

well as heterologous expression of OMP P1 in E. coli unequivocally demonstrate

that this membrane protein is necessary and sufficient to engage CEACAMs. In

line with the species specificity of H. influenzae, OMP P1 shows high selectivity for

human, but not other mammalian CEACAMs. Furthermore, OMP P1 variants

derived from a diverse set of H. influenzae strains with defined CEACAM-binding

characteristics display an identical CEACAM-binding pattern when expressed in E.

coli strongly arguing that OMP P1 dictates CEACAM recognition by Haemophilus

influenzae.

Clearly, our results are on first sight in conflict with prior data, which suggested

that OMP P5 is the CEACAM-binding adhesin of H. influenzae (Hill et al., 2001).

However, already these investigators noted that deletion of the ompP5 gene only

reduces, but does not abrogate the CEACAM-binding capacity of the pathogens

(Hill et al., 2001). In light of our finding, that OMP P1 is the proper CEACAM-

binding surface protein of Hinf, an alternative explanation for the phenotype of Hinf

OMP P5 mutants is needed. For example, the effect of the ompP5 deletion on

CEACAM binding could be indirect via an influence on OMP P1 expression or

localization. Indeed, examination of the outer membrane profile from an OMP P5-

deficient strain revealed that in the absence of OMP P5 the abundance of OMP P1

is affected. In particular, we found lower levels of OMP P1 in outer membrane

preparations from an OMP P5-deficient strain compared to wildtype H. influenzae.

Page 68: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

56

This observation provides an immediate explanation for the reduced CEACAM-

binding of an OMP P5 mutant, and unifies the prior reports with our novel findings.

The reduced presence of OMP P1 in H. influenzae ∆OMP P5 might be connected

to the known role of OMP P5 homologues from other Gram-negative bacteria,

such as E. coli OmpA, Porphyromonas gingivalis Pgm6, or Pseudmonas

aeruginosa OprF to regulate the stability of the bacterial outer membrane,

presumably via interactions with peptidoglycan (Confer and Ayalew, 2013; Gotoh

et al., 1989a; Gotoh et al., 1989b; Nagano et al., 2005; Rawling et al., 1998;

Wang, 2002).

Strong support to our conclusion that OMP P1 is the CEACAM-binding adhesin

comes from the chance observation of a Hinf isolate, NTHi strain 7502, which

expresses abundant OMP P5, but does not bind to any CEACAM. As it turns out,

NTHi strain 7502 is a natural mutant of OMP P1 harboring a missense mutation in

the OMP P1 reading frame. Interestingly, Swords et al. noted reduced epithelial

cell binding and invasion by this NTHi strain in an independent earlier study

(Swords et al., 2000). Together with the gain-of-function upon OMP P1 expression

in E. coli, OMP P1 is necessary and sufficient to engage human CEACAMs.

Interestingly, H. influenzae OMP P1 shares ~ 39% amino acid identity with the

FadL protein of E. coli, an outer membrane protein involved in the uptake of long

chain fatty acids (LCFA). FadL is characterized by a long -barrel consisting of 14

antiparallel -strands connected by several extended extracellular loops. Despite

the large diameter of the FadL monomer, the crystal structure reveals a ~40

residue N-terminal extension, which folds into a compact, plug-like structure

occupying the inner lumen of the -barrel. Therefore, FadL exists as a closed pore

and LCFA transport seems to require conformational rearrangements in both the

N-terminal plug and the -barrel wall (Hearn et al., 2009; van den Berg, 2005). It is

not known if H. influenzae OMP P1 is also involved in LCFA transport. In previous

studies, OMP P1-deficient strains were analyzed in animal models of invasive Hib

or Hinf-aeg disease, where no differences in the growth and pathogenic potential

compared to wildtype bacteria were observed, when these bacteria were injected

intraperitoneally (Hanson et al., 1989; Munson and Hunt, 1989; Segada et al.,

2000). This indicates that OMP P1 is not essential for bacterial survival and growth

under such in vivo conditions. However, given the fact the OMP P1 can serve as

Page 69: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

57

an adhesin, which selectively binds to human, but not other mammalian

CEACAMs, it is in retrospect not surprising that an OMP P1-deficient strain might

not show a phenotype in animal models of H. influenzae infection.

In other -barrel shaped CEACAM-binding adhesins, such as the colony opacity-

associated (Opa) proteins of pathogenic Neisseriae, several extracellular loops are

highly variable between different isolates (Robertson and Meyer, 1992). Despite

their sequence variation, a short amino acid motif, GxI/V/LxQ, found in HV-2 of

N. meningitidis Opa proteins, appears to be critical for receptor engagement (de

Jonge et al., 2003). Interestingly, this motif can be found in OMP P1 loop 3 of

some H. influenzae strains (see e.g. strain H86; Figure II-6B). However, as only a

fraction of the CEACAM-binding H. influenzae strains harbors such a GxI/V/LxQ

motif, this sequence is clearly not necessary for engaging the host cell receptor.

Therefore, further refinement of the structure-activity relationship of OMP P1 and

ideally an atomic resolution of the ligand-receptor complex might allow

appreciation of this highly species-specific interaction between the bacterial

surface protein and the host receptor. Nevertheless, the unequivocal identification

of the bacterial adhesin and its functional determinants provides a solid ground to

devise strategies to interfere with Haemophilus – host tissue interaction.

II.5 Materials and methods

Haemophilus influenzae strains and growth conditions

The H. influenzae wild type strains and mutants used in this study and their

original sources are listed in Table S1. The four strains KN1–KN4 were originally

isolated from ear effusions of pediatric patients. Selected for their colony

morphology and their requirement of factors X and V, the identity of the strains

was confirmed by 16S rRNA gene sequencing. All H. influenzae strains were

grown on brain-heart infusion agar supplemented with Levinthal base (Hill et al.,

2001). Prior to infection or receptor binding assays, the bacteria were precultured

in brain-heart Levinthal broth until the mid-log phase (OD600 = 1).

Cell culture and generation of stable cell lines

Human embryonic kidney epithelial 293T cells (293 cells; ACC-635, German

collection of microorganisms and cell cultures, DSMZ, Braunschweig, Germany)

Page 70: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

58

were maintained in Dulbecco’s modified Eagle’s medium (DMEM) containing 10%

calf serum. Transfection with pcDNA CEACAM1-4L HA or the empty control vector

(pcDNA) was done by calcium phosphate co-precipitation as described (Muenzner

et al., 2005). HeLa (human cervix) epithelial cells, a kind gift of T.F. Meyer (Max-

Planck Institut für Infektionsbiologie, Berlin, Germany), were stably transduced

with lentiviral particles encoding either CEACAM1 ∆CT-GFP or GFP alone as

previously described (Kuespert et al., 2011). HeLa as well as A549 epithelial cells

(human alveolar lung adenocarcinoma cells; ATCC CCL87) were cultured in

DMEM containing 10% fetal calf serum at 37 °C with 5% CO2 and subcultured

every 2–3 days (HeLa cells) or cultured until confluency was reached and seeded

in new culture vessels 16h prior to infection (A549 cells).

Binding assays with soluble CEACAMs

The production of recombinant GFP-tagged CEACAM proteins by 293 cells and

the subsequent bacterial binding assays were performed as described (Kuespert

and Hauck, 2009; Kuespert et al., 2007). Briefly, soluble recombinant GFP-tagged

CEACAM proteins, contained in cell culture supernatant, were clustered with

polyclonal anti-GFP antibodies (tag-tools GmbH, Konstanz, Germany) overnight at

4 °C. Then, 10 × 108 bacteria were added in 1 ml supernatant containing the

indicated GFP-fusion proteins and incubated for 2 h at 37 °C under gentle rotation.

Thereafter, bacteria were washed twice with PBS and samples were processed

either for Western blotting with GFP antibodies or flow cytometry to detect

bacteria-associated fluorescence.

Expression of outer membrane proteins in E. coli

Chromosomal DNA was used as template to amplify Hinf ompP1 or ompP5. The

respective genes were then cloned into the NcoI and XhoI site of pET28a

(Novagen, Madison, Wisconsin). The pET28a vectors encoding OMP P1 or OMP

P5 were transformed in E. coli Rosetta(DE3) (Novagen, Madison, Wisconsin),

which was induced for protein expression by IPTG for 3 hours. All E. coli strains

were cultured at 37 °C in Luria-Bertani (LB) supplemented with kanamycin.

ompP1 complementation

Chromosomal restoration of ompP1 was performed according to Lichtenegger et

al. (Lichtenegger et al., 2014). The ompP1 gene was linked to a kanamycin

resistance gene cassette via SOEing (Splicing by Overlap Extension) PCRs. After

Page 71: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

59

transformation of P1-deficient Rd strain, the defective ompP1 gene (described in

(Kraiss et al., 1998)) was replaced by a functional one and the resistance to

kanamycin was used as positive selectable marker. Table S2 lists the primers

used.

Gentamicin protection assay

The amount of internalized bacteria after infection of epithelial cells was

determined by gentamicin protection assay (Roth et al., 2013). Concisely, 3 × 105

cells/well were seeded in 24-well plates coated with gelatin and were infected with

bacteria at a multiplicity of infection (MOI) = 100. Two hours later, extracellular

bacteria were killed by 1 h incubation with 100 µg/ml gentamicin in cell culture

medium. Internalized bacteria were freed during cell lysis by saponin treatment

and the viable amongst them were enumerated after plating, on appropriate agar,

the suitable dilutions.

Analysis of bacterial invasion by flow cytometry

Prior to infection, bacteria were labeled with 50 µg/ml fluorescein isothiocyanate

(FITC) in PBS (30 min at 37 °C). Labeled bacteria were used to infect cells (MOI =

100) for 2 h. Cells were processed and analyzed for intracellular bacteria by flow

cytometry as described earlier (Pils et al., 2006).

Scanning electron microscopy

A549 cells were infected with Hinf Rd wildtype strain, Hinf ∆P1-, or Hinf ∆P5

mutants for 2 h. Fixation and processing of the samples for scanning electron

microscopy was described (Pils et al., 2012). The micrographs were pseudo-

colored with Adobe Photoshop CS4 (Adobe Systems Gmbh, München, Germany).

Immunofluorescence staining

HeLa cells, seeded in 24-well plates, were infected for 2 h with different E. coli

strains at an MOI = 100. After their fixation, the samples were differentially stained

for extracellular and intracellular bacteria as described (Roth et al., 2013). As a

result, extracellular bacteria were marked by both Cy3 and Cy5, whereas

intracellular bacteria are only stained with Cy5.

Page 72: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

60

Outer membrane proteins isolation

Proteins of the Hinf outer membrane were isolated as reported previously (Roier et

al., 2012). For E. coli, an identical method was used following IPTG induction.

Immunoblotting and used antibodies

Western blotting was performed as described earlier (Hauck et al., 2001) using a

monoclonal antibody (mAb) against GFP (clone JL-8; Clontech, Palo Alto,

California), mAb against tubulin (clone E7; DSHB, University of Iowa), or using

polyclonal mouse sera against OMP P1 (anti-P1) or against OMP P5 (anti-P5)

(Roier et al., 2014). CEACAM1-4L-HA expression in 293 cells was confirmed with

mouse monoclonal antibody against the HA-tag (clone TT7; tag-tools GmbH,

Konstanz, Germany) and with a mouse mAb against CEACAMs (CEACAM1,

CEACAM3, CEA, and CEACAM6) (clone D14HD11; Aldevron, Freiburg,

Germany).

Statistical analysis

Pairwise comparisons were made using Student’s t-test (SigmaStat software,

Systat Software Inc., San Jose, California). In all cases, a p value < 0.05 was

considered significant.

II.6 Acknowledgements

We are indebted to Stefan Schild and Sandro Roier (Universität Graz, Austria) for

providing us with the anti-P1 and anti-P5 serum. We are thankful to Monika Huff-

Nagel (practicing pediatrician; Konstanz, Germany) for contributing primary

isolates of NTHi (strains KN1 – KN4), to Susanne Feindler-Boeckh for technical

support and to Michael Laumann (EM Service, University of Konstanz, Germany)

for help with scanning electron microscopy. A.K.T. is recipient of a fellowship from

the Deutsche Akademische Austauschdienst (DAAD) and an associate fellow of

the RTG1331. S.L. was supported by the ERA-NET FWF I 662-B09. This work

was supported by DFG grant Ha 2856/6-2 to C.R.H.

Page 73: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

61

II.7 Supporting information

Figure S1. A. Growth curves of Hinf Rd wildtype and its mutants in cell culture medium. The growth of Hinf Rdwildtype strain and its isogenic P5 or P1 mutant was monitored by reading the optical density at600 nm (OD600) in Dulbecco’s modified Eagle’s medium supplemented with 10% fetal calf serum.Data shown are the mean of three independent experiments. B. CEACAM1-GFP or GFP expression in stable HeLa cell lines. HeLa cells were stably transducedto express either GFP alone or CEACAM1-GFP. Whole cell lysates (WCL) of virus-transducedHeLa cells were probed with GFP antibodies to confirm the expression of either GFP orCEACAM1-GFP (upper panel). The similar amount of cell lysate in the different samples wasdemonstrated by Western blotting with monoclonal anti-tubulin antibody (lower panel).

Figure S2. The outer membrane P5 is a heat-modifiable protein. 10 µg of outer membranepreparations of Hinf OMP P5-expressing Escherichia coli (Eco-P5-Rd) or Hinf Rd were boiled afteraddition of sample buffer (95ºC for 10 min) or left at room temperature. The samples, boiled (+) ornot (-), were immunoblotted with anti-P5 mouse serum. The arrow heads point to the ~27 kDa- and~35 kDa-bands characteristic of native (fast migrating) and denatured (slow migrating) OMP P5,respectively. The asterisk indicates a non-specific cross-reactive band from E. coli, presumablyOMPA

Page 74: Identification and characterization of the novel CEACAM

Hinf binds human CEACAMs via P1 to colonize mucosa

62

Hinf strain Source

Rd KW20 A. Wright Tuffs University

2019 3198 7502 9274

M. A. Apicella University of Iowa

KN1 KN2 KN3 KN4

This study

H46 H51 H86

R. Mutters Philipps-Universität Marburg

Hinf b Eagan

M. Herbert John Radcliffe Hospital, Oxford

86-028NP

R. S. Jr. Munson The Ohio State University

Hinf-aegyptius ATCC® 11116™

DSMZ-German Collection of Microorganisms and Cell Cultures

Hinf Rd mutant Description Source

∆P1 ∆P2 ∆P4 ∆P5 ∆Tbpx

ompP1::cat ompP2::cat ompP4::bla ompP5::cat HI1217::bla

(Kraiss et al., 1998) (Andersen et al., 2003) (Reidl and Mekalanos, 1996) (Lichtenegger et al., 2014) (Schlor et al., 2003)

Name Sequence (5’ - 3’) Product

Hinf P5-NcoI-forward ACGCCGCCATGGATGAAAAAAACTGCAATCGC OMP P5

Hinf P5-XhoI-reverse ACGCTGCTCGAGCGCGTTATTTAGTACCGTTTACCGCG

Hinf P1-NcoI-forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC OMP P1

Hinf P1-XhoI-reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG

P1-up-forward TTCCGCGTGGCTTGATCAACA

P1-KanR for Rd ∆P1 P1+

P1-down-Kan-reverse GTGTGCTTTATTATGCTAAATTTTTTCGTGAAGAAGGTGTTGCTGAC

Kan-P1-down-forward GTCAGCAACACCTTCTTCACGAAAAAATTTAGCATAATAAAGCACAC

Kan-down-reverse- TCTTGTGCAATGTAACATCAGAGAACGGATTAGAAACTATAATTTAAGT

Down-Kan-forward ACTTAAATTATAGTTTCTAATCCGTTCTCTGATGTTACATTGCACAAGA

Down-reverse AAAGGCAAGGTATCTATAATTTCG

Table S1. List of Hinf strains and mutants used in this study

Table S2 Primers used in this study

Page 75: Identification and characterization of the novel CEACAM

63

CHAPTER III

The outer membrane protein P1 of

Haemophilus influenzae utilizes its

extracellular loops to grasp human

CEACAM

Arnaud Kengmo Tchoupa1 and Christof R. Hauck1,2

1Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany

2Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz,

Germany

In preparation

Page 76: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

64

III.1 Abstract

Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) are a

group of immunoglobulin-related vertebrate glycoproteins expressed by various

cell types throughout the human body, including epithelial cells. Haemophilus

influenzae (Hinf), a Gram-negative colonizer of the nasopharyngeal mucosal

surface, exploits its outer membrane protein (OMP) P1 in order to intimately bind

CEACAMs present on the apical side of nasopharyngeal epithelial cells. Currently,

the molecular determinants in OMP P1, which are involved in providing the binding

interface to human CEACAMs, are largely unknown. Herein, we report that OMP

P1 is structurally conserved within the Pasteurellaceae family of Hinf. However,

Hinf OMP P1 has evolved unique features, absent in other Pasteurellaceae

species, enabling this adhesin to tightly bind CEACAMs. Moreover, the structural

homolog of Hinf OMP P1 in Escherichia coli (FadL) is unable to recognize

CEACAMs. Chimeric proteins between OMP P1 and FadL as well as deletion

mutants of OMP P1 indicated the involvement of a combination of several surface-

exposed loops of Hinf OMP P1 in CEACAM recognition. Most importantly,

restricting the mobility of OMP P1 extracellular loops abrogated the association of

OMP P1 expressing Hinf and E. coli with CEACAMs. Thus, our data strengthen

Hinf as exclusive CEACAM-binding Pasteurellaceae species and suggest a novel

CEACAM-binding clasp constituted by surface-located loops of OMP P1.

III.2 Introduction

Human mucosal surfaces are constantly exposed to incoming bacteria. To

establish themselves within the resident mucosal microbiota, the incoming bacteria

must withstand both mucosal immunity and mechanical cleansing (St. Geme III,

2002). Interestingly, these microorganisms possess specialized adhesive

structures, so-called adhesins, which recognize eukaryotic cell surface receptors

and thereby mediate the intimate attachment of the bacteria to the host tissues

(Abraham et al., 1998). The relevance of these adhesins is not limited to providing

the bacteria with a strong foothold on the mucosa. Indeed, the exquisite interaction

between the adhesins and the eukaryotic receptors can mediate signaling

cascades to ensure bacterial persistence (Tchoupa et al., 2014).

Page 77: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

65

As a prominent example of convergent evolution, some human-specific

pathogens, such as Neisseria gonorrhoeae, Moraxella catarrhalis or Haemophilus

influenzae (Hinf), have evolved structurally unrelated adhesins in order to target

carcinoembryonic antigen-related cell adhesion molecules (CEACAMs), a

subgroup of the immunoglobulin superfamily glycoproteins expressed by a wide

range of cells, including epithelial cells (Kuespert et al., 2006). CEACAMs are

targeted by an extended “lollipop”-like trimeric autotransporter adhesin in

Moraxella catarrhalis: the ubiquitous surface protein (Usp) A1 (Hill and Virji, 2003).

In contrast, Neisseria gonorrhoeae (Ngo) and Neisseria meningitidis (Nme) bind to

human CEACAM receptors via 8-stranded β-barrel proteins with four extracellular

loops: the “opacity-associated” (Opa) proteins (Kuespert et al., 2011; Roth et al.,

2013).

