yersinia-phagocyte interactions during early infection

71
Yersinia-phagocyte interactions during early infection Linda Westermark Department of Molecular Biology Umeå Center for Microbial Research (UCMR) Laboratory for Molecular Infection Medicine Sweden (MIMS) Umeå University Umeå 2013

Upload: others

Post on 03-Feb-2022

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Yersinia-phagocyte interactions during early infection

Yersinia-phagocyte interactions during early infection

Linda Westermark

Department of Molecular Biology

Umeå Center for Microbial Research (UCMR) Laboratory for Molecular Infection Medicine Sweden (MIMS) Umeå University

Umeå 2013

Page 2: Yersinia-phagocyte interactions during early infection

Copyright © Linda Westermark

ISBN: 978-91-7459-713-4 ISSN: 0346-6612 New series: 1591

Cover picture: Dendritic cell and Y. pseudotuberculosis Printed by: Department of Chemistry Printing Service Umeå, Sweden 2013

Page 3: Yersinia-phagocyte interactions during early infection

To all my loved ones

Follow your heart wherever it may lead you…

Page 4: Yersinia-phagocyte interactions during early infection
Page 5: Yersinia-phagocyte interactions during early infection

i

TABLE OF CONTENTS

TABLE OF CONTENTS i

ABSTRACT iii

PUBLICATIONS AND MANUSCRIPTS iv

ABBREVIATIONS v

SAMMANFATTNING PÅ SVENSKA vii

INTRODUCTION 1

1. Yersinia 1

1.1 The Yersinia genus - Species and diseases 1

1.2 Type III secretion system 2

1.2.1 The T3SS apparatus 2

1.2.2 The Yop translocators 3

1.2.3 The Yop effectors 3

1.2.4 The Yop regulators 5

1.2.5 Regulation of T3SS 6

1.3 Adhesins 6

1.3.1 Invasin 7

1.3.2 YadA 7

1.3.3 Ail 7

1.4 Significance of studying Y. pseudotuberculosis 8

2. Phagocytes 9

2.1 Internalization by phagocytosis and micropinocytosis 9

2.2 Monocytes 10

2.2.1 Monocyte subtypes and functions 10

2.2.2 How to study monocytes? 10

2.3 Macrophages 11

2.3.1 Macrophage subtypes and functions 11

2.3.2 Macrophage sensing of pathogens 11

2.3.3 How to study macrophages? 11

2.4 Neutrophils 12

2.4.1 PMN subtypes and functions 13

2.4.2 PMN sensing of pathogens 14

2.4.3 How to study PMNs? 14

2.5 Dendritic cells 14

2.5.1 DC subtypes and functions 15

2.5.2 DC sensing of pathogens 16

2.5.3 How to study DCs? 17

3. Yersinia infection 19

3.1 Infection route and bacterial dissemination 19

3.1.1 Enteric Yersinia infection in humans 19

3.1.2 Enteric Yersinia infection models in mice 19

Page 6: Yersinia-phagocyte interactions during early infection

ii

3.2 Immune responses during enteric Yersinia infection in mice 22

3.2.1 Yersinia and macrophages 22

3.2.2 Yersinia and neutrophils 23

3.2.3 Yersinia and dendritic cells 24

3.2.4 Yersinia and B and T lymphocytes 24

3.3 Relevance of using mice as a model for human bacterial diseases 25

AIMS 27

RESULTS & DISCUSSION 28

4. PAPER I – Cell type-specific effects of Yersinia pseudotuberculosis virulence effectors 28

4.1 Y. pseudotuberculosis inhibits dendritic cells maturation by the action of YopJ 28

4.2 Y. pseudotuberculosis resists internalization by dendritic cells through the action of YopE 28

4.3 The cell specific effect of YopH depends on the mechanism of internalization 30

5. PAPER II – Immune response to diphtheria toxin-mediated depletion complicates the use of the

CD11c-DTRtg model for studies of bacterial gastrointestinal infections. 33

5.1 DTx-mediated depletion of dendritic cells in the CD11c-DTRtg mouse model induces neutrophilia,

which results in lower Y. pseudotuberculosis infection 33

6. PAPER III – Yersinia pseudotuberculosis efficiently avoids polymorphonuclear neutrophils during

early infection 36

6.1 Y. pseudotuberculosis yopH and yopE mutants, but not the wt strain, interact with PMNs during

early infection 36

6.2 PMN depletion increase the virulence of Y. pseudotuberculosis 37

6.3 Undepleted immature PMNs compensate for the loss of mature PMNs in PMN-depleted mice 39

CONCLUSIONS 41

PAPER I 41

PAPER II 41

PAPER III 41

ACKNOWLEDGEMENTS 42

REFERENCES 44

PAPERS I-III 60

Page 7: Yersinia-phagocyte interactions during early infection

iii

ABSTRACT

Pathogenic Gram-negative Yersinia species preferentially target and inactivate

phagocytic cells of the innate immune defense by translocation of effector Yersinia

outer proteins (Yops) into the cells via a type III secretion system. This indicates

that inactivation and avoidance of the early innate immune response is an efficient

way for Yersinia species to avoid elimination and to cause diseases ranging from

mild gastroenteritis (Y. pseudotuberculosis and Y. enterocolitica) to plague (Y.

pestis). In this project, we aimed to study the interaction between

enteropathogenic Y. pseudotuberculosis and phagocytic cells during early infection.

In situ interaction studies on infected intestinal tissues showed that Y.

pseudotuberculosis mainly interacts with dendritic cells (DCs) in lymphoid tissues of

the intestine during initial infection. After massive recruitment of

polymorphonuclear neutrophils (PMNs) to the infected tissues, wild-type (wt)

bacteria also interacted with this phagocyte. In contrast to the wt, mutants lacking

the anti-phagocytic effectors YopH and YopE are avirulent in mice and unable to

spread systemically. Interestingly, our interaction assay showed that these mutants

not only interacted with DCs, but also with PMNs during the initial stage of

infection. Thus, indicating that Y. pseudotuberculosis can avoid interaction with

PMNs during early infection and that this is Yop-dependent. In a phagocytosis assay

Y. pseudotuberculosis was demonstrated to inhibit internalization by DCs in a YopE-

dependent manner, while both YopH and YopE were shown to be involved in the

blocking of phagocytosis by macrophages and PMNs. Thus, indicating that YopH

has cell type-specific effects. To further investigate the role of DCs during initial

stages of infection, a mouse DC depletion model (CD11c-DTRtg

) was applied.

However, the DTx-mediated depletion of DCs in CD11c-DTRtg

mice induced

neutrophilia and the model could not give a definite answer to whether DCs play

an important role in either restricting or stimulating progression of Y.

pseudotuberculosis infection. To investigate involvement of PMNs during early

infection mice were injected with the depleting antibody α-Ly6G. In absence of

PMNs wt, as well as yopH and yopE mutants became more virulent, which further

supports the importance of these Yops for the ability of Y. pseudotuberculosis to

disseminate from the initial infection sites in the intestine to cause systemic

disease.

In summary, our studies show that inhibiting internalization and maturation of DCs

and avoiding phagocytosis by and interaction with macrophages and PMNs during

the early stages of infection are important virulence strategies for Y.

pseudotuberculosis to be able to colonize tissues, proliferate and disseminate

systemically.

Page 8: Yersinia-phagocyte interactions during early infection

iv

PUBLICATIONS AND MANUSCRIPTS

This thesis is based on the following publications and manuscript, which are

referred to in the text by their roman numerals (I-III).

I. Cell-type-specific effects of Yersinia pseudotuberculosis virulence

effectors.

Anna Fahlgren*, Linda Westermark*, Karen Akopyan, Maria Fällman.

Cell Microbiol. 2009 Dec;11(12):1750-67. (*Both authors contributed

equally)

II. Immune response to diphtheria toxin-mediated depletion complicates

the use of the CD11c-DTR(tg) model for studies of bacterial gastrointestinal infections. Linda Westermark, Anna Fahlgren, Maria Fällman.

Microb Pathog. 2012 Sep;53(3-4):154-61.

III. Yersinia pseudotuberculosis efficiently avoids polymorphonuclear

neutrophils during early infection Linda Westermark, Anna Fahlgren, Maria Fällman.

Manuscript

Page 9: Yersinia-phagocyte interactions during early infection

v

ABBREVIATIONS

Ail Attachment and invasion locus

BM-DC Bone marrow-derived dendritic cell

BM-M Bone marrow-derived macrophage

BM-PMN Bone marrow-derived neutrophil

cDC Conventional dendritic cell

CFU Colony forming unit

DC Dendritic cell

DNA Deoxyribonucleic acid

DTx Diphtheria toxin

DTR Diphtheria toxin receptor

GAP GTPase-activating protein

iDC Inflammatory dendritic cell

IFN Interferon

IL Interleukin

LPS Lipopolysaccharide

IVIS In vivo imaging system

LcrV Low calcium response V

M1 Classically activated macrophage

M2 Alternatively activated macrophage

M cell Microfold cell

MHCII Major histocompatibility complex II

MLN Mesenteric lymph node

MoDC Monocyte-derived dendritic cell

Mreg Regulatory macrophage

NET Neutrophil extracellular trap

NLR NOD-like receptor

PBMC-DC Peripheral blood monocyte-derived dendritic cell

pDC Plasmacytoid dendritic cell

p.i. Post infection

PMN Polymorphonuclear neutrophil

PAMP Pathogen-associated molecular pattern

PPs Peyer’s patches

PRR Pattern recognition receptor

PTPase Protein tyrosine phosphatase

T3SS Type III secretion system

TAM Tumor-associated macrophage

TGF Transforming growth factor

TipDC TNF- and iNOS-producing dendritic cell

TLR Toll-like receptor

TNF Tumor necrosis factor

Page 10: Yersinia-phagocyte interactions during early infection

vi

YadA Yersinia adhesin A

Yop Yersinia outer protein

YpkA Yersinia protein kinase A

Ysc Yersinia secretion protein

wt wild- type

Page 11: Yersinia-phagocyte interactions during early infection

vii

SAMMANFATTNING PÅ SVENSKA

Bakterier i Yersinia-genuset infekterar djur och människor och kan orsaka

sjukdomar som sträcker sig från mild gastroenterit (Y. pseudotuberculosis and Y.

enterocolitica) till pest (Y. pestis). De tarmrelaterade Yersinia-infektionerna orsakas

av förtäring av kontaminerad föda och karaktäriseras av diarré, kräkningar,

magsmärtor och feber. Dessa infektioner är vanligtvis självbegränsande hos friska

människor, men kan leda till dödliga, systemiska infektioner hos möss.

När Y. pseudotuberculosis and Y. enterocolitica infekterar via den orala vägen

invaderar de lymphoida vävnader i tarmen via M celler. När bakterierna når

Peyer’s patches och de lymfoida follicklarna i cecum (motsvarar blindtarmen hos

människa) stöter de på immunförsvaret. Försvaret utgörs initialt av makrofager och

dendritiska celler, som har till huvuduppgift att eliminera inkräktare via fagocytos

("cellätande") och starta det specifika försvaret via antigenpresentation. Under

infektionen rekryteras också neutrofiler till infektionsplatsen för att hjälpa

makrofagerna i elimineringen. Väldigt få bakterier kan motstå immunförsvaret i

tarmvävnaden och många infektioner elimineras därför redan här.

Sjukdomsalstrande arter av Yersinia kan dock undvika eliminering genom att stänga

av viktiga immunförsvarsmekanismer (inklusive fagocytos och

inflammationsrespons) och därefter sprida sig vidare till mesenteriska lymfnoder,

mjälte och lever för att orsaka systemisk infektion. Yersinias förmåga att undvika

immunförsvaret är beroende av dess så kallade typ III sekretionssystem (T3SS). Vid

kontakt med en immuncell använder Yersinia sekretionssystemet för att överföra

Yopar (Yersinia outer proteins) till immuncellen. Yoparna stänger av immuncellen

genom att tex blockera fagocytos och mognad, samt orsaka celldöd.

YopH och YopE har identifierats som de två viktigaste Yoparna för att undvika

fagocytos hos makrofager och neutrofiler. YopH är ett effektivt protein tyrosin

fosfatas som stänger av proteiner som är involverade i signalsystemet som är

kopplat till fagocytiska receptorer. YopE har så kallad GAP-aktivitet och kan stänga

av Rho GTPaser som styr många cellulära funktioner, bland annat aktindynamik

som är viktigt för fagocytos. YopJ är ett serin/threonin acetyltransferas som stänger

av olika signalvägar i immuncellerna och påverkar till exempel celldöd och

cytokinproducering i infekterade celler.

I vår första studie visade vi att Y. pseudotuberculosis kan stänga av

mognadsprocessen hos dendritiska celler med YopJ och att bakterien kan undvika

fagocytos. De anti-fagocytiska Yoparna hos Y. pseudotuberculosis visade sig ha

cellspecifika effekter eftersom bakterien använder både YopH och YopE för att

undvika fagocytos hos makrofager och neutrofiler, medan endast YopE har en

effekt mot dendritiska celler. Detta visade sig beror på att dendritiska celler har en

Page 12: Yersinia-phagocyte interactions during early infection

viii

annorlunda mekanism för att internalisera bakterier, som inte involverar

receptorsignalering.

I den andra studien fortsatte vi undersöka hur Y. pseudotuberculosis interagerar

med dendritiska celler genom att använda en musmodell (CD11c-DTRtg

) i vilken

man kan ta bort dessa celler genom att behandla mössen med difteritoxin. Tyvärr

visade det sig att stora mängder neutrofiler kom till vävnaden där de dendritiska

cellerna tagits bort och att dessa sedan eliminerade bakterierna när de försökte

infektera mössen. Genom att förskjuta infektionen två dagar, tills de flesta

neutrofilerna hade försvunnit kunde vi konstatera att dendritiska celler

förmodligen inte spelar en särskilt viktig roll under de tidiga stadierna av Y.

pseudotuberculosis-infektion. Vi drog också slutsatsen att denna modell måste

användas varsamt vid infektionsstudier och att sådana inte kan initieras förrän

immunresponsen har gått tillbaka efter behandlingen.

I den tredje studien tittade vi på hur Y. pseudotuberculosis interagerar med

immunceller i de lymfoida vävnaderna i tarmen under de första stadierna av oral

infektion. Det visade sig att vildtypsbakterier initialt bara interagerar med

dendritiska celler och först senare med neutrofiler. Mutanter som saknar de anti-

fagocytiska Yoparna, YopH eller YopE interagerar med dendriska celler lika ofta som

vildtypen, men interagerar också med neutrofiler. Detta skulle kunna förklara

varför yopH-och yopE-mutanter inte kan sprida sig systemiskt och orsaka sjukdom.

För att studera detta vidare injicerade vi möss med antikroppar som tar bort

neutrofiler och undersökte hur detta påverkade infektionen. Det visade sig att

vildtypen, men framför allt mutanterna blev bättre på att infektera mössen om

neutrofiler inte fanns. Resultaten visar att Y. pseudotuberculosis interagerar med

fagocytiska celler i tarmen under de första stadierna av oral infection, men att

Yoparna gör bakterien tillräckligt effektiv för att kunna undvika immunförsvaret,

sprida sig systemiskt och orsaka en infektion med dödlig utgång. Om bakterien

däremot saknar någon av de viktiga Yoparna blir den stoppad och kan inte sprida

sig systemiskt. Neutrofiler verkar vara ansvariga för denna blockering eftersom

mutanterna kan sprida sig snabbare i möss som saknar neutrofiler.

Sammanfattningsvis visar våra studier att Y. pseudotuberculosis genom att

förhindra internalisering och mognad av dendritiska celler och genom att undvika

fagocytos och interaktion med makrofager och neutrofiler i ett tidigt stadium under

infektionen, kan kolonisera vävnader, föröka sig och sprida sig vidare till inre organ.

Page 13: Yersinia-phagocyte interactions during early infection

1

INTRODUCTION

1. Yersinia

1.1 The Yersinia genus - Species and diseases

The Yersinia genus consists of Gram-negative, rod-shaped bacteria. The genus is

named after the Swiss bacteriologist Alexandre Yersin, who independently of

Shibasaburo Kitasato successfully isolated and described Y. pestis in Hong Kong in

year 1894, during the third plague pandemic [1]. The Yersinia genus includes in

total 15 identified species, represented among environmental, commensal, as well

as pathogenic bacteria. Y. pestis, Y. pseudotuberculosis, and Y. enterocolitica are

the extensively studied and characterized human pathogenic Yersinia species. They

harbor a 70-kb virulence plasmid (pYV) found in all disease causing strains, but not

in the other Yersinia strains [2]. Y. frederiksenii, Y. intermedia, Y. kristensenii, Y.

bercovieri, Y. mollaretii, Y. rohdei, Y. ruckeri, and Y. aldovae are considered to be

non-pathogenic to humans. They are sometimes referred to as Y. enterocolitica-like

bacteria, and have not been studied to the same extent. However, the majority of

these species have been isolated from humans and animals, and it is suggested that

some of them may cause disease with virulence factors different from Y.

enterocolitica [3].

Y. pestis is the causative agent of plague and is responsible for several severe

pandemics and the death of millions of people throughout history. Even though

large plague pandemics do not arise today, small outbreaks of plague still occur in

parts of Asia, Africa and America. Nowadays, Y. pestis is also considered to be a

major treat as a bioweapon due the serious disease it causes and to the occurrence

of antibiotic resistant species. Y. pestis spread via bites from infected Xenopsylla

fleas or aerosols and cause bubonic, septicemia or pneumonic plague depending

on the way of transmission. Unless treated with antibiotics, the mortality rate is

very high [2].

The enteric Yersinia species, Y. pseudotuberculosis and Y. enterocolitica are the

most commonly encountered. They can be found in aquatic environments, soil and

animals, and cause the disease yersiniosis worldwide. Yersiniosis is a

gastrointestinal disease characterized by symptoms like fever, abdominal pain,

vomiting, and diarrhea. Infections are spread through contaminated food and

water, and are usually self-limiting in humans [2]. However, enteric Yersinia

infections have been linked to human inflammatory autoimmune diseases, e.g.

Crohn’s disease and reactive arthritis [4, 5], and Y. pseudotuberculosis infections

have also been connected to Far East scarlet-like fever and Kawasaki disease [6, 7].

Page 14: Yersinia-phagocyte interactions during early infection

2

1.2 Type III secretion system

The ability of bacteria to deliver virulence effectors into host cells is essential for

infection, survival and pathogenesis for many Gram-negative bacteria. Bacteria use

eight different so called secretion systems to transport proteins over their cell wall.

The cell wall of Gram-negative bacteria consists of two membranes, the inner and

outer membrane. Many pathogenic Gram-negative bacteria use a type III secretion

system (T3SS) to transfer proteins from their cytoplasm, over both membranes, to

the cytosol of target host cells [8]. In Yersinia the secretion system was first

discovered in 1990 by Cornelis and coworkers [9], but it was not denoted a T3SS

until Reeves and Salmond discovered the homology between proteins in the

Yersinia secretion system and proteins expressed by plant pathogens [10]. The

whole Yersinia T3SS, including proteins of the secretion apparatus, translocation

proteins, secreted proteins and regulatory proteins are encoded on a virulence

plasmid present in all pathogenic species [11].

In Yersinia the proteins secreted by the T3SS are called Yersinia outer membrane

proteins or Yops, and they can be divided into translocators (YopB , YopD, and

LcrV), effectors (YopH, YopE, YopJ/P, YopK, YopM, YopT, and YpkA/YopO), and

regulators (YopK/YopQ and YopE). The name was originally given because the Yops

were discovered before the secretion system, and were then found localized in the

outer membrane of Y. pseudotuberculosis and Y. enterocolitica [12, 13].

1.2.1 The T3SS apparatus

The T3SS apparatus is often structurally referred to as a syringe, injectisome, or a

needle-like nanomachine. The Ysc-T3SS apparatus of Yersinia is no exception. The

apparatus is build up by more than 20 different proteins forming a membrane

embedded basal structure, an external needle protruding from the bacterial

surface, and a tip complex that caps the needle [14, 15]. In Yersinia the basal

structure is build up by a number of Yersinia secretion proteins (Ysc), e.g. the

scaffold proteins YscCDJ and the export apparatus YscRSTUV. The needle is formed

by YscF and the tip complex contains LcrV.

