regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 armstrong vs. clone 13:...

172
Regulation of anti-viral immunity by dendritic cells and natural killer cells by Achire Nathalia Mbanwi A thesis submitted in conformity with the requirements for the degree of Doctor of Philosophy Department of Immunology University of Toronto © Copyright by Achire Nathalia Mbanwi 2015

Upload: others

Post on 07-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

Regulation of anti-viral immunity

by dendritic cells and natural killer cells

by

Achire Nathalia Mbanwi

A thesis submitted in conformity with the requirements

for the degree of Doctor of Philosophy

Department of Immunology

University of Toronto

© Copyright by Achire Nathalia Mbanwi 2015

Page 2: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

ii

Regulation of anti-viral immunity by dendritic cells and natural killer cells

Achire Nathalia Mbanwi

Doctor of Philosophy

Department of Immunology

University of Toronto

2015

Abstract

4-1BB and its ligand, 4-1BBL, are costimulatory members of the tumor necrosis factor receptor

(TNFR)/TNF family. The 4-1BB/4-1BBL pathway has been shown to potently enhance T cell

responses in models of viral infection and cancer, thereby identifying this pathway as a target for

immunotherapy. In addition to T cells, 4-1BB is expressed on dendritic cells (DCs), which also

express 4-1BBL. The precise nature of 4-1BB/4-1BBL expression on DCs is not known and the

endogenous function of 4-1BB/4-1BBL on this cell type has remained largely elusive. Here, I

show that LPS-activated bone marrow-derived DCs express both 4-1BB and 4-1BBL, and that

these molecules constitutively interact on DCs. Survival, upregulation of key costimulatory

molecules, cytokine/chemokine production, antigen presentation, and the ability of DCs to

generate primary influenza-specific CD8 T cell responses were normal in the absence of 4-1BB

and/or 4-1BBL on DCs. Thus, the biological implications of 4-1BB/4-1BBL interactions on DCs

remain to be elucidated.

It was previously demonstrated in the Watts lab that the 4-1BB pathway on T cells becomes

desensitized during persistent infection with lymphocytic choriomeningitis virus (LCMV) clone

13 through loss of the signaling adapter TRAF1. In the course of these studies, it was found that

Page 3: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

iii

2 months following LCMV clone 13 infection, a time point at which virus is largely cleared from

the spleen and other organs, C57BL/6 mice showed dramatic splenic atrophy. Here, I show that

this atrophy persists for up to 10 months following initial infection and that it does not occur

with the acutely infecting strain, LCMV Armstrong. I go on to show that splenic atrophy is

mediated in part by natural killer (NK) cells, and implicate NKG2D-MULT1 interactions as

contributing to late-stage B cell depletion. Compared to isotype control antibody treatment, type

I interferon receptor 1 (IFNAR1) blocking antibody treatment at the onset of splenic atrophy

increased the proportion of NK cells in the spleen, and was correlated with exacerbated splenic

atrophy. Lastly, splenic atrophy following LCMV clone 13 infection was associated with delayed

clearance of a bacterial pathogen. These findings implicate NK cells in the pathophysiological

mechanisms underlying splenic atrophy following viral infection.

Page 4: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

iv

Acknowledgments

First and foremost, I would like to thank my supervisor, Dr. Tania Watts, for the opportunity to

take on graduate studies in her lab. Over the years, she has been a source of support and

encouragement, and her enthusiasm for science kept me motivated and excited about the work. I

thank her for providing her decades of experience in asking the right questions, designing

experiments, and scientific writing, but also for granting me some freedom in pursuing my own

ideas. I am certainly grateful that I was able to attend 4 conferences throughout my graduate

studies, especially the 100th

annual American Association of Immunologists conference in

Honolulu, Hawaii.

I thank my committee members, Dr. Dana Philpott and Dr. Juan Carlos Zúñiga-Pflücker, for

their thought-provoking questions and ideas during committee meetings, and for being kind and

approachable.

I would also like to recognize the past and present members of the Watts lab for their technical

assistance and expertise, but also for providing encouragement and emotional support when

things went wrong.

Last, but not least, I would like to express my deepest gratitude to my family and friends for their

incredible love and support throughout my graduate studies. When experiments worked, they

were there for the good times. When experiments failed, they were there for the bad times. I

thank them most importantly for always believing in me and pushing me out of my comfort

zone, and for providing that support network that no one can do without. Special thanks to my

parents, Eta and Bershu, my aunt Bertha, and my two little brothers, Sikem and Matthew.

Page 5: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

v

Table of Contents

Abstract ii

Acknowledgments iv

Table of Contents v

List of Tables xi

List of Figures xii

List of Publications xiv

Abbreviations xv

Chapter 1: Introduction 1

1.1 Overview 2

1.2 The innate immune response in anti-viral immunity 2

1.2.1 Dendritic cells 3

DC development and homeostasis 4

DC subsets 4

Functions of DCs 5

1.2.2 DCs in anti-viral immunity 6

1.2.3 Therapeutic implications of DCs: DC vaccination 7

1.2.4 Natural killer cells 8

NK cell development and homeostasis 8

NK cell subsets 9

Page 6: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

vi

NK cell receptors and ligands 10

Functions of NK cells 14

1.2.5 NK cells in anti-viral immunity 15

1.3 The spleen as a secondary lymphoid organ involved in anti-viral immunity 16

1.3.1 Development of the spleen 16

1.3.2 Structure and function of the spleen 18

1.4 Splenic atrophy due to viral infection and other causes 20

1.4.1 Splenic atrophy in sickle cell anemia and autoimmunity 21

1.4.2 Splenic atrophy due to viral infection: what are the mechanisms? 21

1.4.3 Compromised immunity due to complications associated with the spleen 23

1.5 Murine models of viral infection: LCMV 23

1.5.1 LCMV virus 24

1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24

1.5.3 The innate immune response to LCMV infection 27

1.5.4 The role of NK cells during LCMV infection 28

1.5.5 Type I Interferons during LCMV infection 32

1.5.6 Splenic architecture and remodeling following LCMV infection 34

1.5.7 Resolution of inflammation following LCMV infection 36

1.6 Murine models of viral infection: influenza 37

1.6.1 Influenza virus 37

1.6.2 Innate immune responses 38

Page 7: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

vii

1.6.3 Adaptive immune responses 39

1.7 TNF/TNFR family 39

1.7.1 Overview 40

1.7.2 4-1BB/4-1BBL 40

1.7.3 Role of 4-1BB/4-1BBL on T cells 41

1.7.4 Role of 4-1BB/4-1BBL on non-T cells 43

1.7.5 Role of 4-1BB/4-1BBL on DCs 43

1.7.6 4-1BB signaling 44

1.8 Thesis Rationale 47

Chapter 2: 4-1BB and 4-1BBL constitutively interact on LPS-activated dendritic cells

but dendritic cell-intrinsic 4-1BB/4-1BBL interactions are dispensable for

cytokine production and anti-viral CD8 T cell priming 48

2.1 Abstract 49

2.2 Introduction 49

2.3 Materials and Methods 50

2.3.1 Mice 50

2.3.2 DC cultures and surface markers 51

2.3.3 Blocking studies 51

2.3.4 DC survival assay 52

2.3.5 Cytokine production 52

2.3.6 In vitro antigen uptake and presentation 52

2.3.7 DC vaccination 53

Page 8: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

viii

2.3.8 Flow cytometry 54

2.3.9 Lentiviral transductions and confocal microscopy 54

2.3.10 Statistical Analysis 54

2.4 Results 54

2.4.1 Chacterization of DCs lacking 4-1BB and 4-1BBL 54

2.4.2 4-1BBL is not readily detectable in the presence of 4-1BB on LPS-activated

DCs 56

2.4.3 The surface expression of 4-1BBL can be regulated extrinsically 56

2.4.4 Evidence of constitutive interaction between 4-1BB and 4-1BBL on DCs 59

2.4.5 Indistinguishable in vitro survival of WT and DKO DCs 63

2.4.6 4-1BB and/or 4-1BBL do not impact the level of the cytokines IL-2, IL-6,

IL-12, IL-18, and TNF, nor the chemokines MCP-1 and RANTES in LPS-

activated DCs 63

2.4.7 4-1BB and 4-1BBL are dispensable for antigen presentation by DCs

in vitro 63

2.4.8 DC-intrinsic 4-1BB and 4-1BBL are largely dispensable for T cell priming

against a viral antigen in vivo 66

2.5 Discussion 72

Chapter 3: Natural killer cells contribute to splenic atrophy observed months following

LCMV clone 13 infection 76

3.1 Abstract 77

3.2 Introduction 77

3.3 Materials and Methods 79

Page 9: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

ix

3.3.1 Mice and infections 79

3.3.2 In vivo antibody treatments 79

3.3.3 Flow cytometry and antibodies 80

3.3.4 Immunofluorescence microscopy 80

3.3.5 Statistical analysis 81

3.4 Results 81

3.4.1 Persistent splenic atrophy following infection with LCMV clone 13 81

3.4.2 Atrophic spleens are lymphopenic, but show T cell/B cell segregation within the

white pulp 84

3.4.3 NK cells contribute to splenic atrophy following LCMV clone 13

infection 86

3.4.4 IFNAR1 blocking antibody treatment increases the proportion of NK cells, and

exacerbates splenic atrophy 89

3.4.5 Splenic atrophy delays clearance of a bacterial pathogen 92

3.4.6 The proportion of LTi is similar between aged naive and LCMV-

infected mice 94

3.5 Discussion 96

Chapter 4: Discussion and Future Directions 101

4.1 Overview 102

4.2 Role of 4-1BB/4-1BBL on DCs: What unique transcripts are induced by 4-1BB/

4-1BBL on DCs and does this impact secondary immune responses? 102

4.3 4-1BB signaling in DCs 104

4.4 Potential bidirectional signaling by 4-1BB and 4-1BBL on DCs 105

Page 10: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

x

4.5 Splenic atrophy following LCMV clone 13 infection 106

4.6 Can the spleen regenerate following splenic atrophy? 109

4.7 Conclusion 111

Chapter 5: References 113

Page 11: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xi

List of Tables

Chapter 1

Table 1.1 Major mouse and human NK cell receptors 11

Page 12: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xii

List of Figures

Chapter 1

Figure 1.1: Structure of the spleen 19

Figure 1.2: Signaling downstream of 4-1BB on T cells 46

Chapter 2

Figure 2.1: LPS-induced upregulation of MHC-I, CD80, CD86, and CD40 is normal

in the absence of 4-1BB and 4-1BBL 55

Figure 2.2: 4-1BBL is not readily detectable in the presence of 4-1BB on LPS-activated

DCs 57

Figure 2.3: The surface expression of 4-1BBL can be regulated extrinsically 58

Figure 2.4: Citrine expression is detectable on the surface of 4-1BBL-citrine-transduced

WT and 4-1BB-/-

DCs 60

Figure 2.5: Evidence of constitutive interaction between 4-1BB and 4-1BBL on DCs 61

Figure 2.6: 4-1BB and 4-1BBL are dispensable for DC survival in vitro 64

Figure 2.7: 4-1BB and/or 4-1BBL are not required for IL-2, IL-6, IL-12, IL-18, MCP-1,

RANTES, and TNF production by DCs 65

Figure 2.8: 4-1BB and 4-1BBL are dispensable for DC antigen presentation in vitro 67

Figure 2.9: DC-intrinsic 4-1BB and 4-1BBL are largely dispensable for T cell priming

against a viral antigen in vivo 69

Chapter 3

Figure 3.1: Persistent splenic atrophy following infection with LCMV clone 13 82

Figure 3.2: Atrophic spleens are lymphopenic, but show T cell/B cell segregation within the

white pulp 85

Page 13: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xiii

Figure 3.3: NK cells contribute to splenic atrophy 87

Figure 3.4: IFNAR1 blocking antibody treatment increases the proportion of NK cells, and

exacerbates splenic atrophy 90

Figure 3.5: Splenic atrophy delays clearance of a bacterial pathogen 93

Figure 3.6: The proportion of LTi is similar between aged naive and LCMV-infected

mice 95

Chapter 4

Chapter 4.1: 4-1BB/4-1BBL constitutive interaction on bone marrow derived DCs

in vitro 103

Chapter 4.2: NK cells contribute to splenic atrophy following LCMV clone 13

infection 107

Page 14: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xiv

List of Publications

1. Mbanwi AN, Lin GH, Sabbagh L, Watts TH. 4-1BB and 4-1BBL constitutively interact

on LPS-activated dendritic cells but dendritic cell-intrinsic 4-1BB/4-1BBL interactions

are dispensable for cytokine production and anti-viral CD8 T cell priming.

Manuscript in preparation.

2. Mbanwi AN, Wang C, Geddes K, Philpott DJ, Watts TH. Natural killer cells contribute

to splenic atrophy observed months following LCMV clone 13 infection.

Manuscript in preparation.

3. Mbanwi AN, Watts TH. 2014. Costimulatory TNFR family members in control of viral

infection: outstanding questions. Semin Immunol 26: 210-9

4. Lin GH, Edele F, Mbanwi AN, Wortzman ME, Snell LM, Vidric M, Roth K, Hauser AE,

Watts TH. 2012. Contribution of 4-1BBL on radioresistant cells in providing survival

signals through 4-1BB expressed on CD8(+) memory T cells in the bone marrow. Eur J

Immunol 42: 2861-74

Page 15: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xv

Abbreviations

(i)NKT – (invariant) natural killer T cell

(i)Treg – (inducible) regulatory T cell

4-1BBL – ligand of 4-1BB

7-AAD – 7-aminoactinomycin D

Ad - adenovirus

ADCC – antibody-dependent cellular cytotoxicity

AICD – activation-induced cell death

AIDS – acquired immunodeficiency syndrome

AP – alkaline phosphatase

APC – allophycocyanin

APC – antigen presenting cell

ARM – LCMV Armstrong

BAFF – B cell-activating factor of the tumor necrosis factor family

Bcl – B cell lymphoma

BIR – baculovirus inhibitor of apoptosis protein repeat

BM – bone marrow

BrdU – 5’-bromo-2’-deoxyuridine

CCL – CC chemokine ligand

CCR – CC chemokine receptor

CD – cluster of differentiation

Page 16: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xvi

cDC – conventional dendritic cell

cDNA – complementary DNA

CDP – committed dendritic cell progenitor

CDR – complementarity determining region

CFSE – carboxyfluoroscein succinimidyl ester

CFU – colony forming unit

cIAP – cellular inhibitor of apoptosis

CKI – casein kinase I

Cl13 – LCMV clone 13

CMV – cytomegalovirus

CT – computed tomography

CTL – cytotoxic T lymphocyte

CXCL – chemokine (C-X-C) motif ligand

CXCR – chemokine (C-X-C) motif receptor

DAPI – 4’,6-diamino-2-phenylindole

DC – dendritic cell

DC-SIGN – dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin

DD – death domain

DKO – double knockout

DLN – draining lymph node

DNA – deoxyribonucleic acid

Page 17: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xvii

dpi – days post-infection

dsRNA – double-stranded ribonucleic acid

ELISA – enzyme-linked immunosorbent assay

ERK – extracellular signal-regulated kinase

ES – embryonic stem

Fc – fragment crystallizable

FDC – follicular dendritic cell

FITC – fluorescein isothiocyanate

FLT3(L) – FMS-like tyrosine kinase 3(ligand)