The pivotal role of the Opa-mediated CEACAM-binding for successful colonization

has been documented by two outstanding studies with mice transgenic for human

CEACAM1 or human CEA, respectively (Johswich et al., 2013; Muenzner et al.,

2010). Muenzner and colleagues nicely demonstrated that the enhanced capability

of N. gonorrhoeae to colonize the urogenital tract of mice expressing human CEA

versus wild-type mice depended on the presence of the Opa adhesin on the side

of the microbes (Muenzner et al., 2010). Similarly, all the meningococci, that

persisted within the nasopharyngeal mucosa of mice transgenic for human

CEACAM1, expressed CEACAM1-specific Opa proteins (Johswich et al., 2013).

Moreover, the genetic depletion of all four opa genes in the meningococcal strain

H44/76 precluded this bacterium from colonizing the human CEACAM1-transgenic

mice (Johswich et al., 2013).

Hinf, a human-specific inhabitant of the respiratory tract, seems to rely, at least

partly, on CEACAM-binding in order to efficiently colonize the nasopharynx in

animal models (Bookwalter et al., 2007). However, that study failed to provide

unambiguous evidence that Hinf adhesins directly interact with CEACAMs present

in the nasopharynx of chinchilla (Bookwalter et al., 2007). Recently, we uncovered

the necessity and the sufficiency of the outer membrane protein (OMP) P1 in

CEACAM-binding by Haemophilus influenzae (Tchoupa et al., 2015). OMP P1 was

predicted to build a β-barrel structure with 14 strands and 7 surface-exposed

loops. The most prominent loops of OMP P1 corresponded to the highly variable

Page 78: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

66

regions of this protein, as revealed by multiple sequence alignment and protein

modeling (Tchoupa et al., 2015). However, a detailed understanding of the

contribution of these extracellular loops to the interaction between OMP P1 and

CEACAM is lacking.

In this study, we report that, irrespective of the conserved structure of OMP P1

within the family Pasteurellaceae, the targeting of human CEACAM is restricted to

the species Haemophilus influenzae. Detailed investigations on structure-activity

relationships with OMP P1 mutants demonstrate that several flexible extracellular

loops allow OMP P1 to engage human CEACAMs. Furthermore, compromising

the mobility of OMP P1 extracellular loops by chemical crosslinking disrupted the

interaction of Haemophilus influenzae or OMP P1 expressing E. coli with

CEACAMs. Our findings provide the first insights on CEACAM-binding

determinants of OMP P1 and suggest a novel clamp mode of association between

the flexible, surface-exposed loops of the Hinf adhesin and host receptors of the

CEACAM family.

III.3 Results

CEACAM-binding is restricted to OMP P1 of Hinf within the Pasteurellaceae

family

Hinf OMP P1 targets members of the human CEACAM family (Tchoupa et al.,

2015). Several OMP P1 variants derived from Hinf strains have been analyzed in

detail, with OMP P1 derived from Hinf biogroup aegyptius (Hinf-aeg) (strain ATCC

11116) showing the most promiscuous binding to multiple CEACAMs, namely,

CEACAM1, CEA and CEACAM3 (Tchoupa et al., 2015). We wondered whether

OMP P1 and/or OMP P1-like proteins with potential CEACAM-binding properties

were present in related Gram-negative bacteria. Accordingly, the blastp tool was

used, under standard settings, to search for homologues / orthologues of Hinf-aeg

OMP P1 (P1-Hinf-aeg). Obviously, the best hits were from OMP P1 of other Hinf

strains (data not shown). However, several other members of the Pasteurellaceae

family exhibited OMP P1-like proteins highly similar to P1-Hinf-aeg, with OMP P1

of Haemophilus haemolyticus M21639 reaching ~75% amino acid identity (data

not shown). Therefore, we predicted a conserved structure of OMP P1 in

Page 79: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

67

Pasteurellaceae. To confirm this assumption, we took advantage of the known

crystal structure of FadL, the long-chain fatty acid transporter (van den Berg et al.,

2004) and OMP P1 homolog in Escherichia coli. The crystal structure of FadL was

used as a scaffold to align amino acid sequences from OMP P1 of Haemophilus

haemolyticus (P1-Hhae), H. parainfluenzae (P1-Hpara), H. pittmaniae (P1-Hpitt),

Mannheimia heamolytica (P1-Mhae) and P1-Hinf-aeg (Figure III-1A). Of note, this

alignment was performed with P1 sequences lacking signal peptides, based on

mature P1 sequence published elsewhere (Munson and Grass, 1988). Obviously,

apart from the loops 1, 3, 4 and 7, which are highly variable within the Hinf species

with respect to the amino acid composition and length (Tchoupa et al., 2015),

Pasteurellaceae OMP P1 exhibited a striking conservation (Figure III-1A). This

conservation was supported by sequence similarity ranging from ~60% for P1-

Mhae to 74% for P1-Hhae, compared to P1-Hinf-aeg (Figure III-1B). Importantly,

the phylogenetic tree deduced from the multiple amino acid alignment of P1 and

FadL sequences revealed that P1-Mhae (the P1 protein with the lowest similarity

to P1-Hinf-aeg) rather clustered with FadL, whereas the other P1 proteins (P1-

Hinf-aeg, P1-Hhae, P1-Hpara and P1-Hpitt) clustered together (Figure III-1C).

Page 80: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

68

Figure III-1. The structure of OMP P1 is highly conserved within the Pasteurellaceae family. A. OMP P1 amino acid sequences of Hinf-aeg (P1-Hinf-aeg), Haemophilus haemolyticus (P1-Hhae), H. parainfluenzae (P1-Hpara), H. pittmaniae (P1-Hpitt) and Mannheimia heamolytica (P1-Mhae) were aligned using the solved crystal structure of E. coli OMP FadL (FadL-Eco) and the program Expresso within T-Coffee web server. Dashes (-) are inserted to maximize the alignment. The colors indicate the accuracy of the alignment from indigo (<20%) to green (20 – 30%), yellow (30 – 40%) to light orange (40 – 60%) and dark orange (60 – 80 %) to red (>80%). Surface-exposed loops (loop 1 — 7) are delineated by black arrows above the sequences. B. The percent identity or similarity of the sequences described in (A) with P1-Hinf-aeg. C. The phylogenetic tree was constructed from the Clustal Omega alignment of the sequences described in (A) and edited using the Interactive Tree Of Life (iTOL) web tool. The branch length represents the mean number of amino acid substitution per site.

Page 81: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

69

Urged by the similarity of their OMP P1 sequences with P1 Hinf-aeg, we tested a

couple of Pasteurellaceae family members for CEACAM-binding. GFP-tagged

amino-terminal domains of human CEACAM1 (hCEA1-N) or CEACAM8 (hCEA8-

N, negative control) were used in bacterial binding assays (Figure III-2A). Clearly,

no interaction with hCEA1-N could be detected for H. parainfluenzae, H.

haemolyticus or M. haemolytica (Figure III-2B). In clear contrast, Hinf-aeg

exhibited a strong interaction with hCEA1-N (Figure III-2B). Consistent with the

failure of the parent strain to target CEACAMs, E. coli ectopically expressing the

OMP P1 of H. parainfluenzae was not able to bind soluble CEACAMs nor it could

adhere to CEACAM-expressing epithelial cells (data not shown). Combined, these

data strongly suggest that P1 proteins of Hinf species have evolved distinct

features, absent in P1 from closely related species, enabling them to specifically

target human CEACAMs.

Figure III-2. CEACAM-binding properties are restricted to Haemophilus influenzae in thefamily Pasteurellaceae. A. Similar amounts of GFP-fused amino-terminal domain of humanCEACAM1 (hCEA1-N) or human CEACAM8 (hCE8-N) were harvested as cell culture supernatants(supe) of 293 cells and probed with anti-GFP antibodies B. hCEA1-N or hCEA8-N produced asdescribed in (A) was used in bacterial binding assays with H. parainfluenzae (upper left panel), H.haemolyticus (upper right panel), M. heamolytica (lower left panel) and Hinf-aeg (lower right panel).Bacteria incubated in plain cell culture medium were also included as negative controls (blacklines). CEACAM-binding was analyzed by flow cytometry.

Page 82: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

70

Extracellular loops located at either end of OMP P1 are necessary for

CEACAM-binding

To understand the structural basis of OMP P1 CEACAM-binding, we modelled the

three-dimensional architecture of P1-Hinf-aeg according to the known crystal

structure of FadL (Figure III-3A). Overall, the 3-dimensional orientation of OMP P1

and FadL appeared to be highly similar (Figure III-3A). However the most

prominent extracellular loops of P1 (L1, L3, L4 and L7) were divergent (Figure

III-3B). Interestingly, non-CEACAM binding E. coli overexpressing FadL remains

unable to interact with CEACAMs (data not shown). Conversely, the heterologous

expression of P1-Hinf-aeg in E. coli (Eco P1-Hinf-aeg) confers CEACAM-binding

capabilities reminiscent of the parent Hinf-aeg strain (Tchoupa et al., 2015). These

strong discrepancies between CEACAM recognition by FadL or OMP P1 make

FadL suitable for gain-of-function approaches. Accordingly, to rapidly narrow down

the OMP P1 regions involved in CEACAM binding, we generated chimeras

between OMP P1 and FadL by either replacing the first 4 (P1-FadL) or the last 3

loops of FadL (FadL-P1), respectively, by the corresponding sequences from P1-

Hinf-aeg (Figure III-3C). P1-FadL or FadL-P1 where overexpressed in non-

CEACAM binding E. coli (Eco P1-FadL or Eco FadL-P1) and the bacteria were

subsequently incubated with equivalent amounts of GFP-tagged N-terminal

domain of human CEA (hCEA-N) or hCEA8-N (Figure III-3D). After washing, no

interaction with hCEA-N was detected for Eco P1-FadL or Eco FadL-P1 (Figure

III-3E). However, Eco P1-Hinf-aeg strongly bound hCEA-N (Figure III-3E).

Together, these results clearly indicate that the CEACAM-binding determinants of

OMP P1 are located on either moiety of the protein.

Page 83: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

71

Figure III-3. A unique loop is not responsible of the targeting of CEACAMs by OMP P1. A. The solved structure of FadL (left panel) was retrieved from the database RCSB PDB (http://www.rcsb.org/pdb/home/home.do) and used to predict the three-dimensional architecture of Hinf-aeg OMP P1 (right panel). B. In the model of OMP P1, described in (A), highly variable portions of P1 are highlighted in red C. Schematic representations of FadL, P1 and the chimeras FadL-P1 and P1-FadL. Predicted extracellular loops are colored in black for FadL and in white for P1. D. Equivalent amounts of GFP-tagged N-terminal domain of human CEA (hCEA-N) or human

Page 84: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

72

CEACAM8 (hCE8-N) in cell culture supernatants (supe) were verified by immunoblotting with anti-GFP antibodies. E. After overexpression of the indicated proteins in E. coli, the binding to hCEA-N or hCEA8-N was investigated by flow cytometry. The black lines indicate bacteria incubated with plain cell culture medium.

The highly variable loops of OMP P1 are instrumental for CEACAM-

recognition

The chimeras between OMP P1 and FadL have strongly suggested that a single

extracellular loop of OMP P1 may not be sufficient and that loops located at either

end of OMP P1 may be necessary for CEACAM binding. To determine individually,

which loop contributes to the binding interaction, we replaced single loops of OMP

P1 by a short antibody epitope PEYFK (Figure III-4A). Accordingly, we generated

seven OMP P1 mutants lacking distinct extracellular loops (OMP P1-∆L1 – OMP

P1-∆L7) and expressed them individually in E. coli. Similar to the wild-type OMP

P1, all the 7 mutants were detected in outer membrane preparations of E. coli

(Figure III-4B). CEACAM-binding assays revealed that, with the exception of

buried loops L2 and L5, all other loops of OMP P1 contributed to the interaction

with CEACAM (Figure III-4C).

Strikingly, the most prominent extracellular loop of P1-Hinf-aeg (L3) exhibited a

stretch, PEPQK, reminiscent of the antibody epitope PEYFK. Given the possible

practical advantages granted by the insertion of such a tag in a functional protein,

we substituted the proline-214 and the glutamine-215 residues of P1-Hinf-aeg by

tyrosine and phenylalanine, respectively. The mutant generated (P1-PQ-214-215-

YF) was expressed in E. coli outer membrane (Figure III-4D). Much to our

surprise, these double mutations, not predicted by protein modeling to change the

molecular architecture of P1-Hinf-aeg (data not shown), severely impaired its

binding to hCEA-N (Figure III-4E). Taken together, our data suggest that many

extracellular loops of OMP P1 are involved in the interaction with CEACAMs.

Page 85: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

73

Figure III-4.The prominent extracellular loops of OMP P1 are necessary for CEACAM-binding. A. OMP P1 and the seven mutants carrying a specific loop deletion are schematically indicated. B. E. coli transformed with the empty pET28 vector (pET28) or pET28 carrying either OMP P1 (P1) or one of the 7 variants with loop substitutions (P1 ∆L1 — P1 ∆L7) were induced with IPTG. Their outer membrane proteins were subsequently immunoblotted with anti-P1 serum (upper panel). Similar amounts of protein in the different samples were verified by Coomassie staining of the membrane (lower panel). C. The ability of the wild-type P1 and its seven mutants to bind CEACAMs was estimated in pull-down assays with GFP-tagged soluble proteins. No binding is represented by “─”, weak binding by “+”, strong binding by “++”, and a very good by “+++”. D. OMP preparations of E. coli transformed with the empty pET28 vector (Eco pET28) and E. coli expressing P1 of Hinf-aeg (Eco P1-Hinf-aeg) or P1-Hinf-aeg mutant PQ-214-215-YF (Eco P1- PQ-214-215-YF) were probed with anti-P1 serum (upper panel). The membrane was also Coomassie stained to verify the equivalent amounts of protein in the different samples (lower panel). E. Flow cytometry was used to analyze the binding of Eco P1-Hinf-aeg or Eco P1- PQ-214-215-YF to the indicated CEACAMs. The fold change in MFI after hCEA-N–incubation compared to hCEA8-N is indicated for each sample.

Page 86: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

74

Restriction of loop flexibility in OMP P1 abrogates CEACAM-binding

The structure of Neisseria gonorrhoeae Opa60 revealed that the loops of this OMP

are extraordinary flexible (Fox et al., 2014). In agreement with the assumptions of

Fox and colleagues, we speculated that extracellular loops of β-barreled

CEACAM-binding protein, including OpaCEA and OMP P1, function like a

mechanical clamp to tightly encircle their receptor from multiple angles. To

rigorously test this hypothesis, we took advantage of a 1.14 nm-wide crosslinking

agent bis-(sulfosuccinimidyl)-suberate (BS3) to constrain loop flexibility of OpaCEA

or OMP P1. Alternatively, the bacteria were also incubated with succinimidyl-2-

(biotinamido)ethyl-1,3-dithiopropionate (NHS-SS-Biotin). NHS-SS-Biotin, as well

as BS3, likely formed amide bonds with the primary amino groups (-NH2) of

multiple lysine residues present on loop 1, 3, 4, 5 and 7 of P1-Hinf-aeg or loops 1,

2 and 3 of OpaCEA. Interestingly, biotinylated OpaCEA-expressing Neisseria

gonorrhoea or Hinf-aeg was still able to bind GFP-tagged N-terminal domain of

human CEACAM3 (hCEA3-N) but not as good as the untreated bacteria (Figure

III-5A and B). Conversely, pre-treatment with BS3 completely blocked the

recognition of hCEA3-N by OpaCEA or P1-Hinf-aeg (Figure III-5A and B). Similar

amounts of hCEA3-N and hCEA8-N, used in the binding assays, were verified by

Western blotting (Figure III-5C).

Importantly, BS3 treatment did not abrogate CEACAM-binding of Moraxella

catarrhalis (Figure III-5D), a pathogen, which employs a linear amino acid motif in

the stalk of its UspA1 protein for receptor engagement (Hill et al., 2005).

Furthermore, BS3 consistently blocked the interaction between P1-Hinf-aeg and

CEACAM in the E. coli background (Figure III-5E); whereas, the uropathogenic E.

coli AfaE-III, targeting CEACAM via the multimeric immunoglobulin-like protein

AfaE-III (Anderson et al., 2004), still bound hCEA3-N after treatment with BS3

(Figure III-5E). Collectively, these results demonstrate that OMP P1 binding is

sensitive to agents compromising the flexibility of extracellular loops and suggest a

novel clamp mode of interaction between the flexible, extracellular loops of the

Hinf adhesin and host surface receptors of the CEACAM family.

Page 87: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

75

Figure III-5. Loop flexibility in OMP P1 is essential to CEACAM recognition. A. OpaCEA expressing Neisseria gonorrhoeae (Ngo OpaCEA) was left untreated (left panel) or incubated with 2 mM of NHS-SS-Biotin (middle panel) or crosslinker (BS3) (right panel) prior binding assays with the indicated soluble GFP-fused N-terminal domains of human CEACAM3 (hCEA3-N) or CEACAM8 (hCEA8-N). Bacterium-associated fluorescence, as measured by flow cytometry, was used as readout to quantify the interaction of the bacteria with CEACAMs. The fold change in bacterial MFI upon incubation with hCEA3-N in comparison to hCEA8-N is indicated for each sample. The black lines indicate bacteria incubated with plain cell culture medium. B. Similar assays as described in (A) were performed with Hinf-aeg. C. Comparable levels of hCEA3-N or hCEA8-N, in cell culture supernatants (supe) used in A and B, were verified by immunoblotting with monoclonal anti-GFP antibody. D. Cell culture supernatants, containing similar amounts of GFP alone or CEACAM3 N-terminal domain (hCEA3-N) fused to GFP were incubated with bacteria pretreated or not with BS3. The CEACAM-binding of Hinf-aeg (top panel) or Moraxella catarrhalis (bottom panel) was detected with anti-GFP antibodies. E. Eco P1-Hinf-aeg (top panel) or Eco AfaE-III (bottom panel) was treated or not with BS3 prior binding assays with hCEA1-N or hCEA8-N. GFP signals characteristic of CEACAM-binding were detected with anti-GFP antibodies.

Page 88: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

76

Modelling of the OMP P1-CEA complex

Fifteen years ago, Virji and colleagues, in a seminal study, have characterized one

of the two β-sheets of CEACAM1 N-terminal domain (the CFG-face) as the

primary target of CEACAM-binding Hinf (Virji et al., 2000). Furthermore, mutational

analyses identified amino acid residues Y-34 and I-91 as necessary for the

interaction of most Hinf strains with CEACAM1 (Virji et al., 2000). On the other

hand, our current findings underline the importance of P-214 and Q-215 residues

in Hinf OMP P1-recognition of CEACAMs (Figure III-4E). Therefore, to get an

impression of the bimolecular complex P1-Hinf-aeg – CEACAM1-N, the webserver

SwarmDock (http://bmm.cancerresearchuk.org/~SwarmDock/) was used. The

residues P-214 and Q-215 or Y-34 and I-91 were set as active sites in P1-Hinf-aeg

or CEACAM1-N, respectively. The predicted three-dimensional atomic architecture

of the bimolecular complex P1-Hinf-aeg – CEACAM1-N (Figure III-6) makes highly

plausible that many flexible loops of OMP P1 are engaged in the interaction with

CEACAM1-N and warrants further studies to co-crystalize OMP P1 and

CEACAM1-N.

Figure III-6. A putative model of the bimolecular complex CEACAM1-N – P1-Hinf-aeg.The interaction between the amino-terminal domain of human CEACAM1 (red) and the outermembrane protein P1 of Hinf-aeg (blue) was modeled using the docking server SwarmDock.