The similarity between the needle complex and a syringe has generated a generally

accepted model that effectors are directly injected from the bacterial cytosol into

the cytoplasm of the targeted host cell. Nevertheless, there are no experimental

evidences showing that the effectors are secreted through the needle structure.

Instead a study visualizing the presence of Yop translocators and effectors on the

bacterial surface before target cell contact challenge this injectisome model. This

study proposes a two-step translocation model in which the T3SS substrates are

first secreted to the bacterial surface and thereafter translocated across the target

cell membrane upon contact [16].

Page 15: Yersinia-phagocyte interactions during early infection

3

1.2.2 The Yop translocators

Upon contact with host cells, Yop translocators (YopB and YopD) are assembled

into a translocon between the tip complex (LcrV) of the needle and the host cell

membrane. The translocon will form a pore in the host cell membrane and work as

a gateway for translocation of effector proteins [14].

1.2.2.1 YopB and YopD

YopB and YopD are transmembrane proteins responsible for forming the

translocation pore in the host cell membrane [17]. The two proteins bind each

other and are both required for pore formation to occur [18, 19]. Thereby, YopB

and YopD mediate the translocation of effector Yops into the host cell [20, 21].

Both yopB and yopD mutants are avirulent in mice [22, 23].

1.2.2.2 LcrV

Besides YopB and YopD, LcrV (Low calcium response V or the V antigen) has been

shown to be required for translocation of effector Yops [24-26]. LcrV has been

localized at the bacterial surface and can be found at the tip of the YscF needle [27,

28]. The protein is involved in the release of the pore forming proteins YopB and

YopD from the bacterium and in determining the size of the pore, but is not a

structural part of the Yop translocation pore [29, 30]. LcrV is necessary for the

virulence of Yersinia [31]. It is a protective antigen for infection by all three human

pathogenic Yersinia species and has been extensively studied as a vaccine

candidate against Y. pestis [32, 33].

1.2.3 The Yop effectors

The T3SS apparatus and the Yop translocators provide the means for infection, but

the real action is performed by the proteins secreted by the system – the Yop

effectors. In Yersinia there are six common Yop effectors: YopH, YopE, YopT YopJ/P,

YopM, and YpkA/YopO. After translocation into the host cell cytoplasm, the

effector proteins initiate and maintain infection by interfering with important host

cell functions, e.g. cell signaling, phagocytosis, and inflammatory responses [34].

1.2.3.1 YopH

YopH is a very potent and fast acting protein tyrosine phosphatase (PTPase) [35,

36]. After translocation YopH localizes to peripheral focal adhesion complexes in

the host cell [37, 38] and targets proteins involved in receptor signaling and

adaptor proteins, such as FAK, p130Cas, Fyb (also called ADAP and SLAP-130), and

SKAP-HOM [37-40]. Thereby, YopH inhibits Ca2+

signalling in neutrophils and

disrupts focal adhesion complexes resulting in inhibition of phagocytosis and the

associated oxidative burst in macrophages and neutrophils [37, 38, 41, 42]. YopH

has also been shown to target LAT and SLP-76 in T lymphocytes and suppress the

Page 16: Yersinia-phagocyte interactions during early infection

4

activation of B and T lymphocytes [43, 44]. Thus, YopH might have different targets

in different cell types. Certainly, this PTPase performs many important actions

during infection and subsequently, a yopH mutant is avirulent in mice [45, Paper

III]. The role of YopH in the strategies of Yersinia to avoid the immune defense will

be further discussed in section 3.2.

1.2.3.2 YopE

YopE is a GTPase-activating protein (GAP) that acts on GTPases of the Rho family

[46-48], which are important regulators of actin dynamics in eukaryotic cells [49].

In vitro YopE has been reported to stimulate GTP hydrolysis of Cdc42, RhoA and

Rac1 and thereby inhibit these proteins [46, 47]. However, inside cells YopE

preferentially inactivates Rac1 and RhoA, but not Cdc42 [48]. The GAP activity of

YopE is required for the anti-phagocytic capacity and virulence of Yersinia [46]. The

role of YopE as a regulator of Yop secretion and in the strategies of Yersinia to

avoid the immune defense will be further discussed in sections 1.2.4.2 and 3.2,

respectively.

1.2.3.6 YopT

The cysteine protease YopT inactivates Rho family GTPases by removing lipid

modifications [50]. In vitro, YopT can efficiently cleave recombinant RhoA, Rac and

Cdc42, while RhoA is the preferred substrates inside infected cells [51, 52]. YopT

has been reported to contribute to the anti-phagocytic ability of Y. enterocolitica

against phagocytic cells [51, 53, 54]. However, a Y. enterocolitica yopT mutant is as

virulent as the corresponding wt strain in mice [55, 56], showing that YopT is not

essential for causing disease. Further supporting this is the fact that serotype O3

strains of Y. pseudotuberculosis, which do not express YopT because of a deletion

on the virulence plasmid, are still virulent in mouse infection models [57]. A

comparative study of YopT and YopE showed that YopE is more efficient than YopT

in blocking phagocytosis, MAPK and NFκB signaling pathways and interleukin-8 (IL-

8) production [58]. Taken together, these observations suggest that YopT function

might be redundant due to the similar function of YopE.

1.2.3.5 YpkA/YopO

YpkA (YopO in Y. enterocolitica) is a multifunctional protein that can function as a

serine/threonine kinase and has both a Rho-GTPase binding domain and an actin

binding domain. [59-63]. YpkA can disrupt the actin cytoskeleton and thereby

contribute to the anti-phagocytic ability of Yersinia against macrophages [54, 64,

65]. However, our investigations do not show any contribution of YpkA to the anti-

phagocytic capacity of Y. pseudotuberculosis against either macrophages or DCs

(Fällman and Westermark, unpublished data). Also when it comes to the

importance of YpkA for virulence in mice results differ, and ypkA mutants have

Page 17: Yersinia-phagocyte interactions during early infection

5

been reported to be both avirulent [59, 65, 66] and as virulent as wt bacteria [55,

67].

1.2.3.3 YopJ/P

YopJ, or YopP as the protein is named in Y. enterocolitica, is a serine/threonine

acetyltransferase [68, 69]. By inhibiting several signaling pathways YopJ interferes

with apoptosis and cytokine production in infected cells [70-74]. The molecular

targets of YopJ/P have been identified as MAPK kinases (MKKs) and IkB kinase-β

(IKKβ) [73]. YopJ/P has been shown to be important for virulence of

enteropathogenic Yersinia [55, 75, 76], but dispensable for virulence of Y. pestis

[77, 78]. However, others have reported similar results for YopJ in Y.

pseudotuberculosis, i.e. that deletion of YopJ had no effect on virulence [66, 79].

The discrepancy regarding the importance of YopJ/P for virulence might be

explained by the difference in cytotoxic activity reported for YopJ/P proteins from

different Yersinia species and even serogroups. High or increased cytotoxicity of

YopJ/P is suggested to lead to proinflammatory responses and avirulence [76, 80,

81]. The role of YopJ/P in the strategies of Yersinia to avoid the immune defense

will be further discussed in section 3.2.

1.2.3.4 YopM

The functional role of YopM has been somewhat a mystery for a long time. One

reason might be that YopM is the only Yop effector that lacks an enzymatic activity

[57]. YopM is a leucin rich repeat protein that has been reported to have a nuclear

localization in host cells [82-84]. This protein functions as an adaptor protein by

forming a complex with and activating two cytoplasmic kinases, RSK1 and PRK2 in

J774 macrophages and HEK 293 cells [85]. YopM interferes with the immune

response of the host by changing the cytokine response, i.e. induce expression of

the anti-inflammatory IL-10 [86]. Interaction of YopM with RSK1 and PRK2 is

required for the induction of IL-10. Recently, YopM was shown to directly inhibit

caspase-1 in macrophages, which is an important participant in the inflammasome

[87]. This finding might explain why YopM is an important virulence factor and that

a yopM mutant is avirulent in mice [55, 88-90]. The role of YopM in the strategies

of Yersinia to avoid the immune defense will be further discussed in section 3.2.

1.2.4 The Yop regulators

1.2.4.1 YopK/Q

YopK (YopQ in Y. enterocolitica) is secreted and translocated together with the

effector Yops into the targeted host cell. However, this Yop does not have a direct

activity against the host cell itself, but instead a regulatory role for the Yop

secretion and translocation. This function was discovered as a yopK mutant delivers

increased levels of Yop effectors into infected cells [91]. YopK interacts with the

Page 18: Yersinia-phagocyte interactions during early infection

6

pore-forming protein YopD and regulates the ratio of YopB and YopD in the target

cell membrane. Consequently, a yopK mutant forms larger pores, which results in

the over-translocation phenotype [92]. The interaction of YopK with Rack1 in the

host cell is important for full anti-phagocytosis and appears to direct the action of

the effector Yops to the correct locations in the cell [92]. A precise and controlled

Yop delivery seems to be very important for Yersinia virulence, since a yopK mutant

is avirulent in mice despite its over-translocation phenotype [55, 92-95]. The role of

YopK in the strategies of Yersinia to avoid the immune defense will be further

discussed in section 3.2.

1.2.4.2 YopE

In addition to its role in anti-phagocytosis, YopE has been reported to be involved

in the regulation of Yop translocation together with YopK. Similar to a yopK mutant,

a yopE mutant delivers increased levels of Yop effectors into infected cells [48, 96].

Since both yopE and yopK mutants over-translocates Yop effectors, it has been

suggested that they work together for a controlled Yop translocation [96]. The role

of YopE in the strategies of Yersinia to avoid the immune defense will be further

discussed in section 3.2.

1.2.5 Regulation of T3SS

To be successful in virulence bacteria need to express the right set of proteins at

the right time, and at the same time be able to control the use of these proteins. In

Yersinia the T3SS is regulated on many different levels. The expression of the T3SS

and its effector proteins is regulated by temperature, calcium concentration and

host cell contact. The T3SS expression is repressed at 26°C and high calcium

concentration, but expressed at 37°C, low calcium concentrations and upon host

cell contact [77, 97-103]. Secretion and translocation of Yop effectors are regulated

by host cell contact and by two of the effector proteins themselves (YopE and

YopK, also mentioned in section 1.2.4) [21, 103]. In addition, some Yops have

chaperones, which further control the secretion [104].

1.3 Adhesins

During different stages of infection it is required for invading bacteria to be able to

adhere to host cells. Adhesion is mediated by outer membrane proteins called

adhesins. Pathogenic Yersinia species express a number of different adhesins. This

section will present three of these adhesins, expressed by the enteropathogenic

Yersinia species: invasin, yersinia adhesin A (YadA) and attachment and invasion

locus (Ail) [2].

Page 19: Yersinia-phagocyte interactions during early infection

7

1.3.1 Invasin

Invasin is encoded on the chromosome and has its optimal expression at 26°C and

basic conditions or under acidic conditions at 37°C [105]. Invasin is thought to be

present on the bacterial surface during the initial phase of infection, when Yersinia

enters the small intestine [106] and to facilitate the entry into the intestinal tissue

by binding to β1-integrins expressed on the apical surface of microfold cells (M

cells) in the follicle associated epithelium [107-110]. Studies have shown that

invasin promotes the initial steps of host colonization and penetration of the

intestinal epithelium, but have no effect on establishment of a systemic infection

as an invasin mutant can cause fatal infection [111-113]. Invasin can promote Yop

delivery upon contact with host cells [114-116]. However, the interaction between

invasin and β1-integrins is not only beneficial for the bacteria, since it can also

mediate uptake by macrophages [117, 118, Paper I].

1.3.2 YadA

YadA (Yersinia adhesin A) is encoded on the virulence plasmid and exclusively

expressed at 37°C in high levels [119]. Thus, YadA is the major adhesin expressed

after enteric Yersinia has crossed the intestinal epithelial barrier. This protein has

multiple functions. As an adhesin it binds extracellular matrix proteins (collagen,

fibronectin, and laminin) and cells (including macrophages and epithelial cells)

[120] through indirect binding to β1-integrins via a bridging extracellular matrix

molecule [121]. YadA is also the major contributor to Yersinia serum resistance, i.e.

the ability to avoid killing by the complement system [122]. The dense layer of

YadA on the bacterial surface is thought to function as a physical barrier that

prevents the complement systems from opsonization and membrane attack

complex formation. Further, YadA binds two host serum factors responsible for

regulating the activation of the complement system: C4 binding protein and factor

H. C4 binding protein is a negative regulator of the classical pathway [123] and

factor H regulates the alternative pathway by stimulating the cleavage of C3b to its

inactive form iC3b [124, 125]. Thus, YadA enables Yersinia to avoid killing by both

the classical and the alternative complement pathways. A yadA mutant is poor in

colonization and persistence in the PPs of mice [109, 111, 113]. However, this

adhesin does not play an important role for the overall infection since the YadA

deficient YPIII strain of Y. pseudotuberculosis is not attenuated and cause lethal

disease in mice [93, 126].

1.3.3 Ail

Ail (Attachment and invasion locus) is chromosomally encoded and expressed at

37°C under aerobic conditions (Pierson and Falkow 1993). Ail mediates epithelial

cell binding and invasion like invasin, but only to certain cell types, such as HEp-2,

HEC1B, and CHO cells (Miller and Falkow 1988). This might reflect a restricted

Page 20: Yersinia-phagocyte interactions during early infection

8

expression of Ail receptors. Similar to YadA, Ail is involved in serum resistance by

binding C4 binding protein and factor H [123, 127, 128], and thereby inhibits both

the classical and the alternative complement pathways.

1.4 Significance of studying Y. pseudotuberculosis

According to statistics from the Swedish Institute for Communicable Disease

Control approximately 300-800 cases of enteric Yersinia infections per year were

reported in Sweden 2003-2012, corresponding to 3-9 cases per 100,000 habitants

per year. This low prevalence of disease might raise the question why study Y.

pseudotuberculosis? First of all, Y. pseudotuberculosis is an optimal model for

studies of microbial pathogenesis and host responses. It is easy to handle and

cultivate, which is a basic requirement for scientific studies of a bacterium. Its

genome is sequenced and genetic manipulations of this bacterium are greatly

simplified since many of the virulence factors expressed by this pathogen are

encoded on the virulence plasmid. Moreover, scientists interested in studying

bacterial virulence accentuate the close relationship with the more dangerous Y.

pestis, but also that Y. pseudotuberculosis share common virulence traits with

other pathogens e.g. the T3SS. For others, Y. pseudotuberculosis is a good model to

study host-microbe interactions, as it offers a virulence model in mouse for both

acute and persistent infection. Additionally, the effector proteins are directed

against important immune cell functions and mutants can be used to study these.

Taken together, Y. pseudotuberculosis is a good model organism for studying

various aspects of pathogenicity and immune response, including adhesion,

effector protein delivery into host cells, immune evasion, and immune cell

functions.

Page 21: Yersinia-phagocyte interactions during early infection

9

2. Phagocytes

Phagocytes are cells with phagocytic capacity i.e. the ability to internalize particles.

Internalization of larger particles can either occur by phagocytosis or

macropinocytosis. The innate immune system includes a number of phagocytes:,

monocytes, macrophages, neutrophils, and dendritic cells [129].

To sense their surroundings phagocytes express a number of different receptors

collectively called pattern-recognition receptors (PRRs). These receptors recognize

conserved molecules called pathogen-associated molecular patterns (PAMPs) from

invading fungi, viruses, bacteria, and parasites [130].

2.1 Internalization by phagocytosis and micropinocytosis

Phagocytosis, which means cell eating, is a highly conserved and complex

mechanism for particle internalization. Phagocytosis is often mentioned in the

context of elimination of foreign particles and microorganisms, but is also used to

remove dangerous “self” cells and debris generated during development, tissue

turnover and repair [129]. Phagocytosis is a multi-step, receptor-mediated, actin-

dependent process that proceeds in a zipper-like or sinking manner [131].

Internalization through phagocytosis often requires and is initialized by recognition.

The recognition is mediated by direct or indirect binding of the particle or

microorganism to surface receptors on the phagocytes. Direct binding occurs

between surface structures on the phagocytized object and a phagocytic receptor

on the phagocyte, for example interaction between sulfated and mannosylated

sugars and the mannose receptor. In contrast, indirect recognition occurs through

opsonins, for example IgG antibodies or complement fragments, bindning to Fcγ-

receptors and complement receptors, respectively [129]. After recognition by

receptor ligation, signaling from the phagocytic receptor and subsequent local actin

polymerization is activated. This cause internalization and results in formation of a

phagosome in the cytosol of the phagocytic cell [129]. The phagosome then

undergoes maturation through interaction with early and late endosomes and

lysosomes, to finally transform into a phagolysosome. Inside the phagolysosome

ingested particles and microorganisms are degraded through acidification and

enzymatic activities [132].

Macropinocytosis is a clathrin-independent form of endocytosis that mediates

non-selective uptake of solute molecules, nutrients, antigens and even

microorganisms [133]. Like phagocytosis, macropinocytosis is an actin-dependent

process, but instead of proceeding in a zipper-like manner, it is initiated from

surface membrane ruffles. Another difference is that macropinocytosis can give

rise to large endocytic vacuoles called macropinosomes. After formation, the

macropinosomes undergo maturation and fuse with lysosomes. Normally

Page 22: Yersinia-phagocyte interactions during early infection

10

macropinocytosis is a signal-dependent process that occurs in response to growth

factor stimulation, by for example CSF-1 or EGF, but in antigen-presenting cells this

uptake process is constitutively active [133].

2.2 Monocytes

Monocytes develop from bone marrow precursors and circulate in the blood.

During both steady-state and inflammatory conditions monocytes can migrate

from the blood into tissues and differentiate into different types of macrophages

[134] and dendritic cells (DCs) [135, 136]. Monocytes can also directly mediate

antimicrobial activity in infected tissues by release of antimicrobial substances and

transport of antigens to draining lymph nodes, where they either transfer the

antigens to conventional DCs (cDCs) or differentiate into monocyte-derived DCs

[137]. Thus, monocytes are multifunctional cells with roles in homeostasis, immune

defense, and tissue repair.

2.2.1 Monocyte subtypes and functions

In both humans and mice monocytes are divided into three subsets: classical,

intermediate, and non-classical. I humans expression of CD14 and CD16 is used to

distinguish the different subtypes. Classical monocytes are CD14++

CD16-,

intermediate monocytes are CD14++

CD16+, and non-classical monocytes are

CD14+CD16

++. In mice expression of Ly6C and CD43 is used to separate the

subtypes as classical monocytes are Ly6C++

CD43+, intermediate Ly6C

++CD43

++ and

non-classical Ly6C+CD43

++ [138].

As their differential gene expression patterns indicate, monocyte subsets also differ

in their function. Non-classical monocytes show a patrolling behavior in the blood

vessels and are involved in tissue repair, while classical monocytes is the major

subtype of monocytes being recruited during early inflammatory responses [139].

2.2.2 How to study monocytes?

The high numbers of monocytes in blood and bone marrow makes monocytes

possible to isolate. However, the lack of a monocyte-specific marker complicates

identification of monocytes and enforces the use of combined markers and

preferably flow cytometry.

2.2.2.1 Monocyte depletion models in mouse

Since there are no monocyte-specific markers known, there is also no specific

monocyte depletion model. However, since monocytes express Ly6C, they can be

depleted together with neutrophils by injection of α-Gr-1 antibody (further

discussed in section 2.4.3.2).

Page 23: Yersinia-phagocyte interactions during early infection

11

2.3 Macrophages

Macrophages are professional phagocytes located in nearly all tissues of the body.

Macrophage precursors are released from the bone marrow into the blood

circulation as monocytes, which migrate into tissues and differentiate into mature,

tissue-resident macrophages. Macrophages are often strategically located to

perform their important immune surveillance activities against commensals and

possible invaders. These activities include phagocytosis, antigen presentation,

immune suppression during antimicrobial defense, anti-tumor immunity, and

inflammatory responses [140].

2.3.1 Macrophage subtypes and functions

Generally it is believed that macrophages represent a variety of activated

phenotypes rather than separate subpopulations [140]. The large overlap in

expression of surface markers between different subsets of macrophages have led

to the use of quantification of specific gene expression profiles after cytokine or

microbial stimulation, to characterize this cell type. Consequently, macrophages

have been divided into classically activated (M1), alternatively activated (M2),

regulatory (Mreg), and tumor-associated macrophages (TAMs). The classically

activated macrophages mediate defense against bacteria, protozoa and viruses,

and further play a role in the immune response against tumors, while the

alternatively activated macrophages have anti-inflammatory activity and regulate

wound healing. The regulatory macrophages are regulatory because they secrete

large amounts of IL-10 and the tumor-associated macrophages suppress anti-tumor

immunity [140].