FMO – fluorescence minus one

FOXO – forkhead transcription factor, class O

FRC – fibroblastic reticular cell

FSC – forward scatter

FV – Friend virus

GALT – gut-associated lymphoid tissue

GFP – green fluorescent protein

GITR – glucocorticoid induced tumor necrosis factor receptor related protein

GM-CSF – granulocyte-macrophage colony-stimulating factor

GP – glycoprotein

GPPS – glutamine, sodium pyruvate, penicillin streptomycin

HA – hemagglutinin

Page 18: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xviii

HBV – hepatitis B virus

HCV – hepatitis C virus

HEK – human embryonic kidney

HIV – human immunodeficiency virus

HLA – human leukocyte antigen

Hox11 – homeobox 11

HSC – hematopoietic stem cell

HSV1 – herpes simplex virus 1

i.p. – intraperitoneal

i.v. – intravenous

ICAM – intercellular adhesion molecule

IFITM – interferon-induced transmembrane protein

IFN – interferon

IFNAR1 – type I interferon receptor 1

Ig – immunoglobulin

IκBα – inhibitor of NF-κB

IKK – IκB kinase

IL – interleukin

IRF – interferon-regulatory factor

ISG – interferon-stimulated gene

ITAM – immunoreceptor tyrosine-based activating motif

Page 19: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xix

ITIM – immunoreceptor tyrosine-based inhibitory motif

JNK – c-Jun N-terminal kinase

KIR - killer cell immunoglobulin-like receptor

KLRG1 – killer cell lectin-like receptor G1

LCMV – lymphocytic choriomeningitis virus

LN – lymph node

LPS – lipopolysaccharide

LSECtin – lymph node sinusoidal endothelial calcium-dependent lectin

LSP-1 – lymphocyte-specific protein 1

LT – lymphotoxin

LTi – lymphoid tissue inducer cells

LTo – lymphoid tissue organizer cell

LTβR – lymphotoxin-beta-receptor

MAPK – mitogen activated protein kinase

MARCH – membrane-associated RING-CH

MCMV – murine cytomegalovirus

MCP-1 – monocyte chemoattractant protein-1

MDP – monocyte and dendritic cell progenitor

MDSC – myeloid-derived suppressor cell

MFI – mean fluorescence intensity

MHC – major histocompatibility complex

Page 20: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xx

MHV68 – murine gammaherpes virus 68

MKK – MAP kinase kinase

MKKK – MAP kinase kinase kinase

MLN – mediastinal lymph node

MOI – multiplicity of infection

MULT1 – murine UL16-binding protein-like transcript 1

NA – neuraminidase

NCR1 – natural cytotoxicity receptor 1

NEAA – non-essential amino acids

NFAT – nuclear factor of activated T cells

NF-κB – nuclear factor κB

NIK – NF-κB-inducing kinase

NK – natural killer

NKG2D – natural-killer group 2D

NLR - nucleotide oligomerization domain-like receptor

NP – nucleoprotein

OPT - optical projection tomography

OVA – ovalbumin

PAMP – pathogen-associated molecular pattern

PBS – phosphate buffered saline

PD – programmed death

Page 21: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xxi

pDC – plasmacytoid dendritic cell

PE – phycoerythrin

PFU – plaque forming unit

pi – post-infection

PI3K – phosphatidylinositol-3 kinase

PLN – peripheral lymph node

PP –Peyer’s patch

PRR – pattern recognition receptor

RAE1 – retinoic acid early inducible gene 1

RAG – recombination-activating gene

RALDH – retinoic acid-producing retinaldehyde dehydrogenase

RANK – receptor activator of NF-κB

RANTES – regulated on activation, normal T cell expressed and secreted

RBC – red blood cell

RCMV – rat CMV

RCTL – RMCV C-type lectin-like

RDC – respiratory dendritic cell

RING – really interesting new gene

RIP1 – receptor interacting protein 1

RLR - retinoic acid-inducible gene I-like receptor

RORγt – retinoic acid receptor-related orphan receptor gamma t

Page 22: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xxii

SA – streptavidin

SAP – signaling lymphocytic activation molecule-associated protein

SARS – severe acute respiratory syndrome

SH2 – Src homology 2

SHIP – SH2-containing inositol polyphosphate 5-phosphatase

SHP – Src homology 2-containing tyrosine phosphatase

SIINFEKL – OVA257-264

SIV – simian immunodeficiency virus

SLE – systemic lupus erythematosus

SLO – secondary lymphoid organ

SMP – splanchnic mesodermal plate

SSC – side scatter

ssRNA – single-stranded ribonucleic acid

STAT – signal transducer and activator of transcription

TAM – tyro3 axl mer

TBEV - Tick-borne encephalitis virus

TBS(T) – Tris-buffered saline (Tween-20)

TCF-1 – T cell factor 1

TCR – T cell receptor

Tfh – T follicular helper cell

TGFβ – transforming growth factor β

Page 23: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

xxiii

Th – T helper

TIRAP - Toll/IL-1R domain-containing adapter protein

TLR – toll-like receptor

TNF – tumor necrosis factor

TNFR – tumor necrosis factor receptor

TRAF – tumor necrosis factor receptor associated factor

TRAIL – tumor necrosis factor-related apoptosis-inducing ligand

TRAM - toll/IL-1R domain-containing adapter-inducing interferon-β -related adapter molecule

TRIF - toll/IL-1R domain-containing adapter-inducing interferon-β

UV – ultraviolet

VEGF – vascular endothelial growth factor

VCAM – vascular adhesion molecule

VSV – vesicular stomatitis virus

VV – vaccinia virus

WT – wild-type

α-DG – alpha-dystroglycan

Page 24: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

1

Chapter 1

Introduction

Page 25: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

2

1.1 Overview

The innate immune system represents the first line of defense against the microbial world.

Among innate immune cells, both DCs and NK cells have emerged as key players in

orchestrating the immune response against viral pathogens. In this thesis introduction, I will first

discuss the general biology of these cells, and then highlight some of their roles in anti-viral

immunity. The spleen is the largest secondary lymphoid organ in the body, and the work

presented in this thesis involves anti-viral immune responses generated in this organ. I will

therefore discuss the development and structure of the spleen, as well as its role in immunity. In

chapter 3 of this thesis, I show that persistent infection of mice with LCMV clone 13 results in

sustained splenic atrophy that is mediated in part by NK cells. Splenic atrophy is commonly

associated with autoimmune disease, but has been shown to occur following viral infection in

some models. I will discuss these models and postulate on the mechanisms controlling spleen

size. I will then introduce the models of viral infection used in this thesis, namely LCMV and

influenza infection of mice. Members of the TNFR family, in particular 4-1BB and its ligand 4-

1BBL, have been shown to modulate various aspects of T cell biology following influenza

infection. However, 4-1BB is more widely expressed on cells of the immune system, and in

chapter 2, I investigate 4-1BB and 4-1BBL expression and function on DCs. Therefore, I will

end the thesis introduction with a discussion of the TNFR family, with emphasis on 4-1BB/4-

1BBL, and how these molecules modulate T cell and DC biology, particularly in the context of

viral infection.

1.2 The innate immune response in anti-viral immunity

Viral insult can result in acute infection, e.g. influenza in humans and LCMV Armstrong in

mice, or chronic infection, e.g. human immunodeficiency virus (HIV) in humans, and LCMV

Clone 13 in mice. Moreover, the etiology of one in five (1) of all cancers, as well as several

autoimmune diseases, is also thought to involve viral infection. Both the virulence of the virus

and the magnitude and nature of the immune response against it ultimately determine the

decision between viral clearance and establishment of chronicity. During acute viral infection,

the arms race between the virus and the host results in either elimination of the virus and

generation of immune memory, or death of the host. If the virus cannot be eliminated and does

not kill the host, then both the virus and the immune response must strike a balance between

Page 26: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

3

factors that enhance immunity, in order to keep viral replication in check, and factors that slow

down immunity, in order to avoid destruction of self tissues (1, 2). Viral genes that mediate

virulence and cell tropism, as well as multiple immune and non-immune cell types expressing

membrane-bound and soluble molecules act in concert to mediate these outcomes.

Upon viral infection, the innate immune system rapidly induces barriers that control viral

dissemination. Whereas the adaptive immune system generates antigen-specific receptors using

gene rearrangement, the innate immune system uses an array of germ-line encoded receptors

with limited diversity to recognize common features of pathogens (3). Viral components such as

genomic DNA, single-stranded RNA (ssRNA), double-stranded RNA (dsRNA), RNA with 5’-

triphosphate ends and viral proteins are recognized both inside and outside the cell by 3 main

categories of pattern recognition receptors (PRRs): Toll-like receptors (TLRs), retinoic acid-

inducible gene I (RIG-I)-like receptors (RLRs), and nucleotide oligomerization domain (NOD)-

like receptors (NLRs) (4). TLRs are expressed on the cell surface or in endosomal compartments

and signal via adapter proteins such as MyD88, Toll/IL-1R (TIR) domain-containing adapter-

inducing interferon-β (TRIF), TIR domain-containing adapter protein (TIRAP), and TRIF-

related adapter molecule (TRAM), to activate NF-κB and MAPK pathways, and induce the

production of IFNs and other cytokines (5, 6). RLRs are a family of cytoplasmic PRRs that also

drive IFN production and proinflammatory cytokines via signaling pathways involving NF-κB,

IRF3 and IRF7 (7-9). NLRs are a large family of cytosolically expressed PRRs among which are

members that induce the inflammasomes, and regulate maturation of IL-1β and IL-18 via

activation of caspase-1 (7, 10, 11). These receptors are expressed by both immune and non-

immune cells and overall, their signaling leads to secretion of type I IFNs, and other

proinflammatory cytokines and chemokines, as well as increased expression of costimulatory

molecules such as CD80, CD86, and CD40, which are required along with antigen-specific

signals to initiate adaptive immune responses mediated by T and B cells (4).

The cells that make up the innate immune system are highly diverse; they include granulocytes,

such as neutrophils, basophils and eosinophils, lymphocytes such as NK and NKT cells, and

antigen-presenting cells (APCs), such as macrophages and DCs. This thesis focuses on the role

of DCs and NK cells in anti-viral immunity, and these cell types will be discussed further.

1.2.1 Dendritic cells

Page 27: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

4

DC development and homeostasis

DCs are specialized APCs that are essential mediators of immunity and tolerance. For his

discovery of DCs, Ralph Steinman received the 2011 Nobel Prize in Medicine and Physiology.

Monocytes can develop some of the features of DCs under conditions of inflammation in vivo or

when cultured with cytokines in vitro, however, they are not precursors for lymphoid organ DCs

in the steady state (12-16). In the bone marrow (BM), monocyte and DC progenitors (MDPs)

diverge to become monocytes and committed DC progenitors (CDPs), which then differentiate

into pre-DCs. Committed pre-DCs leave the BM and migrate to lymphoid and non-lymphoid

tissues via the blood, where they differentiate into the 2 main populations of DCs in the

lymphoid tissues as well as the non-lymphoid tissue CD103+ DCs. This developmental pathway

is highly dependent on Flt3-Flt3L interactions (16). DC homeostasis is maintained by a balance

of 3 factors: continuous input of pre-DCs from the blood, a limited amount of DC division in

situ, and cell death (16). 5% of DCs in lymphoid tissues are dividing at any time (17-19), and in

the steady state this division is regulated by Flt3 (20, 21) and by LTβR signaling, especially on

CD8- spleen DCs (22). DC half-life varies from 5-7 days in the spleen, lymph nodes (LNs), liver

and kidney, and can be as long as 25 days in the lung (17, 19).

DC subsets

DCs are largely divided into conventional DCs (cDCs) and plasmacytoid DCs (pDCs), and both

types can be found in lymphoid tissues, such as spleen and LN, and in non-lymphoid tissues,

such as skin, liver, and lung (16). The cDC subsets in the spleen include the CD8+CD205

+

subset, which is localized primarily in the T cell area and the marginal zone (23), and the CD8-

33D1+ subset, which is found in the red pulp and bridging channels (16). The former is the

primary cell type which captures and cross-presents antigens from apoptotic cells (24-27), while

the latter is more efficient in processing antigens for presentation by MHC-II (28, 29). LNs

contain similar subsets of DCs (CD8+CD205

+ and CD8

-CD11b

+) (24, 30, 31), as well as

migratory DCs that enter the LNs from tissues in a CCR7-dependent manner (32). Antigens can

also enter the LNs directly through the lymph. It is thought that LN-resident DCs that acquire

antigen directly from the lymph are the first to present peptides to naive CD4 T cells, which

results in T cell priming and IL-2 production. Migratory DCs which pick up antigen in the

Page 28: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

5

periphery and transport it to LNs then further activate these CD4 T cells to induce differentiation

into effector cells (33).

The differential expression of CD303, CD1C and CD141 divides human DCs into different

subsets (34). Human CD141+ DCs share with mouse CD8

+ DCs the high capacity to mediate

cross-presentation (35, 36) and the CD1C+ subset most closely resembles the mouse CD8

- DC

subset (37, 38). Like mouse pDCs (CD11clo

B220+Ly6C

+PDCA-1

+), human CD303

+ pDCs

secrete large amounts of type I IFNs in response to viral infection (39-42). Studies on human

DCs are hampered by the low frequency of circulating DCs, and therefore most studies have

been conducted using peripheral blood monocytes treated with cytokines to differentiate into

DCs. These in vitro conditions might induce selective DC subsets that exhibit markers and

functions distinct from their physiological counterparts (43).

Functions of DCs

CD8+ DCs are potent producers of IL-12, polarize Th1 responses via production of IL-12, IL-18,

TNF and type I IFNs, and have been shown to be the most important DC subset involved in

cross-presentation. CD8-

DCs produce less IL-12 and polarize Th2 responses when there are

comparatively less of the aforementioned cytokines and in the presence of IL-10. DC-secreted

IL-6 is involved in the differentiation of Th17 cells, and the differentiation of other CD4 T cell

lineages, such as T follicular helper cells (Tfh) and T regulatory cells (Tregs), are also influenced

by DC cytokines (44-46). Both CD8+ and CD8

- DCs are capable of priming efficient CD8 T cell

responses (47-49).

DCs are present in nearly every tissue of the body, especially near portals of entry, where they

survey the environment and act as sentinels of invading pathogens. In addition to initiating

immune responses, DCs are also responsible for inducing central and peripheral tolerance to self-

antigens. In the steady state, most DCs exist in an “immature” or naive state, which is

characterized by low expression of costimulatory molecules, low surface MHC-II, and a high

capacity for phagocytosis and endocytosis. When T cells recognize self-antigen on DCs in this

state, they are either anergized, deleted, or differentiate into Tregs via secretion of cytokines such

as TGFβ (45, 50). The importance of DCs in this process is highlighted by the finding that mice

lacking both cDCs and pDCs develop systemic autoimmunity (51).

Page 29: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

6

Following recognition of molecular patterns associated with infection and/or injury/stress, DCs

are activated and undergo a process termed maturation, in which they alter their expression of

chemokine receptors and migrate to lymphoid tissues, upregulate costimulatory molecules and

MHC-II, secrete immunomodulatory cytokines and chemokines, and downregulate their ability

to mediate phagocytosis/endocytosis. CD4 and CD8 T cells that recognize specific antigen on

activated DCs are themselves activated, differentiate, and acquire effector functions (45, 50).

Other cell types such as CD4 T cells can further activate DCs and potentiate immune responses

by upregulating CD40L, which in turn signals through CD40 on DCs. This CD40-mediated

reciprocal activation (DC licensing) has been shown to be important for the ability of DCs to

prime CD8 T cells (52-55). In a recent study (56), it was shown that LTβR signaling in DCs

mediated by lymphotoxin (LT) expressed on activated CD4 T cells also contributes to DC

licensing. LTβR signaling in the DCs induced type I IFN expression, which enhanced the ability

of DCs to stimulate clonal expansion of CD8 T cells. CD40 signaling, on the other hand, was

shown to be more important for the ability of DCs to activate IFNγ production by CD8 T cells

(56, 57). DCs also contribute to humoral immunity via directly presenting unprocessed antigens

to B cells, and by activating helper CD4 T cells (58-61).

Overall, the nature of the signals provided by different microbes, the resulting

cytokine/chemokine milieu, the subset of responding DCs, and the interactions of DCs with other

immune cells ultimately dictates the nature of the immune response.

1.2.2 DCs in anti-viral immunity

As discussed above, DCs enhance immunity to pathogens, including viral pathogens. Due to

their presence at every possible entry site to the body, cDCs are amongst the first cells

encountered by most viruses (62). Among the most studied receptors for viral recognition on

human DCs is DC-SIGN (63, 64). This receptor binds to N-linked glycan substituents on viral

spikes, thereby enabling viral entry (65). It is not clear whether this receptor is actually a pattern

recognition receptor, or whether viruses simply utilize this receptor for entry (62). DEC-205 is

another interesting receptor mediating viral entry because of the enhanced ability of antigens

targeted to this receptor to induce priming of naive T cells (66, 67). Although DEC-205 has been

shown to bind to CpG oligonucleotides (68), and to recognize ligands expressed on dying cells

(69), the precise nature of its natural ligands are not known (69, 70). Other receptors include

Page 30: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

7

CD4 and CXCR4 used by HIV, langerin, Fcγ receptors, mannose receptors, and heparan sulfate

(71-73). The fate of viruses after entry can depend on the receptors to which they bind.

In addition to producing large amounts of type I IFNs in response to viral recognition by

endosomal TLRs, pDCs are also capable of cross-presenting viral antigens and rapidly priming

CTLs (74). Most studies in vivo indicate a negative correlation between pDC activity and

severity of viral disease (62, 75). Due to their central role in viral defense, several viruses

downregulate the activity and number of DCs. Viral replication in DCs can directly inhibit their

ability to mature and upregulate key surface markers involved in APC function, and can also

affect the ability of hematopoietic progenitors to develop into DCs (76). Viruses can also affect

the ability of DCs to polarize a Th1 response by downregulating production of cytokines such as

IL-12, thereby favouring a Th2 response (77-79). Th2 responses are known to have detrimental

effects during viral infection by promoting inflammation, allergy, and fibrosis (62). Since DCs

migrate from portals of entry to the body to LNs, some viruses also exploit this mechanism to

disseminate within the host. Lentiviruses especially get concentrated into and/or onto DCs at

mucosal sites, and are not fully digested and presented, but instead shuttled to LNs, where they

infect target cells (80-82).

1.2.3 Therapeutic applications of DCs: DC Vaccination

Owing to their ability to act as natural adjuvants and initiate immune responses, DCs have been

utilized in vaccination strategies aimed at eliciting immune responses against tumors and viruses.