Page 89: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

77

III.4 Discussion

An unbiased study has identified the outer membrane protein (OMP) P1 as the

CEACAM-binding adhesin of Hinf (Tchoupa et al., 2015). OMP P1 is predicted to

build a 14-stranded -barrel with 7 extracellular loops. The most prominent

surface-exposed loops of OMP P1 (L1, L3, L4 and L7) were also the most variable

amongst Hinf strains. Although this high versatility of their CEACAM-binding

adhesin, all the OMP P1-proficient Hinf strains analyzed to date were able to

interact with CEACAMs (Tchoupa et al., 2015). Therefore, we hypothesized that

OMP P1 might have a conserved function in all the members of the

Pasteurellaceae family.

In this study, by aligning the sequences of OMP P1 from several Pasteurellaceae

family members against the crystal structure of E. coli FadL, we have

demonstrated that the OMP P1 is structurally conserved in Pasteurellaceae.

However, except Hinf, no additional Pasteurellaceae species was found to bind

several human CEACAMs tested. Even though the unique features that confer a

CEACAM-binding to Hinf OMP P1 are still to be identified, structure-activity

investigations undertaken here highlight the relevance of several flexible loops for

the recognition of CEACAM by OMP P1.

Neisseria meningitidis is another inhabitant of the human nasopharynx interacting

with CEACAMs via the opacity-associated (Opa) proteins (Kuespert et al., 2011).

Within the Neisseria genus, the CEACAM-binding capability is not restricted to

species with a high pathogenic potential, such as N. meningitidis and N.

gonorrhoeae, but also shared by strictly commensal species, including N.

lactamica and N. subflava (Toleman et al., 2001). Contrastingly, Haemophilus

influenzae is the unique species of the genus Haemophilus that is able to interact

with human CEACAMs. Indeed, our current data reveal that less pathogenic

Haemophilus species (H. parinfluenzae and H. haemolyticus) are unable to bind

CEACAMs. Therefore, it is enticing to think that the recognition of CEACAMs by

Hinf may provide the bacterium with a selective advantage to outcompete other

Haemophilus species of the same ecological niche. Further studies with Hinf OMP

P1 as well as P1 of commensal species of Haemophilus, all expressed in a

heterologous background (e.g.: Escherichia coli), would elucidate in suitable

Page 90: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

78

animal models the relevance of P1 – CEACAM interaction for a successful

colonization.

An additional human-restricted colonizer of the respiratory tract, which tightly

interacts with CEACAMs, is Moraxella catharralis (Hill and Virji, 2003). Moraxella

catharralis utilizes a stretch of 20 amino acids, within the stalk of its UspA1

adhesin, to bind CEACAMs with a high affinity (Conners et al., 2008). Consistently,

Moraxella catharralis strains deprived of this CEACAM-binding motif in their

UspA1 proteins (e.g.: strain O35E) are unable to interact with CEACAMs (Brooks

et al., 2008). However, such a linear motif is so far unknown in Hinf OMP P1. The

only non-CEACAM binding Hinf strain we have identified so far (strain 7502) is

totally deficient in OMP P1 (Tchoupa et al., 2015). Furthermore, our previous work

demonstrated that Hinf strains targeting one, two or three members of the

CEACAM family exhibited highly variable extracellular loops irrespective of their

CEACAM-binding profiles (Tchoupa et al., 2015).

Non-CEACAM binding OMP P1 of H. parainfluenzae (P1-Hpara) differs

significantly from P1-Hinf-aeg within the most prominent loops (L1, L3, L4 and L7).

Importantly, the deletion of each of these loops disrupts the interaction of P1-Hinf-

aeg with CEACAMs. The necessity of multiple loops to target the same receptor

may be puzzling at the first sight; however, other -barrel shaped CEACAM-

binding adhesins, such as the neisserial Opa proteins, combined the hypervariable

regions of their loops 2 and 3 (HV-1 and HV-2, respectively) to build the CEACAM-

binding interface (de Jonge et al., 2003). Intriguingly, the combination HV-1 — HV-

2 is so specific that a chimeric protein, made of HV-1 and HV-2 originated from

two different CEACAM-binding Opa proteins, is unable to interact with CEACAMs

(de Jonge et al., 2003). In a similar fashion, several variable loops of P1-Hinf-aeg

may cooperate in order to bind CEACAMs. Nevertheless, it is plausible that each

loop does not contribute to a similar extend to the CEACAM-binding. The loss of

the CEACAM-recognition after the substitution of the residues Pro-214 and Gln-

215 in the loop 3 of P1-Hinf-aeg supports this hypothesis. Furthermore, some of

the variations in the extracellular loops appears to be under positive selection

(Mes and van Putten, 2007). Especially in OMP P1 loop 1 and loop 3, both of

which are required for CEACAM binding, particular amino acids seem to be

Page 91: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

79

preferred (Mes and van Putten, 2007), which could be due to their involvement in

receptor recognition.

Considering their size, 5 extracellular loops of Hinf OMP P1 (L2, L3, L4, L5 and

L6) are reminiscent of E. coli homolog FadL loops (van den Berg et al., 2004)

whereas two loops (L1 and L7) are relatively huge in P1. Since the flexibility of a β-

barrel outer membrane protein’s loop is often linked to its length (Fox et al., 2014),

the diverse conformations sampled by the prominent loops 1 and 7 may be

instrumental in stabilizing the complex OMP P1-CEACAM. Accordingly, chimeras

lacking L1 or L7 of P1, or P1 mutants depleted in L1 or L7 are unable to target

CEACAMs. More than the (variable) sequences of the huge loops of P1, the

various conformations conferred by the loop flexibility may promote the recognition

of the CEACAM receptor, as it was speculated for Opa proteins (Fox et al., 2014).

By restricting the flexibility of OpaCEA and P1-Hinf-aeg with chemical cross-linkers,

we abrogated their interaction with CEACAMs. Interestingly, biotinylated OpaCEA

and P1-Hinf-aeg exhibited a reduced interaction with CEACAMs, probably due to

the steric hindrance generated by the 2.34 nm-spacer arm of the biotin used.

Haemophilus influenzae constantly faces the challenge of (i) varying the surface-

exposed regions of its outer membrane protein P1 to escape the immune

response of its human host while (ii) preserving the advantage inherent in the

interaction of P1 with its human receptor CEACAM. This constraint may build the

framework within which the amino acid sequences of OMP P1 extracellular loops

are evolving. Therefore, co-crystal structures of Hinf OMP P1 variants in

bimolecular complexes with an identical CEACAM family member may represent

the turning point in the development of innovative, adhesion disrupting, therapeutic

strategies to combat Haemophilus influenzae.

III.5 Materials and methods

Multiple sequence alignment

OMP P1 sequences of Hinf-aeg ATCC 11116, Haemophilus parainfluenzae ATCC

33392, H. haemolyticus M19501, H. pittmaniae HK85, Mannheimia heamolytica

PKL10, as well as the FadL sequence of E. coli K12 were retrieved from Uniprot

(http://www.uniprot.org/). The PDB file of the crystal structure of E. coli FadL was

Page 92: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

80

downloaded from the protein data bank RCSB (http://www.rcsb.org/pdb/home

/home.do). The different P1 and FadL sequences were aligned using the Expresso

application of the T-Coffee web server (http://tcoffee.crg.cat/apps/tcoffee/do:

expresso) and the solved structure of FadL. The phylogenic tree with the different

P1 or FadL OMPs was edited with iTOL (http://itol.embl.de/).

Bacterial strains and growth conditions

Hinf-aeg ATCC 11116, H. parainfluenzae ATCC 33392, Mannheimia haemolytica

ATCC 33396 and Moraxella catarrhalis ATCC 25238 were obtained from German

Collection of Microorganisms and Cell Cultures (DSMZ, Braunschweig, Germany).

H. haemolyticus NCTC 10659 was purchased from PHE (Public Health England,

Salisbury, UK). OpaCEA-expressing, non-piliated Neisseria gonorrhoeae MS11-

B2.1 (strain N309) is a generous gift of Prof. Thomas F. Meyer (Max-Planck

Institut für Infektionsbiologie, Berlin, Germany). Escherichia coli AfaE-III is the

courtesy of Prof. Ulrich Dobrindt (University of Münster, Münster, Germany). With

the exception of Neisseria gonorrhoeae and Escherichia coli AfaE-III, all the

bacteria were grown on brain-heart infusion agar supplemented with 10%

Levinthal base. Neisseria gonorrhoeae was grown on GC agar plates

supplemented with vitamins while Escherichia coli AfaE-III grew on Luria-Bertani

(LB) agar plates. All the bacteria were grown at 37°C in 5% CO2.

Outer membrane proteins isolation

After IPTG induction, proteins of the E. coli outer membrane were isolated as

described elsewhere (Roier et al., 2012).

Expression of outer membrane proteins in E. coli.

Chromosomal DNA of Hinf-aeg or E. coli K12 was used as template to amplify

ompP1 or fadL, respectively. The respective genes were then cloned into the NcoI

and XhoI site of pET28a (Novagen). E. coli Rosetta (DE3) (Novagen) was

subsequently transformed with the pET28a vectors encoding FadL or OMP P1,

and induced for protein expression by IPTG for 3 hours. LB medium supplemented

with kanamycin was used to grow all the transformed E. coli strains at 37°C.

ompP1 mutagenesis

OMP P1 chimeras and mutants were generated via SOEing PCRs (Horton et al.,

1990). For the chimeras, the primers were designed to fuse in-frame the N-

Page 93: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

81

terminal moiety of P1 with the C-terminal moiety of FadL (P1-FadL) or inversely

(FadL-P1). In loop-depleted P1 mutants, the FIVE-epitope tag (PEYFK) coding

sequence was used as overlap extension to replace every single loop. In the P1-

PQ-214-215-YF mutant, SOEing PCRs were also used to replace the Pro-214 and

Gln-215 residues in P1-Hinf-aeg by Tyr and Phe, respectively. The Table 2 lists

the primers used.

Name Sequence (5’ - 3’) Product

P1-NcoI_forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC

P1-FadL P1-FadL-reverse CATTTCAGGCAGGTTCAGCGTCAGATTACCTTTTTTACCTTCTTTGATG P1-FadL-forward CTGACGCTGAACCTGCCTGAAATG FadL- XhoI-reverse GCGCGGCTCGAGTCAGAACGCGTAGTTAAAGTTAGTACC FadL- NcoI-forward GGACGCCATGGATGAGCCAGAAAACCCTGTTTAC

FadL-P1 FadL-P1-reverse

GTAATCTGGCAATTTAAGGGTTAAATAACCCGATTGCGTTGCGCCACCTGTC

FadL-P1-forward TTAACCCTTAAATTGCCAGATTAC P1-XhoI_reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG P1-NcoI_forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC

P1 ∆Loop1

P1 ∆Loop1-reverse GCCCTCTATCTTGAAGTACTCGGGTACATCACCATTCATATTAATTC P1 ∆Loop1-forward CCCGAGTACTTCAAGATAGAGGGCGGCTCAGCTTC P1-XhoI_reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG P1-NcoI_forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC

P1 ∆Loop2

P1 ∆Loop2-reverse CAAGTCAGTTTTGAAGTACTCGGGTTCACTTTTTAGACCGAAATTGACATTCATTCC

P1 ∆Loop2-forward CCCGAGTACTTCAAAACTGACTTGAGTGCTATC P1-XhoI_reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG P1-NcoI_forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC

P1 ∆Loop3

P1 ∆Loop3-reverse GGGCAAATACTTGAAGTACTCGGGAACACTATCCGCAATAATACC P1 ∆Loop3-forward CCCGAGTACTTCAAGTATTTGCCCTCTAAAGACACATCTG P1-XhoI_reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG P1-NcoI_forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC

P1 ∆Loop4

P1 ∆Loop4-reverse ACCTTTTTTGAAGTACTCGGGGGCAGTGCGGTCAGTAAAATCAATGTCC P1 ∆Loop4-forward CCCGAGTACTTCAAAAAAGGTGATTTAACCCTTACATTGCC P1-XhoI_reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG P1-NcoI_forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC

P1 ∆Loop5

P1 ∆Loop5-reverse TTCTTTATCCTTGAAGTACTCGGGGCTGGCATGTAATTTTGTTAAACG P1 ∆Loop5-forward CCCGAGTACTTCAAGGATAAAGAATTGCAATACAATAATAAC P1-XhoI_reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG P1-NcoI_forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC

P1 ∆Loop6

P1 ∆Loop6-reverse GGTATCTGGCTTGAAGTACTCGGGTTGATCGTAAGCAATACCCGCGCGTAAGG

P1 ∆Loop6-forward CCCGAGTACTTCAAGCCAGATACCGATCGCACTTGGTATAG P1-XhoI_reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG P1-NcoI_forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC

P1 ∆Loop7

P1 ∆Loop7-reverse TGCATTTGTCTTGAAGTACTCGGGTACTTCTTTAAAGTGAACTTTTTTGCC

P1 ∆Loop7-forward CCCGAGTACTTCAAGACAAATGCAAATTATACTTCTCAAGCACATGCAAATC

P1-XhoI_reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG P1-NcoI_forward GCGCCGCCATGGATGAAAAAATTTAATCAATCTC

P1-PQ-214-215-

YF

P1-PQ-YF-reverse ATACTCAGCAATTGTTTTGAAGTATTCTGGCAGTACTGTGGATAC P1-PQ-YF-forward TACTTCAAAACAATTGCTGAGTATTTGAC P1-XhoI_reverse GCGCGGCTCGAGCGCGTTAGAAACTATAATTTAAGTTTAAACCGTAAAG

Table 2. Primers used in this study

Page 94: Identification and characterization of the novel CEACAM

Multiple P1 loops form the CEACAM-binding interface

82

Binding assays with soluble CEACAMs

Soluble GFP-tagged CEACAM ectodomains were produced by 293 cells and

subsequently used in bacterial binding assays as described elsewhere (Kuespert

and Hauck, 2009; Kuespert et al., 2007). Shortly, soluble CEACAM-N-GFP

proteins, contained in cell culture supernatant, were clustered overnight with

polyclonal anti-GFP antibodies at 4°C. Afterwards, 10 × 108 bacteria were added

in 1 ml supernatant containing the indicated GFP-fusion proteins and incubated for

2 h at 37 °C under gentle rotation. After extensive washing with PBS, the samples

were either immunoblotted with a monoclonal GFP antibody or processed for flow

cytometry to detect bacteria-associated fluorescence. In some cases, prior to

bacterial pull-down with CEACAMs, the bacteria were treated with either 2 mM

bis(sulfosuccinimidyl) suberate (BS3) (Thermo Scientific) or succinimidyl-2-

(biotinamido)ethyl-1,3-dithiopropionate (NHS-SS-Biotin) (Thermo Scientific)

according to the manufacturer’s instructions.

Western blotting and used antibodies

Immunoblotting was performed as reported earlier (Hauck et al., 2001) using a

monoclonal antibody (mAb) against GFP (clone JL-8; Clonetech) or polyclonal

mouse sera against OMP P1 (anti-P1) (Roier et al., 2014).

Page 95: Identification and characterization of the novel CEACAM

83

CHAPTER IV

The Haemophilus influenzae adhesin

OMP P1 is regulated by host-derived

fatty acids

Arnaud Kengmo Tchoupa1 and Christof R. Hauck1,2

1Lehrstuhl für Zellbiologie, Universität Konstanz, Konstanz, Germany, and 2Konstanz Research School Chemical Biology, Universität Konstanz, Konstanz,

Germany

In preparation.

Page 96: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

84

IV.1 Abstract

Haemophilus influenzae (Hinf) is a human-restricted colonizer of the upper

respiratory tract. Hinf outer membrane protein (OMP) P1 specifically binds to

carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) present on

the apical surface of human epithelial cells. Strikingly, OMP P1 is a homolog of

long-chain fatty acid (LCFA) transporter FadL of Escherichia coli, which forms a

14-stranded β-barrel in the outer membrane. Whereas the role of FadL as LCFA

transporter and the regulation of FadL expression by environmental LCFAs have

been comprehensively deciphered during the last decades, a role of OMP P1 in

LCFA transport by Hinf is unknown. By employing an isogenic OMP P1 mutant, we

report that LCFAs drive Hinf growth in an OMP P1-independent fashion. However,

LCFAs appear to be potent stimulators of OMP P1 expression. Increased levels of

OMP P1 were functionally relevant as LCFA-treated bacteria showed improved

CEACAM-binding in vitro. As a result, Hinf invasion into CEACAM-expressing

epithelial cells was strongly increased upon exposure to host-derived lipids.

Interestingly, Hinf were found to be taken up by primary human granulocytes in an

opsonin-independent manner. Most importantly, the bacteria fed with arachidonic

acid and recognizing granulocyte CEACAM3 were clearly more phagocytosed.

Thus, our data provide the first evidence of the regulation of a bacterial adhesin by

host-derived lipid signals and provide impetus to study the effects of free fatty

acids on Hinf pathogenesis.

IV.2 Introduction

In Gram-negative bacteria, the outer membrane (OM) is the outermost layer of the

bacterial cell and the dynamic interface between the bacterium and its surrounding

environment, including higher animals and plants. The OM is of prime importance

for the bacterial survival as it has been linked to diverse functions such as the

uptake of nutrients (Nikaido, 2003), the adaptation in host niches (Lin et al., 2002)

and the resistance to antibacterial molecules (Pages et al., 2008). Our improved

understanding of OM importance is fuelled by detailed insights into its structure

and composition. The OM is an asymmetric bilayer with phospholipids in its inner

leaflet and lipopolysaccharides (LPS) in its outer leaflet. Whereas lipoproteins are

Page 97: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

85

anchored to the inner leaflet of the OM, β-barrel proteins are integral constituents

of the lipid bilayer (Nikaido, 2003).

β-barrel OM proteins (OMPs) build channels, through which hydrophilic molecules,

including most nutrients, move from the extracellular milieu into the periplasm.

OMPs allowing influx of structurally unrelated solutes are referred to as non-

specific channels or porins. On the contrary, for the specific channels, the

presence of binding sites for incoming molecules within the β-barrel facilitates the

diffusion of defined ligands (Nikaido, 2003). Specific channels are involved in the

ligand gated transport of various molecules, including small metal ions (Hohle et

al., 2011), a large variety of sugars (Nikaido, 2003) and fatty acids (van den Berg,

2005).

In Escherichia coli, more than four decades ago, Hill and Angelmaier

characterized a mutant of E. coli that was proficient in the degradation system of

long-chain fatty acids (LCFAs), but unable to metabolize the octadecanoic, oleic

acid. These authors suggested therefore the existence of a gene product, which

facilitates the diffusion of fatty acids through the outer membrane. (Hill and

Angelmaier, 1972). Six years later, this gene was identified and named fadL (Nunn

and Simons, 1978). The fadL gene product (FadL), a protein that locates within the

outer membrane of E. coli, was shown to be essential for long-chain fatty acid

transport (Black et al., 1987).

FadL belongs to the fatty acid degradation (fad) regulon genes, which are induced

when LCFAs (with a chain >C12) are present in the bacterial growth medium.

Explicitly, after their transport through the OM by FadL, the LCFAs cross the inner

membrane via a hitherto unknown mechanism (presumably by free diffusion). The

cytosolic LCFAs are activated into their acyl-CoA thioesters by the acyl-CoA

synthetase FadD, which is loosely associated with the inner leaflet of the inner

membrane (Black et al., 1992). The acyl-CoA thioesters bind to FadR, a repressor

of the entire fad regulon. The ensuing structural changes in FadR, upon acyl-CoA

binding, dissociate the protein from its operator sites and increase the fad regulon

expression (Feng and Cronan, 2012). Besides, the fad regulon is also modulated

by the global transcription factor ArcA (Cho et al., 2006) and the global regulator

CRC-cAMP complex (Feng and Cronan, 2012).