2.3.2 Macrophage sensing of pathogens

To be able to fulfill their function as sentinel cells, macrophages express a large

variety of PRRs, including Toll-like receptors (TLRs), C-type lectin receptors,

scavenger receptors, mannose receptors, retinoic acid-inducible gene 1-like

helicase receptors and NOD-like receptors (NLRs) [140]. In macrophages these

receptors are not only involved in the recognition of PAMPs on invading

microorgansims, but also foreign substances and dead or dying cells. In addition,

macrophages express complement and Fc receptors that bind to opsonizing

molecules, i.e. C3b and antibodies bound to the surface of invading

microorganisms and facilitate phagocytosis [140].

2.3.3 How to study macrophages?

Macrophages are present in many different tissues, but as for DCs the numbers are

quite low. In mouse PPs, MLN and spleen the number of macrophages is around 1%

of the total number of leukocytes [111]. Isolation procedures are therefore

inefficient, expensive, and time consuming. Alternative methods, such as use of

Page 24: Yersinia-phagocyte interactions during early infection

12

macrophage cell lines, in vitro differentiation from precursor cells and depletion

models are therefore widely used to study macrophages. Expression of the

glycoprotein F4/80 is commonly used to identify macrophages. Further, differential

expression of F4/80 in combination with CD11b, CD18, CD68 and Fc receptors is

used to distinguish macrophages from DCs [140].

2.3.3.1 Macrophage cell lines

The J774 and Raw264.7 cells are derived from tumors in BALB/c mice and are

macrophage-like cell lines widely used for macrophage studies. Both cell lines are

easy to culture and can be used to study phagocytosis and cytokine production

[141].

2.3.3.2 Macrophage differentiation models in mouse and human

Mouse macrophages can be differentiated from bone marrow progenitors by

stimulation with the growth factor M-CSF [142]. Human blood monocytes can be

stimulated with interferon-γ (IFN-γ) and lipopolysaccharide (LPS) to obtain M1

macrophages and IL-4 or IL-10 to obtain M2 macrophages [143].

2.3.3.3 Macrophage depletion models in mouse

Recent years several DTx-mediated macrophages depletion models have been

developed. Examples are the CD11b-DTRtg

model that depletes CD11b expressing

cells, including macrophages and dendritic cells [144] and the lysM-Cre/DTR model

that depletes wound macrophages [145]. These models show high increase of

neutrophils as a response to the DTx-mediated depletion [144, 145], suggesting

that the models must be used with care to avoid over-interpretation of obtained

results. DTx-mediated depletion models are further discussed in section 2.5.3.3 and

5.1. Macrophages can also be depleted by injection of liposomal clodronate [146].

However, this depletion method targets all phagocytic cells and is therefore not

specific to macrophages.

2.4 Neutrophils

Neutrophils or polymorphonuclear neutrophils (PMNs), are professional

phagocytes and absolutely crucial for the immune defense against invading

microorganisms. PMNs are released from the bone marrow, circulate in blood and

are usually the first leukocytes to be recruited to sites of inflammation. The

number of circulating PMNs differs between humans and mice. In humans, PMNs

correspond to 50–70% of the circulating leukocytes, whereas in mice PMNs only

accounts for 10–25% of the circulating leukocytes [147, 148]. The important role of

PMNs in the immune defense is evident in both humans and mice, as individuals in

total absence of or with significant decreased numbers of PMNs die or suffer from

severe immunodeficiency [149].

Page 25: Yersinia-phagocyte interactions during early infection

13

2.4.1 PMN subtypes and functions

Previously neutrophils have been considered to be a homogeneous cell type

responsible for eliminating invading microorganism. Studies however indicate that

neutrophils might exist in multiple subtypes, with both pro-inflammatory and anti-

inflammatory properties [150-152]. The subtypes likely have diverse roles in

infection, inflammation and cancer immunology. Nevertheless the origin of these

subpopulations needs to be determined, to evaluate whether it is one cell type at

different developmental stages or that has been stimulated differently, or whether

there are truly different neutrophils subtypes [149, 153]. In the following sections,

the focus will be on the role of PMNs during infection.

Upon invasion by pathogenic microorganisms circulating PMNs are recruited to

sites of infection by initially sensing the increased expression of P-selectins and E-

selectins on the surface of the blood vessel endothelial cells. The interaction of P-

selectins and E-selectins with their glycosylated ligands leads to tethering of

neutrophils, followed by rolling, adhesion, crawling, and finally transmigration into

the tissue. Chemokine gradients along the endothelium will guide the PMNs to

preferential sites for transmigration, which occur either paracellular, i.e. between

two endothelial cells or transcellular, i.e. through an endothelial cell. Inside the

tissue, the PMNs follow gradients of chemokines, leading them to the infection site

[149, 153].

PMNs can eliminate pathogens by multiple mechanisms such as phagocytosis,

degranulation, and neutrophil extracellular traps (NETs) [149]. Upon phagocytosis

by PMNs, internalized pathogens are killed through NADPH oxygenase-dependent

mechanisms (reactive oxygen species) or antibacterial proteins, such as cathepsins,

defensins, lactoferrin and lysozyme [149]. The mechanism of phagocytosis is

described in more detail in section 2.1.

PMNs have four different types of granules, or secreted vesicles, that are formed

during their maturation [149]. Each type of granula is filled with anti-microbial

peptides and proteins. The primary azurophilic granules contain myeloperoxidase,

the secondary specific granules contain lactoferrin, the tertiary gelatinase granules

contain matrix metalloproteinase 9 (also known as gelatinase B), and the secretory

granules contain alkaline phosphatase. In contact with pathogens the secretory

vesicles are released first, followed by tertiary, secondary, and primary granules.

PMNs use degranulation to defeat both intracellular and extracellular pathogens by

releasing the granula content either into phagosomes or the external environment

[149].

Highly activated PMNs can also use NETs to eliminate extracellular pathogens. NETs

are composed of the DNA from the PMN itself, to which histones and anti-

Page 26: Yersinia-phagocyte interactions during early infection

14

microbial proteins and enzymes from the granules are attached. The NETs trap the

pathogens, unable them to spread and facilitate internalization by other

phagocytes. In addition, the attached anti-microbial proteins are also thought to

cause direct killing [149].

2.4.2 PMN sensing of pathogens

For a long time PMNs were ignored in the pattern recognition receptor signaling

field, as they were thought to be transcriptionally inactive and limited to respond

to receptor agonists due to their short lifespan. Now it is known that they express a

wide variety of PRRs, including TLRs, C-type lectin receptors, retinoic acid-inducible

gene-like helicase receptors and NLRs. The receptors enable PMNs to sense and

respond to a broad range of microbial and endogenous ligands by changing their

functions and production of cytokines, chemokines and lipid mediators [154].

2.4.3 How to study PMNs?

The high levels of PMNs in blood enable studies of human and mouse PMNs by

isolation of primary cells. Mouse PMNs can also be isolated from bone marrow. The

glycoprotein Ly6G is the best known and most specific marker for PMNs.

2.4.3.1 PMN cell lines

The myeloid leukemia cell line, HL-60, is arrested at the myeloblast differentiation

stage and can be differentiated into PMN-like cells by treatment with various

chemical agents, including retinoic acid and dimethylsulphoxide [155-157].

2.4.3.2 PMN depletion models in mouse

PMNs can be depleted in mice by injection of antibodies. Two monoclonal

depletion antibodies with different specificities are available. α-Ly6G (clone 1A8)

recognize Ly6G and is therefore considered to be specific to PMNs [158]. α-Gr-1

(clone RB6-8C5) mainly detects Ly6G, but also recognizes Ly6C, expressed by PMNs

and monocytes and therefore performs a more unspecific depletion [159, 160].

Due to the higher specificity of α-Ly6G, this antibody is suggested as a better

alternative for PMN depletion than α-Gr-1. PMN-depletion will be further discussed

in section 6.

2.5 Dendritic cells

Roughly defined, DCs are white blood cells that can internalize, process and

present antigens to activate T lymphocytes. However, DCs are so much more than

what this simple definition implies. Since the discovery in 1973 by Ralph Steinman

and Zanvil Cohn many different subtypes and functions have been identified.

Today, DCs are recognized as sentinels of the immune system, as professional

Page 27: Yersinia-phagocyte interactions during early infection

15

antigen-presenting cells and as central actors in the orchestration of the immune

response during tolerance and inflammation [161].

2.5.1 DC subtypes and functions

DCs develop from progenitors in the bone marrow and constitute a very

heterogeneous group of cells with different development, organ localization and

cell surface marker expression, and are therefore complex to categorize. Presently,

DCs can be divided into two major types: conventional DCs (cDCs, also termed

classical DCs) and non-conventional DCs [162]. Non-conventional DCs include

plasmacytoid DCs (pDCs) and monocyte-derived DCs (MoDCs). In addition to cDCs,

pDCs and MoDCs, a DC-like cell type termed follicular DCs belong to this family of

cells [163], although based on appearance rather than function since they are no

“true” DCs and will therefore not be further discussed here.

2.5.1.1 Conventional DCs

The cDCs are characterized as highly phagocytic DCs, specialized in antigen-

processing and antigen-presentation [162]. Briefly, cDCs are subdivided into

migratory and lymphoid-tissue-resident DCs. These subsets can be further divided

based on their surface expression of for example CD4 and CD8. Migratory DCs

include DC subsets in the skin, lungs, intestine, liver, and kidneys. These DCs have

high ability to capture antigens and can migrate from peripheral tissues to

lymphoid organ to present antigens to T lymphocytes and elicit an adaptive

immune response. The lymphoid-tissue-resident DCs reside in the lymphoid organs,

such as lymph nodes, spleen and thymus and lack the ability to migrate [162].

Migration of migratory cDCs to lymphoid organs occurs after capture of antigen

and leads to a mixture of migratory and lymphoid cDCs in the lymphoid tissues

[164].

In the mouse intestine, three different subtypes of DCs have been identified based

on their expression of CD11b and CD8 in combination with CD11c, the pan-DC

marker, and their different functions. CD11b+CD8

- myeloid DCs are located to the

subepithelial dome of the PPs and produce high levels of IL-10 upon stimulation.

These DCs prime T lymphocytes to secrete high levels of IL-4 and IL-10. CD11b-CD8

+

lymphoid DCs are located to the interfollicular regions and produce IL-12p70 upon

stimulation. The double negative population, CD11b-CD8

- DCs are located to both

the subepitheial dome and the interfollicular region and also produce IL-12p70

upon stimulation. Both CD11b-CD8

+ and CD11b

-CD8

- DCs prime T lymphocytes to

produce IFN-γ. [165]

2.5.1.2 Plasmacytoid DCs

pDCs were identified in the 1950s, but first described as plasmacytoid T

lymphocytes and plasmacytoid monocytes based on their plasma cell morphology

Page 28: Yersinia-phagocyte interactions during early infection

16

and expression of either the T lymphocyte marker CD4 or the myeloid cell markers

MHC class II, CD36 and CD68. Not until the 1990s this cell type was characterized as

a dendritic cell [166]. In contrary to the ‘dendritic’ morphology of cDCs, pDCs have

spherical morphology resembling antibody-secreting plasma cells. Regarding

surface markers, pDCs can be distinguished from cDCs by their expression of B220,

Siglec-H, and Bst2 in mice and of BDCA2 (CD303) in humans [167]. pDCs circulate in

the blood stream and lymph vessels and migrate to T lymphocyte areas of

lymphoid organs via high endothelial venules upon stimulation [164]. Due to the

ability of pDCs to produce large amount of type I interferons (IFN) they are

important mediators of antiviral immunity upon viral infection. The IFN production

is induced by sensing of viral nucleic acids through TLR 7 and 9 [166]. In contrast to

cDCs, pDCs are poor T lymphocyte activators. This is due to less efficient capture,

processing and loading of antigens onto MHCII. However, upon activation the

antigen presentation capacity of pDCs increases with increased expression of

MHCII. Because pDCs produce large amounts of IFN and other cytokines, they can

still regulate inflammation and link innate with adaptive immunity by affecting NK-

cells, T lymphocytes, B lymphocytes and other DCs. [168].

2.5.1.3 Monocyte-derived DCs

Monocytes can give rise to DCs in peripheral tissues (e.g. intestine, kidneys, lungs,

and skin) under both steady-state and inflammatory conditions. Such DCs are

called monocyte-derived DCs (MoDCs) and have the ability to internalize antigens

and migrate to draining lymph nodes. [162]. Under inflammatory conditions

monocytes differentiate into MoDCs referred to as inflammatory DCs (iDCs) or TNF-

and iNOS-producing DCs (TipDCs). This development has been observed in infection

models involving bacteria, viruses and parasites [139].

2.5.2 DC sensing of pathogens

The extreme plasticity of DCs enables them to adjust their responses depending on

the nature of the encountered stimulus. DCs have for example been suggested to

be able to distinguish between the presence of an infection contra a cytokine-

mediated inflammation since microbial stimuli, but not inflammatory cytokines,

can stimulate DCs to produce IL-2 [169]. DCs use a broad receptor repertoire to

sense their surroundings and recognize microbial components. The PRRs expressed

by DCs are TLRs, NLRs, RIG-I-like receptors, C-type lectins and mannose receptors

each activated by distinct PAMPs at the cell membrane or within intracellular

vesicles [164, 170]. The binding of a PAMP to a PRR initiates signaling cascades that

eventually result in DC activation and upregulation of MHCII and co-stimulatory

molecules.

Page 29: Yersinia-phagocyte interactions during early infection

17

2.5.3 How to study DCs?

DCs are present in all tissues and in blood. However, the low prevalence makes

isolation procedures inefficient, expensive, and time consuming. DCs constitute

approximately 1% of the total number of leukocytes in PPs and MLN and 2% in

spleen [111]. Alternative methods, such as in vitro differentiation from precursor

cells and depletion models are therefore widely used to study DCs. For cDCs

integrin alpha X chain protein CD11c is the most widely used marker. For intestinal

cDCs the alpha E integrin CD103 is also used.

2.5.3.1 DC cell lines

The best available DC cell line is D1, a growth factor-dependent cell line originating

from C57BL/6 spleen cells through long-term GM-CSF treatment. Unstimulated D1

cells resemble immature DCs, based on the expression of CD11c, MHCII and co-

stimulatory molecules CD80, CD86, and CD40. Stimulation of D1 with

microorganisms, LPS or the cytokines TNFα and IL-1β induce maturation and

upregulation of MHCII, CD86, and CD40 [171].

2.5.3.2 DC differentiation models in mouse and human

Mouse cDCs can easily be differentiated from bone marrow progenitors or

peripheral blood monocytes by stimulation with the growth factor GM-CSF in

absence or presence of IL-4. Mouse pDCs and cDCs can be generated by

stimulation of total bone marrow cultures with Flt3L [172]. GM-CSF differentiated

DCs have been shown to resemble inflammatory DCs and Flt3L differentiated DCs

steady-state resident DCs [173]. From humans, blood monocytes are the most

commonly used precursor cell. Immature myeloid cDCs can be differentiated by

stimulation of blood monocytes with GM-CSF and IL-4. Additional stimulation of

these blood monocytes with transforming growth factor-β (TGF-β) will generate

Langerhans cell-like subsets of DCs [172].

2.5.3.3 DC depletion models in mouse

The CD11c-DTR mouse model is currently the most common model used to study

effects of DC depletion. Mice are insensitive to diphtheria toxin (DTx, an AB toxin)

due to a receptor polymorphism [174]. This was used to construct the CD11c-DTR

mouse, which has a primate DTx receptor (DTR) expressed under the control of the

DC specific CD11c promoter (Itgax) [175]. This set-up allows for conditional knock-

out of cells expressing high levels of CD11c by treatment with DTx [176]. The model

has been quite extensively characterized and a number of problems have been

identified. I) Not all DC subtypes are depleted. Langerhans cells and CD11cint

pDCs

are not affected by the DTx-treatment [177, 178]. This might however be an

advantage if these cells are of interest. II) Other cell types except from DCs are

depleted. The DTx-treatment has been shown to deplete splenic F4/80-CD11c

+

Page 30: Yersinia-phagocyte interactions during early infection

18

marginal zone and metallophilic macrophages [179]. III) The CD11c-DTRtg

mice

cannot be treated with more than two injections of DTx without dying from the

treatment. This results in a limitation of the depletion to four days. The sensitivity

of the CD11c-DTRtg

mice to DTx is thought to be caused by aberrant DTR expression

on non-immune cells, such as gut epithelial cells [175, 179]. Thus, long-term

depletion experiments require radiation chimeras, in which wt mice are

reconstituted with CD11c-DTR bone marrow to construct wt mice with CD11c-DTRtg

hematopoietic stem cells [176]. Two improved CD11c depletion models have been

constructed, the CD11c.DOG and CD11c.LuciDTR [180, 181]. In these models the

DTR has been introduced by a bacterial artificial chromosome and is expressed

under the control of the full CD11c promoter [180, 181]. This is thought to

circumvent aberrant DTR expression on non-immune cells seen in CD11c-DTRtg

mice and makes long-term depletion possible. DTx-mediated depletion models for

specific DC subsets are also available as Langerin.DTR, BDCA2.DTR, SiglecH.DTR,

Clec9a.DTR, and CD205.DTR mice [182]. DTx-mediated depletion will be further

discussed in section 5.1.

Before the introduction of DTx depletion models, injection of liposomal clodronate

has been used to deplete DCs in mice. Clodronate induce suicide by inhibiting the

metabolism of the targeted cells [146]. However, since depletion by clodronate

liposomes requires internalization, this depletion technique is not specific for DC.

Other phagocytic cells, such as macrophages will also be depleted [183]. Depletion

of DCs in mice by injection of depleting antibodies has generally been restrained

because of lack of phenotypic markers specific to individual DC populations and

inefficient susceptibility to antibody-dependent phagocytosis and cytotoxicity in

peripheral tissues. Blood pDCs are an exception as they are efficiently depleted

upon injection of pDCs specific monoclonal antibodies [177].

Page 31: Yersinia-phagocyte interactions during early infection

19

3. Yersinia infection

3.1 Infection route and bacterial dissemination

3.1.1 Enteric Yersinia infection in humans

Enteric Yersinia infections most commonly occur after ingestion of contaminated

food or water. Dairy products, raw meat, and vegetables have been reported as

infection sources [184-186]. Yersinia can replicate and thrive at low temperatures

and thus continue to grow if contaminated food is stored in the refrigerator.

In humans, enteric Yersinia infections often resemble acute appendicitis. Bacteria

are most often found in the mesenteric lymph nodes causing mesenteric

lymphadenitis or localized to the appendix, terminal ileum or ascending colon [187,

188]. Histological examination of infected MLNs and appendices reveal granuloma

formation with central necrosis and microabscess formation [187, 188]. In addition,

ulceration of the intestinal and appendicular mucosa can be observed [188].

Enteric Yersinia infections in humans are usually self-limited and cleared at the

point of MLNs. However, these infections can become persistent and in

immunocompromised patients the bacteria can spread systemically and cause

serious conditions [189-192]. People carrying the HLA-B27 antigen have also been

shown to be extra sensitive to developing reactive arthritis upon enteric Yersinia

infection [193, 194].

3.1.2 Enteric Yersinia infection models in mice

In enteric Yersinia infection models in mice, the infection is initiated through

ingestion of contaminated water or force feeding of concentrated bacterial

suspensions by gavage. In mice, the disease progression is determined by the dose

of infection. In low dose infections, corresponding to 106-10

7 colony forming units

(CFUs) or lower, the infected mice can be divided into three groups depending on if

they clear the infection, develop a persistent or systemic infection (Fahlgren et al.

unpublished data). High dose infection, i.e. 108-10

9 CFUs, results in systemic spread

of the bacteria and subsequent lethal infection of the mice [93, 94, 126, 195, 196].

The susceptibility to Yersinia infection differs between different mouse strains.