In the clinical setting, peripheral blood monocytes from the patient are differentiated into DCs ex

vivo using a cocktail of cytokines, including GM-CSF and IL-4, loaded with tumor-specific or

virus-specific antigens, matured with DC activation-inducing cytokines or pathogen-associated

molecular patterns (PAMPs), and then injected back into the patient with the hopes of inducing

tumor-specific effector T cells capable of reducing tumor mass, or virus-specific T cells capable

of either protecting a naive individual from serious infection or therapeutically reducing viral

load (62, 83). Clinical studies from the past two decades have explored different DC vaccine

preparations, DC activators, antigen preparations, and routes of DC injection. Thus far, only

Sipuleucel-T, which consists of the patient’s enriched blood APCs cultured with a fusion protein

of prostatic acid phosphatase (an antigen expressed in prostate cancer tissue) and GM-CSF, has

been approved by the US Food and Drug Administration for treatment of metastatic prostate

Page 31: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

8

cancer (84). When injected into patients, this drug was shown to extend median survival by

around 4 months (84). There are two ongoing phase III clinical trials evaluating the efficacy of

monocyte-derived ex vivo generated DC vaccines. These trials are testing DC vaccination in

patients with newly diagnosed brain tumors (NCT00045968; Northwest Therapeutics) and in

patients with advanced kidney cancer (NCT01582672; ADAPT trial, Argos Therapeutics) (84).

DC vaccination for the treatment of human viral disease is still being evaluated in animal models

and clinical trials. Studies in animal models of HIV using DCs loaded with HIV-1 viral lysate,

envelope glycoproteins, or inactivated virus have shown that these DCs can mount a potent

immune response against HIV-1 (85-88). In a recent clinical trial, it was shown that autologous

monocyte-derived-DCs pulsed with autologous heat-inactivated whole HIV increased HIV-

specific T cell responses and significantly reduced viral load following combination

antiretroviral therapy interruption (89). What has proven difficult in the field of DC vaccination

is developing efficient protocols that generate large amounts of DCs with the appropriate

maturation status (62).

1.2.4 Natural killer cells

NK cell development and homeostasis

NK cells belong to the family of innate lymphoid cells (ILCs) that are characterized by their

lymphoid morphology, the absence of recombination-activating gene (RAG)-dependent antigen

receptor rearrangement and the absence of myeloid/DC phenotypical markers. Due to their

cytotoxicity and production of IFNγ, NK cells are classified as group 1 ILCs, in contrast to group

2 ILCs, which can produce type 2 cytokines, i.e. IL-5 and IL-13, and group 3 ILCs, which can

produce IL-17 and/or IL-22 (90). NK cells develop and mature in the BM and have been shown

to play a role in the early control of viral infection, tumor immunosurveillance, solid organ and

stem cell transplantation, reproduction, autoimmunity, and asthma (91). The earliest NK cell

progenitors are defined by their potential to respond to IL-15 via upregulation of CD122 and

CD132, and the absence of lineage-specific surface antigens (92, 93). In vitro studies of NK cell

development have also implicated IL-7, stem cell factor, and Flt3L (93), and studies with the

OP9 stromal cell culture system have shown that NK cells can develop in the presence of

specific Notch ligands, such as members of the Delta and jagged families (94-96). The earliest

Page 32: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

9

NK cell defining markers are NK1.1 and NKG2D, followed by the Ly49 receptors (see 1.2.4

“NK cell receptors and ligands”) (97). NK cell receptors must undergo an “education” process in

order to acquire functionality. For example, failure to engage inhibitory receptors during

development can result in peripheral NK cells that are hyporesponsive (98-102); however, the

responsiveness of these cells can be continuously modulated when their activating or inhibitory

receptors are ligated (103). Developing immature NK cells do not possess the ability to produce

cytokines and kill other cells, and these functions are acquired late in development following

maturation. Many cytokines, including IL-2, IL-4, IL-7, IL-10, IL-12, IL-15, IL-18, IL-21, type I

IFN, IFNγ and TGFβ have been shown to influence NK cell maturation (104-107). Although NK

cell development has been best characterized in the BM, the presence of phenotypically

immature NK cells in other tissues such as the thymus suggests that these cells can also mature

outside the BM (108). IL-15 especially has an important role during NK cell development and

continues to impact the homeostasis and survival of peripheral NK cells (109). DCs and

macrophages mediate transpresentation of IL-15 to NK cells in vivo (110-112), and IL-15

functions by increasing the expression of pro-survival Bcl-2 family members (113-115), and at

the same time suppressing the transcription factor FOXO3A and the pro-apoptotic molecule BIM

(114). NK cells are widespread throughout most lymphoid and non-lymphoid tissues, with a

prevalence of around 3% of total lymphocytes in the mouse spleen, and from 2-18% of total

lymphocytes in human peripheral blood (116). BrdU and deuterium-enriched glucose labeling

studies have demonstrated that the half-life of NK cells in the spleen is around 7-17 days and

around 12 days in human blood (92, 117-119). Most splenic NK cells in adult mice do not divide

rapidly under normal conditions (117); however, NK cells do proliferate rapidly in lymphopenic

environments, such as following irradiation or transfer into both RAG-1 and γc-deficient mice

(which lack B, T and NK cells) (117, 120, 121).

NK cell subsets

At least four subsets of mouse NK cells have been described based on their expression of CD11b

and CD27. NK cells differentiate from CD11blo

CD27lo

to CD11blo

CD27hi

to CD11bhi

CD27hi

to

the most mature CD11bhi

CD27lo

(122). The double-positive and most mature subsets show a

comparable capacity to kill target cells and produce IFNγ. The CD11blo

CD27hi

NK cells are

predominantly found in the LNs and BM, the CD11bhi

CD27lo

more so in the spleen, liver, lung,

Page 33: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

10

and blood, and the double-positive are more homogeneously distributed (123). The chemokine

receptors, chemokines and adhesion molecules regulating the trafficking of NK cells between

these tissues are multifold. In humans, NK cells can be divided into CD56dim

and CD56bright

NK

cell subsets, which differ in their homing properties (124). The lack of CD56 expression on

mouse NK cells, and the differences in CD11b/CD27 expression on mouse and human NK cells

has made it difficult to make direct comparisons, however it is believed that the CD56bright

human NK cell subset most resembles the CD11blo

mouse NK cell subsets (91).

NK cell receptors and ligands

NK cells recognize their targets via an array of cell-surface activating and inhibitory receptors

and their activation is ultimately determined by the balance of signals of the two kinds of

receptors (Table 1.1). Activating receptors can detect stress-induced self ligands and infectious

non-self ligands. These ligands generally show limited expression on healthy cells, and are

upregulated following cellular pathology. NK cell recognition of these ligands underlies

“induced-self” recognition (125). Activating receptors can associate with adapter molecules that

have immunoreceptor tyrosine-based activation motifs (ITAMs), such as FcεRIγ, CD3ζ and

DAP12. Upon ligand binding and tyrosine phosphorylation of the ITAM, tyrosine kinases such

as Syk and ZAP70 are recruited via their SH2 domains and mediate downstream events,

including calcium influx, degranulation and transcription of chemokines and cytokines (126).

Below, I will briefly discuss the activating receptors NKG2D and the natural cytotoxicity

receptors (NCRs).

NKG2D is a type II transmembrane glycoprotein that is expressed as a disulfide-linked

homodimer. There are two isoforms of NKG2D generated by alternative splicing in mice:

NKG2D-L (longer isoform), which recruits the adapter molecule DAP10, and NKG2D-S

(shorter isoform), which recruits either DAP10 or DAP12 (127, 128). NKG2D can be expressed

as both isoforms in mice, and can therefore signal via both DAP10 and DAP12, whereas human

NKG2D is only expressed as the long isoform that signals via DAP10 (129). DAP12 signals via

an ITAM in its cytoplasmic tail; however, DAP10 contains a Tyr-x-x-Met motif (where x

represents any amino acid), which binds either Grb2 or the p85 subunit of phosphatidylinositol-3

Page 34: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

11

This table was modified and reproduced from (130)

Page 35: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

12

kinase (PI3K) following phosphorylation (131, 132). NKG2D recognizes multiple ligands in

both mice and humans that are homologous to MHC-I. Human NKG2D ligands include MICA,

MICB and six ULPB/RAET1 proteins, and mouse ligands include five isoforms of RAE-1, 3

isoforms of H60, and MULT1 (133).

Normal healthy cells generally show no or very little expression of NKG2D ligands, and these

ligands are only upregulated following infection, transformation, injury, or other forms of stress.

These processes lead to the activation of cellular stress pathways such as the DNA damage

response and the heat shock pathway, but also trigger NF-κB activation, which have all been

shown to regulate the expression of NKG2D ligands (133). There are stringent regulatory

mechanisms at the transcriptional, translational, and post-translational level controlling the

expression of these ligands, ensuring that only unwanted cells are removed. For example,

MULT1 transcripts are abundant in several normal tissues, especially in the thymus (134, 135),

but do not accumulate at the cell surface due to the protein’s ubiquitination and lysosomal

degradation during steady state conditions (136). This process is regulated by 2 closely related

ubiquitin ligases, membrane-associated RING-CH (MARCH) 4 and 9 (137). Downregulation of

MULT1 is prevented in response to stress imparted by heat shock and UV irradiation (136). In

chapter 3, I show that MULT1 is upregulated on B cells following LCMV clone 13 infection.

The activating natural cytotoxicity receptors (NCRs) are type I integral proteins belonging to the

Ig-superfamily. NCRs, including NCR1/NKp46 in mice, and NKp46, NKp30 and NKp44 in

humans, signal via ITAM-containing adapter molecules (138). Several ligands for these

receptors have been identified (see Table 1.1 for receptor/ligand pairs), including viral

hemagglutinin (139), heparan sulfate proteoglycans (140) and nuclear factor BAT3, which can

be released from tumor cells (141). NCRs have been implicated in anti-tumor immunity as NCR

deficiency compromises NK cell cytotoxicity against tumor targets in vivo (142, 143).

Inhibitory receptors that recognize self-MHC molecules (classical and non-classical) or non-

MHC self molecules act to ensure tolerance to self. Viruses have evolved mechanisms to avoid T

cell-mediated killing by downmodulating surface MHC-I expression; however, by so doing they

trigger NK cell activation, as inhibitory receptors on NK cells recognize MHC-I proteins and

other self molecules indicating a healthy cell. In this way, NK cells are thought to recognize

“missing self” on hematopoietic cells (144, 145). Inhibitory receptors signal via immunoreceptor

Page 36: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

13

tyrosine-based inhibitory motifs (ITIMs) in their cytoplasmic tails. Phosphorylation of the

tyrosine in the ITIM results in the recruitment of phosphatases, including the tyrosine-specific

phosphatases SHP (SH2-containing protein-tyrosine phosphatase)-1 and SHP2, and the

phospholipid-specific phosphatase SHIP (SH2-containing inositol polyphosphate 5-phosphatase)

(126). These phosphatases decrease the phosphorylation of several intracellular signaling

proteins, including FcεRIγ, ZAP70, Syk, PLCγ, LAT, SLP76, and Vav-1, to downmodulate or

prevent NK cell effector functions (126).

The MHC-I-specific inhibitory receptors include members of the C-type lectin-like Ly49 dimers

in mice, members of the killer cell immunoglobulin-like receptors (KIRs) in humans, and the C-

type lectin-like CD94-NKG2A heterodimers in both species. MHC-I recognition by Ly49

receptors depends on the presence of a peptide in the MHC-I binding groove (146-148), and the

level of Ly49 expression is influenced by the level of MHC-I expression in the mouse (149, 150)

(see Table 1.1 for Ly49/H-2 receptor/ligand interactions). Of interest to the finding of

constitutive interaction between 4-1BB and 4-1BBL on DCs presented in chapter 2 of this thesis,

is that Ly49 receptors can interact with MHC-I ligands in cis on the same membrane, as well as

in trans between cells. Cis interactions can reduce the capacity for trans interactions, thereby

reducing the signaling threshold required for activation (151, 152). There are also activating

Ly49 receptors, e.g. Ly49D and Ly49H, which associate with DAP12 to signal. In contrast to the

other Ly49 receptors, which bind H-2 molecules, the activating Ly49H receptor binds m157, a

viral glycoprotein expressed on MCMV-infected cells (153, 154). Although KIRs differ

structurally from Ly49 receptors, these families of receptors have analogous functions in humans

and mice, respectively, and are examples of convergent evolution (155). KIRs consist of type I

transmembrane proteins with 2 or 3 Ig-like domains, and either a short or long cytoplasmic tail

(156, 157) (see Table 1.1 for KIR/HLA receptor/ligand interactions). Similar to Ly49 receptors,

inhibitory KIRs signal through an ITIM, and activating KIRs associate with DAP12 to mediate

signaling (158). KIRs and Ly49 receptors, as well as their MHC-I ligands, are highly

polymorphic and are expressed in a mono-allelic manner (159, 160) by a stochastic mechanism

of gene regulation (161), resulting in distinctive subsets of NK cells that have unique properties

and activation thresholds. The CD94-NKG2 heterodimers bind to the non-classical MHC-I

proteins, HLA-E in humans and Qa1b in mice. These ligands could enable the heterodimers to

indirectly monitor the expression of classical MHC-I on target cells as HLA-E and Qa1b bind

Page 37: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

14

peptides derived from the leader sequence of classical MHC-I proteins (162, 163).

CD94/NKG2A heterodimers are inhibitory due to the ITIM contained in the cytoplasmic tail of

NKG2A, whereas CD94/NKG2C/E heterodimers can associate with DAP12 and are thought to

mediate activation (158). In contrast to the Ly49 receptors and KIRs, the CD94/NKG2 receptors

have limited polymorphism and are not stably expressed, as their expression can be modulated

by several factors, including the cytokine milieu (126, 164).

Non-MHC-I inhibitory receptors include the NKR-P1 receptors, which recognize Clr ligands in

mice, NKR-P1A, which recognizes LLT1 in humans, and 2B4, which recognizes CD48 in both

species. The C-type lectin-like NKR-P1 receptors can be either activating or inhibitory and bind

to a family of genetically linked C-type lectin-like ligands, Clr proteins (165). LLT1 is a

functional homologue of mouse Clr in humans (166, 167). The receptor/ligand interactions and

subsequent signaling within the NKR-P1/Clr system were recently reviewed in (165), and the

inhibitory NKR-P1B receptor is briefly discussed in section 1.2.5 of this thesis. 2B4, and its

ligand CD48, are members of the CD2 family of Ig-related proteins. The cytoplasmic tail of 2B4

contains an immunoreceptor tyrosine-based switch motif, which can recruit tyrosine

phosphatases, i.e. SHP-1 and SHP-2, and the adapter proteins SAP, EAT2 or ERT, upon tyrosine

phosphorylation (126, 168, 169). Signals mediated by 2B4 can be either activating or inhibitory,

depending on the isoform of the receptor, the recruited adapter proteins, the species, as well as

the stage of NK cell differentiation and activation (126, 158).

Functions of NK cells

NK cell activation is itself modulated by the cytokine microenvironment and also contributes to

the modulation of other cell types. Cytokines such as Type I IFN, IL-2, IL-12, IL-18 and IL-15

produced by other cell types are potent activators of NK cell effector function (91). Upon

activation, NK cells kill target cells via exocytosis of cytoplasmic granules containing perforin

and granzymes, FasL-mediated induction of apoptosis, antibody-dependent cellular cytotoxicity

(ADCC), and secrete cytokines such as IFNγ and TNF (91). NK cells also interact with other

cells of the immune system. IFNγ and TNF secreted by NK cells can promote the maturation of

DCs, which in turn can further activate NK cells via DC-derived IL-12 or IL-18 (170-172).

However, NK cells have also been shown to kill DCs, thereby influencing DC homeostasis

(173). In addition to NK cell-derived IFNγ promoting the priming of Th1 cells in the inflamed

Page 38: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

15

LNs (174, 175), NK cells have also been shown to kill activated T cells under certain conditions,

which will be discussed in section 1.5.4. In addition to direct lysis, NK cell acquisition of MHC-

II molecules from DCs has been shown to reduce CD4 T cell responses (176). In the Fas-

deficient mouse model, NK cells were shown to suppress autoreactive B cells in vitro, and NK

cell depletion in vivo worsened the severity of autoimmunity (177). Overstimulated macrophages

have also been shown to be targets of NK cell cytotoxicity (178). In sum, there is evidence that

in addition to their well known role as part of the innate activation of the immune system, NK

cells are also important in immune system downregulation, by eliminating activated cells.

The above mentioned innate effector functions of NK cells are well appreciated; however, recent

evidence in mice and humans suggests that NK cells also exhibit adaptive features, such as

antigen-specificity, clonal expansion, and the ability to generate long-lived memory cells that re-

expand and protect the host following re-exposure. Although the precise phenotype of these

memory NK cells remains elusive, they have been defined as showing relatively increased

expression of Ly49H, Ly6C, CD43, and KLRG1, and decreased expression of CD27 in the

MCMV system (179).

1.2.5 NK cells in anti-viral immunity

NK cells contain viral replication by killing infected cells, thereby controlling viral spread early

following infection, prior to the effector functions of adaptive immunity. A protective effect of

NK cells has been described in HSV-1, influenza virus, ectromelia poxvirus, vaccinia virus

(VV), HIV, hepatitis C virus (HCV), and hepatitis B virus (HBV) infections, and especially in

murine cytomegalovirus (MCMV) infection (180, 181). NK cell depletion is associated with

increased susceptibility to MCMV, and NK cell adoptive transfer with resistance to MCMV

infection (182, 183). Influenza infection of NK cell-depleted mice or of mice with defects in NK

cell activity results in delayed clearance of virus from lungs, and enhanced morbidity and

mortality (184, 185). Simian immunodeficiency virus (SIV) infection of rhesus macaques and

population genetic studies of HIV infection in humans have also suggested a role for NK cells in

modulating the outcome of HIV disease (186-190). Further evidence supporting a protective role

of NK cells comes from the multiple examples of viruses elaborating mechanisms to evade the

NK cell response. For example, MCMV encodes genes (e.g. MCMV m145, m152, m155) that

downregulate the expression of NKG2D ligands, thereby preventing NK cell activation, and also

Page 39: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

16

encodes genes (e.g. MCMV m144) that act as MHC-I homologs, thereby binding/signaling

inhibitory receptors on NK cells (191). Upon infection with rat CMV (RCMV), infected cells

downregulate Clr-b, which is a C-type lectin-like protein that binds to the inhibitory NK cell

receptor NKR-P1B. Thus, Clr-b downregulation sensitizes infected cells to killing by NK cells.