Page 98: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

86

Undeniably, the major breakthrough in the study of FadL was the elucidation of its

three-dimensional architecture (van den Berg et al., 2004). FadL is a 14-stranded

β-barrel protein, whose lumen is occluded by an amino-terminal hatch domain

functioning as a plug (van den Berg et al., 2004). A detailed understanding of the

FadL structure suggested that the LCFAs are transported by FadL via a so-called

lateral diffusion (Hearn et al., 2009). During lateral diffusion, LCFAs utilize the

extracellular part of FadL to circumvent the LPS polar barrier. Once inside the β-

barrel, LCFAs do not diffuse across the channel, but rather use a lateral opening in

the wall of FadL to get into the lipid bilayer from where they diffuse freely into the

periplasm (Hearn et al., 2009).

Almost a quarter century ago, Paul Black, struck by the similarity between FadL

and Haemophilus influenzae (Hinf) outer membrane protein (OMP) P1, has

hypothesized that OMP P1 may be involved in LCFAs transport (Black, 1991). He

speculated that, similar to FadL, OMP P1 expression may be induced by LCFAs or

some other nutrients (Black, 1991). However, no experimental evidences

supported his assumptions. Recently, OMP P1 was identified to play a role in Hinf

pathogenesis (Tchoupa et al., 2015). By binding to the carcinoembryonic antigen-

related cell adhesion molecules (CEACAMs), which are immunoglobulin-like

glycoproteins expressed by epithelial cells in the human respiratory tract and on

other mucosal surfaces, OMP P1 promotes the intimate attachment of Hinf to its

host cells (Tchoupa et al., 2015).

These novel insights into the pathophysiological importance of OMP P1 have

revived the need to understand, whether OMP P1 is involved in uptake of LCFAs

by Hinf and to what extent environmental LCFAs regulate OMP P1 levels. Here we

report that enrichment of Hinf growth medium with LCFAs accelerated not only

bacterial growth, but also upregulated the OMP P1 expression. The elevated

levels of OMP1 in the outer membrane were functionally relevant, as the bacteria

grown in LCFA-supplemented media exhibited an increased interaction with their

cognate receptors and showed increase invasiveness into CEACAM-expressing

epithelial cells. Our investigations suggest that Haemophilus pathogenicity is

guided by environmental cues present inside the host.

Page 99: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

87

IV.3 Results

Haemophilus influenzae growth is improved by long-chain fatty acids

The Rd strain of Hinf was previously shown to grow well in a lipid-depleted version

of the otherwise lipid-rich BHI (brain-heart infusion) medium (Wang and Cronan,

2003). Furthermore, the fatty acid composition of the bacteria remained identical

irrespective of the medium where they were grown (lipid-depleted or not) (Wang

and Cronan, 2003). In that context, we wondered whether Hinf modifies its growth

speed in response to increased concentration of fatty acids in the medium.

Therefore, Hinf strain Rd was grown in BHL broth (BHI + levinthal base)

supplemented with oleic acid, a monounsaturated octadecanoic acid (18:1, cis-9).

Clearly, Rd division rate was accelerated in the presence of oleic acid (Figure

IV-1A). Furthermore, this effect was concentration-dependent as the multiplication

of the bacteria increased with the amount of oleic acid added in the medium

(Figure IV-1A). The obvious stimulation of Hinf growth by oleic acid strongly

suggests that this LCFA is crossing the outer membrane and contributes to

bacterial metabolism. In E. coli, FadL is the principal LCFA transporter (Lepore et

al., 2011). Given the striking similarity between E. coli FadL and Hinf OMP P1, we

analyzed growth of the OMP P1-deficient Rd mutant (Hinf Rd ∆P1) in BHL broth

enriched with oleic acid. Similar to the wild-type Rd strain, the OMP P1 mutant

grew faster upon supplementation of medium with LCFA (Figure IV-1B). Taken

together these data demonstrate that LCFAs are able to boost the growth of Hinf

in an OMP1-independent manner.

Page 100: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

88

OMP P1 is upregulated by long-chain fatty acids

Apart from the stimulation of bacterial growth, addition of LCFAs in the medium

has been consistently shown to induce expression for most genes involved in fatty

acid degradation, including fadL (Dirusso and Black, 2004; Overath and Raufuss,

1967). Consequently, we compared the expression levels of the FadL homolog in

Hinf (OMP P1) upon bacterial growth in BHL broth enriched with oleic acid.

Interestingly, OMP P1 was more abundant on the surface of Hinf Rd once the

medium was supplemented with oleic acid (Figure IV-2A). Next, we sought to

determine if the upregulation of OMP P1 was a general feature shared by all the

LCFAs. Accordingly, arachidonic acid, a polyunsaturated eicosanoic acid (20:4,

all-cis-5, 8, 11, 14), was added in the BHL broth. In accordance with OMP P1

levels after oleic acid-treatment, incubation of the bacteria in arachidonic acid

tremendously augmented their OMP P1 expression (Figure IV-2B). These

observations, made with the Hinf lab strain Rd, were confirmed using a non-

typeable Hinf (NTHi) clinical isolate (NTHi H86) which showed increased OMP P1

levels with arachidonic acid (Figure IV-2B). Combined, these results strongly

suggest that many LCFAs, if not all, induce OMP P1 expression in Hinf. Moreover,

typeable and non-typeable Hinf are responsive to exogenous LCFAs.

Figure IV-1. Oleic acid stimulates Hinf growth in an OMP P1-independent manner.A. Hinf Rd growth was monitored by reading the optical density at 600 nm (OD600) in BHL brothsupplemented or not with oleic acid. B. As in (A), the growth of Hinf Rd OMP P1-deficient mutant(Hinf Rd ∆P1) was also monitored in the presence or not of oleic acid.

Page 101: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

89

Lipid-induced expression of OMP P1 enhances the binding of Hinf to human

CEACAMs

To date, the only experimentally validated role of Hinf OMP P1 is the binding to

human CEACAM family members in order to promote tight adhesion to host cells

(Tchoupa et al., 2015). In contrast to previous assumptions by Hill and colleagues

(Hill et al., 2001), we could not detect a direct CEACAM-binding function for OMP

P5, but found that OMP P5 facilitates the proper surface display of OMP P1 within

the outer membrane (Tchoupa et al., 2015). LCFAs and their effect on OMP P1

levels provided us with new tools to critically test our previous findings. After

arachidonic acid-treatment and intensive washing, Hinf Rd, its OMP P5 mutant or

its OMP P1 mutant was tested for CEACAM-binding with cell culture supernatants

enriched with similar amount of soluble GFP-tagged amino-terminal domain of

human CEACAM1 (hCEA1-N) or human CEACAM8 (hCEA8-N, negative control)

(Figure IV-3A). OMP P1 mutant (∆P1) failed to recognize hCEA1-N whereas Rd

wild-type strain (WT) and OMP P5-deficient mutant (∆P5) clearly bound hCEA1-N

Figure IV-2. Long-chain fatty acids increase OMP P1 levels of typeable and non-typeableHinf. A. Hinf Rd treated or not with 3.6 mM oleic acid were analyzed by flow cytometry followingstaining with -P1 serum in combination with a Dylight® 488-conjugated secondary antibody. B.After incubation with 1mM arachidonic acid, the OMP P1 levels in Hinf Rd (left panel) or NTHi H86(right panel) were analyzed as in (A). The DMSO-treated bacteria (black lines) were included ascontrols.

Page 102: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

90

(Figure IV-3B). Interestingly, upon arachidonic acid-treatment, the wild-type strain

engaged more hCEA1-N while the interaction of OMP P5 mutant with hCEA1

remained unchanged (Figure IV-3B). Consistent with their respective CEACAM-

binding capabilities, OMP P5-deficient mutant only showed a modest OMP P1

induction by LCFAs, whereas the wild-type Rd strain massively responded to

LCFAs by increasing its OMP P1 levels (Figure IV-3C). Taken together, these

results indicate that: (i) the enhanced CEACAM-binding of Hinf in response to

LCFA-treatment is due to increased levels of OMP P1 as a mutant genetically

deficient in OMP P1 is unable to interact with CEACAMs before and after LCFA

stimulation, and (ii) OMP P5 seems to play a role in the regulation of OMP P1

levels in the outer membrane since the OMP P5 mutant failed to upregulate its

OMP P1 levels in a manner comparable to the wild-type strain.

Figure IV-3. LCFA-treatment improves the CEACAM-binding in wild-type Hinf but not in the OMP P5-deficient mutant. A. Soluble GFP-tagged N-terminal domain of human CEACAM1 (hCEA1-N) or CEACAM8 (hCEA8-N) were harvested in cell culture supernatants of 293 cells. Similar amounts were verified by immunoblotting with anti-GFP antibodies. B. hCEA1-N and hCEA8-N, produced as described in (A) were incubated with Hinf Rd wild-type (WT), its OMP P5- (∆P5) or OMP P1-deficient mutant (∆P1) treated or not with arachidonic acid beforehand. After washing, the interaction with CEACAMs was determined upon anti-GFP Western Blot. C. OMP P1 levels in P5 mutant (Hinf Rd ∆P5, left panel) and wild-type (Hinf Rd, right panel), after incubation with arachidonic acid and binding assays with CEACAMs, were analyzed by flow cytometry upon staining with anti-P1 antibodies. The black lines indicate the staining of the DMSO-treated bacteria. The fold change in mean fluorescence (MFI) of LCFA-treated bacteria in comparison to DMSO is indicated for each sample.

Page 103: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

91

LCFA-treated Haemophilus influenzae invade epithelial cells more efficiently

Hinf adhesion to and invasion into epithelial cells were shown to correlate with

OMP P1 expression levels and CEACAM-binding (Tchoupa et al., 2015). By using

the clinical isolate NTHi H86, we wanted to determine if the increased OMP P1

levels upon incubation with LCFAs induce a better internalization of the bacterium

in a CEACAM-dependent manner. First, we tested the effect of LCFAs on NTHi

H86 recognition by soluble CEACAMs. In a gratifying similarity with the Rd strain,

NTHi H86 showcased an improved interaction with hCEA1-N upon treatment with

LCFAs (Figure IV-4A). To confirm the relevance of increased binding to soluble

CEACAMs in a cellular context, NTHi H86 treated or not with LCFAs was stained

with FITC. After FITC-labelling, the bacteria were used to infect 293 cells

transiently expressing HA-tagged full-length CEACAM1-4L or 293 cells transfected

with an empty control plasmid. Four hours post-infection, the fluorescence emitted

by the extracellular bacteria was quenched with trypan blue and the amount of

internalized FITC-labeled bacteria estimated by flow cytometry. Obviously, NTHi

H86 was unable to invade 293 control cells even after incubation with LCFAs

(Figure IV-4B). In clear contrast, the bacteria readily invaded CEACAM1-

expressing cells. However, the LCFA-treatment prior to infection significantly

improved invasiveness of NTHi H86 (Figure IV-4B). Collectively, these data

provide strong evidence that new OMP P1 molecules expressed upon addition of

LCFAs into the bacterial medium are functionally relevant, and promote a stronger

binding of the bacteria with their human receptor CEACAM and a better invasion

of epithelial cells in a CEACAM-dependent manner.

Page 104: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

92

Figure IV-4. Long-chain fatty acids increase the CEACAM-dependent invasiveness of Hinf. A. The CEACAM-binding of NTHi H86 was analyzed by flow cytometry after the treatment of the bacterium with DMSO (left panel) or arachidonic acid (right panel). The fold change in MFI of the bacteria binding to hCEA1-N (red line) is compared to hCEA8 (blue line, negative control). As an additional control, bacteria incubated with the plain cell culture medium were also included (black line). B. Upon incubation with either DMSO or arachidonic acid, NTHi H86 was FITC-labelled and used to infect 293 cells transiently expressing the full-length HA-tagged CEACAM1 (CEACAM1) or control 293 cells transfected with an empty vector (pcDNA3.1). Four hours post-infection, the infected cells were taken into suspension and trypan blue was utilized to quench the fluorescence of extracellular bacteria. 293 cells fluorescence, proportional to the quantity of internalized bacteria, was determined by flow cytometry. Relative values of the bacterial uptake are compared to DMSO-treated bacteria engulfed by CEACAM1-expressing cells. The data represent means ± standard deviation of three independent experiments (**P < 0.01).

Increased OMP P1 levels elicit a stronger innate immune response against

the bacteria

The airway epithelium expresses numerous inflammatory mediators in response to

colonizing Hinf. These cytokines induce the influx of various white blood cells,

including granulocytes (Rao et al., 1999). Human granulocytes possess a

Page 105: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

93

receptor, namely CEACAM3, which is able to induce the opsonin-independent

phagocytosis of bound bacteria (Roth et al., 2013). Recently, we reported that

Haemophilus influenzae biogroup aegyptius (Hinf-aeg) was binding to granulocyte

CEACAM3 (Tchoupa et al., 2015). We wondered whether CEACAM3-recognition

was contributing to efficient engulfment of Hinf by granulocytes. First, we

confirmed the interaction of Hinf-aeg with epithelial CEACAM1 and CEA, and with

granulocyte CEACAM3 ((Tchoupa et al., 2015); Figure IV-5A). Consistent with our

results with other Hinf strains, upon incubation with arachidonic acid, Hinf-aeg

clearly exhibited an improved interaction with the GFP-fused N-terminal domains

of CEACAM1, CEACAM3 and CEA (Figure IV-5B).

Figure IV-5. Hinf-aeg fed with LCFA augments its interaction with epithelial and granulocyte CEACAMs. A. Hinf-aeg was incubated with the indicated GFP-fused amino-terminal CEACAM domains (hCEAX-N). After extensive washing, the bacterial pellets were immunoblotted with anti-GFP antibodies (upper panel). Equivalent amounts of different hCEAX-N in cell culture supernatants (supe) were confirmed by western blotting (lower panel). B. Identical cell culture

Page 106: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

94

supernatants as in (A) were used to perform binding assays with Hinf-aeg that was pre-treated with DMSO or arachidonic acid. The interactions of bacteria with hCEAX-GFP were analyzed by flow cytometry. The fold change in MFI is indicated for each GFP-tagged N-terminal CEACAM domain and compares the CEACAM-binding of the LCFA-treated bacteria and the DMSO-treated bacteria

Next, FITC-labelled Hinf-aeg and the non-CEACAM3-binding NTHi H86 were used

to infect isolated human granulocytes. After 30 min of infection, the fluorescence

emitted by extracellular bacteria were quenched with trypan blue and the

fluorescence of the granulocytes upon infection was used as read-out to estimate

the amount of phagocytosed bacteria. Strikingly, the CEACAM3-binding Hinf-aeg

was more engulfed by the granulocytes in comparison to NTHi H86 (Figure IV-6A).

Moreover, in strict accordance with its better interaction with the soluble

CEACAMs, LCFA-treated Hinf-aeg was more phagocytosed by isolated human

granulocytes (Figure IV-6A and Figure IV-6B). Together, these data indicate that

CEACAM3 may play a major role in the opsonin-independent phagocytosis of

Hinf-aeg by granulocytes. Thus, increased OMP P1 levels upon LCFA-treatment

can be both beneficial (better binding to epithelial CEACAMs) and detrimental

(improved CEACAM3-recognition and faster engulfment by granulocytes).

Figure IV-6. Non-opsonic engulfment of Hinf-aeg by human granulocytes is potentiated bybacterial treatment with LCFA. A. Primary human granulocytes were infected for 30 min witheither Hinf-aeg or NTHi H86 at an MOI of 200. Prior to infection, the bacteria were incubated withDMSO or arachidonic acid and subsequently stained with FITC. After the infection, thefluorescence of extracellular and granulocyte-attached bacteria was quenched with trypan blue.Cell-associated fluorescence, correlating with the amount of taken up bacteria, was analyzed byflow cytometry. Uptake of DMSO-treated Hinf-aeg by human granulocytes was set to 100%. B.Phagocytosis assays, performed as described in (A), were extended to primary humangranulocytes from two additional donors. The isolated granulocytes were infected with DMSO- orLCFA-treated Hinf-aeg.

Page 107: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

95

IV.4 Discussion

Gram-negative bacteria, such as E. coli, are able to use fatty acids as a sole

source of carbon and as an energy source (Nunn and Simons, 1978). The

incoming fatty acids are either catabolized by the bacteria via the β-oxidation

pathway, or incorporated into complex membrane lipids (Overath and Raufuss,

1967). Whenever needed, the bacteria are also able to synthesize unsaturated

fatty acids. Given the high energy cost inherent in such a biosynthesis, the

bacteria must have evolved a finely tuned system to sense available exogenous

fatty acids and stop neosynthesis of fatty acids. In E. coli, the FadR protein

represses the entire set of fatty acid degradation (fad) regulon genes and activates

the fatty acid biosynthetic pathway (Feng and Cronan, 2012). Once the incoming

LCFAs are activated to acyl-CoAs, they bind FadR and abrogate its dual function

as repressor and activator (Iram and Cronan, 2005). Noticeably, the Hinf FadR

protein exhibits only poor acyl-CoA binding affinities (Iram and Cronan, 2005).

These findings question if the regulation of fatty acid degradation genes in Hinf in

response to exogenous fatty acids mimics the situation in E. coli.

In this study, we report that the E. coli FadL homolog, the Hinf outer membrane

protein P1 is induced by addition of LCFAs to the bacterial medium. Though OMP

P1 is not necessary for the increased growth of the bacteria in response to LCFA

supplementation, it nevertheless appears to be part of a fatty acid regulon.

Increased OMP P1 levels improve the exquisite interaction of Hinf with human

CEACAMs. Noteworthy, the genetic depletion of OMP P5 compromises the

increase of OMP P1 surface expression in response to LCFAs, strengthening our

previous hypothesis that OMP P5 may play a role in outer membrane integration

of OMP P1 (Tchoupa et al., 2015). As a consequence of LCFA-induced OMP P1

expression, Hinf invades epithelial cells more efficiently. At the same time, for Hinf

strains expressing a CEACAM3-binding OMP P1, the upregulation of OMP P1 by

LCFAs results in a more efficient phagocytosis by human granulocytes.

Strikingly, Hinf OMP P1 shares a high similarity with E. coli FadL (~39 % amino

acid identity). Three-dimensional modeling of OMP P1, based on the crystal

structure of FadL, predicts that OMP P1 possesses an opening within its β-barrel

(Tchoupa et al., unpublished observations). This opening was shown to be critical

for the lateral transport of LCFAs by FadL in E. coli (Lepore et al., 2011). Although

Page 108: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

96

our data demonstrate that the growth of OMP P1-deficient Hinf Rd mutant is

enhanced upon addition of oleic acid in the bacterial medium, we cannot exclude

that OMP P1 is involved in LCFA transport. Indeed, thanks to their amphipathic

character, LCFAs can freely diffuse across membranes without the need of a

protein-mediated transport (van den Berg et al., 2004). Once the bacteria, in

experimental settings are flooded with high amount of LCFAs, the free diffusion

may become the major mechanism of fatty acid uptake (Brash, 2001). In our

experimental settings, we supplemented the lipid-rich BHL broth with 3.6 mM oleic

acid. Further investigations should take advantage of the lipid-depleted variant of

Hinf growth medium (as described by (Wang and Cronan, 2003)) to test the effect

of increasing amounts of LCFAs on the growth of the P1-deficient Hinf Rd mutant

in comparison with the wild-type strain. Furthermore, physiological concentrations

of LCFAs (e.g.: within the micromolar range for arachidonic acid (Brash, 2001))

should be taken into account when deciphering the role of OMP P1 as a fatty acid

transporter in Hinf.