BALB/c mice are more susceptible to Yersinia infection than C57BL/6 mice [197,

198]. The higher resistance of C57BL/6 mice is thought to be caused by a more

rapid development of a Yersinia-specific Th1 response and difference in

macrophage activation [198-200], and do not seem to be linked to a particular

gene locus, such as Ity or H-2 as has been reported for the intracellular pathogens

Salmonella, Listeria, and Mycobacteria [197]. In persistent Yersinia infection, FVB/n

mice are the most susceptible, followed by BALB/c and C57BL/6 (Fahlgren et al,

unpublished data).

Page 32: Yersinia-phagocyte interactions during early infection

20

Ingested Yersinia passes through the stomach and initially colonizes the intestine

(Fig. 1). During the first stages of infection, Yersinia enters the Payer’s patches (PPs)

of the small intestine through M cells located in the follicle associated epithelium

[110, 201, 202]. The entry is facilitated by the adhesins expressed by enteric

Yersinia (see section 1.3), together with the phagocytic capacity of the M cells [107,

109]. The invasion of PPs results in formation of a small number of monoclonal

microcolonies, indicating that very few bacteria initially invade the gut epithelium

and establish the infection of the PPs [203]. Early localization of enteric Yersinia in

mouse cecum (corresponding to the human appendix) [67, 196, Paper III], suggests

that bacteria enter the lymphoid follicles of this organ in a similar way as the PPs.

Upon entry into the lymphoid follicles of the intestine, Yersinia encounters the

immune defense (Fig. 1). By the actions of the Yops, which are translocated into

attacking immune cells, Yersinia can avoid elimination by the immune system, stay

extracellular and proliferate in tissues [204]. Proliferation results in an

inflammatory reaction and formation of local microabscesses and ulceration of the

overlying epithelium [204].

Figure 1. Entry and immune cell encounter by Y. pseudotuberculosis in Peyer’s patches and

lymphoid follicles of cecum.

After entry into the lymphoid follicles of the small intestine, Yersinia can

disseminate further to mesenteric lymph nodes (MLNs). This dissemination has

been suggested, but not experimentally proven, to be facilitated by migrating

phagocytic cells [57]. It has also been suggested that bacteria might disseminate

from the PPs not only via the lymphatic system but also by invasion of blood

vessels [110]. In MLN, Y. pseudotuberculosis has been reported to have an

Page 33: Yersinia-phagocyte interactions during early infection

21

extracellular localization nearby neutrophils and macrophages in necrotic areas of

the MLN, and nearby B and T lymphocytes in less inflamed areas [205]. From the

MLN the bacteria can spread to spleen and liver to cause systemic disease

eventually resulting in death of the host. Systemic disease caused by wild-type (wt)

Yersinia usually occurs 3-7 days post oral infection in mice [206, Paper III]. In

similarity to PPs, Yersinia forms monoclonal microcolonies in both spleen and liver

[203] Y. pseudotuberculosis has been shown to disseminate to spleen and liver

already 30 minutes after orogastric inoculation [195]. Noteworthy, the bacteria

involved in this fast systemic infection were also cleared fast and a second

colonization of spleen and liver occurred later. The same study challenges the

believe that enteric Yersinia disseminates systemically via PPs and MLN, by using

signature tagged mutagenesis to show that Y. pseudotuberculosis can disseminate

directly to spleen and liver from a replicating pool in the intestine, without passing

the PPs and MLN. The low number of clones in spleen and liver compared with the

intestine further suggests that a severe bottle-neck limits the systemic

dissemination.

Most of the Yop effector proteins are essential for systemic dissemination of

enteric Yersinia species and many Yop effector mutants are therefore avirulent in

mice (Fig. 2). A mutant lacking the PTPase YopH, is most avirulent and rarely spread

beyond the PPs and the lymphoid follicles of cecum [55, 67, Paper III]. Mutants

lacking either of the GAP YopE, the multifunctional YopM, or the Yop translocation

regulator YopK, are slightly less affected and can spread to the MLN [55, 67, 86, 94,

Paper III]. A Y. enterocolitica yopE mutant is more virulent in mice than a Y.

pseudotuberculosis yopE mutant, which might be due to the presence of YopT,

which can compensate for the loss of YopE activity [55]. Mutants deficient in the

YpkA or the YopJ/P can spread systemically to spleen and liver, but are often

cleared at these locations before them can cause full disease [55, 66, 75]. However,

a Y. pseudotuberculosis yopJ mutant can cause death like the wt strain [66, 79].

Figure 2. Dissemination of wt and yop mutants during oral enteric Yersinia infection in mice

Page 34: Yersinia-phagocyte interactions during early infection

22

3.2 Immune responses during enteric Yersinia infection in mice

Upon enteric Yersinia infection, inflammatory cells are recruited to infected tissues

and a Th1 cytokine response with increased IFN-γ production is initiated [206, 207].

The level of tumor necrosis factor-α (TNF-α) also increases in infected tissues, but

Th2 cytokine (IL-4 and IL-5) levels are low. Thus, indicating that a cellular immune

defense is important against enteric Yersinia species. Pathogenic Yersinia species

have however evolved a number of different strategies to avoid the attack from the

immune defense. These strategies include anti-phagocytosis, inhibition of immune

cell maturation, altered cytokine production, and induction of apoptosis. The

effector Yops are responsible for many of the actions, and they act in synergy to

turn off the immune response.

3.2.1 Yersinia and macrophages

The large distribution of resident macrophages in different tissues results in early

interaction between enteric Yersinia and macrophages in the intestine upon

infection. The infection also leads to increase macrophages levels in infected

tissues [206, 208]. Yersinia targets macrophages through translocation of effector

Yops [111, 209], which have large effects on macrophage functions (Fig. 3). In

macrophages YopH and YopE inhibit phagocytosis [42, 54, 117, 210, Paper I]. Some

Yersinia strains use YopT as an additional anti-phagocytic Yop [54]. Besides

inhibiting phagocytosis, YopH and YopE also inhibit exogenous MHCII antigen

presentation in macrophages. However, macrophages can compensate for this

inhibition by using an autophagy-dependent mechanism instead [211]. YopH also

interfere with the oxidative burst of macrophages [212]. Yersinia inhibits

production of different cytokines in macrophages, e.g. TNF-α, IL-1β through the

action of YopJ and YopE [72, 213, 214]. YopJ/P also induces macrophage apoptosis

[70, 215].

Yersinia infection trigger caspase-1 activation by two different pathways in

macrophages: One pathway responds to the T3SS, and the other pathway is

induced by YopJ [216-219]. The caspase-1 activation (by both YopJ and T3SS) is at

the same time counteracted by YopE, which inhibits caspase-1-mediated

maturation and release of IL-1β through its action on Rac1 [214] and YopM, which

directly inhibit caspase-1 [87]. YopK has also been shown to inhibit inflammasome

activation triggered by the T3SS [220]. Production of IL-1β is one of the most

important inflammatory responses upon infection, since this cytokine is involved in

the regulation of both innate and adaptive immune responses [221].

Studies on macrophages suggest that the immune system can recognize and

distinguish between pathogenic Yersinia and Yersinia lacking a functional T3SS. The

pore-forming proteins YopB and YopD induce NFκB- and type I IFN-regulated genes

in macrophages, independently from TLR, Nod1/2, and caspase-1 [219]. It is

Page 35: Yersinia-phagocyte interactions during early infection

23

suggested that this response informs the host cell of the pathogenic challenge,

leading to a unique response, different from the response to bacteria lacking T3SS.

Moreover, the presence of a functional T3SS during phagocytosis decreases

survival of internalized Yersinia inside macrophages [222, 223]. Consequently, the

T3SS is not only beneficial for Yersinia in the contact with macrophages.

Yersinia is considered to be an extracellular bacterium [204]. In contrast, in vitro

studies with bone marrow-derived macrophages have reported that Y.

pseudotuberculosis can survive inside macrophages and that the survival is

dependent on the response regulator PhoP [224, 225]. These studies however used

bacteria pre-grown at 26 or 28°C, hence without a T3SS primed to translocate Yops

during phagocytosis. We could not confirm these results with a similar

experimental setup, but with bacteria induced for T3SS at 37°C (unpublished data).

Our data further support the observation that plasmid-cured Yersinia survives

better in macrophages than wt [222].

3.2.2 Yersinia and neutrophils

Yersinia infection trigger PMN release into the blood and infiltration of PMNs into

different tissues, such as PPs, MLN and spleen [110, 196, 206, 208, 226, Paper III].

Interestingly, the PMN increase in blood is lower upon infection with avirulent Yop

mutants compared to wt bacteria [208, Paper III]. Thus, indicating that systemic

infection trigger the increased release of PMNs from the bone marrow upon

Yersinia infection.

PMNs are the major target cell for the Yersinia effector Yops [111, 209]. This is

further supported by data showing that depletion of PMNs results in a reduction of

the overall levels of Yop translocation into cells [111]. Inside PMNs the effector

Yops interfere with several important functions (Fig. 3). YopH and YopE are

responsible for inhibiting phagocytosis and oxidative burst by PMNs [54, 227, Paper

I]. During Y. enterocolitica infection YopT plays an additional role in the anti-

phagocytosis against PMNs [54]. Y. pseudotuberculosis can inhibit the formation of

NETs, but which Yop that is responsible for this has not yet been elucidated [94].

Others have reported that Y. enterocolitica (strain E40) expressing YadA, induce

formation of NETs, adhere (presumably to collagen in the NETs) and are killed,

while an YadA-deprived strain only induce the NET formation but do not adhere

and subsequently survives. [228]. In contrast to DCs and macrophages, PMNs are

resistant to YopJ/P-induced apoptosis [229].

The role of PMNs during Yersinia infection has been investigated in several

depletion studies with depletion of Gr-1+ cells, including PMNs and monocytes.

Infection of Gr-1-depleted mice indicates an important role for Gr-1+

cells in the

immune response against enteric Yersinia [230]. Wt bacteria, but also avirulent Y.

Page 36: Yersinia-phagocyte interactions during early infection

24

pseudotuberculosis yopK and Y. pestis yopM mutants become more virulent in mice

after Gr-1-depletion [94, 230-232].

3.2.3 Yersinia and dendritic cells

It is not surprising that Yersinia and other pathogens target DCs, as they play a

central role in the regulation of both innate and adaptive immune responses

against invading microorganisms. DCs are also one of the first obstacles that enteric

Yersinia meets in the intestine (paper III) and must bypass to disseminate

systemically. DCs are targeted in vivo by Y. pseudotuberculosis and Y. enterocolitica

through translocation of the effector Yops [111, 209, 233]. A study on splenic DCs

further show that the DC subtypes are differentially targeted by the Yops, which

are most frequently translocated into CD8+, compared with CD4

+ and CD4

-CD8

- DCs

[233]. Thus, Yersinia infection results in a change in the balance between the DC

subsets in spleen as their antigen uptake and degradation, cytokine production,

and death are differently affected.

Inside DCs the Yops interfere with several important immune functions (Fig. 3).

During Y. enterocolitica infection YopH, YopE, and YopT have been reported to

inhibit bacterial internalization by DCs [53, 234], while during Y. pseudotuberculosis

infection YopE is solely responsible for the anti-phagocytic effect (Paper I). YopJ/P

inhibits DC maturation and T lymphocyte stimulation by blocking antigen uptake

and processing [235, 236], and upregulation of co-stimulatory molecules CD80,

CD83 and CD86, and MHCII [234, 237, Paper I]. Moreover, YopJ/P is responsible for

suppressing the production of several cytokines by DCs, e.g. keratinocyte-derived

chemokine (KC), TNF-α, IL-10, and IL-12 [237], and for inducing apoptosis in DCs

[234, 237] by both caspase-dependent and independent mechanisms [81]. Taken

together, several Yops collaborate to inactivate DCs and prevent their activation of

T lymphocytes.

3.2.4 Yersinia and B and T lymphocytes

By inhibiting antigen presentation and inducing apoptosis of dendritic cells and

macrophages, Yersinia indirectly affects the adaptive immune response. However,

these pathogens have also evolved direct strategies to attack B and T lymphocytes

(Fig. 3). The Yop effector proteins are translocated into B and T lymphocytes, but

less frequently than into the innate immune cells [111, 209]. By the act of YopH,

which dephosphorylates tyrosine-phosphorylated components associated with the

antigen receptor signaling complex, Yersinia directly interfere with B and T

lymphocyte antigen receptor-mediated activation. As a result, B lymphocytes

become unable to up-regulate the co-stimulatory molecule B7.2 in response to

antigen stimulation and T lymphocytes become unable to flux calcium and produce

cytokines [43].

Page 37: Yersinia-phagocyte interactions during early infection

25

Both CD4+ helper and CD8

+ cytotoxic T lymphocytes are involved in the immune

response during enteric Yersinia infection [207, 238, 239]. Increased levels of IFN-γ

and low levels of IL-4 and IL-5, suggest that a Th1 response is initiated [206, 207].

Figure 3. Effects of Yersinia effector Yops on different immune cells.

3.3 Relevance of using mice as a model for human bacterial diseases

Mice are the main experimental model used in immunological studies and their use

have generated a great insight into mechanisms of the immune system [240]. Mice

are widely used experimental models because of their genetic and physiological

similarities to humans, and because their genome can be easily manipulated and

analyzed in various disease and therapy studies. However there are also many

differences between these species, so is it relevant to use this animal as model for

human diseases?

The genome sequencing of mouse and human revealed that both species have

approximately 30,000 genes and that only around 300 genes appear to be unique

to one of the species [241]. Hence, genetically we are more alike than one might

think. However, despite the genetic similarities the immune systems of mice and

humans have evolved differently and even though the overall structure is quite

similar, there are significant differences in both the innate and adaptive immune

response regarding for example development, activation, and response to

challenges [240]. One example is the ratio between lymphocytes and PMNs in the

Page 38: Yersinia-phagocyte interactions during early infection

26

blood. In humans, PMNs compromise the major cell type and accounts for 50-70%

of the total leukocytes in blood, while lymphocytes account for 30-50%. In mice the

ratio is reversed, with 75-90% lymphocytes and 10-25% PMNs [147, 148]. The

functional consequence of this difference is however not clarified.

Recently, a study comparing transcriptional responses to inflammatory conditions

concluded that the genomic responses in mouse models poorly mimic human

inflammatory diseases [242]. The study compared gene expression profiles of

blood leukocytes during different inflammatory conditions, including burn, trauma

and endotoxemia in humans and corresponding mouse models. The results of the

study showed that the transcriptional responses to the different inflammatory

conditions in humans had a higher correlation to each other, than to the

corresponding mouse model and additionally that the responses in mouse models

significantly differed from each other. Thus, underlining that mouse disease models

might poorly reflect the human disease. However, it should be noted that the study

only included one mouse strain and that the human participants were under

medical care, which might have affected the results.

Thus, it is important to consider the limitations of mouse models and caution is

necessary when correlating results from mouse studies to human diseases. Some

important questions to ask are: I) Are mice a natural host of the studied pathogen?

II) Does the disease progress in a similar way and over a similar time course in mice

as in humans? III) Does the mouse model reproduce the major clinical symptoms of

the human disease? IV) Are the same tissues and/or cells affected in the mouse

model as in humans? V) Are the same genes and molecular pathways involved?

[243]

Despite the discrepancies between humans and mice, studies of enteric Yersinia

infections using mouse as virulence model are relevant. The progression of enteric

Yersinia infection in mice resembles yersiniosis in humans in many aspects.

Although not allowing direct correlation to infection in humans, mouse studies help

to identify virulence parameters such as presentation of disease, bacterial loads

and dissemination, extension of tissue damage and to some extent immunological

responses. It is however important to consider the choice of mouse strain, infection

doses and route of infection when interpreting results and relevance for human

pathogenesis. Interpretation of immunological effects also requires a careful

consideration of potential differences between mice and men, which when

possible, can be overcomed by additional in vitro cell culture studies using human

cells.

Page 39: Yersinia-phagocyte interactions during early infection

27

AIMS

The major aim of this thesis was to study the interaction between

Y. pseudotuberculosis and phagocytic cells during early infection. The more specific

aims were:

Investigate the effect of the effector Yop proteins on DC maturation.

Investigate the effect of the anti-phagoctic effectors YopH and YopE on

DCs.

Study the role of DCs during early Y. pseudotuberculosis infection.

Study the role of PMNs during early Y. pseudotuberculosis infection and

for avirulence of yopH and yopE mutants.

Page 40: Yersinia-phagocyte interactions during early infection

28

RESULTS & DISCUSSION

4. PAPER I – Cell type-specific effects of Yersinia pseudotuberculosis virulence effectors

Pathogenic Yersinia species use a T3SS for translocation of a variety of effector

proteins into host immune cells. The actions of the effector proteins against

immune cell functions of macrophages and PMNs have been more extensively

studied than the effects in DCs. In this study we therefore aimed to investigate how

the Yops of Y. pseudotuberculosis affect two key mechanisms of DCs - maturation

and internalization.

4.1 Y. pseudotuberculosis inhibits dendritic cells maturation by the action of YopJ

To investigate the effect of the different Yop effectors on DC maturation, bone

marrow-derived DCs (BM-DCs) were differentiated from mouse bone marrow

progenitors and infected with different Y. pseudotuberculosis strains. Flow

cytometry analysis of the expression of the co-stimulatory molecules CD80, CD83,

and CD86 showed that wt Y. pseudotuberculosis could inhibit DC maturation (Paper

I, Fig. 1C). In contrast, the virulence plasmid-cured strain, which lack the T3SS and

its effector proteins, induced DC maturation. Thus, inhibition of DC maturation was

T3SS dependent. Infection with different single yop mutants revealed that YopJ was

responsible for the inhibition of DC maturation as the yopJ mutant was the only

mutant that induced maturation. Mutants in YopH, YopE and YopM inhibited DC

maturation as efficiently as wt, indicating that these Yop effectors are not involved

in the inhibition of DC maturation. These data confirm previous findings that Y.

pestis and Y. enterocolitica also inhibit DC maturation by the action of YopJ [53,

234, 237, 244, 245]. In conclusion, the human pathogenic species of Yersinia use

the same mechanism to prevent DC maturation.

4.2 Y. pseudotuberculosis resists internalization by dendritic cells through the action of YopE

Internalization is a central defense mechanism employed by immune cells to

eliminate invading microorganisms. Pathogenic Yersinia species can efficiently

avoid internalization and thereby elimination by macrophages and PMNs, which

has been extensively studied in macrophages cell-lines, isolated peritoneal

macrophages, and blood PMNs [42, 117, 210]. At the initiation of this study the

effects of YopH and YopE against DCs were however not elucidated. This together

with the discovery that DCs are one of the major target cells for Yop translocation

Page 41: Yersinia-phagocyte interactions during early infection

29

[246] urged us to investigate the anti-phagocytic capacity of Y. pseudotuberculosis

against DCs.

BM-DCs were differentiated from mouse bone marrow and infected with Y.

pseudotuberculosis wt, virulence plasmid-cured strain, yopH and yopE mutants.

Internalization was evaluated by double immunofluorescent staining, which

discriminates between extra- and intracellular bacteria. Wt bacteria could block

internalization by BM-DCs, while the virulence plasmid-cured strain could not and

was subsequently more frequently internalized (Paper I, Fig. 1D). Interestingly,

infections with the yopH and yopE mutants revealed that YopE is solely responsible

for the anti-phagocytosis against DCs. The internalization assay showed that the

yopH mutant (expressing YopE) could resist internalization to the same level as wt,

while the yopE mutant (expressing YopH) was internalized as the virulence plasmid-

cured strain. Previous data on the anti-phagocytosis against macrophages and

PMNs were also confirmed in bone marrow-derived macrophages (BM-Ms) and

PMNs (BM-PMNs) showing that both YopH and YopE contribute to the anti-

phagocytic capacity of Y. pseudotuberculosis in these cells (Paper I, Fig. 2).

To investigate if this cell type specific feature of Y. pseudotuberculosis was

restricted to DCs from mice or a general effect, the internalization experiments

were performed on DCs derived from human peripheral blood monocytes (PBMC-

DCs) and isolated human blood PMNs (hPMNs). Similar results were seen for the

human DCs and PMNs suggesting that the missing effect in anti-phagocytosis of

YopH against DCs is a general effect (Paper I, Fig. 3B). Further, the data indicate

that YopH is especially important for resisting internalization by more professional

phagocytes, i.e. macrophages and PMNs.

Simultaneously to our study, Adkins et al. investigated the anti-phagocytic capacity

of Y. enterocolitica and concluded that YopH and YopE are both involved in the

inhibition of internalization by DCs for this member of the Yersinia genus [53].