However, RCMV encodes a decoy gene product (RCTL) that has homology to the Clr ligands

and when expressed, protects infected cells from NK cell killing via interaction with NKR-P1B

(192). Cell-surface expression of Clr-b has also been shown to be downregulated in murine cell

lines and BM-derived macrophages by infection with two poxviruses, ectromelia virus and VV

(193). The precise mechanisms underlying Clr-b downregulation following viral infection are not

known but could involve the activation of cellular stress pathways, which have been shown to

cause downregulation of Clr-b at the cell surface (194). It is also possible that the virus encodes

proteins that affect Clr-b protein trafficking and/or turnover.

NK cells have also been shown to perform regulatory activities during viral infection, thereby

dampening anti-viral immunity, and promoting viral persistence (180). These functions of NK

cells might have evolved to control virus-mediated immunopathology and will be discussed in

the section on the role of NK cells during LCMV infection (1.5.4).

In sum, the activation of innate immune cells, such as DCs and NK cells, and their subsequent

effector functions result in early viral recognition, killing of infected cells, and initiation of

antigen-specific adaptive immune responses. However, it is becoming increasingly appreciated

that these innate cells can also serve a regulatory function and can continue to be key players

throughout the course of viral infection (195, 196).

1.3 The spleen as a secondary lymphoid organ involved in anti-viral immunity

Immune responses against pathogens, such as viruses, are orchestrated in secondary lymphoid

organs. The spleen is the largest secondary lymphoid organ and is situated in the left cranial

abdomen, below the diaphragm and connected to the stomach. Its main functions are to remove

aging erythrocytes, filter soluble and particulate antigens from blood, and initiate immune

responses to blood-borne microorganisms (197).

1.3.1 Development of the spleen

Page 40: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

17

The formation of the splanchnic mesodermal plate (SMP) at E12 is the initial event in the

development of the spleen. The SMP acts as an anlage to organize further formation of the

spleen (198). When formation of the SMP is defective, as occurs in mice that have a mutation in

the dominant hemimelia gene (199), or the cells that form it fail to proliferate, spleen

development halts. Several transcription factors such as homeobox 11 (Hox11), Wilm’s tumor 1,

Pbx1, Nkx3.2 and Pod1 are absolutely necessary for spleen formation (200-204). Genetic

experiments have shown that Pbx1 interacts with Hox11, and appears to be required earlier than

Nkx3.2 and Pod1 (204).

Progenitors of the erythroid and myeloid lineages are the first to colonize the spleen. On day

E13.5, lymphoid tissue inducer cells (LTi) are detectable in the spleen (205), and the first

hematopoietic stem cells (HSCs) appear at day E14.5 (206). The process of lymphoid

organogenesis has perhaps been best studied in LNs. During LN and Peyer’s patch (PP)

development, stromal lymphoid tissue organizer cells (LTo) expressing LTβR engage with

LTα1β2 expressed by LTi to activate classical and alternative NF-κB, resulting in the expression

of VCAM-1 and ICAM-1 on LTo and the production of CCL19, CCL21, and CXCL13. These

signals, along with TNFR1 signaling, increase attraction and retention of LTi and other

hematopoietic cells in the developing LN anlagen and induce further LTα1β2 expression, creating

a positive feedback loop that sustains LN development (207, 208).

While the development of LNs has been fairly well studied, the picture is less clear in the spleen.

Although LTi are present in neonatal spleen, they do not express LTα1β2, and the earliest

formation of white pulp and compartmentalization of red and white pulp in the spleen is LTα1β2-

independent. However, postnatal development of the white pulp, which involves the influx of T

cells, depends on LTα1β2 expression by B cells, which occurs starting around 4 days after birth

(209). At midgestation, spleen mesenchymal cells include LTo (209-212), but whether these

cells function like their LN counterparts in organizing the formation of lymphoid compartments

is not well known (213).

Overall, there are many gaps in the knowledge of mechanisms governing the development and

organization of the spleen, but it is clear that the molecular interactions governing these

processes are distinct from those involved in the generation of LNs and PPs (198). For example,

aly/aly mice, which have a point mutation in the alternative NF-κB-inducing kinase (NIK) (214),

Page 41: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

18

or mice deficient in LTα, lack all LNs and PPs, but still have a spleen, albeit with a disturbed

structure (215, 216).

1.3.2 Structure and function of the spleen

The spleen is a highly organized organ that is separated into areas of red pulp and white pulp

(Fig. 1.1). The red pulp largely consists of macrophages that are involved in iron metabolism and

removing old RBC and bacteria from the blood, and plasmablasts/plasma cells producing

antibodies. The white pulp, or the lymphoid region, of the spleen consists of distinct T and B cell

areas, surrounded by the marginal zone. Hematopoietic cells from the blood enter the white pulp

via the marginal zone, which consists of SIGNR1/MARCO-expressing marginal zone

macrophages, SIGLEC1-expressing marginal zone metallophilic macrophages, marginal zone B

cells, and DCs. The main difference in the structure of the mouse and human spleen is that

humans have an inner and outer marginal zone, which is surrounded by a large perifollicular

zone containing macrophages (198, 217).

The organization of the white pulp is maintained by specific chemokine gradients and cell-cell

interactions. B cells express the chemokine receptor CXCR5, which mediates their migration to

CXCL13, produced by follicular DCs (FDCs) and other stromal cells in the B cell follicles (207).

T cells and DCs express the chemokine receptor CCR7, which mediates their migration to the

ligands CCL19 and CCL21, produced by stromal cells in the T cell zone (218). The expression

of these chemokines is regulated by LTβR and TNFR1 signaling between stromal cells and

hematopoietic cells; disorganization of the white pulp occurs in the absence of these receptors

(219). These molecules are involved in the activation of transcription factors in the NF-κB

family. For example, triggering LTβR results in the expression of CCL21 in the spleen via

activation of the alternative IKKα/NIK-dependent NF-κB pathway (208).

Owing to their expression of an array of pattern recognition receptors and scavenger receptors,

marginal zone macrophages are potently involved in the uptake of bacteria and viruses in the

bloodstream, and produce cytokines such as type I IFNs. Marginal zone B cells are specialized to

detect blood borne pathogens and quickly differentiate into IgM-producing plasma cells, and

along with migratory and splenic DCs, can migrate to the white pulp to function as APCs.

Activated T cells and B cells meet at the T-B border to initiate adaptive immune responses. The

Page 42: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

19

This figure was adapted from (217, 220)

Page 43: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

20

precise mechanisms governing lymphocyte exit from the white pulp are unclear but likely

involve the modulation of cell surface chemokine receptors (198, 217).

The central function of the spleen in protection from encapsulated bacteria was highlighted in

studies of splenectomized mice and humans. Although the spleen is involved in anti-viral

immunity, it seems to be less vital than other secondary lymphoid tissues, such as LNs, in

mediating clearance of certain viruses. LCMV Armstrong or LCMV WE infection of mice

lacking a spleen, Hox11-/-

mice, resulted in barely detectable levels of virus and viral clearance

by day 15 at the latest. However, LCMV infection of aly/aly mutant mice, which lack all

secondary lymphoid organs (e.g. LNs and PPs), except the spleen, resulted in life-long viral

persistence. Similarly, infection of aly/aly mice with VV and vesicular stomatitis virus (VSV)

resulted in significantly delayed viral clearance and no production of neutralizing IgG,

respectively, whereas these infections in Hox11-/-

mice resulted in no abnormalities following

VV infection, and only slightly delayed antibody production following VSV infection. Aly/aly

mice succumbed to lethal encephalitis following VSV infection due to the lack of a neutralizing

IgG response, while Hox11-/-

and WT mice were protected from encephalitis. It should be noted

that although aly/aly mice contain a spleen, it exhibits a disturbed structure, which certainly

complicates the interpretation of studies with these mice (221). In contrast to these studies

however, infection of mice lacking splenic macrophages with acute LCMV and VSV was shown

to impair viral control, suggesting that the spleen or at least cell types in the spleen are important

in the defense against these viral pathogens (222, 223). It is possible that other factors in Hox11-/-

mice or differences in strains and doses of virus might account for these differences. Thus, the

indispensable role of the spleen in certain bacterial infections is clear, however its absolute

requirement during anti-viral immunity is less so.

1.4 Splenic atrophy due to viral infection and other causes

Splenic atrophy refers to the wasting of the spleen, and can result in hyposplenism, reduced

splenic function, or asplenia, a non-functional spleen. Splenic atrophy can result in compromised

immunity and compromised blood filtering. In humans, splenic atrophy and

hyposplenism/asplenia are commonly assessed by CT scan of the abdomen, a technetium-99

scan, and the presence of Howell-Jolly bodies, which are nuclear remnants in RBC normally

removed by the spleen. Acquired splenic atrophy in humans is most commonly associated with

Page 44: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

21

sickle cell anemia (224, 225), but also with autoimmune conditions (226, 227), such as celiac

disease (228-231), systemic lupus erythematosus (SLE) (227, 232) and Sjögren’s syndrome (227,

233). Although viral infections can induce transient splenomegaly due to an active immune

response in the spleen, infection with severe acute respiratory syndrome (SARS) (234) and

H5N1 (235) in humans, as well as coxsackievirus B3 (236), certain strains of parainfluenza virus

(237), murine gammaherpesvirus 68 (MHV68) (238, 239), and tick-borne encephalitis virus

(TBEV) (240) infections in mice have been associated with atrophy of spleen tissue.

1.4.1 Splenic atrophy in sickle cell anemia and autoimmunity

The pathophysiological mechanisms underlying splenic atrophy are not understood in most

cases. In sickle cell anemia, it is thought to occur due to repeated attacks of vasooclusion and

infarction within the splenic microvasculature, eventually leading to autosplenectomy in some

individuals (224). Splenic atrophy in intestinal disorders such as celiac disease has been

associated with folate deficiency (226), but also with autoimmune reactions leading to loss of

cells in the spleen. Celiac disease is frequently associated with a number of other autoimmune

diseases such as Sjögren’s syndrome and SLE, and it is not entirely clear whether splenic atrophy

predisposes to autoimmunity, or vice versa (228, 241, 242). Marginal zone B cells and DCs play

a role in the tolerance of autoantigens, and favour the expansion of Tregs (243-245). If these

cells are reduced in number or have compromised function as can occur following splenic

atrophy/hyposplenia, it is conceivable that this can promote the development of autoimmunity

(229). In SLE, splenic atrophy and hyposplenism have been linked to immune complex

saturation, leading to Fc-receptor blockage, as well as microthrombosis in the spleen related to

anti-phospholipid antibodies (227, 232).

1.4.2 Splenic atrophy due to viral infection: what are the mechanisms?

Splenic atrophy following viral infection is also poorly understood and has been attributed to the

cytopathicity of the virus itself, the resulting production of proinflammatory cytokines such as

TNF, the effector functions of T cells, and activation of the sympathetic nervous system.

Atrophy of the white pulp of the spleen following SARS infection in humans and parainfluenza

strain YN infection of mice is thought to be due to direct injury of the splenocytes by the virus

(234, 237). In the SARS study, they also found that the extent of damage to the immune cells

Page 45: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

22

was related to the length of the disease, in that a longer duration conferred more severe damage.

In contrast, splenic atrophy induced by infection of mice with coxsackievirus B3 and TBEV was

shown to occur in the absence of detectable virus in the cells being depleted (236, 240). Splenic

atrophy in the absence of direct viral cytopathicity of immune cells might be mediated by viral

infection of cell types required for lymphoid tissue homeostasis or viral factors that directly

interfere with immune cell production/maturation, diverted homing of lymphocytes to

nonlymphoid organs, autoreactive phenomena triggered by infection, or the depletion of immune

cells by the immune response generated by the virus.

In the murine models of splenic atrophy discussed above, the spleen was not the only organ

affected, as an involuted thymus and smaller LNs were also evident in some models (236, 237,

240). Loss of spleen size was also accompanied by a reduction in body weight, although this was

considered not to explain atrophy of the spleen, as loss in bodyweight was disproportional with

the loss in organ weight (236). Atrophic spleens were shown to be approximately 2-3 fold

smaller than uninfected controls in size (236, 237) and number of immune cells (236, 240), and

the kinetics of atrophy was quite fast following infection, with an onset of atrophy between 5-6

days following coxsackievirus B3 infection (236). Splenic atrophy following parainfluenza strain

YN and TBEV infection was assessed at only one time point, 7 and 13 days post-infection (dpi),

respectively (237, 240). In those studies where the number of specific cell types in the spleen

was assessed, it was shown that T cell, B cell and null subsets were affected (236, 239, 240).

Atrophy was irreversible as these infections ultimately killed their hosts (236, 237, 240). This,

however, was not the case with MHV68 infection of IFNγR-/-

mice (239).

MHV68 infection of WT and IFNγR-/-

mice results in similar viral clearance from the lung, and

both groups of mice do not succumb to the infection. However, unlike infection of WT mice,

infection of IFNγR-/-

mice leads to extensive and sustained splenic atrophy, as well as destruction

of splenic architecture. These pathological changes in the spleen were attributed to CD8 T cells,

in that CD8 T cell depletion prevented these changes. How the absence of IFNγ causes the CD8

T cells to induce splenic atrophy is not clear, but is thought to involve the production of perforin

and other factors normally controlled or inhibited in the presence of IFNγR. Virus replication in

splenocytes followed by a lysis of infected cells was eliminated as the mechanism underlying

splenic atrophy in this model, as blocking viral replication at 6 dpi did not impact splenic atrophy

Page 46: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

23

that became evident at 14 dpi. This study was taken out to 70 dpi, a time point at which IFNγR-/-

mice still had spleens that were around 2-3 times smaller than WT (238, 239).

Stress responses following viral infection and stroke, associated with increased activation of the

sympathetic nervous system and increased levels of glucocorticoid hormones, are also associated

with splenic/thymic atrophy, as these steroids are anti-inflammatory and can trigger apoptosis in

immune cells through activation of caspases (240, 246, 247).

These studies of splenic atrophy suggest that spleen size is largely determined by the number of

cells within the spleen, and that it is affected by the cytopathicity of the virus, the effector

functions of immune cells such as T cells, cytokine production, the integrity of the vasculature

supporting the spleen, and the stress response. The exact mechanisms underlying splenic atrophy

or its causes are not clear in any of these studies, leaving investigators only to speculate. In

chapter 3 of this thesis, I investigate the mechanisms underlying splenic atrophy following

persistent LCMV clone 13 infection, which will be discussed in section 1.5.

1.4.3 Compromised immunity due to complications associated with the spleen

Pathology of the spleen due to splenic atrophy, hyposplenism, or asplenia is associated with an

increased risk of infection to encapsulated bacteria such as Streptococcus pneumonia, Neisseria

meningitidis, and Haemophylus influenzae. This is due to defective/reduced activity of

opsonizing molecules such as properdin and tuftsin, and because of reduced numbers of natural

antibody producing-IgM-memory B cells in the marginal zone of the spleen, which are

responsible for mediating T-independent responses against these bacteria. Treatments include

prophylactic antibiotic therapy and immunization against these pathogens (248). Increased

susceptibility to the gram-negative bacteria Salmonella has also been documented (249, 250).

It is not entirely clear how much spleen tissue is required to protect against infection (251). Some

studies have investigated this; however, it seems that the precise mass/dimensions probably

depend on the nature of the infection and the immune response against it. In chapter 3 of this

thesis, I investigate the ability of LCMV-infected mice experiencing splenic atrophy to clear

Salmonella infection.

1.5 Murine models of viral infection: LCMV

Page 47: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

24

1.5.1 LCMV virus

LCMV is a non-cytopathic rodent pathogen that was first isolated by Charles Armstrong in 1934,

and is the prototypical member of the arenaviridae family (252). It is a spherical, enveloped virus

that consists of two ssRNA segments, designated L (7.2 kb) and S (3.4 kb). The L segment

encodes the viral RNA-dependent RNA polymerase and a RING finger protein called Z, which is

thought to act as a matrix protein (253) and regulate transcription (254). The S segment encodes

the two main structural proteins: nucleoprotein (NP) and the glycoprotein (GP) precursor (GP-

C). GP-C is post-translationally cleaved to GP-1 and GP-2. GP-1 is thought to mediate binding

to the cellular receptor for LCMV, α-dystroglycan (αDG) (255-257). The outcome of LCMV

infection in mice depends on the size of the viral inoculum, the strain of the virus, age of the host

and route of infection.

1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence

There are several strains of LCMV that differ in viral pathogenesis and persistence; of interest to

the present work are LCMV Armstrong (ARM) and LCMV clone 13 (Cl13). The acutely

infecting strain, ARM, is the parental strain, while the persistent genetic variant Cl13 was

isolated from the spleens of 2-month old BALB/c WEHI mice that had been infected with ARM

at birth (258). These carrier mice can make antibody responses to the virus, but diminished CTL

responses, due to the thymic deletion of virus-specific CTL during development (259-263). The

absence of a functional anti-LCMV CTL response in these neonatally ARM-infected mice leads

to life-long infection.