Whereas the physiological role of OMP P1 as LCFA transporter is still unclear,

recent findings have provided strong evidence that OMP P1 specifically interact

with human CEACAMs (Tchoupa et al., 2015). The relevance of OMP P1 in the

pathogenesis of Haemophilus influenzae is strengthened by a previous report

demonstrating that a natural OMP P1 deficient mutant, the NTHi strain 7502,

exhibits a compromised adherence to and invasion of bronchial epithelial cells

(Swords et al., 2000). Obviously, OMP P1-proficient strains of Hinf strongly attach

to CEACAM-expressing epithelial cells (Tchoupa et al., 2015; Virji et al., 2000).

Taking into account the findings presented in this manuscript, it is interesting to

mention that bacterial or viral infections of the mucosal surface are able to

stimulate the release of various pro-inflammatory cytokines initiating local

inflammations (Rao et al., 1999). It is a long standing observation that

inflammatory processes can trigger massive release of a LCFA from mammalian

cells, the arachidonic acid (Hammarstrom et al., 1975). Free arachidonic acid

molecules are converted in various bioactive eicosanoids, including

prostaglandins, leukotrienes and lipoxins (Harizi et al., 2008). Increased amounts

of eicosanoids during inflammation are, depending on the context of their

production, either pro-inflammatory or anti-inflammatory (Harizi et al., 2008).

Furthermore, a pro-inflammatory milieu also leads to a strong upregulation of

Page 109: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

97

CEACAMs, in particular CEACAM1 on epithelial and endothelial cells (Muenzner

et al., 2001, Muenzner et al., 2002). Therefore, inflammation might simultaneously

increase the presence of the Hinf adhesin as well as the abundance of the

pathogen’s receptor on the host side, which might tip the balance from harmless

colonization of a spatially restricted niche on the mucosa to invasive behavior of

the pathogen and systemic disease.

Interestingly, OMP P1 is not the only outer membrane protein of Haemophilus

influenzae, whose expression can be modulated by environmental cues. Classic

examples are provided by the transferrin binding protein (Tbp) A and B. The OMPs

TbpA and TbpB coordinately bind iron-saturated human transferrin in order to

extract and translocate iron into the bacterial periplasm (Gray-Owen and

Schryvers, 1993). The genes tbpA and tbpB are expressed as a single

transcriptional unit under the control of a ferric uptake regulatory (Fur-like)

repressor protein and their expression is suppressed by environmental iron (Rao

et al., 1999).

Importantly, arachidonic acid-derived leukotrienes, also produced by epithelial

bronchial cells (Jame et al., 2007), are powerful chemotactic agents of

granulocytes (Murray et al., 2003). Ex vivo studies with isolated human

granulocytes shed light on the interaction of infiltrating granulocytes with invading

bacteria. Clearly, the opsonization of the bacteria with antibodies and complement

components is essential for the granulocyte-mediated killing of NTHi (Langereis

and Weiser, 2014). However, our current data demonstrate that various NTHi

strains are readily phagocytosed by granulocytes in an opsonin-independent

manner. It is still unclear if such engulfment of NTHi will lead to their destruction.

Importantly, opsonin-independent recognition of Neisseria gonorrhoeae via the

granulocyte-specific CEACAM3 receptor was consistently reported to lead to

uptake and elimination of gonococci by human granulocytes (Buntru et al., 2011;

Roth et al., 2013; Schmitter et al., 2004). Concordantly, the results presented in

this manuscript speak for a better recognition of CEACAM3-binding Hinf-aeg by

the granulocytes in comparison to the non-CEACAM3-binding NTHi H86 strain.

However, due to the presence of multiple CEACAMs in human granulocytes, the

differential uptake might also reflect differences in the binding affinities to human

CEACAMs by the used Hinf strains. Therefore, experiments with human

Page 110: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

98

granulocytes in the presence of blocking antibodies against CEACAM3 or with

myeloid cell lines with genetic disruption of the CEACAM3 gene might help to

clarify this issue.

Nevertheless, the observed increased uptake of LCFA-fed Hinf-aeg by

granulocytes fits into a plausible scenario where the arachidonic acid released

upon bacterial attachment to the epithelium (via CECAM1 or CEA) would

simultaneously increase OMP P1 expression by the bacteria while alarming the

granulocytes. The CEACAM3-mediated bacterial recognition could lead to the

elimination of invading bacteria and tip the balance in favor of a local and

contained infection. However, some strains might escape this defence mechanism

by expressing non-CEACAM3-binding adhesin, increasing the chances of

disseminated disease. This concept is in accordance with a recent report where N.

gonorrohoeae strains isolated from patients with disseminated disease harbored

CEACAM-binding adhesins, which lacked CEACAM3-recognition (Roth et al.,

2013). As some strains of the biogroup aegyptius of Hinf are able to cause life-

threatening systemic diseases (Harrison et al., 2008), it would be of prime

importance to investigate whether these strains interact or not with granulocyte

CEACAM3.

In conclusion, we have shown that Hinf increases its OMP P1 levels in response to

fatty acids, such as arachidonic acid. Whereas the molecular mechanisms

underpinning this OMP P1 upregulation are still unclear, our findings suggest that

the Hinf can sense lipid environmental cues to regulate its adhesin. Given the

presence of arachidonic acid at virtually all the mucosal surfaces of human body

and the high structural conservation of OMP P1 / FadL in Gram-negative bacteria,

sensing and responding to lipid signals might represent a widespread mechanism

contributing to the efficient colonization of highly specific niches by the bacteria.

IV.5 Materials and methods

Haemophilus influenzae strains and growth conditions

Hinf Rd and its mutants used in this study were described previously (Tchoupa et

al., 2015). Hinf-aeg ATCC® 11116 was purchased from German Collection of

Microorganisms and Cell Cultures (DSMZ, Braunschweig, Germany). NTHi H86

Page 111: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

99

was generously provided by Prof. Reidl (University of Graz, Graz, Austria). The

bacteria were all grown on brain-heart infusion agar enriched with 10% Levinthal

base (BHL).

Growth curves

The growth kinetics were determined for the bacteria grown in BHL broth

supplemented or not with 3.6 or 7.2 mM oleic acid (Sigma-Aldrich). 10 × 108

bacteria were added to 8 mL of BHL broth. Cultures were grown at 37°C with

constant shaking (200 rpm) and the absorbance at 600 nm was measured every

30–60 minutes.

Analysis of surface-exposed OMP P1 levels by flow cytometry

The bacteria were incubated for 3 h at 37 °C with 3.6 mM oleic acid (Sigma-

Aldrich), 1 mM arachidonic acid (Sigma-Aldrich) or the solvent DMSO (Roth,

Karlsruhe, Germany). PBS was then used to wash the bacteria and anti-OMP P1

mouse serum (diluted 1:200) was directly added to intact bacteria. The samples

were incubated for 90 min at 4 °C under gentle rotation. After staining with Dylight®

488-conjugated anti-mouse IgG antibody (Jackson Immuno-Research, West

Grove, PA), the bacteria were washed with PBS and analyzed with the BD™ LSR

II flow cytometer.

CEACAM-binding assays

Soluble GFP-fused amino-terminal CEACAM domains were produced by 293 cells

and used for bacterial pull-down assays as previously described (Tchoupa et al.,

2015). In short, soluble recombinant GFP-tagged CEACAM N-terminal domains,

enriched in cell culture supernatants, were clustered with polyclonal anti-GFP

antibodies (overnight, at 4 °C). 10 × 108 bacteria were afterwards added in 1 ml

supernatant containing the indicated GFP-fusion proteins and incubated for 2 h at

37 °C under constant rotation. After washing with PBS, the bacteria were either

immunoblotted with a monoclonal antibody against GFP or bacteria-associated

fluorescence was detected by flow cytometry. In some cases, prior to bacterial

pull-down with CEACAMs, 6 × 109 bacteria were incubated in BHL broth

supplemented with 100 µM of arachidonic acid (Sigma-Aldrich) for 35 min at 37 °C

and washed twice with PBS.

Page 112: Identification and characterization of the novel CEACAM

Host-derived fatty acids modulate P1 levels

100

Western blotting

Immunoblotting was performed as described earlier (Hauck et al., 2001) using a

monoclonal antibody (mAb) against GFP (clone JL-8; Clonetech).

Cell culture

Cells of the HEK (Human Embryonic Kidney) 293T cell line (293 cells; ACC-635,

German Collection of Microorganisms and Cell Cultures, DSMZ, Braunschweig,

Germany) were grown in Dulbecco’s modified Eagle’s medium (DMEM) enriched

with 10% calf serum at 37° C with 5% CO2 and subcultured every 2–3 days. 293

cells were transfected calcium phosphate co-precipitation with 5–8 µg of plasmid

DNA for each 10-cm culture dish.

Quantification of bacterial internalization by flow cytometry

DMSO- or LCFA-treated bacteria were labeled with fluorescein isothiocyanate

(FITC) as described (Tchoupa et al., 2015) and used to infect cells (MOI = 200) for

2 h. The amount of intracellular bacteria was estimated by flow cytometry as

described earlier (Pils et al., 2006).

Statistical analysis

Pairwise comparisons were conducted using Student’s t-test (SigmaStat software,

Systat Software Inc., San Jose, California). P value < 0.05 was considered

significant.

Page 113: Identification and characterization of the novel CEACAM

General Discussion

101

General Discussion

Haemophilus influenzae type b (Hib) conjugated vaccine has nearly eradicated Hib

invasive infections in countries where its use is widespread (Agrawal and Murphy,

2011). Due to its components, the Hib capsular polysaccharide PRP (poly-ribose

ribitol phosphate) attached to a carrier protein, the Hib conjugate vaccine is

ineffective against unencapsulated Haemophilus influenzae (NTHi), representing

most of the circulating strains in the human population. The enormous genetic and

phenotypic diversity of NTHi strains is the major obstacle for vaccine development

against unencapsulated Hinf. Interestingly, notwithstanding their heterogeneity,

most of the NTHi isolates, as well as encapsulated strains, are able to bind the

amino-terminal domain of the human glycoprotein CEACAM1 (Virji et al., 2000).

An antigenic variable outer membrane protein, the OMP P5, was suggested to be

the bacterial ligand used by Hinf to target CEACAMs (Hill et al., 2001). Importantly,

the ompP5 gene is present in all Hinf isolates whose genome sequences are

available (Euba et al., 2015). Therefore, we hypothesized that identification of a

conserved CEACAM-binding motif within the OMP P5 may represent a major

breakthrough to allow novel preventive and curative measures against Hinf.

In this study, multiple sequence alignment of Hinf OMP P5 from strains with

different CEACAM-binding profiles did not provide any clue as to why NTHi strain

7502 is unable to target CEACAMs. Moreover, OMP P5 of various CEACAM-

binding Hinf strains, when expressed in non-CEACAM binding E. coli, was not

sufficient to promote the recognition of human CEACAMs. These initials

observations raised uncertainty about OMP P5 as the Hinf CEACAM-binding

ligand. Therefore, we screened mutant and wild-type Hinf strains to finally identify

OMP P1 as the bona fide adhesin recognizing human CEACAMs. Our results on

first sight were in conflict with former work, which suggested a direct interaction

between OMP P5 and CEACAMs (Hill et al., 2001). However, this previous study

already conceded that: (i) the non-CEACAM binding strain NTHi H305 expresses

the OMP P5; (ii) genetic depletion of OMP P5 weakens the interaction of

CEACAM-binding Hinf with soluble CEACAMs but not with CEACAM-expressing

cells and (iii) the adhesion of OMP P5-deficient Hinf to CEACAM-expressing cells

was inhibited by an anti-CEACAM antibody (Hill et al., 2001). Combined, these

observations strongly suggested that another Hinf factor, whose surface levels

Page 114: Identification and characterization of the novel CEACAM

General Discussion

102

might be affected by OMP P5-depletion, was responsible for CEACAM-targeting

by the bacteria. Consistent with this plausible hypothesis, our data demonstrate

reduced levels of OMP P1 in OMP P5 mutant, providing a reasonable explanation

for the altered CEACAM-binding phenotype upon OMP P5 deletion. Fluctuations in

OMP P1 levels upon knock out of another outer membrane protein are not

unusual. For instance, Cope and co-workers noticed increased production of OMP

P1 in a OMP P2-depleted Hib mutant. Importantly, the complementation of OMP

P2 in the OMP P2-deficient mutant brings back the OMP P1 synthesis to the wild-

type levels (Cope et al., 1990). Conversely, we did not observe major changes in

OMP P2 levels upon OMP P1 deletion. However, a Hinf strain engineered to

express more OMP P1 in comparison to the otherwise isogenic wild-type strain,

exhibits less OMP P2 compared to the wild-type strain (Hanson et al., 1989). It is

likely that OMP P1 together with other outer membrane proteins builds the so-

called OMP islands where the absence or the overabundance of one OMP may

results in remodeling of OMP islands’ composition in order to preserve their

stability.

Lately, the strain NTHi strain 375 was characterized as a non-CEACAM-binding

strain notwithstanding its well-expressed OMP P5 protein sharing 95% amino acid

identity with OMP P5 of Hinf Rd strain (Euba et al., 2015). Furthermore, the

massive attachment of the Rd strain to CEACAM1-expressing cells was reduced,

not abrogated, upon deletion of OMP P5 (Euba et al., 2015). Whereas the residual

expression of OMP P1 in Rd ∆P5 mutant explains why this mutant is able to

adhere to cells in a CEACAM-dependent manner, it is unclear why NTHi 375 does

not bind CEACAMs. NTHi 7502, the only strain of our Hinf collection unable to

recognize CEACAMs, does not express any OMP P1 because of a mutation in its

ompP1 gene creating a premature stop codon (Tchoupa et al., 2015). In contrast,

NTHi 375 ompP1 gene is intact. It would be interesting to analyze the amount of

OMP P1 expressed by NTHi 375. High expression levels of OMP P1 in NTHi 375

would establish NTHi 375 as the first OMP P1-proficient Hinf strain unable to bind

CEACAMs, a magnificent tool in order to decrypt the molecular requirements for

OMP P1 – CEACAM interaction.

There are numerous examples, where the deletion of a particular OMP results in

readjusted levels of a different OMP. Therefore, it is essential to combine loss-of-

Page 115: Identification and characterization of the novel CEACAM

General Discussion

103

function assays upon OMP deletion with gain-of-function experiments after OMP

overexpression. Accordingly, our findings demonstrate that non-CEACAM-binding

E. coli is converted to a CEACAM-binding strain upon OMP P1 overexpression.

Thus, Hinf OMP P1 is not only necessary but also sufficient for CEACAM-

targeting. Of note, OMP P1 is only the second CEACAM-binding adhesin, after the

neisserial Opa proteins, to have been rigorously tested in a heterologous

background (Gray-Owen et al., 1997; Roth et al., 2013). The misidentification of

OMP P5 as the Hinf CEACAN-binding ligand (Hill et al., 2001) underlines the

importance of unbiased assays evaluating the sufficiency of an OMP to target its

postulated receptor.

The OMP P1, one of the major Hinf outer membrane proteins, is highly

immunogenic. Its use as a vaccine has been hampered by the lack of cross-

protection provided against heterologous strains. Indeed, chinchilla immunized

with recombinant OMP P1 from a given strain are protected against this strain but,

not against a strain with a different P1 (91% amino acid identity) (Bolduc et al.,

2000). This failure is due to the variability of the immunogenic and surface-

exposed regions of OMP P1 amongst Hinf strains (Bolduc et al., 2000).

Importantly, the atomic resolution structure of E. coli FadL (van den Berg et al.,

2004), the homolog of OMP P1, has greatly contributed to the understanding of

OMP P1 molecular architecture as FadL can be used to model OMP P1 (Mes and

van Putten, 2007). OMP P1 is predicted to build a 14-stranded β-barrel with 7

extracellular loops. By combining protein modeling and multiple sequence

alignment, we assigned the P1 immunogenic regions, previously known as

variable regions I, II, III and IV, to the loops 1, 3, 4 and 7, respectively. Although

these loops significantly differ from one Hinf strain to another, they are essential

for CEACAM-binding as their deletion result in loss of interaction. Interestingly, the

Opa protein, another β-barreled CEACAM-targeting adhesin, also uses two

hypervariable regions (HV1 and HV2) located on its loops 2 and 3 in order to bind

CEACAMs (Bos et al., 2002). Notwithstanding its variability, HV2 of

meningococcal Opa proteins harbors a conserved amino acid motif, GxI/V/LxQ,

that is critical for CEACAM recognition (de Jonge et al., 2003). This motif is

present in OMP P1 loop3 of only a small fraction of Hinf isolates, suggesting its

dispensability in CEACAM-binding by Hinf.

Page 116: Identification and characterization of the novel CEACAM

General Discussion

104

The relevance of Hinf attachment to epithelial CEACAMs in vivo was previously

investigated using the chinchilla as a model (Bookwalter et al., 2007). Bookwalter

and colleagues showed that animals pretreated with anti-human CEACAM

antibodies were less colonized by Hinf and suggested therefore a direct interaction

between Hinf and chinchilla CEACAM1 (cCEACAM1) for nasopharyngeal

colonization (Bookwalter et al., 2007). Given the inexistence of direct evidence for

the binding of Hinf to cCEACAM1 and the relatively poor similarity (56% identity)

between chinchilla and human CEACAM1 amino-terminal domains, it is necessary

to rigorously test Hinf for cCEACAM1-binding. Our experimental setup, with GFP-

tagged CEACAM N-terminal domains, may be very useful for such a purpose and

has enabled us already to demonstrate that Hinf interacts with human CEACAM1,

but not with CEACAM1 from murine, bovine or canine origin. This apparent

species-specificity of CEACAM-targeting was also observed in other human-

restricted, CEACAM-binding pathogens (Neisseria, gonorrhoeae, Neisseria

meningitidis and Moraxella catarrhalis) that also selectively target human, but not

other mammalian CEACAM1 orthologues (Voges et al., 2010).

CEACAM homologs are present in many vertebrates, including cartilaginous fish,

frogs, lizards and humans, but absent in avian species (Chang et al., 2013). In

each species where they are expressed, CEACAMs have undergone a rapid

evolution to acquire or lose functions in response to the environmental selection

pressure and their recognition by pathogens is thought to constitute a driving force

for their evolution (Kammerer and Zimmermann, 2010). Our current results speak

for a co-evolution of Hinf OMP P1 with its human receptor(s). Although OMP P1 is

well-conserved in the Pasteurellaceae family, it is unclear which genetic alteration

has enabled only Hinf to recognize and exploit CEACAMs. It is plausible that

antigenic variations occurring on the prominent extracellular loops of OMP P1 may

have, by chance, rendered a Hinf strain able to interact with CEACAMs. The

ensuing advantages in terms of colonization and survival within the host might

have enabled the CEACAM-binding Hinf strains to gradually outcompete the non-

CEACAM-binding isolates. However, the importance of CEACAM-targeting for Hinf

colonization has to be explored in vivo with suitable animal models (Johswich et

al., 2013; Muenzner et al., 2010) or with human volunteers (Poole et al., 2013)

taking in account the species-specificity of OMP P1 – CEACAM interaction. The

only non-CEACAM binding Hinf strain we have characterized to date, NTHi strain

Page 117: Identification and characterization of the novel CEACAM

General Discussion

105

7502, was isolated from a patient with chronic obstructive pulmonary disease

(COPD) (Swords et al., 2000). Owing to the complex pathophysiology of COPD, it

is a mystery if NTHi 7502 was responsible of one exacerbation episode of the

disease or if this isolate is able to colonize a healthy, human airway.