Thus, suggesting that Y. enterocolitica YopH has another role against DCs than Y.

pseudotuberculosis YopH. It should however be noted that this study rather

investigate the internalization by DCs than the anti-phagocytosis by Yersinia since it

analyses percent DCs with intracellular bacteria instead of investigating percent

internalized bacteria, which also consider the ability of the different mutants to

adhere to the cells. Nevertheless, difference in functional importance between

homologous proteins within the Yersinia genus is nothing new. The YopJ protein of

Y. pestis and Y. pseudotuberculosis, and the YopP protein of Y. enterocolitica is one

example. These proteins are differentially expressed and secreted and thereby play

diverse roles during infection with different Yersinia species [76].

Page 42: Yersinia-phagocyte interactions during early infection

30

4.3 The cell specific effect of YopH depends on the mechanism of internalization

The first step in investigating the mechanism behind the cell specificity of YopH was

to analyze the presence of YopH and YopE inside DCs and macrophages. BM-DCs

and BM-Ms were infected with a multiple yop mutant overexpressing either YopH

or YopE. Western blot analysis of intracellular fractions from the two cell types

confirmed that both YopH and YopE were translocated into DCs at similar levels as

into macrophages (Paper I, Fig. 4). Thus, YopH is present inside DCs upon bacterial

contact, but does not contribute to the inhibition of internalization.

The second step in the investigation was to analyze the expression and localization

of YopH target proteins inside DCs. The expression of Fyb and p130Cas was

investigated by Western blot, which revealed that BM-DCs have a markedly lower

expression of both proteins compared with BM-Ms (Paper I, Fig. 5A). However, in

BM-DCs the p130Cas antibody recognized a protein of ~105-110 kDa in size, which

might represent the Cas homologous proteins HEF-1/Cas-L and Sin/Efs [247]. Next,

BM-DCs and BM-Ms were stained to visualize the cellular location of Fyb. In J774

macrophages Fyb is localized to the cell periphery, in areas with high actin

dynamics [248]. In BM-Ms, Fyb showed a similar cellular distribution as in J774

macrophages, with Fyb especially enriched in F-actin rich areas in the cell periphery

(Paper I, Fig. 5B). The immunofluorescence confirmed the low expression of Fyb

detected in BM-DCs by Western blot and further showed that Fyb was diffusely

distributed in the cytoplasm of BM-DCs. Thus, both expression and cellular

localization of the YopH target protein Fyb is strikingly different in BM-DCs

compared with BM-Ms.

The third step in the investigation of the cell type specific effect of YopH was to

analyze the mechanism of internalization used by BM-DCs and BM-Ms. During the

analysis of internalization we observed that the number of attached bacteria and

the number of cells with attached bacteria were significantly higher for BM-Ms

than for BM-DCs. Thus, indicating that macrophages possess a higher capacity for

binding Y. pseudotuberculosis. This was further supported by a kinetic assay

showing that BM-Ms both bind higher number of bacteria but also manage to

internalize them faster than the BM-DCs (Paper I, Fig. 6). While BM-Ms constantly

internalize a high percentage of the attached bacteria, the internalization by BM-

DCs gradually increased over time. β1-integrins on the surface of macrophages and

PMNs mediate the phagocytosis of Y. pseudotuberculosis by binding to the

bacterial surface protein invasin [41]. The internalization of a virulence plasmid-

cured Y. pseudotuberculosis strain lacking invasin by BM-Ms was accordingly

decreased due to affected bacterial binding to the cell (Paper I, Fig. 6). In contrast

the internalization of the same strain by BM-DCs was not affected, showing that

Page 43: Yersinia-phagocyte interactions during early infection

31

the uptake by DCs is independent of the interaction between β1-integrins and

invasin.

Next, live cell microscopy in combination with scanning and transmission electron

microscopy was used to further investigate the mechanism of bacterial

internalization by the two cell types. These advanced microscope techniques

revealed that macrophages mainly internalize Y. pseudotuberculosis through a

zipper-like phagocytosis mechanism resulting in bacteria inside tight phagosomes

(Paper I, Fig. 7A, 7C-G, 8A-B and 8G-H). In contrast, DCs mainly use

micropinocytosis, a random, ruffle-like mechanism resulting in more spacious

phagosomes (Paper I, Fig. 7B, 7H-L, 8E-F and 8I-J). Thus, macrophages and DCs use

different mechanisms to internalize Y. pseudotuberculosis (Fig. 4).

Figure. 4. Y. pseudotuberculosis internalization and anti-phagocytosis against macrophages

and DCs. A) Binding of virulence plasmid-cured Y. pseudotuberculosis (YPIII) to the surface of

a macrophage via invasin-β1-integrin interactions stimulates internalization by a zipper-like

mechanism. A wt bacterium blocks internalization by translocation of anti-phagocytic

effectors YopH and YopE into the macrophage. B) Virulence plasmid-cured Y.

pseudotuberculosis interacting with DCs is internalized by random, ruffle-like

macrophinocytosis. A wt bacterium translocates the anti-phagocytic effectors YopH and

YopE into DCs. Because this internalization mechanism occurs independently of local

Page 44: Yersinia-phagocyte interactions during early infection

32

receptor-mediated responses YopH cannot inhibit internalization. YopE can still influence the

cytoskeletal dynamics and subsequently block internalization.

In summary, these results show that the inability of YopH to inhibit internalization

of Y. pseudotuberculosis by DCs is dependent on the mechanism of internalization.

YopH is translocated into DCs, but since macropinocytosis occurs without the

involvement of β1-integrins and strict receptor mediated signalling mechanisms,

YopH does not have any signalling molecules to act on and cannot prevent

phagocytosis.

Page 45: Yersinia-phagocyte interactions during early infection

33

5. PAPER II – Immune response to diphtheria toxin-mediated depletion complicates the use of the CD11c-DTRtg model for studies of bacterial gastrointestinal infections.

The CD11c-DTRtg

mouse model has been frequently used to investigate the effects

of DC depletion during steady-state and inflammatory conditions [181, 249-251]. In

CD11c-DTRtg

mice, a primate diphtheria toxin receptor (DTR) is expressed under the

control of the CD11c promoter, which allows depletion of CD11chigh

cells by

treatment with diphtheria toxin DTx [175]. In this study we aimed to use the

CD11c-DTRtg

model to investigate the role of DCs during early Y. pseudotuberculosis

infection.

5.1 DTx-mediated depletion of dendritic cells in the CD11c-DTRtg mouse model induces neutrophilia, which results in lower Y. pseudotuberculosis infection

DTx-treated CD11c-DTRtg

mice and PBS-treated wt BALB/c mice were infected with

bioluminescent Y. pseudotuberculosis and the infection was monitored for three

days by in vivo biophotonic imaging using IVIS®Spectrum. Remarkably, Y.

pseudotuberculosis infection of DC-depleted mice resulted in significantly reduced

levels of bacterial colonization at day one and three post infection (p.i.) compared

with undepleted mice (Paper II, Fig. 1). Thus, suggesting that depletion of one of

the most important immune cells might be beneficial for the host and that DCs play

an important role in the establishment of enteric Yersinia infection. However,

analysis of the immune response in uninfected DTx treated CD11c-DTRtg

mice

revealed an induction of the immune response also during steady-state conditions

(Paper II, Fig. 2A-B). The immune response included a massive recruitment of PMNs

to PPs and spleen one to two days after DTx-mediated DC depletion, followed by

an increase of macrophages four days after DTx treatment. This immune response

was not observed in DTx-treated wt mice, showing that the response was not

towards the injected DTx and supporting that the immune response was induced in

response to the DC depletion (Paper II, Fig. 2C). The triggered immune cell

recruitment was also accompanied by increase of KC (homolog to human IL-8) in

uninfected DTx-treated CD11c-DTRtg

mice (Paper II, Fig. 2C). Interestingly, when the

mice were infected by intra-peritoneal injections of bacteria instead of orally, the

observed difference in bacterial colonization between undepleted and DC-depleted

mice was diminished (Paper II, Fig. 3). This result shows that the route of infection

affects the outcome of disease in this model. Further, it indicates that Y.

pseudotuberculosis is more sensitive to the DTx-mediated immune response when

entering via the gastrointestinal route compared with direct systemic spread.

The analyses of the DTx-mediated immune response by immunohistochemistry,

flow cytometry, and a cytokine assay indicated that the response was transient and

Page 46: Yersinia-phagocyte interactions during early infection

34

only triggered by the first DTx-treatment (Paper I, Fig. 2). This means that most of

the PMNs had disappeared at day four after the first DTx-treatment (corresponding

to day three p.i. in infected mice). Hence, to overcome the most activated immune

response, we postponed the onset of infection until three days after the first DTx-

treatment and investigated the bacterial colonization one day p.i.. With the new

experimental setup the difference in bacterial colonization between DC-depleted

and undepleted mice was markedly reduced (Paper II, Fig. 4). Thus, further

supporting that Y. pseudotuberculosis colonization in DC-depleted mice was

restricted as a direct result of the DTx-mediated immune response. Y. enterocolitica

has previously been reported to be restricted to enter into already infected PPs,

presumably due to initiated host immune responses against already invading

bacteria [203]. This is in consistency with our results and might explain why Y.

pseudotuberculosis has colonization problems in DTx-treated CD11c-DTRtg

mice.

CD11c-DTRtg

, CD11c.DOG, and CD11c.LuciDTR mice have all shown increased levels

of neutrophils after DTx-treatment [180, 181, 252] + Paper II), which complicates

the use of this type of depletion model in infection studies (Paper II). Autenrieth et

al. suggest that the observed neutrophilia in DTx-treated CD11c.DOG mice is due to

dysregulation of PMN homeostasis in the absence of DCs [252]. However, the

results in our study do not support this observation. First, PMNs were recruited to

PPs two days after DTx treatment in uninfected CD11c-DTRtg

mice even though

PMNs are usually not present in PPs under steady-state conditions. Secondly, the

increase of PMNs was transient and was not further triggered by the second DTx-

treatment. If DCs would be responsible for regulating PMN homeostasis, PMNs

would be continuously recruited upon DC depletion.

In summary, in addition to the previously reported problems for the CD11c-DTRtg

model regarding for example the limited number of DTx-treatments and depletion

of other cell types, we report that DTx-mediated depletion of DCs induces a

transient PMN response that affects bacterial gastrointestinal infection (Fig. 5). Due

to this response we suggest that infection studies in CD11c-DTRtg

mice and other

DTX-mediated models should not be initiated before the immune status has

returned to normal after the initial DTx-treatment. Consequently, the CD11c-DTRtg

model is best used in the chimeric form, which tolerates repeated DTx-treatments

and thereby long-term depletion. Thus, chimeras can be used in infection studies

where infection is initiated when the immune system has stabilized after depletion.

Page 47: Yersinia-phagocyte interactions during early infection

35

Figure 5. DTx treatment and the subsequent DC depletion results in neutrophilia in CD11c-

DTRtg

mice. The recruited PMNs can eliminate Y. pseudotuberculosis infecting via the oral

route.

Page 48: Yersinia-phagocyte interactions during early infection

36

6. PAPER III – Yersinia pseudotuberculosis efficiently avoids polymorphonuclear neutrophils during early infection

The knowledge about the host cell interactions during the very initial stages of Y.

pseudotuberculosis infection in mice is limited. In this study we therefore aimed to

investigate Yersinia-phagocyte interactions during early infection.

6.1 Y. pseudotuberculosis yopH and yopE mutants, but not the wt strain, interact with PMNs during early infection

Upon entry into the intestinal lymphoid tissues enteric Yersinia encounters

phagocytes. Macrophages and PMNs have been reported to increase at the site of

infection, while the number of DCs are more or less unaffected [110, 111, 196, 206,

208, 226, 253]. In a previous study, we showed that depending on which phagocyte

Y. pseudotuberculosis encounters it needs to be differently equipped to avoid

internalization (Paper I). To avoid phagocytosis by macrophages and PMNs Y.

pseudotuberculosis requires the action of both the PTPase YopH and the GAP YopE,

while YopE alone is enough to withstand internalization by DCs. The importance of

YopH and YopE for avoidance of internalization is also reflected in the virulence of

the corresponding mutants. A yopH mutant is highly avirulent and rarely

disseminates from the intestine, while a yopE mutant can disseminate to the MLN

[55, 67].

To investigate the cell type specific feature of YopH further, we developed an ex

vivo interaction assay in mice. Interaction between Y. pseudotuberculosis and

different phagocytes upon bacterial entry into the lymphoid follicles of the

intestine was investigated by double immunofluorescence using tissue from

infected mice day one and three p.i.. Bioluminescent Y. pseudotuberculosis wt,

yopH and yopE mutant strains were used in the infection, enabling efficient

collection and analysis of PPs and cecums positive for bacteria by in vivo

biophotonic imaging using IVIS®Spectrum. Cecum was finally chosen as the tissue

for analysis, due to a low prevalence of bacteria (especially yopH and yopE

mutants) in PPs at early stages of infection and corresponding results for wt

bacteria in the two tissues (data for PPs not shown). The interaction between single

bacteria and phagocytes in situ was investigated using tissues frozen at day one

and three p.i.. The double immunofluorescence showed that wt Y.

pseudotuberculosis mainly interacted with DCs at day one p.i. (Paper III, Fig. 1A).

The interaction with macrophages was remarkably low and no interaction occurred

with the PMNs recruited to the infected tissue at this time point. At day three p.i.

the level of interaction of wt bacteria with DCs and macrophages resembled day

one p.i. (Paper III, Fig. 1B). However, at this stage wt bacteria also interacted with

the massive number of PMNs recruited to the infected tissue. Interestingly,

infection with the virulence deficient yopH and yopE mutants resulted in a

Page 49: Yersinia-phagocyte interactions during early infection

37

strikingly different pattern of bacteria-phagocyte interaction. At day one p.i. the

level of interaction with DCs was similar for yopH and yopE mutants, as that

observed for the wt. However, in contrast to wt bacteria the mutants also

interacted with PMNs at this early time point. At day three p.i. interaction of the

mutants with both DCs and PMNs were more frequently occurring than for wt

bacteria. Taken together the results show that yopH and yopE mutants interact

with PMNs in the intestine both earlier and more frequently than wt. This suggests

that wt bacteria can avoid interactions with PMNs by the action of these Yop

effectors.

6.2 PMN depletion increase the virulence of Y. pseudotuberculosis

To further investigate the role of PMNs during initial intestinal infection and the

way in which Y. pseudotuberculosis avoid these cells, we employed a PMN

depletion model. Mice were depleted of PMNs by intraperitoneal injection of α-

Ly6G antibodies and infected with bioluminescent Y. pseudotuberculosis wt, yopH,

yopE, and yopK mutants. The yopK mutant was included as a control in this

experiment, as the virulence of this mutant has previously been shown to increase

in the absence of PMNs [94]. The infection was monitored by physical

examinations, flow cytometry, and in vivo biophotonic imaging of whole animals

and organs using IVIS®Spectrum.

The physical examination of the mice included visual observation of disease

symptoms such as ruffled fur, diarrhea, hunchback posture, and inactivity. Disease

symptoms were initially observed at day three p.i.. At this time point PMN-

depleted mice infected with wt, yopE, and yopK mutants showed slightly more

symptoms compared with the undepleted mice (Paper III, Table 1). At termination

of the infection at day five p.i. the yopE mutant infected PMN-depleted mice still

had more severe symptoms of disease compared with the undepleted mice. Wt

and yopK mutant infected PMN-depleted mice showed that same level of

symptoms as the undepleted mice at day five p.i.. Undepleted and PMN-depleted

mice infected with yopH mutant did not show any symptoms of disease. Thus, the

physical examinations of the mice indicated that PMN-depleted mice infected with

wt, yopE or yopK mutant were initially more affected by disease than infected

undepleted mice.

To monitor PMN depletion, blood samples were collected and analyzed by flow

cytometry for cellular expression of the PMN marker Gr-1 (Ly6G/6C) and major

histocompatibility complex (MHCII). Uninfected, undepleted mice had stable levels

of Gr-1high

MHCII- cells (corresponding to PMNs) (Paper III, Fig. 2B). In wt infected

undepleted mice the level of Gr-1high

MHCII- cells gradually increased in the blood

during the infection. Interestingly, flow cytometry analysis of blood samples from

mice infected with yopH, yopE and yopK mutants revealed that these mutants

Page 50: Yersinia-phagocyte interactions during early infection

38

induced a significantly lower release of PMNs into the blood circulation compared

with wt (Paper III, Fig. S1). This finding indicates that release of PMNs is triggered

by systemic dissemination of this bacterium, which is in consistency with previous

reported data [208]. This also explains the gradually increasing levels of PMNs in

blood upon wt infection. In our study this conclusion is further supported by the

increase of PMNs in blood from one of the yopH mutant infected mice. This

particular mouse had an unusually observed, but constrained bacterial plaque in

the liver at day five p.i. and subsequently also higher increase of PMNs in blood

compared with the remaining yopH mutant infected mice. All α-Ly6G-treated mice

showed very low levels of Gr-1high

MHCII- cells in the blood (Paper III, Fig. 2B). Thus,

confirming that the α-Ly6G antibodies were injected in sufficient amounts for

depletion of PMNs also during Y. pseudotuberculosis infection.

To monitor infection progression the infected mice were anesthetized and signals

from the bioluminescent bacteria were measured by IVIS. Mice were also dissected

to investigate the bacterial dissemination in organs by IVIS. The external IVIS

analysis indicated that the colonization efficiency of all bacterial strains increased

in the absence of PMNs at day one p.i. (Fig. 6 and Paper III, Fig. 3 and Fig. 4). For

the wt strain the increased colonization efficiency was seen as significantly higher

external bioluminescent signals in PMN-depleted mice compared with undepleted

mice. The frequency of bacterial dissemination to MLN was not affected by PMN

depletion thus indicating that wt bacteria can efficiently resist the PMN-mediated

defense in the lymphoid follicles of the intestine. This was also in consistency with

the interaction data for the wt strain (Paper III, Fig. 1). For the yopH, yopE, and

yopK mutants both increased external bioluminescent signals and increase

bacterial dissemination to MLN were observed in PMN-depleted compared with

undepleted mice. Thus, indicating that PMNs are responsible for restricting these

mutants from dissemination to the MLN.

Previous studies in PMN-depleted mice show that Y. pseudotuberculosis yopK and

Y. pestis yopM mutants become more infectious in the absence of PMNs [94, 231,

232]. Taken together, our and others data show that many Yops contribute to the

PMN-resistant phenotype of Yersinia species and are important for the efficient

PMN escape of wt bacteria. A fully equipped Yersinia is not markedly affected by

the anti-microbial actions of PMNs. Hence, PMN depletion only accelerates the

infection progression slightly. For the avirulent Yop mutants the observed result of

PMN depletion is more pronounced as the absence of PMNs enable these bacteria

to disseminate faster and reach deeper tissues.

Page 51: Yersinia-phagocyte interactions during early infection

39

Figure 6. PMN depletion by injection of α-Ly6G antibodies results in increase initial

colonization by Y. pseudotuberculosis wt bacteria and faster dissemination of avirulent yopH,

yopE and yopK mutants to mesenteric lymph node.

6.3 Undepleted immature PMNs compensate for the loss of mature PMNs in PMN-depleted mice

At day three and five p.i. the physical examination of disease symptoms, and the

IVIS analysis of external bacteria signals and bacterial dissemination in organs

revealed that a difference in the infection progression could no longer be observed

between undepleted and PMN-depleted mice (Paper III, Table 1, Fig. 3 and data not

shown). Thus, suggesting that a compensatory immune response is initiated in the

absence of PMNs. This was confirmed by the flow cytometry analysis performed to

monitor the PMN depletion, which revealed that a Gr-1int

MHCII- cell population

increased in a similar pattern in PMN-depleted mice, as Gr-1high

MHCII- cells

increased in undepleted mice (Paper III, Fig. 2 and Fig. 5A). Further analysis by flow

cytometry showed that the Gr-1int

MHCII- cell population in infected PMN-depleted

mice was mainly immature PMNs (Gr-1int

F4/80-), but also inflammatory monocytes

(Gr-1int

F4/80+) (Paper III, Fig. 5B). Immunohistochemistry showed that these

immature PMNs behaved like mature PMNs and were also recruited to infected

tissues (Paper III, Fig. S2). Flow cytometry also revealed a slight increase of Gr-

1+F4/80

+ monocytes in blood of infected PMN-depleted mice, which was

accompanied by an increase of Ly6C+F4/80

+ inflammatory macrophages in infected

tissues of PMN-depleted mice shown by immunohistochemistry and

immunofluorescence (Paper III, Fig. 5A, Fig. S2, and Fig. S3). Similar results

regarding immature PMNs and macrophages have been observed in the oviducts of

PMN-depleted mice infected with Chlamydia [254]. Taken together these findings

suggest that release of immature PMNs is a general effect in α-Ly6G-treated mice

upon infection and that the expression of Ly6G on these cells is not sufficient for

antibody depletion.