Although ARM and Cl13 differ dramatically in their persistence in the host, in that ARM is

cleared within 1 week, whereas Cl13 can persist for months, they are surprisingly genetically

very similar (264-266). Armstrong and Cl13 differ by only 5 nucleotides, of which 3 result in

amino acid changes. The lysine to glutamine change at position 1079 in the viral polymerase was

shown to selectively increase viral titers in macrophages, but the overall contribution of this

change to viral persistence remains ill-defined (265). The recently identified asparagine (ARM)

to aspartic acid (Cl13) amino acid substitution at position 176 in GP1, present in roughly 50% of

persistent LCMV strains, was shown not to be required for the ability of Cl13 to persist long-

term (267). In contrast, the phenylalanine (F) to leucine (L) substitution at position 260 in GP1,

Page 48: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

25

present in more than 95% of viral isolates mediating persistence, was shown to be essential for

the ability of Cl13 to persist long-term (268, 269). Cl13 binds to αDG, the cellular receptor for

LCMV, with 10-1000x higher affinity than ARM, a difference that is associated with the F260L

amino acid substitution, and has profound functional consequences. Among cells of the immune

system, αDG is preferentially expressed on CD11c+/DEC205

+ splenic DCs, and shows very little

expression on T, B, or CD11b+ cells. Strains and variants of LCMV that bind with high affinity

to αDG are associated with replication in the marginal zone and white pulp of the spleen and

viral persistence, whereas those that bind with lower affinity to αDG are associated with the red

pulp and viral clearance. The high expression of αDG on DCs leads to the higher infectivity and

impairment of these cells by Cl13, resulting in long-term persistence (270-272). Cl13 infection

inhibits DC maturation and upregulation of key surface markers involved in APC function

(MHC-I/II, CD40, CD80, CD86), and upregulates immunosuppressive cytokines and ligands,

thereby reducing the ability of these cells to fully prime T cells (76, 273-276). In contrast, ARM

predominantly infects F4/80+ macrophages and a few DCs (270).

There is evidence that LCMV uses receptors other than αDG to infect cells. αDG-/-

mouse

embryonic stem (ES) cells can be infected by non-immunosuppressive strains such as ARM,

which infects more than 50% of these cells by 48 hours following infection (277).

Immunosuppressive variants, such as Cl13, show little infection of αDG-/-

ES cells after 48 hours

(<10%), indicating a strong dependence of infection on αDG. However, the immunosuppressive

variant PBL364 can infect more than 80% of ES cells lacking αDG by 48 hours following

infection (277). Two members of the TAM (Tyro3 Axl Mer) family of receptors (Tyro3 and

Axl), as well as DC-SIGN and lymph node sinusoidal endothelial calcium-dependent lectin

(LSECtin) were recently suggested to act as receptors for LCMV and a related arenavirus, Lassa

virus (278, 279). Jurkat T cells overexpressing Axl, Tyro3, DC-SIGN, LSECtin, or a control

protein were infected with viruses pseudotyped with the GPs of 4 strains of LCMV, including

ARM and Cl13. It was found that in the presence of these receptors on Jurkat cells, there was 2-3

logs higher infectivity when compared to control. There weren’t any striking differences in the

infectivity of immunosuppressive and non-immunosuppressive strains, indicating that usage of

these receptors does not correlate directly with LCMV virulence (278). Following this report,

Sullivan et al. evaluated the physiological relevance of Axl during in vivo ARM and Cl13

infection. Axl-/-

and WT mice cleared Cl13 from the blood with similar kinetics, and there were

Page 49: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

26

no quantitative or qualitative differences in the CD4 or CD8 T cell responses following both

ARM and Cl13 infection (280). These findings indicate a lack of major in vivo significance of

Axl expression in the establishment or course of an acute or persistent LCMV infection, although

it is possible that the expression of the other TAM receptors, Tyro3 and Mer, compensate for the

lack of Axl. In sum, αDG is still the only receptor identified that correlates with LCMV

pathogenicity, but it is likely that other receptors exist. The in vivo relevance of Tyro3, DC-

SIGN, and LSECtin during LCMV infection remains to be evaluated.

Following ARM infection, a robust anti-LCMV CTL response is generated that is associated

with specific lysis of infected cells, and production of effector molecules, including IFNγ, TNF,

IL-2, perforin, and granzyme B (281-283). CD8 T cells reactive to the immunodominant LCMV

peptides NP396-404, GP33-41, and to a lesser extent GP276-286 in the context of MHC-I H-2Db

(284-

288), can reach a peak expansion of around 107 cells per spleen by day 8 following infection and

can constitute up to 50% of CD8 T cells in the spleen (266, 289, 290). Following expansion and

viral clearance, more than 90-95% of these cells die (290, 291), leaving behind a small

population of memory CD8 T cells (292, 293). In contrast to ARM infection, infection with Cl13

is associated with physical deletion of NP396-404-reactive CD8 T cells (266, 294, 295) and

functional impairment of other viral-specific CD8 T cells, including those reactive to GP33-41 and

GP276-286. Since NP is the most abundantly expressed viral antigen, those CD8 T cells reactive to

NP396-404 are the ones most likely to be stimulated and eventually deleted/exhausted, whereas

those reactive to the less abundant GP are present and functional longer (296, 297). Loss of

function is indirectly correlated with antigen persistence/viral load. This functional impairment

(exhaustion) is associated with a hierarchical loss of function, with the ability to lyse cells and

produce IL-2 lost first, followed by the inability to produce TNF, and lastly IFNγ (266).

In addition to the loss of effector function, T cell exhaustion following persistent LCMV

infection is also associated with the expression of inhibitory cell surface molecules, including

PD-1/PD-L1, LAG-3, Tim-3, 2B4, CD160, and CTLA-4 and immunoregulatory cytokines,

including IL-10 and TGFβ (298). When comparing functional memory CD8 T cells with

functionally impaired viral-specific CD8 T cells by gene array, it was found that the exhausted T

cells showed enhanced expression of the inhibitory molecule PD-1. Blocking PD-1-PD-L1

interactions in vivo was shown to restore the ability of CD8 T cells in chronically infected mice

Page 50: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

27

to proliferate, produce cytokines, kill cells, and decrease viral load (299). Similarly, production

of the immunoregulatory cytokine IL-10 was found to be sustained following persistent LCMV

infection, and IL-10R blockade mediated clearance of an otherwise persistent Cl13 infection

(300, 301). CD4 and CD8 T cells, as well as DCs, macrophages, and NK cells have been

implicated in producing IL-10 during persistent viral infection (302). The above findings have

been corroborated in studies of HIV and HCV infection in humans, suggesting that the

immunoregulatory mechanisms underlying chronic viral infection are shared by distinct viruses

in different hosts. These mechanisms are thought to have evolved to put a break on the T cell

response when virus cannot be cleared, so as to avoid immune-mediated pathology. For example,

PD-L1-/-

mice succumb to Cl13 infection within 8 days due to immunopathological damage

(299).

CD8 T cells are the main cell type involved in viral clearance, as mice lacking CD8 T cells never

purge the virus (303). Several lines of evidence make it evident that CD4 T cells also play a

substantial role. Following ARM infection, virus-specific CD4 T cells, which are primarily Th1

in nature, expand with slower kinetics and to lower numbers than CD8 T cells, and produce IFNγ

and IL-2 (304-307). In the absence of CD4 T cells, mice are capable of clearing ARM infection,

but show evidence of T cell exhaustion and defects in the generation of a stable memory pool

(297, 308, 309). Compared to ARM infection, Cl13 infection induces viral-specific CD4 T cells

that are reduced in number, and show functional defects, such as in production of IFNγ, IL-2 and

TNF (310-312). In this way, similar to CD8 T cells, CD4 T cells also show evidence of

exhaustion during persistent LCMV infection. Nevertheless, these CD4 T cells are critical for

eventual control of the virus, as CD4 T cell depletion during persistent LCMV infection leads to

uncontrolled life-long viremia (313). CD4 T cell help, associated with the production of IL-21,

provided to CD8 T cells and antibody-producing B cells, eventually controls persistent LCMV

infection within 2-3 months in the periphery (314, 315). Signaling through costimulatory

molecules such as OX40 on CD4 T cells has been shown to contribute to control of LCMV clone

13 infection (316).

1.5.3 The innate immune response to LCMV infection

Although much is understood about the role of the adaptive immune response to LCMV

infection, the contribution of innate cell types is relatively poorly defined. DCs are the critical

Page 51: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

28

APCs required to mount an effective CTL response following LCMV infection, in that mice

lacking DCs, but not macrophages and B cells, show defective priming (317). Within 24 hours of

ARM infection, splenic DCs produce cytokines including type I IFN, TNF, and IL-12, and

acquire the ability to stimulate naive LCMV-specific CD8 T cells ex vivo (318, 319). This DC

activation is accompanied by an increased tendency to undergo apoptosis, and cDC numbers in

the spleen decrease around 2-fold by 3 dpi. Both DC activation and apoptosis are IFN-dependent.

The decline in cDCs is accompanied by an increase in the number of pDCs by 3 dpi, which also

upregulate type I IFN production by 24 hours post-infection (pi) (318).

In a recent study, the innate immune response to ARM and Cl13 infection was compared side by

side. This study identified no difference in the early response (within 72 hours) of DCs to both

infections, in that DCs similarly upregulated activation markers and stimulated anti-viral CD8 T

cells, although by day 14 pi, myeloid cells from Cl13-infected mice showed an inability to

effectively stimulate T cell responses. By 7 dpi, there was an increase in a population of

myeloid-derived suppressor cells (MDSCs) during both infections in lymphoid organs and blood,

which presumably arise to dampen immune responses and avoid immunopathology. This

population of MDSCs was greater in the Cl13-infected animals, persisted compared to ARM-

infected animals, and potently suppressed T cell proliferation ex vivo. Depleting these cells with

a Gr-1 antibody enhanced anti-viral CD8 T cell responses, but did not affect viral load (195).

Other studies have also identified the enhanced and sustained presence of immunoregulatory

APCs expressing molecules such as IL-10 and PD-L1 during Cl13 infection (275, 320). These

studies highlight the involvement of the innate immune system both early and throughout the

course of persistent viral infection.

The aforementioned differences in the nature of ARM and Cl13 and the immune responses they

elicit make these viruses excellent models to investigate the processes underlying acute versus

persistent viral infection. Studies of LCMV have advanced our understanding of the pathological

mechanisms underlying chronic viral infections and many of the findings have held true in

human infections, such as HIV, HCV, and HBV.

1.5.4 The role of NK cells during LCMV infection

Page 52: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

29

In general, LCMV infection is relatively NK-resistant, in that NK cells do not directly interfere

with LCMV replication in the mouse (180). Although IFNγ expressed by NK cells has been

implicated in controlling LCMV replication following ARM infection (321), the data

overwhelmingly point to a role for NK cells not in direct control of virus, but in modulation of

anti-LCMV T cell responses, which then impact viral control. By infecting NK cell-deficient

mice and WT NK cell-depleted mice with LCMV WE, an acute strain of LCMV that is more

persistent than ARM, Lang et al. showed that NK cells mediate viral persistence of LCMV by

killing activated LCMV-specific CD8 T cells in a perforin-dependent manner. In the absence of

NK cells, mice were able to clear the virus in several organs (322).

Waggoner et al. took a more detailed approach and compared low, medium, and high dose

infection with Cl13, and concluded that NK cells act as “rheostats” in controlling anti-viral T cell

immunity. Low-dose (5 × 104

PFU) infection generates a robust anti-LCMV T cell response

which results in viral clearance. In this model, NK cell depletion enhanced the CD8 T cell

response, which had no effect on the already effective clearance of virus. Medium dose Cl13 (2 ×

105 PFU) normally results in partial functional exhaustion of T cells and T-cell dependent

immunopathology. In this setting, NK cell depletion reversed the immunopathology because a

greater number of anti-LCMV CD4 and CD8 T cells were able to enhance viral clearance before

the acquisition of severe immune pathology. However, depletion of NK cells prior to a high dose

infection (2 × 106 PFU), which normally results in a persistent infection not associated with

mortality, worsened the outcome, mediating increased immunopathology and mortality. In this

context, NK cells were required to dampen immunopathological T cell responses. Overall, NK

cell downmodulation of anti-viral T cell responses in the context of LCMV infection can be

either beneficial or detrimental, depending on whether the T cell response is mediating viral

clearance or immunopathology. In contrast to Lang’s study, Waggoner et al. point to a direct

perforin-mediated cytolytic effect of NK cells on CD4 T cells, which then negatively impact the

anti-viral CD8 T cell response (323).

These studies looked at the role of NK cells early following infection, in that mice were depleted

of NK cells prior to infection. A recent study evaluated the outcome of NK cell depletion 1 day-4

weeks following Cl13 infection, and found that NK cells continue to promote persistent viral

infection due to downmodulation of virus-specific T cell responses. The effect of NK cell

Page 53: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

30

depletion on viral burden and virus-specific T cell responses was found to be dependent on the

size of the viral inoculum, viral load at the time of depletion, and the presence of CD4 T cells

(196). In contrast to these findings, another report suggested that delayed NK cell depletion

following Cl13 infection did not significantly impact the numbers of virus-specific T cells as

measured by cytokine production (324). It is argued that whether or not late-stage NK cell

depletion impacts anti-viral T cell responses and viral load depends on the viral load and the

extent of immune exhaustion at the time of NK cell depletion. If viral load is high, then anti-viral

T cells are more exhausted and NK cell depletion, which would increase the number of anti-viral

T cells, would not have an impact on viral load as the T cells are exhausted. However, if the viral

burden is lower, then there are still functional T cells that would be spared by NK cell depletion,

which would then mediate viral control (196). In addition to viral persistence, Cook et al. used

an NK cell-depleting antibody to identify a role for NK cells in mediating T cell exhaustion

(324). NK cell depletion prior to Cl13 infection reduced the percentage of transferred P14 T cells

(TCR transgenic CD8 T cells specific for LCMV GP33-41) expressing the inhibitory receptors

PD-1, Lag3 and 2B4, and the per cell expression of these molecules 8 dpi, as well as their

production of IFNγ, TNF and IL-2 following ex vivo re-stimulation on day 29 pi. A role for NK

cells in mediating diminished APC function following Cl13 infection was also identified, in that

APCs isolated from Cl13-infected/NK cell-depleted mice (2-3 dpi) showed an enhanced ability

to stimulate LCMV-specific T cell proliferation ex vivo when compared to APCs isolated from

Cl13 infected/untreated mice (2-3 dpi) (324).

The mechanisms of T cell recognition by NK cells are likely to involve multiple receptor-ligand

interactions. Lang et al. point to a role for NKG2D, in that they found that LCMV infection

upregulates NKG2D and NKG2D ligand expression, and that blocking NKG2D with an antibody

results in viral control, a finding that is similar to NK cell depletion in their model (322). In

contrast, Waggoner et al. found no role for NKG2D in the NK cell-dependent killing of CD4 T

cells, and instead pointed to an interaction between the inhibitory NK cell receptor, 2B4, and its

ligand, CD48 (180, 323). Cl13 infection of 2B4-/-

mice, which have hyperactive NK cells due to

loss of an inhibitory signal, results in reduced T cell responses and a persistent viral infection

that is never cleared (325). The differences in the Lang and Waggoner studies have not been

resolved. They might be due to the differences in the tropism and pathogenicity of the different

LCMV strains used, Cl13 and WE, and differences in the NK ligands upregulated or

Page 54: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

31

downregulated on the T cells by the different viruses. It is also possible that different doses of the

depleting NK cell antibody, anti-NK1.1 (200 μg/mouse in the Lang study versus 25 μg/mouse in

the Waggoner study), resulted in depletion of different subsets of NK cells with distinct activities

during LCMV infection.

The T cell-expressed inhibitory NK cell ligands MHC-I and Qa-1b, as well as the ligands for the

activating receptor NCR1, were recently implicated in the mechanism whereby NK cells

recognize and kill virus-specific CD4 and CD8 T cells following LCMV infection. It was shown

that type I IFN signaling early following LCMV infection can protect anti-viral T cells from

perforin-dependent NK cell-mediated cytotoxicity by upregulating NK cell inhibitory ligands

and downmodulating NK cell activating ligands on T cells. LCMV-specific IFNAR1-/-

T cells (T

cells lacking type I IFN receptor 1) were rapidly eliminated following transfer into LCMV-

infected hosts, however, this depletion was abrogated with prior NK cell depletion (326, 327).

NK cells have also been shown to dampen T cell-mediated immunity in other murine viral

infection models, including MCMV (328, 329), Friend virus (FV) (330), and influenza (331).

These functions of NK cells might have evolved to avoid T cell-mediated immunopathology or

development of autoimmunity.