Modelling of the OMP P1 structure has revealed that it might exhibit a large

opening in its putative outer membrane embedded β-barrel, reminiscent of the

lateral opening through which E. coli FadL transports long-chain fatty acids

(LCFAs) (Hearn et al., 2009). Our preliminary attempts failed to demonstrate an

essential role of Hinf OMP P1 in fatty acid uptake. However, a strong upregulation

of OMP P1 is noticeable upon treatment of inf with fatty acids. Similarly, FadL is

acknowledged to be induced by LCFAs (Feng and Cronan, 2012). Furthermore, E.

coli can: (i) sense the presence of LCFA in its environment; (ii) induce the

expression of its LCFA transporter (FadL) and (iii) shut down its own fatty acid

biosynthesis machinery (Feng and Cronan, 2012). It is currently unclear if this

intricate regulation of fatty acids degradation or neosynthesis is present in Hinf.

Nevertheless, the increased levels of OMP P1 upon addition of LCFAs enable Hinf

to better bind to soluble CEACAMs and to invade epithelial cells more efficiently in

a CEACAM-dependent manner. Consistently, the Hinf P1-deficient mutant is

unable to interact with CEACAMs even after LCFA-treatment. Thus, our data

underline the role of OMP P1 as the Hinf CEACAM-binding ligand while

suggesting that OMP P1 levels can be controlled by host-derived environmental

cues. This is the first example of a bacterial adhesin, whose expression is

modulated by lipid messengers that can be delivered by the host.

In conclusion, our current work provides strong evidence on the necessity and the

sufficiency of OMP P1 in the recognition of human CEACAMs by Haemophilus

influenzae and demonstrates that OMP P5 does not function as a CEACAM-

binding adhesin. OMP P1 utilizes its immunogenic, flexible and surface-exposed

extracellular loops to build the binding interface towards CEACAMs. Besides, the

induction of OMP P1 expression upon supplementation of fatty acids strongly

enhances the interaction of Hinf with CEACAMs and strengthens the finding that

OMP P1 is the bona fide CEACAM-binding adhesin of Hinf. Combined, these data

shed new light on the critical role played by OMP P1 in the crosstalk between

Haemophilus influenzae and its human host. The knowledge gained here is a solid

Page 118: Identification and characterization of the novel CEACAM

General Discussion

106

starting point for further studies pertaining to the relevance of OMP P1 in Hinf

pathogenesis, while paving the way for OMP P1-based preventive and therapeutic

approaches against Haemophilus influenzae.

Page 119: Identification and characterization of the novel CEACAM

Declaration of author’s contributions

107

Declaration of author’s contributions

Chapter I: “Signaling by epithelial members of the CEACAM family – mucosal

docking sites for pathogenic bacteria”. Cell Communication and Signaling 2014:

12:27

Arnaud Kengmo Tchoupa, Tamara Schumacher and Christof R. Hauck.

The manuscript was written by AKT and CRH. AKT, TS and CRH revised the

manuscript.

Chapter II: “Outer membrane protein P1 is the CEACAM-binding adhesin of

Haemophilus influenzae”. Mol Microbiol. 2015 Jul 16. doi: 10.1111/mmi.13134.

Arnaud Kengmo Tchoupa, Sabine Lichtenegger, Joachim Reidl and Christof R.

Hauck.

CRH and JR conceived and supervised the project. AKT and SL performed the

experiments. AKT and CRH wrote the paper.

Chapter III: “The outer membrane protein P1 of Haemophilus influenzae utilizes

its extracellular loops to grasp human CEACAM.” In preparation.

Arnaud Kengmo Tchoupa and Christof R. Hauck.

CRH and AKT designed the study. Experiments were conducted by AKT. AKT

wrote the manuscript.

Chapter IV: “Haemophilus influenzae upregulates its outer membrane protein P1

in response to fatty acids”. In preparation.

Arnaud Kengmo Tchoupa and Christof R. Hauck.

CRH and AKT designed the study. AKT performed the experiments. The

manuscript was written by AKT.

Page 120: Identification and characterization of the novel CEACAM

List of publications

108

List of publications

1. Publications that are included in this thesis

Tchoupa, A.K., T. Schuhmacher, and C.R. Hauck. 2014. Signaling by

epithelial members of the CEACAM family - mucosal docking sites for

pathogenic bacteria. Cell Commun Signal. 12:27.

Tchoupa, A.K., S. Lichtenegger, J. Reidl, and C.R. Hauck. 2015. Outer

membrane protein P1 is the CEACAM-binding adhesin of Haemophilus

influenzae. Molecular microbiology.

2. Publications not part of this thesis

Roth, A., C. Mattheis, A.K. Tchoupa, M. Hupfeld, N. Hartmann, M. Unemo,

C.R. Hauck. Induction of the opsonophagoctosis by CEA-Fc fusion proteins.

In preparation.

Kuespert, K., A. Roth, J. Adrian, A.K. Tchoupa, C. Sprissler, T. Güntert, G.

O. Cory, C.R. Hauck. JNKs are involved in CEACAM3-mediated opsonin-

independent phagocytosis of Neisseria gonorrhoeae. The Journal of

biological chemistry. In revision.

Muenzner, P., A.K. Tchoupa, B. Klauser, T. Brunner, U. Dobrindt, C.R.

Hauck. Uropathogenic E. coli exploit CEA to promote colonization of the

urogenital tract mucosa. Plos Pathogens. In revision.

Page 121: Identification and characterization of the novel CEACAM

References

109

References

Abraham, S.N., A.B. Jonsson, and S. Normark. 1998. Fimbriae-mediated host-pathogen cross-talk. Curr Opin Microbiol. 1:75-81.

Agrawal, A., and T.F. Murphy. 2011. Haemophilus influenzae infections in the H. influenzae type b conjugate vaccine era. Journal of clinical microbiology. 49:3728-3732.

Andersen, C., E. Maier, G. Kemmer, J. Blass, A.K. Hilpert, R. Benz, and J. Reidl. 2003. Porin OmpP2 of Haemophilus influenzae shows specificity for nicotinamide-derived nucleotide substrates. The Journal of biological chemistry. 278:24269-24276.

Anderson, K.L., J. Billington, D. Pettigrew, E. Cota, P. Simpson, P. Roversi, H.A. Chen, P. Urvil, L. du Merle, P.N. Barlow, M.E. Medof, R.A. Smith, B. Nowicki, C. Le Bouguenec, S.M. Lea, and S. Matthews. 2004. An atomic resolution model for assembly, architecture, and function of the Dr adhesins. Molecular cell. 15:647-657.

Apicella, M.A. 2010. Neisseria meningitidis. Churchill Livingstone, Philadelphia. 2737-2752 pp.

Araki, N., M.T. Johnson, and J.A. Swanson. 1996. A role for phosphoinositide 3-kinase in the completion of macropinocytosis and phagocytosis by macrophages. The Journal of cell biology. 135:1249-1260.

Avadhanula, V., C.A. Rodriguez, G.C. Ulett, L.O. Bakaletz, and E.E. Adderson. 2006. Nontypeable Haemophilus influenzae adheres to intercellular adhesion molecule 1 (ICAM-1) on respiratory epithelial cells and upregulates ICAM-1 expression. Infection and immunity. 74:830-838.

Bakaletz, L.O., B.D. Baker, J.A. Jurcisek, A. Harrison, L.A. Novotny, J.E. Bookwalter, R. Mungur, and R.S. Munson, Jr. 2005. Demonstration of Type IV pilus expression and a twitching phenotype by Haemophilus influenzae. Infection and immunity. 73:1635-1643.

Barnett, T.R., L. Drake, and W. Pickle, 2nd. 1993. Human biliary glycoprotein gene: characterization of a family of novel alternatively spliced RNAs and their expressed proteins. Molecular and cellular biology. 13:1273-1282.

Barnich, N., F.A. Carvalho, A.L. Glasser, C. Darcha, P. Jantscheff, M. Allez, H. Peeters, G. Bommelaer, P. Desreumaux, J.F. Colombel, and A. Darfeuille-Michaud. 2007. CEACAM6 acts as a receptor for adherent-invasive E. coli, supporting ileal mucosa colonization in Crohn disease. The Journal of clinical investigation. 117:1566-1574.

Beauchemin, N., and A. Arabzadeh. 2013. Carcinoembryonic antigen-related cell adhesion molecules (CEACAMs) in cancer progression and metastasis. Cancer Metastasis Rev.

Beauchemin, N., P. Draber, G. Dveksler, P. Gold, S. Gray-Owen, F. Grunert, S. Hammarstrom, K.V. Holmes, A. Karlsson, M. Kuroki, S.H. Lin, L. Lucka, S.M. Najjar, M. Neumaier, B. Obrink, J.E. Shively, K.M. Skubitz, C.P. Stanners, P. Thomas, J.A. Thompson, M. Virji, S. von Kleist, C. Wagener, S. Watt, and W. Zimmermann. 1999. Redefined nomenclature for members of the carcinoembryonic antigen family. Experimental cell research. 252:243-249.

Benchimol, S., A. Fuks, S. Jothy, N. Beauchemin, K. Shirota, and C.P. Stanners. 1989. Carcinoembryonic antigen, a human tumor marker, functions as an intercellular adhesion molecule. Cell. 57:327-334.

Page 122: Identification and characterization of the novel CEACAM

References

110

Berger, C.N., O. Billker, T.F. Meyer, A.L. Servin, and I. Kansau. 2004. Differential recognition of members of the carcinoembryonic antigen family by Afa/Dr adhesins of diffusely adhering Escherichia coli (Afa/Dr DAEC). Molecular microbiology. 52:963-983.

Black, P.N. 1991. Primary sequence of the Escherichia coli fadL gene encoding an outer membrane protein required for long-chain fatty acid transport. Journal of bacteriology. 173:435-442.

Black, P.N., C.C. DiRusso, A.K. Metzger, and T.L. Heimert. 1992. Cloning, sequencing, and expression of the fadD gene of Escherichia coli encoding acyl coenzyme A synthetase. The Journal of biological chemistry. 267:25513-25520.

Black, P.N., B. Said, C.R. Ghosn, J.V. Beach, and W.D. Nunn. 1987. Purification and characterization of an outer membrane-bound protein involved in long-chain fatty acid transport in Escherichia coli. The Journal of biological chemistry. 262:1412-1419.

Bolduc, G.R., V. Bouchet, R.Z. Jiang, J. Geisselsoder, Q.C. Truong-Bolduc, P.A. Rice, S.I. Pelton, and R. Goldstein. 2000. Variability of outer membrane protein P1 and its evaluation as a vaccine candidate against experimental otitis media due to nontypeable Haemophilus influenzae: an unambiguous, multifaceted approach. Infection and immunity. 68:4505-4517.

Bookwalter, J.E., J.A. Jurcisek, S.D. Gray-Owen, S. Fernandez, G. McGillivary, and L.O. Bakaletz. 2007. A CEACAM1 homologue plays a pivotal role in nontypeable Haemophilus influenzae colonization of the chinchilla nasopharynx via the OMP P5-homologous adhesin. Infection and immunity.

Bookwalter, J.E., J.A. Jurcisek, S.D. Gray-Owen, S. Fernandez, G. McGillivary, and L.O. Bakaletz. 2008. A carcinoembryonic antigen-related cell adhesion molecule 1 homologue plays a pivotal role in nontypeable Haemophilus influenzae colonization of the chinchilla nasopharynx via the outer membrane protein P5-homologous adhesin. Infection and immunity. 76:48-55.

Bos, M.P., F. Grunert, and R.J. Belland. 1997. Differential recognition of members of the carcinoembryonic antigen family by Opa variants of Neisseria gonorrhoeae. Infection & Immunity. 65:2353-2361.

Bos, M.P., D. Kao, D.M. Hogan, C.C. Grant, and R.J. Belland. 2002. Carcinoembryonic antigen family receptor recognition by gonococcal Opa proteins requires distinct combinations of hypervariable Opa protein domains. Infection and immunity. 70:1715-1723.

Bouchet, V., D.W. Hood, J. Li, J.R. Brisson, G.A. Randle, A. Martin, Z. Li, R. Goldstein, E.K. Schweda, S.I. Pelton, J.C. Richards, and E.R. Moxon. 2003. Host-derived sialic acid is incorporated into Haemophilus influenzae lipopolysaccharide and is a major virulence factor in experimental otitis media. Proceedings of the National Academy of Sciences of the United States of America. 100:8898-8903.

Brash, A.R. 2001. Arachidonic acid as a bioactive molecule. The Journal of clinical investigation. 107:1339-1345.

Brooks, M.J., J.L. Sedillo, N. Wagner, W. Wang, A.S. Attia, H. Wong, C.A. Laurence, E.J. Hansen, and S.D. Gray-Owen. 2008. Moraxella catarrhalis binding to host cellular receptors is mediated by sequence-specific determinants not conserved among all UspA1 protein variants. Infection and immunity. 76:5322-5329.

Page 123: Identification and characterization of the novel CEACAM

References

111

Brummer, J., A. Ebrahimnejad, R. Flayeh, U. Schumacher, T. Loning, A.M. Bamberger, and C. Wagener. 2001. cis Interaction of the cell adhesion molecule CEACAM1 with integrin beta(3). Am J Pathol. 159:537-546.

Buntru, A., K. Kopp, M. Voges, R. Frank, V. Bachmann, and C.R. Hauck. 2011. Phosphatidylinositol-3' kinase activity is critical for initiating the oxidative burst and bacterial destruction during CEACAM3-mediated phagocytosis. The Journal of biological chemistry. 286:9555-9566.

Buntru, A., A. Roth, N.J. Nyffenegger-Jann, and C.R. Hauck. 2012. HemITAM signaling by CEACAM3, a human granulocyte receptor recognizing bacterial pathogens. Archives of biochemistry and biophysics. 524:77-83.

Camacho-Leal, P., and C.P. Stanners. 2008. The human carcinoembryonic antigen (CEA) GPI anchor mediates anoikis inhibition by inactivation of the intrinsic death pathway. Oncogene. 27:1545-1553.

Camacho-Leal, P., A.B. Zhai, and C.P. Stanners. 2007. A co-clustering model involving alpha5beta1 integrin for the biological effects of GPI-anchored human carcinoembryonic antigen (CEA). Journal of cellular physiology. 211:791-802.

Carruthers, M.D., E.N. Tracy, A.C. Dickson, K.B. Ganser, R.S. Munson, Jr., and L.O. Bakaletz. 2012. Biological roles of nontypeable Haemophilus influenzae type IV pilus proteins encoded by the pil and com operons. Journal of bacteriology. 194:1927-1933.

Chan, C.H., and C.P. Stanners. 2004. Novel mouse model for carcinoembryonic antigen-based therapy. Molecular therapy : the journal of the American Society of Gene Therapy. 9:775-785.

Chang, C.L., J. Semyonov, P.J. Cheng, S.Y. Huang, J.I. Park, H.J. Tsai, C.Y. Lin, F. Grutzner, Y.K. Soong, J.J. Cai, and S.Y. Hsu. 2013. Widespread divergence of the CEACAM/PSG genes in vertebrates and humans suggests sensitivity to selection. PloS one. 8:e61701.

Chen, C.J., J. Kirshner, M.A. Sherman, W. Hu, T. Nguyen, and J.E. Shively. 2007. Mutation analysis of the short cytoplasmic domain of the cell-cell adhesion molecule CEACAM1 identifies residues that orchestrate actin binding and lumen formation. The Journal of biological chemistry. 282:5749-5760.

Chen, T., and E.C. Gotschlich. 1996. CGM1a antigen of neutrophils, a receptor of gonococcal opacity proteins. Proceedings of the National Academy of Sciences of the United States of America. 93:14851-14856.

Chen, T., F. Grunert, A. Medina-Marino, and E.C. Gotschlich. 1997. Several carcinoembryonic antigens (CD66) serve as receptors for gonococcal opacity proteins. Journal of Experimental Medicine. 185:1557-1564.

Cheung, P.H., W. Luo, Y. Qiu, X. Zhang, K. Earley, P. Millirons, and S.H. Lin. 1993. Structure and function of C-CAM1. The first immunoglobulin domain is required for intercellular adhesion. The Journal of biological chemistry. 268:24303-24310.

Cho, B.K., E.M. Knight, and B.O. Palsson. 2006. Transcriptional regulation of the fad regulon genes of Escherichia coli by ArcA. Microbiology. 152:2207-2219.

Clementi, C.F., and T.F. Murphy. 2011. Non-typeable Haemophilus influenzae invasion and persistence in the human respiratory tract. Front Cell Infect Microbiol. 1:1.

Confer, A.W., and S. Ayalew. 2013. The OmpA family of proteins: roles in bacterial pathogenesis and immunity. Veterinary microbiology. 163:207-222.

Page 124: Identification and characterization of the novel CEACAM

References

112

Conners, R., D.J. Hill, E. Borodina, C. Agnew, S.J. Daniell, N.M. Burton, R.B. Sessions, A.R. Clarke, L.E. Catto, D. Lammie, T. Wess, R.L. Brady, and M. Virji. 2008. The Moraxella adhesin UspA1 binds to its human CEACAM1 receptor by a deformable trimeric coiled-coil. The EMBO journal. 27:1779-1789.

Cope, L.D., S.E. Pelzel, J.L. Latimer, and E.J. Hansen. 1990. Characterization of a mutant of Haemophilus influenzae type b lacking the P2 major outer membrane protein. Infection and immunity. 58:3312-3318.

de Jonge, M.I., H.J. Hamstra, L. van Alphen, J. Dankert, and P. van der Ley. 2003. Mapping the binding domains on meningococcal Opa proteins for CEACAM1 and CEA receptors. Molecular microbiology. 50:1005-1015.

del Pozo, M.A., N.B. Alderson, W.B. Kiosses, H.H. Chiang, R.G. Anderson, and M.A. Schwartz. 2004. Integrins regulate Rac targeting by internalization of membrane domains. Science. 303:839-842.

Dirusso, C.C., and P.N. Black. 2004. Bacterial long chain fatty acid transport: gateway to a fatty acid-responsive signaling system. The Journal of biological chemistry. 279:49563-49566.

Eades-Perner, A.M., H. van der Putten, A. Hirth, J. Thompson, M. Neumaier, S. von Kleist, and W. Zimmermann. 1994. Mice transgenic for the human carcinoembryonic antigen gene maintain its spatiotemporal expression pattern. Cancer research. 54:4169-4176.

Ergun, S., N. Kilik, G. Ziegeler, A. Hansen, P. Nollau, J. Gotze, J.H. Wurmbach, A. Horst, J. Weil, M. Fernando, and C. Wagener. 2000. CEA-related cell adhesion molecule 1: a potent angiogenic factor and a major effector of vascular endothelial growth factor. Molecular cell. 5:311-320.

Euba, B., J. Moleres, C. Viadas, I. Ruiz de los Mozos, J. Valle, J.A. Bengoechea, and J. Garmendia. 2015. Relative Contribution of P5 and Hap Surface Proteins to Nontypable Haemophilus influenzae Interplay with the Host Upper and Lower Airways. PloS one. 10:e0123154.

Feng, Y., and J.E. Cronan. 2012. Crosstalk of Escherichia coli FadR with global regulators in expression of fatty acid transport genes. PloS one. 7:e46275.

Finlay, B.B., and S. Falkow. 1997. Common themes in microbial pathogenicity revisited. Microbiology and Molecular Biology Reviews. 61:136-169.

Fox, D.A., P. Larsson, R.H. Lo, B.M. Kroncke, P.M. Kasson, and L. Columbus. 2014. Structure of the neisserial outer membrane protein opa60: loop flexibility essential to receptor recognition and bacterial engulfment. J Am Chem Soc. 136:9938-9946.