The incomplete depletion using α-Ly6G prevents the ability to investigate whether

the previously observed effects of increased Yersinia virulence in Gr-1-depleted

Page 52: Yersinia-phagocyte interactions during early infection

40

mice are dependent on PMNs or PMNs in combination with inflammatory

monocytes. However, the increase of inflammatory monocytes and inflammatory

macrophages in PMN-depleted mice in this study indicate that that these cells play

a role during Yersinia infection. Thus, suggesting that mice treated with α-Gr-1 will

be more affected by Yersinia infection than mice treated with α-Ly6G.

Page 53: Yersinia-phagocyte interactions during early infection

41

CONCLUSIONS

PAPER I

Y. pseudotuberculosis inhibits DC maturation through the action of YopJ.

Y. pseudotuberculosis avoids internalization by macrophages and PMNs

through the action of YopH and YopE, while YopE alone can inhibit

internalization by DCs.

The cell type specific feature of YopH is dependent on the different

mechanisms of internalization used by macrophages and DCs.

PAPER II

The CD11c-DTRtg

model is complicated to use for infectious studies due to

DTx-mediated neutrophilia.

The induced immune response of CD11c-DTRtg

mice and other DTx-

mediated depletion models upon DTx-treatment must be carefully

investigated and taken into consideration before initiation of infection to

avoid misinterpretation of obtained results.

PAPER III

Avoiding interaction with PMNs is a key feature of pathogenic Yersinia,

which allows colonization and effective dissemination in mice.

Y. pseudotuberculosis yopH and yopE mutants, but not the wt strain,

interact with PMNs in the lymphoid follicles of the intestine during the

initial stage of oral infection.

PMN-depletion results in more efficient initial colonization by the wt strain,

as well as yopH, yopE, and yopK mutants. In addition, dissemination to MLN

of the virulence attenuated mutants increases in absence of PMNs.

PMN-depletion with α-Ly6G antibody is problematic since immature PMNs

are not depleted and can compensate for the loss of mature PMNs in the

immune response towards invading bacteria.

Page 54: Yersinia-phagocyte interactions during early infection

42

ACKNOWLEDGEMENTS

At the age of fourteen me and my friends were asked about our ambitions in life.

My friends did not have a straight answer, but I immediately said “I am going to be

a scientist”. Wherever my heart will finally lead me in life, I at least did science for 6

years and 4 months (project student + PhD). This time has literary been filled with

blood, sweat and tears, but most of the time a lot of fun and fantastic people.

Thank you Maria for accepting me as your PhD student and making my teenage

dream come true. Thank you for being enthusiastic, for challenging me with

gradually increasing responsibility, for sending me to interesting conferences all

over the world and for cheering me up when my mouse experiments showed

totally unexpected and strange results. Those **** neutrophils! I will never forget

the time I spent in your lab.

My fellow group members during the PhD: Anna, my Baloo and second supervisor

who has taken care of me from day one and taught me (almost) all the importat lab

knowledge I have - From how to infect a mouse to how to make noodles in the

microwave oven. Without your good advices about experiments, writing, and life in

general, this journey would not have been the same. It has been truly fantastic

working with you! Sara, the lab was never the same after you left. I missed our

talks about everything and nothing, you singing in the lab, and even your stuff

taking over my lab bench and your phone ringing in the office when you were not

there. Thank you for accepting to be my toastmaster. Good luck with the “football

team”! Kemal, I am gladly handing over the important job of being the senior PhD

student in the group to you. Thank you for saving me when I locked myself out of

my apartment, twice. Kristina, thank you for supplying me with the pIB30 mutant

and lux-strains, for help during the stressful PMN depletion experiment and for

apples and yarn. Saskia, thank you for interesting talks, leftover PMNs, tasty fika

and a lot of nice chocolate. Nayyer, I really like your “I will kill you!” and “Are you

kidding?”-comments. Never stop using them! Take good care of Anna when I am

gone. Karen, for contributions to my first publication. To my hardworking students,

Hanna, Tönis and Anna vdM. Thank you for doing experiments for me! I hope I did

not scare you, but inspire you as my first supervisors, Gosia and Ingrid, did.

6 years and 4 months is a long time and it makes me sad to leave the Department

of Molecular Biology and all its fantastic people. Thank you group HWW, MFr, ÅF

DMi and VS for interesting journal clubs and project presentations. Maybe I will

come back and terrorize you with more immune response papers and talks in the

future. Thank you to the people in the bunker and at the ”PhD-table” for

company, interesting talks, jokes, and fika through all these years. Thank you to my

partners in crime during the years in the marketing group, during teaching and to

Page 55: Yersinia-phagocyte interactions during early infection

43

all the people making crafting nights and molbiol running competitions possible. An

extra thank you to Mari for fun activities in- and outside work. Let us continue

dreaming of a postdoc in the States! Maria N for pep talks and for saving my ass

during the depletion experiments. To Jenny for great company during the trip to

Athens and course at Spetses. To Rutan for help with FACS and for letting me use

your machine. To Marie A, Bhupender and Ingrid for help with the microscopes. To

Johnny for many acute orderings of antibodies and liquid nitrogen, and for lending

me tools, which I always returned! To Ethel for all paperwork and other important

things, and the media ladies for providing me with endless numbers of plates,

buffers and media. Thank you Marek for computer support and Marianella for

keeping the department cleaner than everyone else. And finally, Sven for being the

funniest boss ever!

My friends during the undergraduate student time and after, Sofia, Reine,

Annasara, Annika, Marie and John. Thank you for dinners and other fun get

togethers. And thank you for continuously inviting me to birthday celebrations

even though I am always busy with something else. Anna, maybe you were the

most cleaver in the class. The one who finished, but decided to work with

something totally different. One thing is at least true - Malmö is too far away!

My family, relatives and friends, this thesis is for you! Now, when it is finally time

for me to move on, you might actually understand what I have done during all

these years. Thank you for being interested, asking questions and worrying about

me during periods of too much work. Thank you for lighting up my life outside

work! And a million thanks to those of you who have helped me take care of

Hedvig. This work would not have been finished in time without you! Mom and

Dad, I obviously would not be where I am today without you. Thank you for the

genetic and social contributions that have made me the person I am, and thanks

for letting me be me. Hedvig, my sweet little Bolonka. The best friend anyone can

have! No one will ever be as happy as you are to see me, independently if I left you

for ten minutes or a month. Thank you for all the unconditioned love, joy and

happiness you give me!

Where ever my heart will lead me now, I am grateful that all of you passed me by!

Page 56: Yersinia-phagocyte interactions during early infection

44

REFERENCES

1. Ligon, B.L., Plague: a review of its history and potential as a biological weapon.

Seminars in pediatric infectious diseases, 2006. 17(3): p. 161-70.

2. Leo, J.C. and M. Skurnik, Adhesins of human pathogens from the genus Yersinia.

Advances in experimental medicine and biology, 2011. 715: p. 1-15.

3. Sulakvelidze, A., Yersiniae other than Y. enterocolitica, Y. pseudotuberculosis, and

Y. pestis: the ignored species. Microbes and infection / Institut Pasteur, 2000. 2(5):

p. 497-513.

4. Karlinger, K., et al., The epidemiology and the pathogenesis of inflammatory bowel

disease. European journal of radiology, 2000. 35(3): p. 154-67.

5. Carter, J.D. and A.P. Hudson, Reactive arthritis: clinical aspects and medical

management. Rheumatic diseases clinics of North America, 2009. 35(1): p. 21-44.

6. Eppinger, M., et al., The complete genome sequence of Yersinia pseudotuberculosis

IP31758, the causative agent of Far East scarlet-like fever. PLoS genetics, 2007.

3(8): p. e142.

7. Tahara, M., et al., Analysis of Kawasaki disease showing elevated antibody titres of

Yersinia pseudotuberculosis. Acta paediatrica, 2006. 95(12): p. 1661-4.

8. Galan, J.E. and H. Wolf-Watz, Protein delivery into eukaryotic cells by type III

secretion machines. Nature, 2006. 444(7119): p. 567-73.

9. Michiels, T., et al., Secretion of Yop proteins by Yersiniae. Infection and immunity,

1990. 58(9): p. 2840-9.

10. Salmond, G.P. and P.J. Reeves, Membrane traffic wardens and protein secretion in

gram-negative bacteria. Trends in biochemical sciences, 1993. 18(1): p. 7-12.

11. Cornelis, G.R., et al., The virulence plasmid of Yersinia, an antihost genome.

Microbiology and molecular biology reviews : MMBR, 1998. 62(4): p. 1315-52.

12. Straley, S.C. and R.R. Brubaker, Cytoplasmic and membrane proteins of yersiniae

cultivated under conditions simulating mammalian intracellular environment.

Proceedings of the National Academy of Sciences of the United States of America,

1981. 78(2): p. 1224-8.

13. Portnoy, D.A., S.L. Moseley, and S. Falkow, Characterization of plasmids and

plasmid-associated determinants of Yersinia enterocolitica pathogenesis. Infection

and immunity, 1981. 31(2): p. 775-82.

14. Ghosh, P., Process of protein transport by the type III secretion system.

Microbiology and molecular biology reviews : MMBR, 2004. 68(4): p. 771-95.

15. Dewoody, R.S., P.M. Merritt, and M.M. Marketon, Regulation of the Yersinia type

III secretion system: traffic control. Frontiers in cellular and infection microbiology,

2013. 3: p. 4.

16. Akopyan, K., et al., Translocation of surface-localized effectors in type III secretion.

Proceedings of the National Academy of Sciences of the United States of America,

2011. 108(4): p. 1639-44.

17. Hakansson, S., et al., YopB and YopD constitute a novel class of Yersinia Yop

proteins. Infection and immunity, 1993. 61(1): p. 71-80.

Page 57: Yersinia-phagocyte interactions during early infection

45

18. Hartland, E.L. and R.M. Robins-Browne, In vitro association between the virulence

proteins, YopD and YopE, of Yersinia enterocolitica. FEMS microbiology letters,

1998. 162(2): p. 207-13.

19. Neyt, C. and G.R. Cornelis, Insertion of a Yop translocation pore into the

macrophage plasma membrane by Yersinia enterocolitica: requirement for

translocators YopB and YopD, but not LcrG. Molecular microbiology, 1999. 33(5): p.

971-81.

20. Rosqvist, R., et al., Translocation of the Yersinia YopE and YopH virulence proteins

into target cells is mediated by YopB and YopD. Contributions to microbiology and

immunology, 1995. 13: p. 230-4.

21. Persson, C., et al., Cell-surface-bound Yersinia translocate the protein tyrosine

phosphatase YopH by a polarized mechanism into the target cell. Molecular

microbiology, 1995. 18(1): p. 135-50.

22. Hartland, E.L., et al., Essential role of YopD in inhibition of the respiratory burst of

macrophages by Yersinia enterocolitica. Infection and immunity, 1994. 62(10): p.

4445-53.

23. Hartland, E.L., A.M. Bordun, and R.M. Robins-Browne, Contribution of YopB to

virulence of Yersinia enterocolitica. Infection and immunity, 1996. 64(6): p. 2308-

14.

24. Nilles, M.L., K.A. Fields, and S.C. Straley, The V antigen of Yersinia pestis regulates

Yop vectorial targeting as well as Yop secretion through effects on YopB and LcrG.

Journal of bacteriology, 1998. 180(13): p. 3410-20.

25. Nilles, M.L., et al., Yersinia pestis LcrV forms a stable complex with LcrG and may

have a secretion-related regulatory role in the low-Ca2+ response. Journal of

bacteriology, 1997. 179(4): p. 1307-16.

26. Persson, C., et al., Localization of the Yersinia PTPase to focal complexes is an

important virulence mechanism. Molecular microbiology, 1999. 33(4): p. 828-38.

27. Mueller, C.A., et al., The V-antigen of Yersinia forms a distinct structure at the tip of

injectisome needles. Science, 2005. 310(5748): p. 674-6.

28. Pettersson, J., et al., The V-antigen of Yersinia is surface exposed before target cell

contact and involved in virulence protein translocation. Molecular microbiology,

1999. 32(5): p. 961-76.

29. Holmstrom, A., et al., LcrV is a channel size-determining component of the Yop

effector translocon of Yersinia. Molecular microbiology, 2001. 39(3): p. 620-32.

30. Sarker, M.R., et al., The Yersinia Yop virulon: LcrV is required for extrusion of the

translocators YopB and YopD. Journal of bacteriology, 1998. 180(5): p. 1207-14.

31. Skrzypek, E. and S.C. Straley, Differential effects of deletions in lcrV on secretion of

V antigen, regulation of the low-Ca2+ response, and virulence of Yersinia pestis.

Journal of bacteriology, 1995. 177(9): p. 2530-42.

32. Brubaker, R.R., The V antigen of yersiniae: an overview. Contributions to

microbiology and immunology, 1991. 12: p. 127-33.

33. Quenee, L.E. and O. Schneewind, Plague vaccines and the molecular basis of

immunity against Yersinia pestis. Human vaccines, 2009. 5(12): p. 817-23.

34. Trosky, J.E., A.D. Liverman, and K. Orth, Yersinia outer proteins: Yops. Cellular

microbiology, 2008. 10(3): p. 557-65.

Page 58: Yersinia-phagocyte interactions during early infection

46

35. Guan, K.L. and J.E. Dixon, Protein tyrosine phosphatase activity of an essential

virulence determinant in Yersinia. Science, 1990. 249(4968): p. 553-6.

36. Andersson, K., et al., YopH of Yersinia pseudotuberculosis interrupts early

phosphotyrosine signalling associated with phagocytosis. Molecular microbiology,

1996. 20(5): p. 1057-69.

37. Black, D.S. and J.B. Bliska, Identification of p130Cas as a substrate of Yersinia YopH

(Yop51), a bacterial protein tyrosine phosphatase that translocates into

mammalian cells and targets focal adhesions. The EMBO journal, 1997. 16(10): p.

2730-44.

38. Persson, C., et al., The PTPase YopH inhibits uptake of Yersinia, tyrosine

phosphorylation of p130Cas and FAK, and the associated accumulation of these

proteins in peripheral focal adhesions. The EMBO journal, 1997. 16(9): p. 2307-18.

39. Black, D.S., et al., The Yersinia tyrosine phosphatase YopH targets a novel

adhesion-regulated signalling complex in macrophages. Cellular microbiology,

2000. 2(5): p. 401-14.

40. Hamid, N., et al., YopH dephosphorylates Cas and Fyn-binding protein in

macrophages. Microbial pathogenesis, 1999. 27(4): p. 231-42.

41. Andersson, K., et al., Yersinia pseudotuberculosis-induced calcium signaling in

neutrophils is blocked by the virulence effector YopH. Infection and immunity,

1999. 67(5): p. 2567-74.

42. Rosqvist, R., I. Bolin, and H. Wolf-Watz, Inhibition of phagocytosis in Yersinia

pseudotuberculosis: a virulence plasmid-encoded ability involving the Yop2b

protein. Infection and immunity, 1988. 56(8): p. 2139-43.

43. Yao, T., et al., Suppression of T and B lymphocyte activation by a Yersinia

pseudotuberculosis virulence factor, yopH. The Journal of experimental medicine,

1999. 190(9): p. 1343-50.

44. Gerke, C., S. Falkow, and Y.H. Chien, The adaptor molecules LAT and SLP-76 are

specifically targeted by Yersinia to inhibit T cell activation. The Journal of

experimental medicine, 2005. 201(3): p. 361-71.

45. Bolin, I. and H. Wolf-Watz, The plasmid-encoded Yop2b protein of Yersinia

pseudotuberculosis is a virulence determinant regulated by calcium and

temperature at the level of transcription. Molecular microbiology, 1988. 2(2): p.

237-45.

46. Black, D.S. and J.B. Bliska, The RhoGAP activity of the Yersinia pseudotuberculosis

cytotoxin YopE is required for antiphagocytic function and virulence. Molecular

microbiology, 2000. 37(3): p. 515-27.

47. Von Pawel-Rammingen, U., et al., GAP activity of the Yersinia YopE cytotoxin

specifically targets the Rho pathway: a mechanism for disruption of actin

microfilament structure. Molecular microbiology, 2000. 36(3): p. 737-48.

48. Aili, M., et al., Functional analysis of the YopE GTPase-activating protein (GAP)

activity of Yersinia pseudotuberculosis. Cellular microbiology, 2006. 8(6): p. 1020-

33.

49. Heasman, S.J. and A.J. Ridley, Mammalian Rho GTPases: new insights into their

functions from in vivo studies. Nature reviews. Molecular cell biology, 2008. 9(9): p.

690-701.

Page 59: Yersinia-phagocyte interactions during early infection

47

50. Shao, F., et al., Biochemical characterization of the Yersinia YopT protease:

cleavage site and recognition elements in Rho GTPases. Proceedings of the

National Academy of Sciences of the United States of America, 2003. 100(3): p.

904-9.

51. Aepfelbacher, M., et al., Characterization of YopT effects on Rho GTPases in

Yersinia enterocolitica-infected cells. The Journal of biological chemistry, 2003.

278(35): p. 33217-23.

52. Fueller, F. and G. Schmidt, The polybasic region of Rho GTPases defines the

cleavage by Yersinia enterocolitica outer protein T (YopT). Protein science : a

publication of the Protein Society, 2008. 17(8): p. 1456-62.

53. Adkins, I., et al., Yersinia outer proteins E, H, P, and T differentially target the

cytoskeleton and inhibit phagocytic capacity of dendritic cells. International journal

of medical microbiology : IJMM, 2007. 297(4): p. 235-44.

54. Grosdent, N., et al., Role of Yops and adhesins in resistance of Yersinia

enterocolitica to phagocytosis. Infection and immunity, 2002. 70(8): p. 4165-76.

55. Trulzsch, K., et al., Contribution of the major secreted yops of Yersinia

enterocolitica O:8 to pathogenicity in the mouse infection model. Infection and

immunity, 2004. 72(9): p. 5227-34.

56. Iriarte, M. and G.R. Cornelis, YopT, a new Yersinia Yop effector protein, affects the

cytoskeleton of host cells. Molecular microbiology, 1998. 29(3): p. 915-29.

57. Viboud, G.I. and J.B. Bliska, Yersinia outer proteins: role in modulation of host cell

signaling responses and pathogenesis. Annual review of microbiology, 2005. 59: p.

69-89.

58. Viboud, G.I., E. Mejia, and J.B. Bliska, Comparison of YopE and YopT activities in

counteracting host signalling responses to Yersinia pseudotuberculosis infection.

Cellular microbiology, 2006. 8(9): p. 1504-15.

59. Galyov, E.E., et al., A secreted protein kinase of Yersinia pseudotuberculosis is an

indispensable virulence determinant. Nature, 1993. 361(6414): p. 730-2.

60. Hakansson, S., et al., The Yersinia YpkA Ser/Thr kinase is translocated and

subsequently targeted to the inner surface of the HeLa cell plasma membrane.

Molecular microbiology, 1996. 20(3): p. 593-603.

61. Barz, C., et al., The Yersinia Ser/Thr protein kinase YpkA/YopO directly interacts

with the small GTPases RhoA and Rac-1. FEBS letters, 2000. 482(1-2): p. 139-43.

62. Dukuzumuremyi, J.M., et al., The Yersinia protein kinase A is a host factor inducible

RhoA/Rac-binding virulence factor. The Journal of biological chemistry, 2000.

275(45): p. 35281-90.

63. Juris, S.J., et al., A distinctive role for the Yersinia protein kinase: actin binding,

kinase activation, and cytoskeleton disruption. Proceedings of the National

Academy of Sciences of the United States of America, 2000. 97(17): p. 9431-6.