Human genetic studies have shown that the presence of certain combinations of NK cell

inhibitory receptor genes in the KIR family and HLA genes correlates with viral control in

cohorts of HCV-infected individuals (332-334), and a better prognosis in HIV patients (335-

337). Co-expression of the inhibitory KIR3DL1 genotype that exhibits high cell surface

expression (KIR3DL1*h) and its ligand, a HLA-B allele with an isoleucine at position 80 (HLA-

Bw480I

), is associated with slower HIV-1 disease progression (337). Co-expression of the

inhibitory KIR2DL3 and its cognate HLA-C ligand is associated with spontaneous resolution of

viremia following HCV infection and is found more frequently in exposed seronegative aviremic

individuals and in HCV-infected individuals that respond favourably to treatment (332, 333,

338). These inhibitory receptors are thought to be protective by enhancing the activity of NK

cells during these infections. The inhibitory KIR3DL1*h/HLA-Bw480I

pairing that is protective

in HIV infection is believed to be associated with high cell surface expression of the inhibitory

KIR, which leads to strong NK cell licensing, that is, an enhanced functional competence (339).

The inhibitory KIR2DL3/HLA-C1 interaction is thought to be more protective in HCV infection

Page 55: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

32

because it mediates a weaker inhibitory signal than other KIR/HLA pairings, thereby being

easily overridden by activating signals (332, 340, 341). One study showed that HIV-exposed

seronegative female sex workers in Abidjan, Côte d’Ivoire, more frequently possessed inhibitory

KIR genes in the absence of their cognate ligand genes, whereas HIV-seropositive workers were

characterized by inhibitory KIR/HLA receptor/ligand pairings (336). Absence of ligands for

inhibitory KIRs was thought to lower the threshold for NK cell activation (336). Indeed, strong

NK cell responses have been associated with inhibition of HIV and HCV replication (332, 342,

343). However, studies of NK cells in LCMV infection suggest that these inhibitory receptors

could also be protective in the human studies because they shut down NK cell responses, which

would otherwise dampen anti-viral immunity and promote viral persistence.

In sum, NK cells were classically known for their role in mediating viral clearance early

following infection, however, mounting evidence from the study of LCMV and other viral

infections suggests that NK cells have an immunoregulatory role that prevents pathology, but

contributes to viral persistence. It seems therefore that depleting NK cells could be a therapeutic

approach in the treatment of chronic viral infections in humans; however, caution must be taken,

as these functions of NK cells presumably evolved to protect the host from immunopathology.

As the Waggoner study showed, depleting NK cells during a high dose Cl13 infection eventually

enhanced immunopathology and mortality (323). More work will be needed before we can

translate these findings into treatments for human disease. These effects of NK cells were largely

shown to be mediated in the first few days following viral infection. In chapter 3 of this thesis, I

identify a novel role for NK cells in the later stages (>3 weeks) of LCMV infection.

1.5.5 Type I Interferons during LCMV infection

Type I IFN production reaches peak titers in the serum 2-3 days pi with ARM (344). Multiple

cell types are induced to produce IFN, including DCs, pDCs, and marginal zone macrophages,

which are thought to be the primary IFN producers (345). IFN expression wanes with the

resolution of ARM infection, but persists following Cl13 infection, and is detectable in DCs out

to day 50 pi (274, 320). The IFNAR-regulated genes, OAS and Mx1, are specifically enriched in

the immunoregulatory APC population expressing high levels of PD-L1 and IL-10 on day 9 post

Cl13 infection, suggesting that prolonged IFN signaling and immunosuppression are linked

(320).

Page 56: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

33

IFNs are involved in DC activation and maturation, directly affect T cell

expansion/differentiation, and have also been shown to activate NK cells following LCMV

infection (344, 346-350). Resistance to IFN α/β and γ impacts the ability of various LCMV

strains to spread within the host, in that relatively IFN-resistant strains such as Cl13 spread, but

IFN-sensitive strains such as ARM, do not (351). IFNs can play a critical role in LCMV

infection by reducing viral loads early, and therefore modifying the extent of CD8 T cell

exhaustion and viral persistence, depending on the strain of LCMV (351). Control of ARM

infection certainly depends on a robust IFN response, in that IFNAR-/-

mice show accelerated

kinetics of virus replication, higher peak viral titers, and a slower kinetics of clearance, with

virus still detectable at day 9 pi (320), and variable clearance between 2-8 weeks pi (351).

The IFN signature induced by Cl13 infection, however, can also contribute to

immunosuppression and viral persistence, as alluded to earlier. Cl13 infects hematopoietic

progenitor cells and impairs their development into DCs, a mechanism that is dependent on type

I IFN/STAT2 (76, 274, 352). It was shown that Flt3L treatment of Cl13-infected mice resulted in

reduced expansion of CD8+ and CD8

- DCs compared to uninfected controls, whereas the same

treatment in IFNAR-deficient mice resulted in normal expansion of CD8+ DCs (76). This

strategy evoked by the virus results in fewer DCs available to prime T cells and generate anti-

viral responses. Blocking IFNAR signaling prior to Cl13 infection reduced expression of

molecules associated with immunosuppression (IL-10, PD-L1), but initially increased viral load,

indicating that early IFN signaling was required for viral control. However, when the immune

response was assessed on day 30+, there was now significantly less viremia in the IFNAR

blocking antibody treated mice. Similar results were obtained when IFN signaling was blocked

therapeutically during late stages of infection (day 25+) (320, 353). These and other data suggest

a dual role for IFNs following Cl13 infection - stimulatory early, but immunosuppressive late.

During chronic viral infection in humans, a combined ribavirin/IFN therapy has been shown to

be effective in the treatment of HCV-infected individuals, although some patients do not respond

to this therapy. A characteristic of these patients is a high IFN signature prior to treatment, which

given the findings in Cl13 infection, might mediate immune dysfunction and resistance to any

benefits bestowed by further addition of IFN (354, 355). An elevated interferon signature in

HCV patients can be associated with limited control of virus replication and development of liver

Page 57: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

34

pathology (356, 357). In line with these data is the finding that HIV infection of humans and

pathological SIV infection of rhesus macaques is associated with an elevated, prolonged IFN

signature, whereas non-pathogenic SIV infections of sooty mangabeys and African green

monkeys shows curtailed IFN expression (358, 359). Blocking IFN early during SIV infection of

rhesus macaques, however, is not beneficial, in that this was recently shown to reduce anti-viral

genes, increase SIV reservoir size, and accelerate CD4 T cell depletion and progression to AIDS.

In this model, administering IFN-α2 initially prevented systemic infection, but continued

treatment was followed by IFN-desensitization and a worse outcome (360).

Taken together, these data suggest that timing of IFN blockade/treatment is a critical factor in

determining the efficacy of therapy during chronic viral infection in mouse and man. IFNs can be

required early following initial infection to control viral load, therefore blocking IFNs at this

time can be detrimental and administering IFNs beneficial. However, persistent IFN signaling

can induce immune dysfunction, therefore blocking IFNs later during the infection, after initial

viral replication has been controlled, can be beneficial, and administering IFN at this time could

enhance immune dysfunction or have no effect, depending on the functionality of the immune

response. These uncertainties highlight the complexity of targeting type I IFNs therapeutically.

In chapter 3 of this thesis, I investigate the effect of IFN blockade on the population of NK cells

in the spleen and splenic atrophy following Cl13 infection.

1.5.6 Splenic architecture and remodeling following LCMV infection

Adaptive immune responses, such as those that occur following viral infection, are characterized

by substantial restructuring of secondary lymphoid organs, such as the spleen and LNs, including

both their size and architecture. Mesoscopic imaging techniques such as optical projection

tomography (OPT) are suited to probe objects of 0.5 to 15 mm in diameter, which are too large

for conventional microscopic imaging (361, 362). OPT analysis of whole-mount fluorescently

labeled murine peripheral LNs (PLNs) was used to obtain quantitative structural 3D information

on PLN structure following acute LCMV infection (362). This study revealed that reorganization

of PLN microarchitecture following acute LCMV infection, was accompanied by a 3-fold

increase in PLN volume and high endothelial venule network length and required LTα1β2-

expressing B cells, but not VEGF-A (362). During infection, the fibroblastic reticular cell (FRC)

Page 58: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

35

network expands by proliferation in order to increase in volume and accommodate more

lymphocytes.

Although immune responses are associated with an increase in the size of lymphoid tissues, their

microarchitecture must be maintained in order to facilitate effective communication between

immune cells, and generate protective T cell and antibody responses. Infection with several

strains of LCMV has been shown to disrupt lymphoid architecture early following infection, in

part due to the CD8 T cell mediated destruction of virally-infected marginal zone macrophages

and FDCs, although organization returns by 3 weeks pi (363). This disrupted architecture is

believed to contribute to the immunosuppressive capabilities of persistent LCMV strains. It was

recently shown that type I IFN production following Cl13 infection also contributes to splenic

disorganization, in that blocking IFN prior to infection prevented disruption of lymphoid tissues

in the spleen (353). Pathological cytokine production following infection with LCMV WE also

contributes to disruption of splenic architecture (364). Blocking CD27 signaling, which was

thought to mediate pathological IFNγ and TNF production by CD4 T cells, abrogated LCMV-

induced immunopathology and actually mediated elimination of an otherwise chronic LCMV

strain, Docile (364). TNF-mediated loss of marginal zone macrophages was also induced by the

parasite Leishmania donovani resulting in lymphoid tissue remodeling and impaired lymphocyte

trafficking into the white pulp (365).

Cl13 can also infect and disrupt the conduit function of FRC, a network of cells which provides a

3-dimensional framework in SLOs on which immune cells travel, interact, and receive survival

signals (366). By immortalizing FRC in vitro, it was shown that these cells also present antigen

in an MHC-I/II-dependent manner and provide activation signals to LCMV-specific CD4 and

CD8 T cells (367). The disrupted function of these cells and their expression of PD-L1 following

Cl13 infection are thought to contribute to immunosuppression and viral persistence (366, 368).

Damage to the FRC following HIV infection is also thought to contribute to progressive

depletion of CD4 and CD8 T cell populations, as these cells are prevented from receiving

survival factors such as IL-7 through the FRC network (369).

Restoration of lymphoid tissue architecture following LCMV infection is mediated in part by

adult LTi. Following LCMV WE infection, LTi proliferate and interact with stromal cells in a

LTβR-LTα1β2-dependent manner, thereby restoring lymphoid organ structure following infection

Page 59: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

36

(370). Thus, similar to their role in ontogeny, LTi are also involved in maintaining the structural

integrity of lymphoid tissues later in life. Embryonic mesenchymal progenitors of the Nkx2-

5+Islet1

+ lineage, which give rise to embryonic mesenchymal cells with lymphoid tissue

organizer activity, were also recently shown to be involved in regenerating FRC and stromal cell

integrity following LCMV WE infection. Although the precise nature of the cells that participate

in the regeneration of stromal networks in the spleen is not known, these authors point to a

potential role of perivascular cells, which were recently suggested to function as mesenchymal

stem cells. These cells are not targeted by LCMV, thereby making them candidates for local

stromal progenitors during tissue regeneration (213).

Remodeling of splenic architecture following infection also occurs with other viruses such as

MCMV. MCMV infection leads to loss of marginal zone macrophages, and specifically

downregulates CCL21 expression by splenic stromal cells in an LT-independent manner,

resulting in aberrant T cell localization within the white pulp (371). This is thought to be a

strategy used by the virus to impede cellular trafficking and cell-cell interactions required to

mount effective immune responses.

In sum, the destruction of lymphoid architecture required to mount anti-LCMV immune

responses is another reason that LCMV continues to replicate and persist throughout the host.

This is a reversible process, in that splenic architecture is eventually regained; however, the

mechanisms underlying the regeneration of lymphoid tissue integrity are just beginning to be

elucidated.

1.5.7 Resolution of inflammation following LCMV infection

Although the expansion phase of the immune response (0-8 dpi) following LCMV infection has

been well characterized, less is known about the resolution of inflammation during the

contraction phase (day 8+). The loss of cell numbers is largely attributed to the apoptosis of CD8

T cells, due to the unavailability of survival cytokines in the absence of viral replication, activity

of multiple death-inducing TNF family receptors, and toxicity due to repeated release of perforin

and IFNγ (372-375). The rate of CD8 T cell contraction following ARM infection is independent

of the dose and duration of infection, the magnitude of expansion, or the amount of antigen

displayed (376). Following clearance of infectious virus, CD8 T cells no longer proliferate;

Page 60: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

37

however, CD4 T cells show delayed contraction compared to CD8 T cells, due to their continual

proliferation in response to residual antigen (305, 377-381).

The prototypical member of the death-inducing TNFR family is TNFR1. ARM infection of

TNFR1-deficient mice revealed that this molecule is not a critical player in the downregulation

of CD8 T cell responses; however, delayed contraction is observed in TNFR1/2 DKO mice (382-

384). TNF deficiency prolonged the effector phase of the CD8 T cell response and increased the

number of memory CD8 T cells following ARM infection due to reduced apoptosis (385).

TNFR1 deficiency alone and in combination with TNFR2 deficiency delayed contraction of CD8

T cells specific for GP33-41 and GP276-286 following Cl13 infection (386). Similar to TNFR1, the

TNFR family member Fas is largely dispensable for the apoptosis of LCMV-specific CD8 T

cells (387, 388); however, combined Fas/BIM deficiency leads to a LN-specific block in

contraction following ARM infection (389). BIM was also shown to play a critical role in the

downregulation of CD8 T cell responses following Cl13 infection, in that BIM deficiency

prevented the apoptosis of NP396-404-specific CD8 T cells (390). Taken together, these data

suggest that the strain of LCMV, as well as the specificity of the CD8 T cells differentially

impact the genes and pathways utilized in the contraction phase of the immune response. How

other cell types are depleted during the contraction phase is not well understood, and is the topic

of chapter 3.

1.6 Murine models of viral infection: influenza

1.6.1 Influenza virus

Pandemic influenza infection of humans claimed between 20 and 50 million lives in the 20th

century, and today seasonal influenza virus remains a significant cause of morbidity and

mortality especially in the immunocompromised, young children, and the elderly (391). The

enveloped influenza virus is a member of the orthomyxoviridae family and contains a genome

composed of 8 segments of negative sense ssRNA surrounded by NP. The major targets of the

anti-influenza antibody response are the envelope glycoproteins hemagglutinin (HA) and

neuraminidase (NA), of which there are several subtypes. HA mediates binding of the virus to

sialic acid residues on target cells and fusion of the viral envelope with the endosome membrane

whereas NA allows the virus to be released from the host cell by cleavage of the sialic acid

Page 61: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

38

groups from glycoproteins (392). Due to the error prone polymerase, the influenza virus

continuously mutates, and viral variants with mutations in HA that abrogate recognition by

neutralizing antibodies are selected in a process termed antigenic drift. Antigenic drift results in

seasonal influenza viruses that require new vaccines. Antigenic shift, the process by which RNA

segments of two viruses reassort, however, can result in novel HA or NA subtypes to which

humans have little or no immunity, potentially resulting in a pandemic.

1.6.2 Innate immune responses

Influenza infection of mice is used as an animal model to study influenza pathogenesis. Influenza

A/HKx31 (X31), an H3N2 virus, and the more virulent A/PR8 (PR8), an H1N1 virus, are

commonly used strains in the laboratory. X31 elicits a mild infection that is cleared within 8

days, whereas PR8 infection results in a more severe disease outcome that can culminate in

death. X31 is a recombinant virus consisting of the H and N segments of the Hong Kong 1968

virus, with the other genes of the PR8 virus, thus the two viruses share common CD8 T cell

epitopes (393, 394). Animal models have shown that influenza infection of epithelial cells in the

respiratory tract is first detected by the innate arm of the immune system. Innate receptors such

as RIG-I and NLRP3 mediate cell-intrinsic recognition by detecting virus that is present within

the cytosol of infected cells. TLRs such as TLR3, which detects dsRNA in virus-infected cells,

and TLR7, which detects viral ssRNA that has been taken up into endosomes of infected cells

mediate cell-extrinsic recognition. Viral recognition leads to the secretion of type I IFNs, pro-

inflammatory cytokines and chemokines such as IL-1, IL-6 and TNF, eicosanoids, and the

upregulation of adhesion molecules required for leukocyte migration. Type I IFNs, produced by

macrophages, pneumocytes, DCs and pDCs, induce an anti-viral state through the expression of

IFN-stimulated genes (ISGs), such as MX proteins, IFITM proteins and PKR, in neighbouring

cells (395).

The release of chemokines leads to the recruitment of other cell types such as neutrophils,

monocytes, NK cells, and memory T cells (396). NK cells produce large amounts of IFNγ and

have been shown to kill influenza-infected epithelial cells. Mice lacking the activating NCR1

receptor on NK cells were shown to have a higher mortality rate following influenza infection

(184). Monocytes, neutrophils and alveolar macrophages help clear infected dead cells, which is

an important mechanism involved in viral clearance (397). Respiratory DCs (RDCs) are also

Page 62: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

39

major players in the innate immune response to influenza infection. Since influenza infection is

mainly restricted to the respiratory tract, it is the antigen bearing RDCs migrating to the LNs that

initiates adaptive immunity in lymphoid tissues. As early as 6 hours following infection, these

DCs upregulate costimulatory molecules, MHC-II, adhesion molecules, and CCR7, in order to

migrate to the LNs and activate influenza-specific CD4 and CD8 T cells (398-402). The CD103+

RDC subset has been identified as the main APC in the initial priming of T cells, after which the

CD11bhi

DCs continue presenting antigen in the LNs. The former subset of DCs efficiently

differentiates CD8 T cells that are associated with migration to the lung, whereas the latter subset

differentiates CD8 T cells that are associated with retention in the LNs (403, 404). DCs in the

lung have also been shown to be important in maintaining CD8 T cell survival in the lung (405).