Foxwell, A.R., J.M. Kyd, and A.W. Cripps. 1998. Nontypeable Haemophilus influenzae: pathogenesis and prevention. Microbiology and molecular biology reviews : MMBR. 62:294-308.

Gaur, S., J.E. Shively, Y. Yen, and R.K. Gaur. 2008. Altered splicing of CEACAM1 in breast cancer: identification of regulatory sequences that control splicing of CEACAM1 into long or short cytoplasmic domain isoforms. Molecular cancer. 7:46.

Gold, P., and S.O. Freedman. 1965. Specific carcinoembryonic antigens of the human digestive system. The Journal of experimental medicine. 122:467-481.

Gotoh, N., H. Wakebe, and T. Nishino. 1989a. Ultrastructural aspects of fragility of Pseudomonas aeruginosa outer membrane devoid of protein F. FEMS microbiology letters. 50:51-53.

Page 125: Identification and characterization of the novel CEACAM

References

113

Gotoh, N., H. Wakebe, E. Yoshihara, T. Nakae, and T. Nishino. 1989b. Role of protein F in maintaining structural integrity of the Pseudomonas aeruginosa outer membrane. Journal of bacteriology. 171:983-990.

Gray-Owen, S.D., and R.S. Blumberg. 2006. CEACAM1: contact-dependent control of immunity. Nat Rev Immunol. 6:433-446.

Gray-Owen, S.D., D.R. Lorenzen, A. Haude, T.F. Meyer, and C. Dehio. 1997. Differential Opa specificities for CD66 receptors influence tissue interactions and cellular response to Neisseria gonorrhoeae. Molecular microbiology. 26:971-980.

Gray-Owen, S.D., and A.B. Schryvers. 1993. The interaction of primate transferrins with receptors on bacteria pathogenic to humans. Microbial pathogenesis. 14:389-398.

Gu, A., Z. Zhang, N. Zhang, W. Tsark, and J.E. Shively. 2010. Generation of human CEACAM1 transgenic mice and binding of Neisseria Opa protein to their neutrophils. PloS one. 5:e10067.

Guignot, J., S. Hudault, I. Kansau, I. Chau, and A.L. Servin. 2009. Human decay-accelerating factor and CEACAM receptor-mediated internalization and intracellular lifestyle of Afa/Dr diffusely adhering Escherichia coli in epithelial cells. Infection and immunity. 77:517-531.

Hammarstrom, S. 1999. The carcinoembryonic antigen (CEA) family: structures, suggested functions and expression in normal and malignant tissues. Semin Cancer Biol. 9:67-81.

Hammarstrom, S., M. Hamberg, B. Samuelsson, E.A. Duell, M. Stawiski, and J.J. Voorhees. 1975. Increased concentrations of nonesterified arachidonic acid, 12L-hydroxy-5,8,10,14-eicosatetraenoic acid, prostaglandin E2, and prostaglandin F2alpha in epidermis of psoriasis. Proceedings of the National Academy of Sciences of the United States of America. 72:5130-5134.

Hanson, M.S., L.D. Cope, and E.J. Hansen. 1989. Expression of the heat-modifiable major outer membrane protein of Haemophilus influenzae type b is unrelated to virulence. Infection and immunity. 57:1639-1646.

Harizi, H., J.B. Corcuff, and N. Gualde. 2008. Arachidonic-acid-derived eicosanoids: roles in biology and immunopathology. Trends in molecular medicine. 14:461-469.

Harrison, L.H., V. Simonsen, and E.A. Waldman. 2008. Emergence and disappearance of a virulent clone of Haemophilus influenzae biogroup aegyptius, cause of Brazilian purpuric fever. Clinical microbiology reviews. 21:594-605.

Hauck, C.R., T. Hunter, and D.D. Schlaepfer. 2001. The v-Src SH3 domain facilitates a cell adhesion-independent association with FAK. Journal of Biological Chemistry. 276:17653-17662.

Hearn, E.M., D.R. Patel, B.W. Lepore, M. Indic, and B. van den Berg. 2009. Transmembrane passage of hydrophobic compounds through a protein channel wall. Nature. 458:367-370.

Hill, D.J., A.M. Edwards, H.A. Rowe, and M. Virji. 2005. Carcinoembryonic antigen-related cell adhesion molecule (CEACAM)-binding recombinant polypeptide confers protection against infection by respiratory and urogenital pathogens. Molecular microbiology. 55:1515-1527.

Hill, D.J., M.A. Toleman, D.J. Evans, S. Villullas, L. Van Alphen, and M. Virji. 2001. The variable P5 proteins of typeable and non-typeable Haemophilus influenzae target human CEACAM1. Molecular microbiology. 39:850-862.

Page 126: Identification and characterization of the novel CEACAM

References

114

Hill, D.J., and M. Virji. 2003. A novel cell-binding mechanism of Moraxella catarrhalis ubiquitous surface protein UspA: specific targeting of the N-domain of carcinoembryonic antigen-related cell adhesion molecules by UspA1. Molecular microbiology. 48:117-129.

Hill, D.J., C. Whittles, and M. Virji. 2012. A novel group of Moraxella catarrhalis UspA proteins mediates cellular adhesion via CEACAMs and vitronectin. PloS one. 7:e45452.

Hill, F.F., and D. Angelmaier. 1972. Specific enrichment of mutants of Escherichia coli with an altered acyl CoA synthetase by tritium suicide. Molecular & general genetics : MGG. 117:143-152.

Hoffmann, C., A. Berking, F. Agerer, A. Buntru, F. Neske, G.S. Chhatwal, K. Ohlsen, and C.R. Hauck. 2010. Caveolin limits membrane microdomain mobility and integrin-mediated uptake of fibronectin-binding pathogens. Journal of cell science. 123:4280-4291.

Hohle, T.H., W.L. Franck, G. Stacey, and M.R. O'Brian. 2011. Bacterial outer membrane channel for divalent metal ion acquisition. Proceedings of the National Academy of Sciences of the United States of America. 108:15390-15395.

Hoiseth, S.K., and J.R. Gilsdorf. 1988. The relationship between type b and nontypable Haemophilus influenzae isolated from the same patient. The Journal of infectious diseases. 158:643-645.

Horst, A.K., W.D. Ito, J. Dabelstein, U. Schumacher, H. Sander, C. Turbide, J. Brummer, T. Meinertz, N. Beauchemin, and C. Wagener. 2006. Carcinoembryonic antigen-related cell adhesion molecule 1 modulates vascular remodeling in vitro and in vivo. The Journal of clinical investigation. 116:1596-1605.

Horton, R.M., Z.L. Cai, S.N. Ho, and L.R. Pease. 1990. Gene splicing by overlap extension: tailor-made genes using the polymerase chain reaction. BioTechniques. 8:528-535.

Huang, J., J.D. Hardy, Y. Sun, and J.E. Shively. 1999. Essential role of biliary glycoprotein (CD66a) in morphogenesis of the human mammary epithelial cell line MCF10F. Journal of cell science. 112 ( Pt 23):4193-4205.

Hultgren, S.J., S. Abraham, M. Caparon, P. Falk, J.W. St Geme, 3rd, and S. Normark. 1993. Pilus and nonpilus bacterial adhesins: assembly and function in cell recognition. Cell. 73:887-901.

Indik, Z.K., J.G. Park, S. Hunter, and A.D. Schreiber. 1995. The molecular dissection of Fc gamma receptor mediated phagocytosis. Blood. 86:4389-4399.

Iram, S.H., and J.E. Cronan. 2005. Unexpected functional diversity among FadR fatty acid transcriptional regulatory proteins. The Journal of biological chemistry. 280:32148-32156.

Jalalvand, F., and K. Riesbeck. 2014. Haemophilus influenzae: recent advances in the understanding of molecular pathogenesis and polymicrobial infections. Current opinion in infectious diseases. 27:268-274.

Jame, A.J., P.M. Lackie, A.M. Cazaly, I. Sayers, J.F. Penrose, S.T. Holgate, and A.P. Sampson. 2007. Human bronchial epithelial cells express an active and inducible biosynthetic pathway for leukotrienes B4 and C4. Clinical and experimental allergy : journal of the British Society for Allergy and Clinical Immunology. 37:880-892.

Johswich, K.O., S.E. McCaw, E. Islam, A. Sintsova, A. Gu, J.E. Shively, and S.D. Gray-Owen. 2013. In Vivo Adaptation and Persistence of Neisseria

Page 127: Identification and characterization of the novel CEACAM

References

115

meningitidis within the Nasopharyngeal Mucosa. PLoS pathogens. 9:e1003509.

Kammerer, R., T. Popp, S. Hartle, B.B. Singer, and W. Zimmermann. 2007. Species-specific evolution of immune receptor tyrosine based activation motif-containing CEACAM1-related immune receptors in the dog. BMC Evol Biol. 7:196.

Kammerer, R., T. Popp, B.B. Singer, J. Schlender, and W. Zimmermann. 2004. Identification of allelic variants of the bovine immune regulatory molecule CEACAM1 implies a pathogen-driven evolution. Gene. 339:99-109.

Kammerer, R., and W. Zimmermann. 2010. Coevolution of activating and inhibitory receptors within mammalian carcinoembryonic antigen (CEA) families. BMC Biology. 8:12.

Kirshner, J., C.J. Chen, P. Liu, J. Huang, and J.E. Shively. 2003. CEACAM1-4S, a cell-cell adhesion molecule, mediates apoptosis and reverts mammary carcinoma cells to a normal morphogenic phenotype in a 3D culture. Proceedings of the National Academy of Sciences of the United States of America. 100:521-526.

Kirshner, J., J. Hardy, S. Wilczynski, and J.E. Shively. 2004. Cell-cell adhesion molecule CEACAM1 is expressed in normal breast and milk and associates with beta1 integrin in a 3D model of morphogenesis. J Mol Histol. 35:287-299.

Klaile, E., T.E. Klassert, I. Scheffrahn, M.M. Muller, A. Heinrich, K.A. Heyl, H. Dienemann, C. Grunewald, R. Bals, B.B. Singer, and H. Slevogt. 2013. Carcinoembryonic antigen (CEA)-related cell adhesion molecules are co-expressed in the human lung and their expression can be modulated in bronchial epithelial cells by non-typable Haemophilus influenzae, Moraxella catarrhalis, TLR3, and type I and II interferons. Respiratory research. 14:85.

Klaile, E., M.M. Muller, C. Kannicht, B.B. Singer, and L. Lucka. 2005. CEACAM1 functionally interacts with filamin A and exerts a dual role in the regulation of cell migration. Journal of cell science. 118:5513-5524.

Klaile, E., O. Vorontsova, K. Sigmundsson, M.M. Muller, B.B. Singer, L.G. Ofverstedt, S. Svensson, U. Skoglund, and B. Obrink. 2009. The CEACAM1 N-terminal Ig domain mediates cis- and trans-binding and is essential for allosteric rearrangements of CEACAM1 microclusters. The Journal of cell biology. 187:553-567.

Korotkova, N., E. Cota, Y. Lebedin, S. Monpouet, J. Guignot, A.L. Servin, S. Matthews, and S.L. Moseley. 2006. A subfamily of Dr adhesins of Escherichia coli bind independently to decay-accelerating factor and the N-domain of carcinoembryonic antigen. The Journal of biological chemistry. 281:29120-29130.

Korotkova, N., Y. Yarova-Yarovaya, V. Tchesnokova, N. Yazvenko, M.A. Carl, A.E. Stapleton, and S.L. Moseley. 2008. Escherichia coli DraE adhesin-associated bacterial internalization by epithelial cells is promoted independently by decay-accelerating factor and carcinoembryonic antigen-related cell adhesion molecule binding and does not require the DraD invasin. Infection and immunity. 76:3869-3880.

Kraiss, A., S. Schlor, and J. Reidl. 1998. In vivo transposon mutagenesis in Haemophilus influenzae. Appl Environ Microbiol. 64:4697-4702.

Kuespert, K., and C.R. Hauck. 2009. Characterizing host receptor recognition by individual bacterial pathogens. In Methods in Molecular Biology. Vol. 470. S. Rupp, K. Sohn, and N. Hauser, editors. Humana Press, Totowa. 57-65.

Page 128: Identification and characterization of the novel CEACAM

References

116

Kuespert, K., S. Pils, and C.R. Hauck. 2006. CEACAMs - their role in physiology and pathophysiology. Current opinion in cell biology. 18:565-571.

Kuespert, K., A. Roth, and C.R. Hauck. 2011. Neisseria meningitidis has two independent modes of recognizing its human receptor CEACAM1. PloS one. 6:e14609.

Kuespert, K., S. Weibel, and C.R. Hauck. 2007. Profiling of bacterial adhesin – host receptor recognition by soluble immunoglobulin superfamily domains. Journal of Microbiological Methods. 68:478-485.

Langereis, J.D., and J.N. Weiser. 2014. Shielding of a lipooligosaccharide IgM epitope allows evasion of neutrophil-mediated killing of an invasive strain of nontypeable Haemophilus influenzae. mBio. 5:e01478-01414.

Laurie, N.A., M.M. Comegys, M.P. Carreiro, J.F. Brown, D.L. Flanagan, K.E. Brilliant, and D.C. Hixson. 2005. Carcinoembryonic antigen-related cell adhesion molecule 1a-4L suppression of rat hepatocellular carcinomas. Cancer research. 65:11010-11017.

Lawson, E.L., D.R. Mills, K.E. Brilliant, and D.C. Hixson. 2012. The transmembrane domain of CEACAM1-4S is a determinant of anchorage independent growth and tumorigenicity. PloS one. 7:e29606.

Lepore, B.W., M. Indic, H. Pham, E.M. Hearn, D.R. Patel, and B. van den Berg. 2011. Ligand-gated diffusion across the bacterial outer membrane. Proceedings of the National Academy of Sciences of the United States of America. 108:10121-10126.

Leung, N., C. Turbide, M. Olson, V. Marcus, S. Jothy, and N. Beauchemin. 2006. Deletion of the carcinoembryonic antigen-related cell adhesion molecule 1 (Ceacam1) gene contributes to colon tumor progression in a murine model of carcinogenesis. Oncogene. 25:5527-5536.

Leusch, H.G., Z. Drzeniek, Z. Markos-Puztai, and C. Wagener. 1991. Binding of Escherichia coli and Salmonella strains to members of the carcinoembryonic antigen family: differential binding inhibition by aromatic glycosides of mannose. Infection & Immunity. 59:2051-2057.

Leusch, H.G., S.A. Hefta, Z. Drzeniek, K. Hummel, Z. Markos-Pusztai, and C. Wagener. 1990. Escherichia coli of human origin binds to carcinoembryonic antigen (CEA) and non-specific crossreacting antigen (NCA). FEBS letters. 261:405-409.

Li, C., C.J. Chen, and J.E. Shively. 2009. Mutational analysis of the cytoplasmic domain of CEACAM1-4L in humanized mammary glands reveals key residues involved in lumen formation: stimulation by Thr-457 and inhibition by Ser-461. Experimental cell research. 315:1225-1233.

Li, Y., H. Cao, Z. Jiao, S.B. Pakala, D.N. Sirigiri, W. Li, R. Kumar, and L. Mishra. 2010. Carcinoembryonic antigen interacts with TGF-{beta} receptor and inhibits TGF-{beta} signaling in colorectal cancers. Cancer research. 70:8159-8168.

Lichtenegger, S., I. Bina, S. Roier, S. Bauernfeind, K. Keidel, S. Schild, M. Anthony, and J. Reidl. 2014. Characterization of lactate utilization and its implication on the physiology of Haemophilus influenzae. International journal of medical microbiology : IJMM. 304:490-498.

Lin, J., S. Huang, and Q. Zhang. 2002. Outer membrane proteins: key players for bacterial adaptation in host niches. Microbes and infection / Institut Pasteur. 4:325-331.

Lobo, E.O., Z. Zhang, and J.E. Shively. 2009. Pivotal advance: CEACAM1 is a negative coreceptor for the B cell receptor and promotes CD19-mediated

Page 129: Identification and characterization of the novel CEACAM

References

117

adhesion of B cells in a PI3K-dependent manner. Journal of leukocyte biology. 86:205-218.

McCaw, S.E., E.H. Liao, and S.D. Gray-Owen. 2004. Engulfment of Neisseria gonorrhoeae: revealing distinct processes of bacterial entry by individual carcinoembryonic antigen-related cellular adhesion molecule family receptors. Infection and immunity. 72:2742-2752.

Mes, T.H., and J.P. van Putten. 2007. Positively selected codons in immune-exposed loops of the vaccine candidate OMP-P1 of Haemophilus influenzae. Journal of molecular evolution. 64:411-422.

Morton, D.J., A. Smith, T.M. VanWagoner, T.W. Seale, P.W. Whitby, and T.L. Stull. 2007. Lipoprotein e (P4) of Haemophilus influenzae: role in heme utilization and pathogenesis. Microbes and infection / Institut Pasteur. 9:932-939.

Muenzner, P., V. Bachmann, J. Hentschel, W. Zimmermann, and C.R. Hauck. 2010. Human-specific bacterial pathogens block shedding of epithelial cells by stimulating integrin activation. Science. 329:1197-1201.

Muenzner, P., V. Bachmann, K. Kuespert, and C.R. Hauck. 2008. The CEACAM1 transmembrane domain, but not the cytoplasmic domain, directs internalization of human pathogens via membrane-microdomains. Cellular microbiology. 10:1074-1092.

Muenzner, P., O. Billker, T.F. Meyer, and M. Naumann. 2002. Nuclear factor-kB directs CEACAM1 receptor expression in Neisseria gonorrhoeae-infected epithelial cells. Journal of Biological Chemistry. 277:7438-7446.

Muenzner, P., M. Naumann, T.F. Meyer, and S.D. Gray-Owen. 2001. Pathogenic Neisseria Trigger Expression of Their Carcinoembryonic Antigen-related Cellular Adhesion Molecule 1 (CEACAM1; Previously CD66a) Receptor on Primary Endothelial Cells by Activating the Immediate Early Response Transcription Factor, Nuclear Factor-kappa B. Journal of Biological Chemistry. 276:24331-24340.

Muenzner, P., M. Rohde, S. Kneitz, and C.R. Hauck. 2005. CEACAM engagement by human pathogens enhances cell adhesion and counteracts bacteria-induced detachment of epithelial cells. Journal of Cell Biology. 170:825-836.

Muller, M.M., E. Klaile, O. Vorontsova, B.B. Singer, and B. Obrink. 2009. Homophilic adhesion and CEACAM1-S regulate dimerization of CEACAM1-L and recruitment of SHP-2 and c-Src. The Journal of cell biology. 187:569-581.

Muller, M.M., B.B. Singer, E. Klaile, B. Obrink, and L. Lucka. 2005. Transmembrane CEACAM1 affects integrin-dependent signaling and regulates extracellular matrix protein-specific morphology and migration of endothelial cells. Blood. 105:3925-3934.

Munson, R., Jr., and S. Grass. 1988. Purification, cloning, and sequence of outer membrane protein P1 of Haemophilus influenzae type b. Infection and immunity. 56:2235-2242.

Munson, R., Jr., and A. Hunt. 1989. Isolation and characterization of a mutant of Haemophilus influenzae type b deficient in outer membrane protein P1. Infection and immunity. 57:1002-1004.

Murphy, T.F., H. Faden, L.O. Bakaletz, J.M. Kyd, A. Forsgren, J. Campos, M. Virji, and S.I. Pelton. 2009. Nontypeable Haemophilus influenzae as a pathogen in children. Pediatr Infect Dis J. 28:43-48.