64. Trasak, C., et al., Yersinia protein kinase YopO is activated by a novel G-actin

binding process. The Journal of biological chemistry, 2007. 282(4): p. 2268-77.

65. Wiley, D.J., et al., The Ser/Thr kinase activity of the Yersinia protein kinase A (YpkA)

is necessary for full virulence in the mouse, mollifying phagocytes, and disrupting

the eukaryotic cytoskeleton. Microbial pathogenesis, 2006. 40(5): p. 234-43.

Page 60: Yersinia-phagocyte interactions during early infection

48

66. Galyov, E.E., S. Hakansson, and H. Wolf-Watz, Characterization of the operon

encoding the YpkA Ser/Thr protein kinase and the YopJ protein of Yersinia

pseudotuberculosis. Journal of bacteriology, 1994. 176(15): p. 4543-8.

67. Logsdon, L.K. and J. Mecsas, Requirement of the Yersinia pseudotuberculosis

effectors YopH and YopE in colonization and persistence in intestinal and lymph

tissues. Infection and immunity, 2003. 71(8): p. 4595-607.

68. Mukherjee, S., et al., Yersinia YopJ acetylates and inhibits kinase activation by

blocking phosphorylation. Science, 2006. 312(5777): p. 1211-4.

69. Mittal, R., S.Y. Peak-Chew, and H.T. McMahon, Acetylation of MEK2 and I kappa B

kinase (IKK) activation loop residues by YopJ inhibits signaling. Proceedings of the

National Academy of Sciences of the United States of America, 2006. 103(49): p.

18574-9.

70. Monack, D.M., et al., Yersinia signals macrophages to undergo apoptosis and YopJ

is necessary for this cell death. Proceedings of the National Academy of Sciences of

the United States of America, 1997. 94(19): p. 10385-90.

71. Palmer, L.E., et al., YopJ of Yersinia spp. is sufficient to cause downregulation of

multiple mitogen-activated protein kinases in eukaryotic cells. Infection and

immunity, 1999. 67(2): p. 708-16.

72. Palmer, L.E., et al., YopJ of Yersinia pseudotuberculosis is required for the inhibition

of macrophage TNF-alpha production and downregulation of the MAP kinases p38

and JNK. Molecular microbiology, 1998. 27(5): p. 953-65.

73. Orth, K., et al., Inhibition of the mitogen-activated protein kinase kinase

superfamily by a Yersinia effector. Science, 1999. 285(5435): p. 1920-3.

74. Schesser, K., et al., The yopJ locus is required for Yersinia-mediated inhibition of NF-

kappaB activation and cytokine expression: YopJ contains a eukaryotic SH2-like

domain that is essential for its repressive activity. Molecular microbiology, 1998.

28(6): p. 1067-79.

75. Monack, D.M., et al., Yersinia-induced apoptosis in vivo aids in the establishment of

a systemic infection of mice. The Journal of experimental medicine, 1998. 188(11):

p. 2127-37.

76. Brodsky, I.E. and R. Medzhitov, Reduced secretion of YopJ by Yersinia limits in vivo

cell death but enhances bacterial virulence. PLoS pathogens, 2008. 4(5): p.

e1000067.

77. Straley, S.C. and W.S. Bowmer, Virulence genes regulated at the transcriptional

level by Ca2+ in Yersinia pestis include structural genes for outer membrane

proteins. Infection and immunity, 1986. 51(2): p. 445-54.

78. Zauberman, A., et al., Disparity between Yersinia pestis and Yersinia enterocolitica

O:8 in YopJ/YopP-dependent functions. Advances in experimental medicine and

biology, 2007. 603: p. 312-20.

79. Meinzer, U., et al., Yersinia pseudotuberculosis effector YopJ subverts the

Nod2/RICK/TAK1 pathway and activates caspase-1 to induce intestinal barrier

dysfunction. Cell host & microbe, 2012. 11(4): p. 337-51.

80. Zauberman, A., et al., Yersinia pestis endowed with increased cytotoxicity is

avirulent in a bubonic plague model and induces rapid protection against

pneumonic plague. PloS one, 2009. 4(6): p. e5938.

Page 61: Yersinia-phagocyte interactions during early infection

49

81. Grobner, S., et al., Yersinia YopP-induced apoptotic cell death in murine dendritic

cells is partially independent from action of caspases and exhibits necrosis-like

features. Apoptosis : an international journal on programmed cell death, 2006.

11(11): p. 1959-68.

82. Evdokimov, A.G., et al., Unusual molecular architecture of the Yersinia pestis

cytotoxin YopM: a leucine-rich repeat protein with the shortest repeating unit.

Journal of molecular biology, 2001. 312(4): p. 807-21.

83. Skrzypek, E., et al., Application of a Saccharomyces cerevisiae model to study

requirements for trafficking of Yersinia pestis YopM in eucaryotic cells. Infection

and immunity, 2003. 71(2): p. 937-47.

84. Benabdillah, R., et al., Identification of a nuclear targeting signal in YopM from

Yersinia spp. Microbial pathogenesis, 2004. 36(5): p. 247-61.

85. McDonald, C., et al., The yersinia virulence factor YopM forms a novel protein

complex with two cellular kinases. The Journal of biological chemistry, 2003.

278(20): p. 18514-23.

86. McPhee, J.B., P. Mena, and J.B. Bliska, Delineation of regions of the Yersinia YopM

protein required for interaction with the RSK1 and PRK2 host kinases and their

requirement for interleukin-10 production and virulence. Infection and immunity,

2010. 78(8): p. 3529-39.

87. LaRock, C.N. and B.T. Cookson, The Yersinia virulence effector YopM binds caspase-

1 to arrest inflammasome assembly and processing. Cell host & microbe, 2012.

12(6): p. 799-805.

88. Kerschen, E.J., et al., The plague virulence protein YopM targets the innate immune

response by causing a global depletion of NK cells. Infection and immunity, 2004.

72(8): p. 4589-602.

89. Mulder, B., et al., Identification of additional virulence determinants on the pYV

plasmid of Yersinia enterocolitica W227. Infection and immunity, 1989. 57(8): p.

2534-41.

90. Leung, K.Y., B.S. Reisner, and S.C. Straley, YopM inhibits platelet aggregation and is

necessary for virulence of Yersinia pestis in mice. Infection and immunity, 1990.

58(10): p. 3262-71.

91. Holmstrom, A., et al., YopK of Yersinia pseudotuberculosis controls translocation of

Yop effectors across the eukaryotic cell membrane. Molecular microbiology, 1997.

24(1): p. 73-91.

92. Thorslund, S.E., et al., The RACK1 signaling scaffold protein selectively interacts

with Yersinia pseudotuberculosis virulence function. PloS one, 2011. 6(2): p.

e16784.

93. Holmstrom, A., et al., Virulence plasmid-encoded YopK is essential for Yersinia

pseudotuberculosis to cause systemic infection in mice. Infection and immunity,

1995. 63(6): p. 2269-76.

94. Thorslund, S.E., et al., Role of YopK in Yersinia pseudotuberculosis resistance

against polymorphonuclear leukocyte defense. Infection and immunity, 2013.

81(1): p. 11-22.

95. Straley, S.C. and M.L. Cibull, Differential clearance and host-pathogen interactions

of YopE- and YopK- YopL- Yersinia pestis in BALB/c mice. Infection and immunity,

1989. 57(4): p. 1200-10.

Page 62: Yersinia-phagocyte interactions during early infection

50

96. Aili, M., et al., Regulation of Yersinia Yop-effector delivery by translocated YopE.

International journal of medical microbiology : IJMM, 2008. 298(3-4): p. 183-92.

97. Forsberg, A. and H. Wolf-Watz, The virulence protein Yop5 of Yersinia

pseudotuberculosis is regulated at transcriptional level by plasmid-plB1-encoded

trans-acting elements controlled by temperature and calcium. Molecular

microbiology, 1988. 2(1): p. 121-33.

98. Hoe, N.P., F.C. Minion, and J.D. Goguen, Temperature sensing in Yersinia pestis:

regulation of yopE transcription by lcrF. Journal of bacteriology, 1992. 174(13): p.

4275-86.

99. Hoe, N.P. and J.D. Goguen, Temperature sensing in Yersinia pestis: translation of

the LcrF activator protein is thermally regulated. Journal of bacteriology, 1993.

175(24): p. 7901-9.

100. Forsberg, A., et al., Molecular cloning and expression of calcium-regulated,

plasmid-coded proteins of Y. pseudotuberculosis. Microbial pathogenesis, 1987.

2(2): p. 123-37.

101. Bolin, I., D.A. Portnoy, and H. Wolf-Watz, Expression of the temperature-inducible

outer membrane proteins of yersiniae. Infection and immunity, 1985. 48(1): p. 234-

40.

102. Pettersson, J., et al., Modulation of virulence factor expression by pathogen target

cell contact. Science, 1996. 273(5279): p. 1231-3.

103. Rosqvist, R., K.E. Magnusson, and H. Wolf-Watz, Target cell contact triggers

expression and polarized transfer of Yersinia YopE cytotoxin into mammalian cells.

The EMBO journal, 1994. 13(4): p. 964-72.

104. Trulzsch, K., et al., Analysis of chaperone-dependent Yop secretion/translocation

and effector function using a mini-virulence plasmid of Yersinia enterocolitica.

International journal of medical microbiology : IJMM, 2003. 293(2-3): p. 167-77.

105. Revell, P.A. and V.L. Miller, A chromosomally encoded regulator is required for

expression of the Yersinia enterocolitica inv gene and for virulence. Molecular

microbiology, 2000. 35(3): p. 677-85.

106. Grassl, G.A., et al., Interaction of Yersinia enterocolitica with epithelial cells: invasin

beyond invasion. International journal of medical microbiology : IJMM, 2003.

293(1): p. 41-54.

107. Clark, M.A., B.H. Hirst, and M.A. Jepson, M-cell surface beta1 integrin expression

and invasin-mediated targeting of Yersinia pseudotuberculosis to mouse Peyer's

patch M cells. Infection and immunity, 1998. 66(3): p. 1237-43.

108. Schulte, R., et al., Translocation of Yersinia entrocolitica across reconstituted

intestinal epithelial monolayers is triggered by Yersinia invasin binding to beta1

integrins apically expressed on M-like cells. Cellular microbiology, 2000. 2(2): p.

173-85.

109. Marra, A. and R.R. Isberg, Invasin-dependent and invasin-independent pathways

for translocation of Yersinia pseudotuberculosis across the Peyer's patch intestinal

epithelium. Infection and immunity, 1997. 65(8): p. 3412-21.

110. Autenrieth, I.B. and R. Firsching, Penetration of M cells and destruction of Peyer's

patches by Yersinia enterocolitica: an ultrastructural and histological study. Journal

of medical microbiology, 1996. 44(4): p. 285-94.

Page 63: Yersinia-phagocyte interactions during early infection

51

111. Durand, E.A., et al., The presence of professional phagocytes dictates the number

of host cells targeted for Yop translocation during infection. Cellular microbiology,

2010. 12(8): p. 1064-82.

112. Pepe, J.C. and V.L. Miller, Yersinia enterocolitica invasin: a primary role in the

initiation of infection. Proceedings of the National Academy of Sciences of the

United States of America, 1993. 90(14): p. 6473-7.

113. Pepe, J.C., et al., Pathogenesis of defined invasion mutants of Yersinia

enterocolitica in a BALB/c mouse model of infection. Infection and immunity, 1995.

63(12): p. 4837-48.

114. Maldonado-Arocho, F.J., et al., Adhesins and host serum factors drive Yop

translocation by yersinia into professional phagocytes during animal infection.

PLoS pathogens, 2013. 9(6): p. e1003415.

115. Mejia, E., J.B. Bliska, and G.I. Viboud, Yersinia controls type III effector delivery into

host cells by modulating Rho activity. PLoS pathogens, 2008. 4(1): p. e3.

116. Boyd, A.P., et al., Yersinia enterocolitica can deliver Yop proteins into a wide range

of cell types: development of a delivery system for heterologous proteins. European

journal of cell biology, 2000. 79(10): p. 659-71.

117. Fallman, M., et al., Yersinia pseudotuberculosis inhibits Fc receptor-mediated

phagocytosis in J774 cells. Infection and immunity, 1995. 63(8): p. 3117-24.

118. Deuretzbacher, A., et al., Beta1 integrin-dependent engulfment of Yersinia

enterocolitica by macrophages is coupled to the activation of autophagy and

suppressed by type III protein secretion. Journal of immunology, 2009. 183(9): p.

5847-60.

119. Hoiczyk, E., et al., Structure and sequence analysis of Yersinia YadA and Moraxella

UspAs reveal a novel class of adhesins. The EMBO journal, 2000. 19(22): p. 5989-

99.

120. El Tahir, Y. and M. Skurnik, YadA, the multifaceted Yersinia adhesin. International

journal of medical microbiology : IJMM, 2001. 291(3): p. 209-18.

121. Eitel, J. and P. Dersch, The YadA protein of Yersinia pseudotuberculosis mediates

high-efficiency uptake into human cells under environmental conditions in which

invasin is repressed. Infection and immunity, 2002. 70(9): p. 4880-91.

122. Biedzka-Sarek, M., R. Venho, and M. Skurnik, Role of YadA, Ail, and

Lipopolysaccharide in Serum Resistance of Yersinia enterocolitica Serotype O:3.

Infection and immunity, 2005. 73(4): p. 2232-44.

123. Kirjavainen, V., et al., Yersinia enterocolitica serum resistance proteins YadA and ail

bind the complement regulator C4b-binding protein. PLoS pathogens, 2008. 4(8): p.

e1000140.

124. Biedzka-Sarek, M., et al., Functional mapping of YadA- and Ail-mediated binding of

human factor H to Yersinia enterocolitica serotype O:3. Infection and immunity,

2008. 76(11): p. 5016-27.

125. Biedzka-Sarek, M., et al., Characterization of complement factor H binding to

Yersinia enterocolitica serotype O:3. Infection and immunity, 2008. 76(9): p. 4100-

9.

126. Costa, T.R., et al., Type III secretion translocon assemblies that attenuate Yersinia

virulence. Cellular microbiology, 2013. 15(7): p. 1088-110.

Page 64: Yersinia-phagocyte interactions during early infection

52

127. Ho, D.K., et al., The Yersinia pseudotuberculosis outer membrane protein Ail

recruits the human complement regulatory protein factor H. Journal of

immunology, 2012. 189(7): p. 3593-9.

128. Ho, D.K., et al., Functional recruitment of the human complement inhibitor C4BP to

Yersinia pseudotuberculosis outer membrane protein Ail. Journal of immunology,

2012. 188(9): p. 4450-9.

129. Groves, E., et al., Molecular mechanisms of phagocytic uptake in mammalian cells.

Cellular and molecular life sciences : CMLS, 2008. 65(13): p. 1957-76.

130. Mogensen, T.H., Pathogen recognition and inflammatory signaling in innate

immune defenses. Clinical microbiology reviews, 2009. 22(2): p. 240-73, Table of

Contents.

131. Aderem, A. and D.M. Underhill, Mechanisms of phagocytosis in macrophages.

Annual review of immunology, 1999. 17: p. 593-623.

132. Tjelle, T.E., T. Lovdal, and T. Berg, Phagosome dynamics and function. BioEssays :

news and reviews in molecular, cellular and developmental biology, 2000. 22(3): p.

255-63.

133. Lim, J.P. and P.A. Gleeson, Macropinocytosis: an endocytic pathway for

internalising large gulps. Immunology and cell biology, 2011. 89(8): p. 836-43.

134. van Furth, R. and Z.A. Cohn, The origin and kinetics of mononuclear phagocytes.

The Journal of experimental medicine, 1968. 128(3): p. 415-35.

135. Randolph, G.J., et al., Differentiation of phagocytic monocytes into lymph node

dendritic cells in vivo. Immunity, 1999. 11(6): p. 753-61.

136. Varol, C., et al., Monocytes give rise to mucosal, but not splenic, conventional

dendritic cells. The Journal of experimental medicine, 2007. 204(1): p. 171-80.

137. Shi, C. and E.G. Pamer, Monocyte recruitment during infection and inflammation.

Nature Reviews Immunology, 2011. 11(11): p. 762-774.

138. Ziegler-Heitbrock, L., et al., Nomenclature of monocytes and dendritic cells in

blood. Blood, 2010. 116(16): p. e74-80.

139. Ingersoll, M.A., et al., Monocyte trafficking in acute and chronic inflammation.

Trends in immunology, 2011. 32(10): p. 470-7.

140. Murray, P.J. and T.A. Wynn, Protective and pathogenic functions of macrophage

subsets. Nature reviews. Immunology, 2011. 11(11): p. 723-37.

141. van Helden, S.F., F.N. van Leeuwen, and C.G. Figdor, Human and murine model cell

lines for dendritic cell biology evaluated. Immunology letters, 2008. 117(2): p. 191-

7.

142. Pfau, J.C., et al., Environmental oxygen tension affects phenotype in cultured bone

marrow-derived macrophages. American journal of physiology. Lung cellular and

molecular physiology, 2004. 286(2): p. L354-62.

143. Leidi, M., et al., M2 macrophages phagocytose rituximab-opsonized leukemic

targets more efficiently than m1 cells in vitro. Journal of immunology, 2009.

182(7): p. 4415-22.

144. Duffield, J.S., et al., Selective depletion of macrophages reveals distinct, opposing

roles during liver injury and repair. The Journal of clinical investigation, 2005.

115(1): p. 56-65.

145. Goren, I., et al., A transgenic mouse model of inducible macrophage depletion:

effects of diphtheria toxin-driven lysozyme M-specific cell lineage ablation on

Page 65: Yersinia-phagocyte interactions during early infection

53

wound inflammatory, angiogenic, and contractive processes. The American journal

of pathology, 2009. 175(1): p. 132-47.

146. Van Rooijen, N. and A. Sanders, Liposome mediated depletion of macrophages:

mechanism of action, preparation of liposomes and applications. Journal of

immunological methods, 1994. 174(1-2): p. 83-93.

147. Doeing, D.C., J.L. Borowicz, and E.T. Crockett, Gender dimorphism in differential

peripheral blood leukocyte counts in mice using cardiac, tail, foot, and saphenous

vein puncture methods. BMC clinical pathology, 2003. 3(1): p. 3.

148. Haley, P.J., Species differences in the structure and function of the immune system.

Toxicology, 2003. 188(1): p. 49-71.

149. Kolaczkowska, E. and P. Kubes, Neutrophil recruitment and function in health and

inflammation. Nature reviews. Immunology, 2013. 13(3): p. 159-75.

150. Pillay, J., et al., A subset of neutrophils in human systemic inflammation inhibits T

cell responses through Mac-1. The Journal of clinical investigation, 2012. 122(1): p.

327-36.

151. Tsuda, Y., et al., Three different neutrophil subsets exhibited in mice with different

susceptibilities to infection by methicillin-resistant Staphylococcus aureus.

Immunity, 2004. 21(2): p. 215-26.

152. Fridlender, Z.G., et al., Polarization of tumor-associated neutrophil phenotype by

TGF-beta: "N1" versus "N2" TAN. Cancer cell, 2009. 16(3): p. 183-94.

153. Kolaczkowska, E. and P. Kubes, Phagocytes & granulocytes. Angiogenic neutrophils:

a novel subpopulation paradigm. Blood, 2012. 120(23): p. 4455-7.

154. Thomas, C.J. and K. Schroder, Pattern recognition receptor function in neutrophils.

Trends in immunology, 2013.

155. Newburger, P.E., et al., Functional changes in human leukemic cell line HL-60. A

model for myeloid differentiation. The Journal of cell biology, 1979. 82(2): p. 315-

22.

156. Breitman, T.R., S.E. Selonick, and S.J. Collins, Induction of differentiation of the

human promyelocytic leukemia cell line (HL-60) by retinoic acid. Proceedings of the

National Academy of Sciences of the United States of America, 1980. 77(5): p.

2936-40.

157. Breitman, T.R., S.J. Collins, and B.R. Keene, Terminal differentiation of human

promyelocytic leukemic cells in primary culture in response to retinoic acid. Blood,

1981. 57(6): p. 1000-4.

158. Daley, J.M., et al., Use of Ly6G-specific monoclonal antibody to deplete neutrophils

in mice. Journal of leukocyte biology, 2008. 83(1): p. 64-70.