1.6.3 Adaptive immune responses

Within 5-7 days pi, antigen-specific CD4 and CD8 T cells home to the lung. Depending on the

target cell encountered, CD8 T cells kill infected cells in a Fas/FasL or perforin/granzyme

dependent manner and/or produce inflammatory cytokines such as TNF and IFNγ (394).

Antigen-specific CD4 T cells are important in the production of anti-influenza antibodies by B

cells, but also produce anti-viral cytokines, can kill virally infected cells, and play a role in the

development of anti-influenza CD8 T cell responses (406-409).

Although the immune response generated against influenza infection is meant to clear the virus

and protect the host, it can also cause severe immunopathology leading to morbidity or mortality

of the host. Activated CD4 and CD8 T cells, as well as macrophages secreting nitric oxide and

oxygen radicals can mediate lung tissue damage (391, 410, 411). Therefore, as with LCMV

infection, the immune system must strike a balance – clear the virus, but spare the host.

Costimulatory members of the TNFR family, such as 4-1BB, GITR, OX40 and CD27, have been

shown to impact multiple stages of the immune response against influenza infection, including

the priming of antigen-specific T cells by DCs, effector and memory T cell proliferation,

survival, and effector function, as well as the development of memory and secondary response to

influenza (412-414). The TNFR family, and specifically, 4-1BB and 4-1BBL, will be further

discussed in the next section.

1.7 TNF/TNFR family

Page 63: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

40

1.7.1 Overview

TNF, the founding member of the TNF superfamily, earned its name following the initial

observations that it could mediate tumor regression (415). The TNF superfamily is now known

to include 30 receptors in the TNFR family and 19 ligands in the TNF family (416). The success

of TNF blockers such as Etanercept (Enbrel; Amgen/Pfizer) in several inflammatory conditions

has garnered much interest in harnessing the immune modifying capabilities of the other

members of this family for therapeutic benefit (417). Reagents that either stimulate or block the

activity of other TNF superfamily members are being evaluated in clinical trials, and a few have

been approved for human use to treat autoimmune/inflammatory conditions and cancer (417).

For example, a depleting anti-CD30 specific antibody has been approved for treatment of

Hodgkin’s lymphoma and anaplastic large-cell lymphoma and an antagonizing human BAFF-

specific antibody has been approved for treatment of SLE (417). TNFR family members are type

I transmembrane proteins, characterized by cysteine-rich extracellular domains, although

following cleavage, they can also exist in soluble form. TNF family members include type II

transmembrane proteins that are also found in both membrane-bound and soluble form, and

similar to their respective receptors, are thought to associate and mediate their effector functions

as trimers. It is the cytoplasmic portion of the TNFRs that broadly divide this family into two

groups: pro-survival and pro-death (415). Members that contain a death domain (DD), such as

Fas, associate with DD-containing proteins to signal apoptosis, whereas members without DDs,

such as 4-1BB, GITR, CD27 and OX40 can associate with TNFR associated factors (TRAFs) to

mediate survival signaling (413). Of course, this simple dichotomy is much more complex, in

that some pro-death members can mediate survival signals under certain conditions, and vice

versa. Here, I will discuss the TNFR family member 4-1BB and its ligand 4-1BBL, which are the

focus of chapter 2.

1.7.2 4-1BB/4-1BBL

Of the TNFR family, 4-1BB (CD137, TNFRSF9) is especially interesting due to the ability of

anti-4-1BB antibodies to potently enhance T cell responses in the context of cancer and viral

infections (418, 419), and induce immunoregulatory activity in models of autoimmunity and

inflammatory disease (413, 420). Although these antibodies have been associated with cell

toxicity (421-423), modifying the dose and timing of administration can reduce toxic side

Page 64: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

41

effects. Several antibodies to 4-1BB are currently being evaluated in clinical trials for cancer

(417, 424, 425).

4-1BB and its ligand, 4-1BBL, are largely pro-survival costimulatory members of the

TNFR/TNF family. Mouse 4-1BB, a type I transmembrane protein, was initially discovered and

cloned in 1989 by screening cDNA that was selectively expressed in activated T cells (426),

followed by the identification of mouse 4-1BBL, a type II transmembrane protein, by Goodwin’s

group in 1993 (427). Human 4-1BB and 4-1BBL were subsequently characterized by other

groups (428-430). The 4-1BB gene is located on chromosome 4 in mice and chromosome 1 in

humans, and the 4-1BBL gene on chromosome 17 in mice and chromosome 9 in humans.

Similar to other transmembrane proteins of the TNFR/TNF family, 4-1BB and 4-1BBL mediate

their activity by associating as trimers (431).

Much research activity has focused on the role of 4-1BB/4-1BBL on multiple subsets of T cells

in anti-viral and anti-tumor immunity, where this pathway has been shown to provide a CD28-

independent costimulatory signal, induce survival signaling via upregulation of survival

molecules such as Bcl-xL and Bfl-1 and downregulation of pro-apoptotic molecules such as BIM,

and enhance cell cycle progression and cytokine production (413, 414, 432, 433). 4-1BB is not

expressed on naive T cells, but NF-κB and MAPK signaling can upregulate 4-1BB expression on

both CD4 and CD8 T cells (413). 4-1BB expression on these cells is very rapid and transient in

vivo, but the duration of 4-1BB expression can be prolonged under conditions were antigen

persists (434-438). 4-1BBL is predominantly expressed on APCs, where it can be regulated by

CD40, IgM, and TLR signaling in vitro (439-441). 4-1BBL expression is also detectable on

hematopoietic progenitors cells and at sites of inflammation on cardiac myocytes, neurons, and

aortic tissue (432).

1.7.3 Role of 4-1BB/4-1BBL on T cells

The study of 4-1BB- and 4-1BBL-deficient mice and the use of agonistic anti-4-1BB antibodies

have highlighted the importance of this pathway in enhancing immunity against viral infections,

such as influenza, LCMV, VSV, VV, FV, MCMV, and MHV68 (413, 414, 432, 442). When 4-

1BBL-/-

mice were infected with influenza X31, it was shown that 4-1BBL was not required for

the primary influenza-specific CD8 T cell response, but for sustaining the number of memory

Page 65: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

42

CD8, but not CD4, T cells and the recall response to influenza (443). Recent data show that

GITR controls the antigen-independent expression of 4-1BB on memory CD8 T cells in the BM

and liver (444), and that 4-1BB-expressing memory CD8 T cells plausibly receive survival

signals from 4-1BBL expressing stromal cells in the BM (445).

In contrast to these studies, a role for 4-1BB/4-1BBL has been identified in the primary T cell

response against viral infection in other models (436, 446-448). Whether this pathway influences

the primary response seems to depend on the severity of infection and persistence of the virus.

For example, during mild respiratory influenza infection with X31, 4-1BB is only transiently

upregulated on lung CD8 T cells and is dispensable for the magnitude of the primary CD8 T cell

response and mouse survival. However, during severe respiratory influenza infection with the

PR8 strain, 4-1BB expression on lung CD8 T cells is sustained, and critical for CD8 T cell

accumulation in the lungs, viral clearance and mouse survival. In this model, 4-1BB acts to keep

the CD8 T cells alive long enough to clear the infection (436). When virus cannot be cleared,

such as in chronic infection with LCMV clone 13, the 4-1BB pathway in CD8 T cells becomes

desensitized due to loss of the signaling adapter TRAF1 (437).

CD4 T cell responses are also modulated by 4-1BB/4-1BBL signaling, however, most studies on

the role of 4-1BB/4-1BBL in anti-viral immunity have identified a role for this pathway

predominantly in CD8 T cells. Indeed, in some models, 4-1BB is induced at a higher level on

CD8 compared to CD4 T cells (449, 450). The role of endogenous 4-1BB/4-1BBL seems to be

mainly in enhancing CD8 T cell survival (436, 443, 451), however, 4-1BB stimulation has also

been shown to modulate T cell proliferation, cytokine production and effector function (413).

Tregs represent a double-edged sword in viral immunity, in that their activities can suppress host

immunity to viral infection and enable viral persistence; however, they can also be deemed

beneficial when dampening immune responses associated with immunopathology (452, 453). In

contrast to naive T cells, where 4-1BB is upregulated upon activation, 4-1BB is constitutively

expressed on Tregs and can be increased by TCR stimulation and IL-2 treatment (454-457).

Anti-4-1BB costimulation has been shown to render effector CD8 T cells resistant to virus-

induced Treg-mediated suppression (458); however, in some models, it is possible that the

effects of 4-1BB are directly on Tregs, as this pathway has been shown to modulate Treg

Page 66: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

43

expansion. However, there are conflicting data on whether 4-1BB on Tregs enhances or

abrogates their suppressive function (454-456, 459).

1.7.4 Role of 4-1BB/4-1BBL on non-T cells

The effects of 4-1BB and 4-1BBL in the aforementioned studies and in the vast majority of

studies have been attributed to the role this pathway plays in T cell biology, however 4-1BB is

broadly expressed on other cell types, including APCs (441, 460-465), NK/NKT cells (466-469),

hematopoietic progenitor cells (470), mast cells (471), and neutrophils (472, 473). How 4-1BB

expression on these cell types is integrated with 4-1BB expression on T cell subsets is not

understood. As this thesis explores the role of 4-1BB and 4-1BBL on DCs, these cells will be

discussed in more detail.

1.7.5 Role of 4-1BB/4-1BBL on DCs

Several stimuli, including LPS, anti-CD40, TNF, IFNγ, IL-12, Zymosan, and GM-CSF can

induce or upregulate 4-1BB expression on DCs either in vitro or ex vivo (441, 460-462), and

although endogenous 4-1BB appears to be dispensable for DC maturation (460), it has been

shown to modulate DC function. Anti-4-1BB treatment of DCs was shown to induce cytokine

(IL-6 and IL-12) production (441, 462), survival molecules (Bcl-xl and Bcl-2) (460, 464), and an

enhanced ability to stimulate T cell proliferation, independent of effects of T cells (462).

However, 4-1BB on DCs has also been suggested to suppress immunity. Anti-4-1BB treatment

at the beginning of LCMV Armstrong infection resulted in activation-induced cell death (AICD)

of CD8 T cells and immunosuppression, whereas delaying the treatment by 3 days enhanced

virus-specific CD8 T cell responses and mediated rapid viral clearance (474). In a follow-up

report, the deleterious effects of early anti-4-1BB treatment were argued to involve targeting of

both T cells and DCs. Cotransfer experiments of virus-specific T cells and other lineages into

WT and 4-1BB-/-

hosts followed by LCMV infection and anti-4-1BB treatment showed that only

cotransfer with DCs mediated AICD of virus-specific CD8 T cells. In vitro, anti-4-1BB

treatment of DCs was shown to mediate STAT3 activation, which has been shown to enhance the

ability of DCs to induce antigen-specific T cell tolerance. The authors proposed that early

signaling by 4-1BB on DCs might be involved in the early events that program T cell contraction

following viral infection (463). In another model supporting a regulatory role for 4-1BB on DCs,

Page 67: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

44

it was shown that anti-4-1BB treatment of mesenteric LN DCs induces RALDH activity and

promotes iTreg development (461).

The above studies reveal the discrepancy in the literature on whether 4-1BB signaling on DCs

enhances immune responses or downmodulates them. The effects of anti-4-1BB on DC activity

are probably model-dependent as timing of administration and cell location were different in the

above studies. It is also not clear what endogenous 4-1BB does on DCs, as the handful of studies

that have looked at the function of 4-1BB on DCs have mainly tested the effects of

supraphysiological signaling of 4-1BB by use of anti-4-1BB antibodies or 4-1BBL-

overexpressing cell lines. Moreover, the DC-intrinsic roles of 4-1BB/4-1BBL molecules during

an ongoing immune response in vivo are not known. In chapter 2 of this thesis, I explore the

endogenous function of 4-1BB and 4-1BBL on BM-derived DCs.

1.7.6 4-1BB signaling

The cytoplasmic tails of 4-1BB and other TNFRs do not possess intrinsic enzymatic activity, and

therefore associate both directly and indirectly with a family of adapter proteins, TRAFs, to

mediate intracellular signaling. This association is directly mediated via TRAF-interacting motifs

consisting of 4-6 amino acids in the cytoplasmic tail (475-477). Although initially identified

downstream of TNFR2 (478) and other members of the TNFR family, it is now well appreciated

that TRAFs effect multiple receptor signaling pathways, including TLRs, NLRs, and TCRs, and

that their aberrant regulation is associated with several human diseases (479). To date, there are 6

identified mammalian TRAFs: TRAF1-6. All members contain a conserved C-terminal TRAF

domain, which consists of an N-terminal coiled-coil region (TRAF-N) and a C-terminal-β-

sandwich (TRAF-C), involved in protein-protein interactions with receptors, TRAFs, and other

signaling molecules (479). Minor structural differences in the TRAF-C domain accounts for

binding to different receptors (480, 481), whereas the TRAF-N region is responsible for

homotypic and heterotypic interactions with other TRAF molecules (479). Apart from TRAF1,

all members also contain an N-terminal RING domain that can potentially mediate E3 ubiqutin

ligase activity (479).

Mouse 4-1BB recruits TRAFs 1 and 2, and mediates both classical and alternative NF-κB

activation, as well as extracellular signal-regulated kinase (ERK)/c-Jun N-terminal kinase

Page 68: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

45

(JNK)/p38 MAPK and delayed PI3K/AKT activation (Fig. 1.2) (414, 482-485). TRAF1 has been

shown to be required for ERK activation-dependent BIM downmodulation downstream of 4-

1BB in T cells (486). Recent results show that the F-actin binding protein LSP-1 is recruited to

the 4-1BB signalosome via TRAF1 and that in the absence of LSP-1, T cells fail to activate ERK

during 4-1BB signaling (487). 4-1BB can activate the alternative NF-κB pathway via TRAF3

degradation, with delayed kinetics compared to activation of the classical NF-κB pathway (485).

TRAF1 was found to restrain costimulation-independent activation of the alternative NF-κB

pathway in T cells, whereas it is essential for early activation of the classical NF-κB pathway and

survival signaling downstream of 4-1BB in T cells (485, 488). In resting T cells, the activation

of the alternative NF-kB pathway is restricted by the E3 ligase activity of cellular inhibitor of

apoptosis (cIAP) 1 and 2, which are brought into proximity with the alternative NF-κB activating

kinase, NIK, through association with TRAF2 (489, 490).

Of interest, Zheng et al. (491) have shown that a heterotrimer of two TRAF2 with one TRAF1

coiled coil region preferentially and asymetrically recruits a single cIAP BIR domain, compared

to a TRAF2 homotrimer. The TRAF1 (TRAF2)2 N domain heterotrimer more efficiently recruits

cIAPs, thus this in turn may allow TRAF2 to more efficiently recruit cIAPs to act as E3 ligases

for NIK degradation in anti-CD3-activated cells than a TRAF2 homotrimer. However, as TRAF1

has limited tissue distribution (492), it clearly isn’t required to restrict NIK in all cells. Thus,

McPherson and colleagues suggested that the fact that TCR stimulation increases the levels of

the NIK substrate, P100, in cells, may be the reason why TRAF1 is needed in addition to TRAF2

to efficiently recruit cIAP molecules (485). Alternatively, TRAF1 is also needed to protect

TRAF2 from degradation downstream of 4-1BB (486) and other TNFR family members (493,

494). Thus, the role of TRAF1 in preventing constitutive NIK activation could be due to it

stabilizing TRAF2 against degradation in activated T cells. Further work will be required to

resolve this issue. Interestingly, in the context of chronic viral infection in mouse and man,

TRAF1 is specifically lost from chronically stimulated PD-1hi

virus-specific CD8 T cells,

thereby desensitizing the 4-1BB pathway (437). TRAF1 degradation was shown to be mediated

at least in part by a TGFβ-dependent mechanism. This effect required protein synthesis

downstream of TGFβ and was sensitive to chloroquine, suggesting that a TGFβ-induced gene

promotes TRAF1 degradation in chronically stimulated T cells, perhaps via autophagy, although

this requires further study.

Page 69: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

46

This figure was modified and reproduced from (414)

Page 70: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

47

4-1BB has recently been shown to interact with Galectin-9, a member of the β-galactoside-

binding family of lectins, at sites in the CDR4 that are independent of the binding sites for anti-4-

1BB or 4-1BBL. This interaction, which is thought to involve 4-1BB and galectin 9 on the same

cell, may facilitate the aggreggation of 4-1BB monomers, dimers, or trimers when ligated by

anti-4-1BB or 4-1BBL, thus mediating productive signaling. In the absence of Galectin-9, the

stimulatory function of anti-4-1BB in in vivo models, and in the in vitro stimulation of T cells,

DCs, and NK cells is impaired (495).

1.8 Thesis Rationale

As outlined in this introductory chapter, innate immune responses orchestrated in secondary

lymphoid organs, such as the spleen, play a key role in modulating the outcome of viral

infection. My original goal was to study the role of 4-1BB and 4-1BBL on innate immune cells,

and how this impacts anti-viral immunity. Chapter 2 asked about the expression and function of

4-1BB/4-1BBL on DCs, and whether these molecules intrinsically modulate the ability of DCs to

generate influenza-specific T cell responses in the spleen. Chapter 3 emerged from a study of the

role of the 41BB signaling pathway during ARM and Cl13 infection, where it was noted that 60

days following Cl13 infection, the spleen remained small even weeks after LCMV was no longer

detectable in the spleen. I went on to analyze this process in-depth and uncovered a role for NK

cells in viral-induced splenic atrophy.