Murray, J., C. Ward, J.T. O'Flaherty, I. Dransfield, C. Haslett, E.R. Chilvers, and A.G. Rossi. 2003. Role of leukotrienes in the regulation of human

Page 130: Identification and characterization of the novel CEACAM

References

118

granulocyte behaviour: dissociation between agonist-induced activation and retardation of apoptosis. British journal of pharmacology. 139:388-398.

Nagano, K., E.K. Read, Y. Murakami, T. Masuda, T. Noguchi, and F. Yoshimura. 2005. Trimeric structure of major outer membrane proteins homologous to OmpA in Porphyromonas gingivalis. Journal of bacteriology. 187:902-911.

Nguyen, T., C.J. Chen, and J.E. Shively. 2014. Phosphorylation of CEACAM1 Molecule by Calmodulin Kinase IID in a Three-dimensional Model of Mammary Gland Lumen Formation. The Journal of biological chemistry. 289:2934-2945.

Nicholson, T.B., and C.P. Stanners. 2006. Specific inhibition of GPI-anchored protein function by homing and self-association of specific GPI anchors. The Journal of cell biology. 175:647-659.

Nikaido, H. 2003. Molecular basis of bacterial outer membrane permeability revisited. Microbiology and molecular biology reviews : MMBR. 67:593-656.

Norskov-Lauritsen, N. 2014. Classification, identification, and clinical significance of haemophilus and aggregatibacter species with host specificity for humans. Clinical microbiology reviews. 27:214-240.

Nunn, W.D., and R.W. Simons. 1978. Transport of long-chain fatty acids by Escherichia coli: mapping and characterization of mutants in the fadL gene. Proceedings of the National Academy of Sciences of the United States of America. 75:3377-3381.

Obrink, B. 1997. CEA adhesion molecules: multifunctional proteins with signal-regulatory properties. Current opinion in cell biology. 9:616-626.

Ordonez, C., A.B. Zhai, P. Camacho-Leal, L. Demarte, M.M. Fan, and C.P. Stanners. 2007. GPI-anchored CEA family glycoproteins CEA and CEACAM6 mediate their biological effects through enhanced integrin alpha5beta1-fibronectin interaction. Journal of cellular physiology. 210:757-765.

Overath, P., and E.M. Raufuss. 1967. The induction of the enzymes of fatty acid degradation in Escherichia coli. Biochemical and biophysical research communications. 29:28-33.

Pages, J.M., C.E. James, and M. Winterhalter. 2008. The porin and the permeating antibiotic: a selective diffusion barrier in Gram-negative bacteria. Nature reviews. Microbiology. 6:893-903.

Patel, P.C., H.S. Lee, A.Y. Ming, A. Rath, C.M. Deber, C.M. Yip, J.V. Rocheleau, and S.D. Gray-Owen. 2013. Inside-out signaling promotes dynamic changes in the CEACAM1 oligomeric state to control its cell adhesion properties. The Journal of biological chemistry. 288(41):29654-29669.

Pils, S., K. Kopp, L. Peterson, J. Delgado-Tascon, N.J. Nyffenegger-Jann, and C.R. Hauck. 2012. The adaptor molecule Nck localizes the WAVE complex to promote actin polymerization during CEACAM3-mediated phagocytosis of bacteria. PloS one. 7:e32808.

Pils, S., T. Schmitter, F. Neske, and C.R. Hauck. 2006. Quantification of bacterial invasion into adherent cells by flow cytometry. Journal of Microbiological Methods. 65:301-310.

Poole, J., E. Foster, K. Chaloner, J. Hunt, M.P. Jennings, T. Bair, K. Knudtson, E. Christensen, R.S. Munson, Jr., P.L. Winokur, and M.A. Apicella. 2013. Analysis of nontypeable haemophilus influenzae phase-variable genes during experimental human nasopharyngeal colonization. The Journal of infectious diseases. 208:720-727.

Page 131: Identification and characterization of the novel CEACAM

References

119

Rahman, M.M., X.X. Gu, C.M. Tsai, V.S. Kolli, and R.W. Carlson. 1999. The structural heterogeneity of the lipooligosaccharide (LOS) expressed by pathogenic non-typeable Haemophilus influenzae strain NTHi 9274. Glycobiology. 9:1371-1380.

Rao, V.K., G.P. Krasan, D.R. Hendrixson, S. Dawid, and J.W. St Geme, 3rd. 1999. Molecular determinants of the pathogenesis of disease due to non-typable Haemophilus influenzae. FEMS Microbiol Rev. 23:99-129.

Rawling, E.G., F.S. Brinkman, and R.E. Hancock. 1998. Roles of the carboxy-terminal half of Pseudomonas aeruginosa major outer membrane protein OprF in cell shape, growth in low-osmolarity medium, and peptidoglycan association. Journal of bacteriology. 180:3556-3562.

Reddy, M.S., J.M. Bernstein, T.F. Murphy, and H.S. Faden. 1996. Binding between outer membrane proteins of nontypeable Haemophilus influenzae and human nasopharyngeal mucin. Infection and immunity. 64:1477-1479.

Reidl, J., and J.J. Mekalanos. 1996. Lipoprotein e(P4) is essential for hemin uptake by Haemophilus influenzae. The Journal of experimental medicine. 183:621-629.

Reidl, J., S. Schlor, A. Kraiss, J. Schmidt-Brauns, G. Kemmer, and E. Soleva. 2000. NADP and NAD utilization in Haemophilus influenzae. Molecular microbiology. 35:1573-1581.

Robertson, B.D., and T.F. Meyer. 1992. Genetic variation in pathogenic bacteria. Trends in Genetics. 8 (12):422-427.

Roier, S., T. Blume, L. Klug, G.E. Wagner, W. Elhenawy, K. Zangger, R. Prassl, J. Reidl, G. Daum, M.F. Feldman, and S. Schild. 2014. A basis for vaccine development: Comparative characterization of Haemophilus influenzae outer membrane vesicles. International journal of medical microbiology : IJMM:10.1016/j.ijmm.2014.1012.1005.

Roier, S., D.R. Leitner, J. Iwashkiw, K. Schild-Prufert, M.F. Feldman, G. Krohne, J. Reidl, and S. Schild. 2012. Intranasal immunization with nontypeable Haemophilus influenzae outer membrane vesicles induces cross-protective immunity in mice. PloS one. 7:e42664.

Rosadini, C.V., S. Ram, and B.J. Akerley. 2014. Outer membrane protein P5 is required for resistance of nontypeable Haemophilus influenzae to both the classical and alternative complement pathways. Infection and immunity. 82:640-649.

Roth, A., C. Mattheis, P. Münzner, M. Unemo, and C.R. Hauck. 2013. Innate recognition by neutrophil granulocytes differs between Neisseria gonorrhoeae strains causing local or disseminating infections. Infection and immunity. 81(7):2358-2370.

Samara, R.N., L.M. Laguinge, and J.M. Jessup. 2007. Carcinoembryonic antigen inhibits anoikis in colorectal carcinoma cells by interfering with TRAIL-R2 (DR5) signaling. Cancer research. 67:4774-4782.

Samineni, S., C. Glackin, and J.E. Shively. 2011. Role of CEACAM1, ECM, and mesenchymal stem cells in an orthotopic model of human breast cancer. Int J Breast Cancer. 2011:381080.

Schlor, S., G. Kemmer, and J. Reidl. 2003. Transposon Tn10. Methods in molecular medicine. 71:211-224.

Schmitter, T., F. Agerer, L. Peterson, P. Muenzner, and C.R. Hauck. 2004. Granulocyte CEACAM3 is a phagocytic receptor of the innate immune system that mediates recognition and elimination of human-specific pathogens. Journal of Experimental Medicine. 199:35-46.

Page 132: Identification and characterization of the novel CEACAM

References

120

Schmitter, T., S. Pils, S. Weibel, F. Agerer, A. Buntru, K. Kopp, and C.R. Hauck. 2007. Opa proteins of pathogenic Neisseriae initiate Src-kinase-dependent or lipid raft-mediated uptake via distinct human CEACAM isoforms. Infection & Immunity. 75:4116-4126.

Screaton, R.A., L. DeMarte, P. Draber, and C.P. Stanners. 2000. The specificity for the differentiation blocking activity of carcinoembryonic antigen resides in its glycophosphatidyl-inositol anchor. The Journal of cell biology. 150:613-626.

Segada, L.M., G.M. Carlone, L.L. Gheesling, and A.J. Lesse. 2000. Characterization of P1-deficient isogenic mutant of Haemophilus influenzae biogroup aegyptius associated with Brazilian purpuric fever. Microbial pathogenesis. 28:145-155.

Servin, A.L. 2005. Pathogenesis of Afa/Dr diffusely adhering Escherichia coli. Clinical microbiology reviews. 18:264-292.

Sievers, F., A. Wilm, D. Dineen, T.J. Gibson, K. Karplus, W. Li, R. Lopez, H. McWilliam, M. Remmert, J. Soding, J.D. Thompson, and D.G. Higgins. 2011. Fast, scalable generation of high-quality protein multiple sequence alignments using Clustal Omega. Mol Syst Biol. 7:539.

Singer, B.B., E. Klaile, I. Scheffrahn, M.M. Muller, R. Kammerer, W. Reutter, B. Obrink, and L. Lucka. 2005. CEACAM1 (CD66a) mediates delay of spontaneous and Fas ligand-induced apoptosis in granulocytes. European journal of immunology. 35:1949-1959.

Singer, B.B., I. Scheffrahn, R. Kammerer, N. Suttorp, S. Ergun, and H. Slevogt. 2010. Deregulation of the CEACAM expression pattern causes undifferentiated cell growth in human lung adenocarcinoma cells. PloS one. 5:e8747.

Singer, B.B., I. Scheffrahn, and B. Obrink. 2000. The tumor growth-inhibiting cell adhesion molecule CEACAM1 (C-CAM) is differently expressed in proliferating and quiescent epithelial cells and regulates cell proliferation. Cancer research. 60:1236-1244.

Singh, B., Y.C. Su, T. Al-Jubair, O. Mukherjee, T. Hallstrom, M. Morgelin, A.M. Blom, and K. Riesbeck. 2014. A fine-tuned interaction between trimeric autotransporter haemophilus surface fibrils and vitronectin leads to serum resistance and adherence to respiratory epithelial cells. Infection and immunity. 82:2378-2389.

Sirakova, T., P.E. Kolattukudy, D. Murwin, J. Billy, E. Leake, D. Lim, T. DeMaria, and L. Bakaletz. 1994. Role of fimbriae expressed by nontypeable Haemophilus influenzae in pathogenesis of and protection against otitis media and relatedness of the fimbrin subunit to outer membrane protein A. Infection and immunity. 62:2002-2020.

Slevogt, H., S. Zabel, B. Opitz, A. Hocke, J. Eitel, D. N'Guessan P, L. Lucka, K. Riesbeck, W. Zimmermann, J. Zweigner, B. Temmesfeld-Wollbrueck, N. Suttorp, and B.B. Singer. 2008. CEACAM1 inhibits Toll-like receptor 2-triggered antibacterial responses of human pulmonary epithelial cells. Nature immunology. 9:1270-1278.

St. Geme III, J.W. 2002. Molecular and cellular determinants of non-typeable Haemophilus influenzae adherence and invasion. Cellular microbiology. 4:191 - 200.

St. Geme III, J.W., and D. Cutter. 1996. Influence of pili, fibrils, and capsule on in vitro adherence by Haemophilus influenzae type b. Molecular microbiology. 21.

Page 133: Identification and characterization of the novel CEACAM

References

121

Stern, A., M. Brown, P. Nickel, and T.F. Meyer. 1986. Opacity genes in Neisseria gonorrhoeae: Control of phase and antigenic variation. Cell. 47:61-71.

Su, Y.C., O. Mukherjee, B. Singh, O. Hallgren, G. Westergren-Thorsson, D. Hood, and K. Riesbeck. 2015. Haemophilus influenzae P4 Interacts With Extracellular Matrix Proteins Promoting Adhesion and Serum Resistance. The Journal of infectious diseases.

Swords, W.E., B.A. Buscher, K. Ver Steeg Ii, A. Preston, W.A. Nichols, J.N. Weiser, B.W. Gibson, and M.A. Apicella. 2000. Non-typeable Haemophilus influenzae adhere to and invade human bronchial epithelial cells via an interaction of lipooligosaccharide with the PAF receptor. Molecular microbiology. 37:13-27.

Tchoupa, A.K., S. Lichtenegger, J. Reidl, and C.R. Hauck. 2015. Outer membrane protein P1 is the CEACAM-binding adhesin of Haemophilus influenzae. Molecular microbiology.

Tchoupa, A.K., T. Schuhmacher, and C.R. Hauck. 2014. Signaling by epithelial members of the CEACAM family - mucosal docking sites for pathogenic bacteria. Cell Commun Signal. 12:27.

Teglund, S., A. Olsen, W.N. Khan, L. Frangsmyr, and S. Hammarstrom. 1994. The pregnancy-specific glycoprotein (PSG) gene cluster on human chromosome 19: fine structure of the 11 PSG genes and identification of 6 new genes forming a third subgroup within the carcinoembryonic antigen (CEA) family. Genomics. 23:669-684.

Teixeira, A.M., J. Fawcett, D.L. Simmons, and S.M. Watt. 1994. The N-domain of the biliary glycoprotein (BGP) adhesion molecule mediates homotypic binding: domain interactions and epitope analysis of BGPc. Blood. 84:211-219.

Toleman, M., E. Aho, and M. Virji. 2001. Expression of pathogen-like Opa adhesins in commensal Neisseria: genetic and functional analysis. Cellular microbiology. 3:33-44.

Turbide, C., T. Kunath, E. Daniels, and N. Beauchemin. 1997. Optimal ratios of biliary glycoprotein isoforms required for inhibition of colonic tumor cell growth. Cancer research. 57:2781-2788.

van den Berg, B. 2005. The FadL family: unusual transporters for unusual substrates. Current opinion in structural biology. 15:401-407.

van den Berg, B., P.N. Black, W.M. Clemons, Jr., and T.A. Rapoport. 2004. Crystal structure of the long-chain fatty acid transporter FadL. Science. 304:1506-1509.

Van Eldere, J., M.P. Slack, S. Ladhani, and A.W. Cripps. 2014. Non-typeable Haemophilus influenzae, an under-recognised pathogen. The Lancet. Infectious diseases. 14:1281-1292.

Virji, M. 2009. Pathogenic Neisseriae: surface modulation, pathogenesis and infection control. Nature reviews. Microbiology. 7:274-286.

Virji, M., D. Evans, J. Griffith, D. Hill, L. Serino, A. Hadfield, and S.M. Watt. 2000. Carcinoembryonic antigens are targeted by diverse strains of typable and non-typable Haemophilus influenzae. Molecular microbiology. 36:784-795.

Virji, M., K. Makepeace, D.J.P. Ferguson, and S.M. Watt. 1996a. Carcinoembryonic antigens (CD66) on epithelial cells and neutrophils are receptors for Opa proteins of pathogenic Neisseriae. Molecular microbiology. 22:941-950.

Virji, M., S.M. Watt, S. Barker, K. Makepeace, and R. Doyonnas. 1996b. The N-domain of the human CD66a adhesion molecule is a target for Opa proteins

Page 134: Identification and characterization of the novel CEACAM

References

122

of Neisseria meningitidis and Neisseria gonorrhoeae. Molecular microbiology. 22:929-939.

Voges, M., V. Bachmann, R. Kammerer, U. Gophna, and C.R. Hauck. 2010. CEACAM1 recognition by bacterial pathogens is species-specific. BMC Microbiology. 10:117.

Voges, M., V. Bachmann, J. Naujoks, K. Kopp, and C.R. Hauck. 2012. Extracellular IgC2-like domains of CEACAMS mediate PI3K sensitivity during uptake of pathogens. PloS One accepted. 7 e39908.

Wang, H., and J.E. Cronan. 2003. Haemophilus influenzae Rd lacks a stringently conserved fatty acid biosynthetic enzyme and thermal control of membrane lipid composition. Journal of bacteriology. 185:4930-4937.

Wang, J., S.D. Gray-Owen, A. Knorre, T.F. Meyer, and C. Dehio. 1998. Opa binding to cellular CD66 receptors mediates the transcellular traversal of Neisseria gonorrhoeae across polarized T84 epithelial cell monolayers. Molecular microbiology. 30:657-671.

Wang, Y. 2002. The function of OmpA in Escherichia coli. Biochemical and biophysical research communications. 292:396-401.

Watt, S.M., A.M. Teixeira, G.Q. Zhou, R. Doyonnas, Y. Zhang, F. Grunert, R.S. Blumberg, M. Kuroki, K.M. Skubitz, and P.A. Bates. 2001. Homophilic adhesion of human CEACAM1 involves N-terminal domain interactions: structural analysis of the binding site. Blood. 98:1469-1479.

Wikstrom, K., G. Kjellstrom, and B. Obrink. 1996. Homophilic intercellular adhesion mediated by C-CAM is due to a domain 1-domain 1 reciprocal binding. Experimental cell research. 227:360-366.

Wong, C., Y. Liu, J. Yip, R. Chand, J.L. Wee, L. Oates, B. Nieswandt, A. Reheman, H. Ni, N. Beauchemin, and D.E. Jackson. 2009. CEACAM1 negatively regulates platelet-collagen interactions and thrombus growth in vitro and in vivo. Blood. 113:1818-1828.

Wu, S., M.M. Baum, J. Kerwin, D. Guerrero, S. Webster, C. Schaudinn, D. VanderVelde, and P. Webster. 2014. Biofilm-specific extracellular matrix proteins of nontypeable Haemophilus influenzae. Pathogens and disease. 72:143-160.

Yokoyama, S., C.J. Chen, T. Nguyen, and J.E. Shively. 2007. Role of CEACAM1 isoforms in an in vivo model of mammary morphogenesis: mutational analysis of the cytoplasmic domain of CEACAM1-4S reveals key residues involved in lumen formation. Oncogene. 26:7637-7646.

Zebhauser, R., R. Kammerer, A. Eisenried, A. McLellan, T. Moore, and W. Zimmermann. 2005. Identification of a novel group of evolutionarily conserved members within the rapidly diverging murine Cea family. Genomics. 86:566-580.

Zhang, H., A. Eisenried, W. Zimmermann, and J.E. Shively. 2013. Role of CEACAM1 and CEACAM20 in an in vitro model of prostate morphogenesis. PloS one. 8:e53359.

Zheng, J., K.K. Miller, T. Yang, M.S. Hildebrand, A.E. Shearer, A.P. DeLuca, T.E. Scheetz, J. Drummond, S.E. Scherer, P.K. Legan, R.J. Goodyear, G.P. Richardson, M.A. Cheatham, R.J. Smith, and P. Dallos. 2011. Carcinoembryonic antigen-related cell adhesion molecule 16 interacts with alpha-tectorin and is mutated in autosomal dominant hearing loss (DFNA4). Proceedings of the National Academy of Sciences of the United States of America. 108:4218-4223.

Page 135: Identification and characterization of the novel CEACAM

References

123

Zhou, H., A. Fuks, G. Alcaraz, T.J. Bolling, and C.P. Stanners. 1993a. Homophilic adhesion between Ig superfamily carcinoembryonic antigen molecules involves double reciprocal bonds. The Journal of cell biology. 122:951-960.

Zhou, H., C.P. Stanners, and A. Fuks. 1993b. Specificity of anti-carcinoembryonic antigen monoclonal antibodies and their effects on CEA-mediated adhesion. Cancer research. 53:3817-3822.