159. Fleming, T.J., M.L. Fleming, and T.R. Malek, Selective expression of Ly-6G on

myeloid lineage cells in mouse bone marrow. RB6-8C5 mAb to granulocyte-

differentiation antigen (Gr-1) detects members of the Ly-6 family. Journal of

immunology, 1993. 151(5): p. 2399-408.

160. Tepper, R.I., R.L. Coffman, and P. Leder, An eosinophil-dependent mechanism for

the antitumor effect of interleukin-4. Science, 1992. 257(5069): p. 548-51.

161. Banchereau, J. and R.M. Steinman, Dendritic cells and the control of immunity.

Nature, 1998. 392(6673): p. 245-52.

162. Kushwah, R. and J. Hu, Complexity of dendritic cell subsets and their function in the

host immune system. Immunology, 2011. 133(4): p. 409-19.

Page 66: Yersinia-phagocyte interactions during early infection

54

163. Aguzzi, A. and N.J. Krautler, Characterizing follicular dendritic cells: A progress

report. European journal of immunology, 2010. 40(8): p. 2134-8.

164. Buckwalter, M.R. and M.L. Albert, Orchestration of the immune response by

dendritic cells. Current biology : CB, 2009. 19(9): p. R355-61.

165. Iwasaki, A. and B.L. Kelsall, Unique functions of CD11b+, CD8 alpha+, and double-

negative Peyer's patch dendritic cells. Journal of immunology, 2001. 166(8): p.

4884-90.

166. Gilliet, M., W. Cao, and Y.J. Liu, Plasmacytoid dendritic cells: sensing nucleic acids in

viral infection and autoimmune diseases. Nature reviews. Immunology, 2008. 8(8):

p. 594-606.

167. Satpathy, A.T., et al., Re(de)fining the dendritic cell lineage. Nature immunology,

2012. 13(12): p. 1145-54.

168. Colonna, M., G. Trinchieri, and Y.J. Liu, Plasmacytoid dendritic cells in immunity.

Nature immunology, 2004. 5(12): p. 1219-26.

169. Granucci, F., et al., Early IL-2 production by mouse dendritic cells is the result of

microbial-induced priming. Journal of immunology, 2003. 170(10): p. 5075-81.

170. Sallusto, F., et al., Dendritic cells use macropinocytosis and the mannose receptor

to concentrate macromolecules in the major histocompatibility complex class II

compartment: downregulation by cytokines and bacterial products. The Journal of

experimental medicine, 1995. 182(2): p. 389-400.

171. Winzler, C., et al., Maturation stages of mouse dendritic cells in growth factor-

dependent long-term cultures. The Journal of experimental medicine, 1997. 185(2):

p. 317-28.

172. Sato, K. and S. Fujita, Dendritic cells: nature and classification. Allergology

international : official journal of the Japanese Society of Allergology, 2007. 56(3):

p. 183-91.

173. Xu, Y., et al., Differential development of murine dendritic cells by GM-CSF versus

Flt3 ligand has implications for inflammation and trafficking. Journal of

immunology, 2007. 179(11): p. 7577-84.

174. Pappenheimer, A.M., Jr., et al., Diphtheria toxin and related proteins: effect of

route of injection on toxicity and the determination of cytotoxicity for various

cultured cells. The Journal of infectious diseases, 1982. 145(1): p. 94-102.

175. Jung, S., et al., In vivo depletion of CD11c+ dendritic cells abrogates priming of

CD8+ T cells by exogenous cell-associated antigens. Immunity, 2002. 17(2): p. 211-

20.

176. Bar-On, L. and S. Jung, Defining in vivo dendritic cell functions using CD11c-DTR

transgenic mice. Methods in molecular biology, 2010. 595: p. 429-42.

177. Bennett, C.L. and B.E. Clausen, DC ablation in mice: promises, pitfalls, and

challenges. Trends in immunology, 2007. 28(12): p. 525-31.

178. Sapoznikov, A., et al., Organ-dependent in vivo priming of naive CD4+, but not

CD8+, T cells by plasmacytoid dendritic cells. The Journal of experimental medicine,

2007. 204(8): p. 1923-33.

179. Probst, H.C., et al., Histological analysis of CD11c-DTR/GFP mice after in vivo

depletion of dendritic cells. Clinical and experimental immunology, 2005. 141(3): p.

398-404.

Page 67: Yersinia-phagocyte interactions during early infection

55

180. Tittel, A.P., et al., Functionally relevant neutrophilia in CD11c diphtheria toxin

receptor transgenic mice. Nature methods, 2012. 9(4): p. 385-90.

181. Hochweller, K., et al., A novel CD11c.DTR transgenic mouse for depletion of

dendritic cells reveals their requirement for homeostatic proliferation of natural

killer cells. European journal of immunology, 2008. 38(10): p. 2776-83.

182. van Blijswijk, J., B.U. Schraml, and C. Reis e Sousa, Advantages and limitations of

mouse models to deplete dendritic cells. European journal of immunology, 2013.

43(1): p. 22-6.

183. Zhang, Y., et al., APCs in the liver and spleen recruit activated allogeneic CD8+ T

cells to elicit hepatic graft-versus-host disease. Journal of immunology, 2002.

169(12): p. 7111-8.

184. Bonardi, S., et al., Detection, semiquantitative enumeration, and antimicrobial

susceptibility of Yersinia enterocolitica in pork and chicken meats in Italy. Journal of

food protection, 2010. 73(10): p. 1785-92.

185. Schiemann, D.A., Yersinia enterocolitica in milk and dairy products. Journal of dairy

science, 1987. 70(2): p. 383-91.

186. Lee, T.S., et al., Prevalence, antibiotic susceptibility, and virulence factors of

Yersinia enterocolitica and related species from ready-to-eat vegetables available

in Korea. Journal of food protection, 2004. 67(6): p. 1123-7.

187. Andrew, J.H., D. Weedon, and N.S. Mair, Terminal ileitis, mesenteric lymphadenitis

and appendicitis due to Yersinia pseudotuberculosis type VA: case report.

Pathology, 1976. 8(3): p. 189-93.

188. El-Maraghi, N.R. and N.S. Mair, The histopathology of enteric infection with

Yersinia pseudotuberculosis. American journal of clinical pathology, 1979. 71(6): p.

631-9.

189. Tripoli, L.C., et al., Disseminated Yersinia enterocolitica. Case report and review of

the literature. Journal of clinical gastroenterology, 1990. 12(1): p. 85-9.

190. Saebo, A. and J. Lassen, Survival and causes of death among patients with Yersinia

enterocolitica infection. A Norwegian 10-year follow-up study on 458 hospitalized

patients. Scandinavian journal of infectious diseases, 1992. 24(5): p. 613-7.

191. Watson, G.T., et al., When Nature Meets Nurture: Persistent Yersinia Infection. The

American journal of medicine, 2013.

192. Hoogkamp-Korstanje, J.A., J. de Koning, and J. Heesemann, Persistence of Yersinia

enterocolitica in man. Infection, 1988. 16(2): p. 81-5.

193. Hannu, T., et al., Reactive arthritis after an outbreak of Yersinia pseudotuberculosis

serotype O:3 infection. Annals of the rheumatic diseases, 2003. 62(9): p. 866-9.

194. Tertti, R., et al., An outbreak of Yersinia pseudotuberculosis infection. The Journal

of infectious diseases, 1984. 149(2): p. 245-50.

195. Barnes, P.D., et al., Yersinia pseudotuberculosis disseminates directly from a

replicating bacterial pool in the intestine. The Journal of experimental medicine,

2006. 203(6): p. 1591-601.

196. Trcek, J., K. Berschl, and K. Trulzsch, In vivo analysis of Yersinia enterocolitica

infection using luxCDABE. FEMS microbiology letters, 2010. 307(2): p. 201-6.

197. Hancock, G.E., R.W. Schaedler, and T.T. MacDonald, Yersinia enterocolitica

infection in resistant and susceptible strains of mice. Infection and immunity, 1986.

53(1): p. 26-31.

Page 68: Yersinia-phagocyte interactions during early infection

56

198. Autenrieth, I.B., et al., Immune responses to Yersinia enterocolitica in susceptible

BALB/c and resistant C57BL/6 mice: an essential role for gamma interferon.

Infection and immunity, 1994. 62(6): p. 2590-9.

199. Tansini, A. and B.M. de Medeiros, Susceptibility to Yersinia pseudotuberculosis

infection is linked to the pattern of macrophage activation. Scandinavian journal of

immunology, 2009. 69(4): p. 310-8.

200. Tumitan, A.R., et al., Pattern of macrophage activation in yersinia-resistant and

yersinia-susceptible strains of mice. Microbiology and immunology, 2007. 51(10):

p. 1021-8.

201. Grutzkau, A., et al., Involvement of M cells in the bacterial invasion of Peyer's

patches: a common mechanism shared by Yersinia enterocolitica and other

enteroinvasive bacteria. Gut, 1990. 31(9): p. 1011-5.

202. Hanski, C., et al., Immunohistochemical and electron microscopic study of

interaction of Yersinia enterocolitica serotype O8 with intestinal mucosa during

experimental enteritis. Infection and immunity, 1989. 57(3): p. 673-8.

203. Oellerich, M.F., et al., Yersinia enterocolitica infection of mice reveals clonal

invasion and abscess formation. Infection and immunity, 2007. 75(8): p. 3802-11.

204. Simonet, M., S. Richard, and P. Berche, Electron microscopic evidence for in vivo

extracellular localization of Yersinia pseudotuberculosis harboring the pYV plasmid.

Infection and immunity, 1990. 58(3): p. 841-5.

205. Balada-Llasat, J.M. and J. Mecsas, Yersinia has a tropism for B and T cell zones of

lymph nodes that is independent of the type III secretion system. PLoS pathogens,

2006. 2(9): p. e86.

206. Handley, S.A., et al., Characterization of oral Yersinia enterocolitica infection in

three different strains of inbred mice. Infection and immunity, 2004. 72(3): p. 1645-

56.

207. Autenrieth, I.B., et al., T lymphocytes mediate protection against Yersinia

enterocolitica in mice: characterization of murine T-cell clones specific for Y.

enterocolitica. Infection and immunity, 1992. 60(3): p. 1140-9.

208. Logsdon, L.K. and J. Mecsas, The proinflammatory response induced by wild-type

Yersinia pseudotuberculosis infection inhibits survival of yop mutants in the

gastrointestinal tract and Peyer's patches. Infection and immunity, 2006. 74(3): p.

1516-27.

209. Koberle, M., et al., Yersinia enterocolitica targets cells of the innate and adaptive

immune system by injection of Yops in a mouse infection model. PLoS pathogens,

2009. 5(8): p. e1000551.

210. Rosqvist, R., et al., The cytotoxic protein YopE of Yersinia obstructs the primary host

defence. Molecular microbiology, 1990. 4(4): p. 657-67.

211. Russmann, H., et al., Alternative endogenous protein processing via an autophagy-

dependent pathway compensates for Yersinia-mediated inhibition of endosomal

major histocompatibility complex class II antigen presentation. Infection and

immunity, 2010. 78(12): p. 5138-50.

212. Bliska, J.B. and D.S. Black, Inhibition of the Fc receptor-mediated oxidative burst in

macrophages by the Yersinia pseudotuberculosis tyrosine phosphatase. Infection

and immunity, 1995. 63(2): p. 681-5.

Page 69: Yersinia-phagocyte interactions during early infection

57

213. Ruckdeschel, K., et al., Yersinia enterocolitica promotes deactivation of

macrophage mitogen-activated protein kinases extracellular signal-regulated

kinase-1/2, p38, and c-Jun NH2-terminal kinase. Correlation with its inhibitory

effect on tumor necrosis factor-alpha production. The Journal of biological

chemistry, 1997. 272(25): p. 15920-7.

214. Schotte, P., et al., Targeting Rac1 by the Yersinia effector protein YopE inhibits

caspase-1-mediated maturation and release of interleukin-1beta. The Journal of

biological chemistry, 2004. 279(24): p. 25134-42.

215. Mills, S.D., et al., Yersinia enterocolitica induces apoptosis in macrophages by a

process requiring functional type III secretion and translocation mechanisms and

involving YopP, presumably acting as an effector protein. Proceedings of the

National Academy of Sciences of the United States of America, 1997. 94(23): p.

12638-43.

216. Zheng, Y., et al., YopJ-induced caspase-1 activation in Yersinia-infected

macrophages: independent of apoptosis, linked to necrosis, dispensable for innate

host defense. PloS one, 2012. 7(4): p. e36019.

217. Lilo, S., Y. Zheng, and J.B. Bliska, Caspase-1 activation in macrophages infected

with Yersinia pestis KIM requires the type III secretion system effector YopJ.

Infection and immunity, 2008. 76(9): p. 3911-23.

218. Zheng, Y., et al., A Yersinia effector with enhanced inhibitory activity on the NF-

kappaB pathway activates the NLRP3/ASC/caspase-1 inflammasome in

macrophages. PLoS pathogens, 2011. 7(4): p. e1002026.

219. Auerbuch, V., D.T. Golenbock, and R.R. Isberg, Innate immune recognition of

Yersinia pseudotuberculosis type III secretion. PLoS pathogens, 2009. 5(12): p.

e1000686.

220. Brodsky, I.E., et al., A Yersinia effector protein promotes virulence by preventing

inflammasome recognition of the type III secretion system. Cell host & microbe,

2010. 7(5): p. 376-87.

221. Fitzgerald, K.A. and L.A. O'Neill, The role of the interleukin-1/Toll-like receptor

superfamily in inflammation and host defence. Microbes and infection / Institut

Pasteur, 2000. 2(8): p. 933-43.

222. Zhang, Y., et al., Type III secretion decreases bacterial and host survival following

phagocytosis of Yersinia pseudotuberculosis by macrophages. Infection and

immunity, 2008. 76(9): p. 4299-310.

223. Zhang, Y., G. Romanov, and J.B. Bliska, Type III secretion system-dependent

translocation of ectopically expressed Yop effectors into macrophages by

intracellular Yersinia pseudotuberculosis. Infection and immunity, 2011. 79(11): p.

4322-31.

224. Grabenstein, J.P., et al., The response regulator PhoP of Yersinia

pseudotuberculosis is important for replication in macrophages and for virulence.

Infection and immunity, 2004. 72(9): p. 4973-84.

225. Pujol, C. and J.B. Bliska, The ability to replicate in macrophages is conserved

between Yersinia pestis and Yersinia pseudotuberculosis. Infection and immunity,

2003. 71(10): p. 5892-9.

Page 70: Yersinia-phagocyte interactions during early infection

58

226. McCoy, M.W., et al., The C-terminal tail of Yersinia pseudotuberculosis YopM is

critical for interacting with RSK1 and for virulence. Infection and immunity, 2010.

78(6): p. 2584-98.

227. Ruckdeschel, K., et al., Differential contribution of Yersinia enterocolitica virulence

factors to evasion of microbicidal action of neutrophils. Infection and immunity,

1996. 64(3): p. 724-33.

228. Casutt-Meyer, S., et al., Oligomeric coiled-coil adhesin YadA is a double-edged

sword. PloS one, 2010. 5(12): p. e15159.

229. Spinner, J.L., et al., Neutrophils are resistant to Yersinia YopJ/P-induced apoptosis

and are protected from ROS-mediated cell death by the type III secretion system.

PloS one, 2010. 5(2): p. e9279.

230. Conlan, J.W., Critical roles of neutrophils in host defense against experimental

systemic infections of mice by Listeria monocytogenes, Salmonella typhimurium,

and Yersinia enterocolitica. Infection and immunity, 1997. 65(2): p. 630-5.

231. Ye, Z., et al., Gr1+ cells control growth of YopM-negative yersinia pestis during

systemic plague. Infection and immunity, 2009. 77(9): p. 3791-806.

232. Ye, Z., et al., Distinct CCR2(+) Gr1(+) cells control growth of the Yersinia pestis

DeltayopM mutant in liver and spleen during systemic plague. Infection and

immunity, 2011. 79(2): p. 674-87.

233. Autenrieth, S.E., et al., Immune evasion by Yersinia enterocolitica: differential

targeting of dendritic cell subpopulations in vivo. PLoS pathogens, 2010. 6(11): p.

e1001212.

234. Trulzsch, K., et al., Yersinia outer protein P inhibits CD8 T cell priming in the mouse

infection model. Journal of immunology, 2005. 174(7): p. 4244-51.

235. Kramer, U. and C.A. Wiedig, Y. enterocolitica translocated Yops impair stimulation

of T-cells by antigen presenting cells. Immunology letters, 2005. 100(2): p. 130-8.

236. Autenrieth, S.E., et al., Yersinia enterocolitica YopP inhibits MAP kinase-mediated

antigen uptake in dendritic cells. Cellular microbiology, 2007. 9(2): p. 425-37.

237. Erfurth, S.E., et al., Yersinia enterocolitica induces apoptosis and inhibits surface

molecule expression and cytokine production in murine dendritic cells. Infection

and immunity, 2004. 72(12): p. 7045-54.

238. Autenrieth, I.B., et al., Immunohistological characterization of the cellular immune

response against Yersinia enterocolitica in mice: evidence for the involvement of T

lymphocytes. Immunobiology, 1993. 187(1-2): p. 1-16.

239. Autenrieth, I.B., et al., The cellular immune response against Yersinia enterocolitica

in different inbred strains of mice: evidence for an important role of T lymphocytes.

Zentralblatt fur Bakteriologie : international journal of medical microbiology, 1993.

278(2-3): p. 383-95.

240. Mestas, J. and C.C. Hughes, Of mice and not men: differences between mouse and

human immunology. Journal of immunology, 2004. 172(5): p. 2731-8.

241. Waterston, R.H., et al., Initial sequencing and comparative analysis of the mouse

genome. Nature, 2002. 420(6915): p. 520-62.

242. Seok, J., et al., Genomic responses in mouse models poorly mimic human

inflammatory diseases. Proceedings of the National Academy of Sciences of the

United States of America, 2013. 110(9): p. 3507-12.

243. Of men, not mice. Nature medicine, 2013. 19(4): p. 379-379.

Page 71: Yersinia-phagocyte interactions during early infection

59

244. Velan, B., et al., Discordance in the effects of Yersinia pestis on the dendritic cell

functions manifested by induction of maturation and paralysis of migration.

Infection and immunity, 2006. 74(11): p. 6365-76.

245. Robinson, R.T., et al., Yersinia pestis evades TLR4-dependent induction of IL-

12(p40)2 by dendritic cells and subsequent cell migration. Journal of immunology,

2008. 181(8): p. 5560-7.

246. Marketon, M.M., et al., Plague bacteria target immune cells during infection.

Science, 2005. 309(5741): p. 1739-41.

247. O'Neill, G.M., S.J. Fashena, and E.A. Golemis, Integrin signalling: a new Cas(t) of

characters enters the stage. Trends in cell biology, 2000. 10(3): p. 111-9.

248. Yuan, M., L. Mogemark, and M. Fallman, Fyn binding protein, Fyb, interacts with

mammalian actin binding protein, mAbp1. FEBS letters, 2005. 579(11): p. 2339-47.

249. Hapfelmeier, S., et al., Microbe sampling by mucosal dendritic cells is a discrete,

MyD88-independent step in DeltainvG S. Typhimurium colitis. The Journal of

experimental medicine, 2008. 205(2): p. 437-50.

250. Loof, T.G., et al., The contribution of dendritic cells to host defenses against

Streptococcus pyogenes. The Journal of infectious diseases, 2007. 196(12): p. 1794-

803.

251. Varol, C., et al., Intestinal lamina propria dendritic cell subsets have different origin

and functions. Immunity, 2009. 31(3): p. 502-12.

252. Autenrieth, S.E., et al., Depletion of dendritic cells enhances innate anti-bacterial

host defense through modulation of phagocyte homeostasis. PLoS pathogens,

2012. 8(2): p. e1002552.

253. Autenrieth, I.B., et al., Defense mechanisms in Peyer's patches and mesenteric

lymph nodes against Yersinia enterocolitica involve integrins and cytokines.

Infection and immunity, 1996. 64(4): p. 1357-68.

254. Frazer, L.C., et al., Enhanced neutrophil longevity and recruitment contribute to the

severity of oviduct pathology during Chlamydia muridarum infection. Infection and

immunity, 2011. 79(10): p. 4029-41.