Page 71: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

48

Chapter 2

4-1BB and 4-1BBL constitutively interact on LPS-activated dendritic cells but

dendritic cell-intrinsic 4-1BB/4-1BBL interactions are dispensable for

cytokine production and anti-viral CD8 T cell priming

Mbanwi AN, Lin GH, Sabbagh L, Watts TH. 4-1BB and 4-1BBL constitutively interact on LPS-

activated dendritic cells but dendritic cell-intrinsic 4-1BB/4-1BBL interactions are dispensable

for cytokine production and anti-viral CD8 T cell priming.

Manuscript in preparation.

Author contributions

Mbanwi AN and THW designed the experiments and wrote the paper.

Mbanwi AN performed all the experiments.

Lin GH performed the initial experiments showing that the anti-4-1BBL antibody TKS-1 only

detects 4-1BBL in the absence of 4-1BB on LPS-activated bone marrow-derived DCs.

Sabbagh L gave technical advice on how to clone the pLJM1-4-1BBL-citrine construct and

transduce cell lines and DCs.

Page 72: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

49

2.1 Abstract

Stimulation of the TNFR family member 4-1BB has been shown to potently activate T cell

responses in models of viral infection and cancer. However, 4-1BB is widely expressed on other

cells of the immune system, including dendritic cells (DCs), which also express its ligand, 4-

1BBL. The precise nature of 4-1BB/4-1BBL expression on DCs is not known and the

endogenous function of 4-1BB/4-1BBL on this cell type has remained largely elusive. Here, we

show that LPS stimulation induced both 4-1BB and 4-1BBL expression on bone marrow-derived

DCs, and that these molecules constitutively interact on LPS-activated DCs. However, this

interaction was shown to be dispensable for DC survival, the upregulation of key costimulatory

molecules, cytokine/chemokine production and the ability to stimulate T cell proliferation in

vitro following LPS stimulation. Using an in vivo DC vaccination approach, we further show that

DC-intrinsic 4-1BB and 4-1BBL do not modulate the ability of DCs to induce clonal expansion

of influenza-specific CD8 T cells. The finding of constitutive interaction between 4-1BB and 4-

1BBL on DCs suggests a role for these molecules in regulating DC function; however, the

precise nature of this regulation remains to be elucidated.

2.2 Introduction

The tumor necrosis factor receptor (TNFR) family member 4-1BB was originally identified as an

inducible molecule on activated T cells (426, 496). Its TNF family member ligand, 4-1BBL, is

mainly expressed on activated antigen-presenting cells (APCs) (427, 441), but has also been

detected on hematopoietic progenitor cells (470, 497, 498). 4-1BB signaling in vitro has been

shown to enhance both CD4 and CD8 T cell activation (413, 499), although in vivo, the pro-

survival effects of 4-1BB are more prominent on CD8 compared to CD4 T cells (443, 451, 486,

500). The anti-tumor and anti-viral effects of 4-1BB agonistic antibodies and 4-1BBL vectors

have been largely attributed to stimulation of 4-1BB on T cells (445, 501, 502), however,

functional 4-1BB is expressed on other cells of the immune system, including DCs (441, 460-

464), NK cells (467, 468), NKT cells (466, 469), monocytes (429), neutrophils (472, 473), B

cells (465) and mast cells (471), although the direct effects of 4-1BB on non-T cells in vivo has

not been rigorously studied.

Page 73: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

50

DCs are key sentinels of the immune system that orchestrate the T cell response via their ability

to present antigens and costimulatory molecules to T cells, as well as by their production of

chemokines and cytokines (50, 503). Several TNFRs including CD40, RANK and LTβR have

been shown to play a role in DC biology (504). 4-1BB has also been reported on DCs (441, 460-

462), yet its activity on this cell type has not been extensively studied. In addition to 4-1BB, 4-

1BBL has been reported to be induced on activated DCs (441), raising the question of whether

intrinsic 4-1BB/4-1BBL interactions regulate DC function. In this report, we investigate the

expression of 4-1BB and 4-1BBL on bone marrow-derived DCs and provide evidence that 4-

1BB and its ligand can constitutively interact on DCs. However, despite the evidence of a

constitutive interaction, we find that 4-1BB/4-1BBL interactions are not required for LPS-

induced upregulation of costimulatory molecules, DC survival, cytokine/chemokine production,

nor the ability of DCs to stimulate T cell proliferation in vitro. Using an in vivo DC vaccination

approach, we also show that DC-intrinsic 4-1BB and 4-1BBL are dispensable for the primary

clonal expansion of influenza-specific CD8 T cells.

2.3 Materials and Methods

2.3.1 Mice

C57BL/6 wild-type (WT) mice were purchased from Charles River Laboratories (Saint-

Constant, QC, Canada). CD45.1 congenic mice and OT-I TCR transgenic mice were obtained

from The Jackson Laboratory (Bar Harbor, ME, USA). 4-1BB-/-

mice extensively backcrossed to

the C57BL/6 background were bred in our facility. These mice were previously provided to us

by Dr. Byoung S. Kwon (National Cancer Center, Ilsan, Korea). 4-1BBL-/-

mice were originally

obtained under a materials transfer agreement from Immunex (Amgen, Thousand Oaks, CA,

USA) and further backcrossed to the C57BL/6 background in our facility. 4-1BB-/-

and 4-1BBL-/-

mice were crossed to generate 4-1BB-/-

4-1BBL-/-

(DKO) mice. Heterozygotes from a C57BL/6

and 4-1BBL-/-

backcross were further crossed to generate WT and 4-1BBL-/-

littermate controls.

Littermate controls were selected either from the same mother or sisters from the same harem

cage. Littermate controls were used when indicated in the figure legends. Mice were maintained

under specific pathogen-free conditions in sterile microisolators at the University of Toronto. All

Page 74: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

51

mouse experiments were approved by the University of Toronto animal care committee in

accordance with the regulations of the Canadian Council on Animal Care.

2.3.2 DC cultures and surface markers

Femurs and tibias were removed from mice (WT, 4-1BB-/-

, 4-1BBL-/-

, and DKO) and bone

marrow cells were harvested by flushing with PBS. Bone marrow cells were seeded at 5 × 105

cells/ml in 100 mm bacteriological Petri dishes. RPMI-1640 media supplemented with 10% heat-

inactivated fetal calf serum, 1X NEAA, 1X GPPS, 50 μM 2-mercaptoethanol, and 40 ng/ml GM-

CSF was used to culture DCs. Fresh media was added on day 3, and on day 6, one half of the old

media was replaced with new media. Nonadherent cells were harvested on days 7-10 for

analysis. DC cultures were always used at a final concentration of 1 × 106

cells/ml. To activate

DCs, immature DCs were stimulated with either 10 ng/ml or 1 μg/ml LPS (026:B6; Sigma-

Aldrich) in 24-well plates or 15 ml round-bottom polypropylene tubes.

Prior to detection of surface markers, WT, 4-1BB-/-

, 4-1BBL-/-

and DKO DCs were treated with

anti-CD16/CD32 Fc block (clone 93) for 15 minutes on ice to prevent antibodies from binding to

the Fc receptors. DCs were then surface stained with a viability dye [LIVE/DEAD Violet

Viability/Vitality Kit (Life Technologies)], and the following antibodies: biotinylated anti-MHC-

I (clone AF6-88.5.5.3), MHC-II-FITC (clone Y3P), CD11c-APC (clone N418), biotinylated

CD80 (clone 16-10A1), biotinylated CD86 (clone GL1), biotinylated CD40 (clone 1C10),

CD45.1-FITC (clone A20), CD45.2-FITC (clone 104), biotinylated 4-1BB (clone 3H3),

biotinylated 4-1BBL (clone TKS-1), biotinylated 4-1BBL (clone 19H3), streptavidin-PE, and

streptavidin-Alexa Fluor 488. Antibodies were either purchased from eBioscience (MHC-I,

CD11c, CD45.1, CD45.2, streptavidin-PE, streptavidin-Alexa Fluor 488) or purified from

hybridomas in our laboratory (CD80, CD86, CD40, MHC-II, 4-1BB, 4-1BBL). Fluorescence

minus one (FMO) controls, 4-1BB-/-

and 4-1BBL-/-

mice were used as staining controls. Surface

marker expression was assessed by flow cytometry.

2.3.3 Blocking studies

293T cells overexpressing 4-1BBL (pLJM1-4-1BBL-citrine) or LPS-activated 4-1BB-/-

DCs

were pre-treated with either PBS, 10 μg/ml 4-1BB-alkaline phosphatase (4-1BB-AP) (439), 10

μg/ml anti-4-1BBL (clone TKS-1), 10 μg/ml anti-4-1BBL (clone 19H3), 10 μg/ml AP, or 10

Page 75: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

52

μg/ml Rat IgG for 10 minutes on ice. Cells were then surface stained with anti-CD11c-APC

and/or either biotinylated anti-4-1BBL (clone TKS-1) or biotinylated anti-4-1BBL (clone 19H3)

followed by secondary streptavidin-PE. FMO controls and/or 4-1BBL-/-

DCs were used as

staining controls. Surface marker expression was assessed by flow cytometry.

2.3.4 DC survival assay

Immature DCs (WT and DKO) were stimulated with 1 μg/ml LPS and their survival rate was

tracked on day 0 prior to LPS treatment and every day for 5 days following LPS treatment. DCs

were surface stained with anti-CD16/CD32 Fc block, a viability dye and antibodies to CD11c

and MHC-II, as described above. Following surface staining, DCs were fixed and permeabilized

using Cytofix/Cytoperm (BD Biosciences), and stained intracellularly with anti-Bcl-xL-Alexa

Fluor

488 (clone 54H6; Cell Signaling Technology) and anti-Bcl-2-FITC (clone 10C4;

eBioscience). DCs were then analyzed by flow cytometry.

2.3.5 Cytokine production

Supernatants were first collected from immature DCs (WT and DKO/4-1BBL-/-

) and stored at -

80°C. These DCs were then stimulated with 1 μg/ml LPS. Supernatants were collected 7, 24, 48

and 72 hours following LPS treatment and stored at -80°C. Thawed supernatants were

appropriately diluted and analyzed for the presence of mouse IL-6, IL-12, IL-18, MCP-1 and

RANTES using a custom FlowCytomix multiplex kit (eBioscience) as per the manufacturer’s

instructions. The presence of IL-2 and TNF in the thawed supernatants was determined using

ELISA kits (mouse TNF-α and mouse IL-2 ELISA Ready-SET-Go!) as per the manufacturer’s

instructions.

2.3.6 In vitro antigen uptake and presentation

Immature DCs were treated with 10, 25, 50 or 100 MOI of replication defective adenovirus 5

(Ad)-SIINFEKL-GFP [a gift from Dr. Jonathan Bramson (McMaster University, Hamilton, ON,

Canada)], or 1, 10, or 50 MOI of Ad-OVA, or various concentrations of SIINFEKL peptide and

stimulated with 1 μg/ml LPS. Adenoviruses were added to DC cultures in 15 ml round-bottom

polypropylene tubes. Tubes were then centrifuged at 2200 rpm at 37°C for 2 hours. After 24 and

48 hours, GFP-transduced DCs were washed and stained with a viability dye and antibodies to

Page 76: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

53

CD11c and MHC-II and analyzed by flow cytometry. Ad-OVA transduced DCs were washed

and stained with a viability dye and antibodies to CD11c, MHC-II and SIINFEKL-H-2Kb (clone

25-D1.16; eBioscience) or mixed with CFSE-labelled OT-I CD8 T cells at a ratio of 104 DCs to

105

OT-I CD8 T cells. SIINFEKL-loaded DCs were also cocultured with CFSE-labelled OT-I

CD8 T cells. CFSE dilution 3-4 days following DC-T coculture was analyzed by flow cytometry.

To generate CFSE-labelled T cells, CD8 T cells were first purified from spleens of naive OT-I

mice using a negative selection mouse CD8 T cell enrichment kit (STEMCELL Technologies).

Purified CD8 T cells were then stained with 1 μM CFSE for 10 minutes at 37°C and washed to

remove excess CFSE.

2.3.7 DC vaccination

Ad-NP expresses influenza NP in the E1 region and was kindly provided by Dr. Jonathan

Bramson at McMaster University, Hamilton, ON, Canada. The NP was derived from influenza

A/PR8 GenBank: J02147.1. 50 MOI of Ad-NP was added to immature DCs (WT and 4-1BBL-/-

littermate controls, and 4-1BB-/-

) in 15 ml round bottom polypropylene tubes. Tubes were

centrifuged at 2200 rpm at 37°C for 2 hours. The next day, 1 μg/ml LPS was added to the

cultures. 24 hours following LPS treatment, DCs were washed three times with PBS, and

resuspended at a concentration of 10 × 106 cells/ml in PBS. 6-week-old recipient mice (DKO)

were intravenously injected with 1.5 × 106

DCs/mouse. On day 7 post DC vaccination, mice

were euthanized and their spleens were harvested. Splenocytes were surface stained with a

viability dye, anti-CD8-PE (clone 53-6.7; eBioscience), and Db/NP366-74 tetramers [obtained from

the National Institute for Allergy and Infectious Diseases tetramer facility (Emory University,

Atlanta, GA, USA)].

For intracellular staining, splenocytes were restimulated with 1 μM NP366-74 peptide for 6 hours

with Golgi Stop (BD Biosciences) at 37°C. For detection of degranulation, 5 μg/ml of anti-

CD107a (clone 1D4B; BD Biosciences) was also added at the beginning of the restimulation

culture. Cells were then surface stained with a viability dye and anti-CD8-PE. Following surface

staining, cells were fixed and permeabilized in Cytofix/Cytoperm solution (BD Biosciences) and

stained intracellularly with anti-IFNγ-PE-Cy7 (clone XMG1.2; eBioscience) and anti-TNF-APC

(clone MP6-XT22; BD Biosciences). FMO controls, unstimulated (no peptide), and naive

Page 77: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

54

(unvaccinated mice) samples were used as staining controls. Samples were then analyzed by

flow cytometry.

2.3.8 Flow cytometry

Samples were analyzed using a FACSCalibur with CellQuest software or FACSCanto II with

FACSDiva acquisition software (BD Biosciences), and FlowJo software (Tree Star, Inc.).

2.3.9 Lentiviral transductions and confocal microscopy

A lentiviral vector (pLJM1) expressing 4-1BBL-citrine fusion protein or the empty vector, along

with a packaging plasmid, was first transfected into HEK293T cells using FuGENE6 (Promega).

48 hours following transfection, viral supernatants from the HEK293T cultures were used to

transduce WT and 4-1BB-/-

DCs on day 7 of DC culture. DC cultures with added viral

supernatants were spun at 2200 rpm at 37°C for 2 hours. Virally transduced DC cultures were

further stimulated with 1 μg/ml LPS, washed and visualized by confocal microscopy 24 hours

later. Cells were visualized in cell culture plates using an inverted Zeiss LSM510 confocal

microscope.

2.3.10 Statistical Analysis

All statistical analyses were performed using Graphpad software (Prism). For comparison of two

groups, p values were obtained using the Student’s t test (unpaired, two tailed, 95% confidence

interval).

2.4 Results

2.4.1 Chacterization of DCs lacking 4-1BB and 4-1BBL

To investigate the expression and function of 4-1BB and its ligand on DCs, we generated DCs

from bone marrow cells by culturing them in the presence of GM-CSF for 7-10 days (505).

Immature DCs derived from either WT, 4-1BB-/-

, or 4-1BBL-/-

bone marrow cells were identified

by CD11c/MHC-II double staining (Fig. 2.1A), and were found to express similar levels of

MHC-I and the costimulatory molecules CD80, CD86, and CD40 (Fig. 2.1B, top panel).

Page 78: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

55

Page 79: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

56

Following stimulation with 1 μg/ml LPS for 24 hours, WT, 4-1BB-/-

, and 4-1BBL-/-

DCs

similarly upregulated the expression of these molecules (Fig. 2.1B, bottom panel), suggesting

that 4-1BB and 4-1BBL are dispensable for LPS-induced upregulation of key DC surface

markers involved in APC function.

2.4.2 4-1BBL is not readily detectable in the presence of 4-1BB on LPS-activated DCs

We next investigated the expression of 4-1BB and 4-1BBL on LPS-activated bone marrow-

derived DCs. 4-1BB and 4-1BBL were not readily detectable on immature DCs (Fig. 2.2A), but

were both upregulated with LPS stimulation (Fig. 2.2B). 4-1BB was upregulated on WT DCs,

and in the absence of 4-1BBL on 4-1BBL-/-

DCs. However, interestingly, 4-1BBL was only

detectable in the absence of its receptor on 4-1BB-/-

DCs, and not on WT DCs. Thus, LPS

stimulation of DCs upregulates both 4-1BB and 4-1BBL, however, 4-1BBL is only detectable in

the absence of 4-1BB, suggesting that 4-1BB either downregulates 4-1BBL or masks its

detection.

2.4.3 The surface expression of 4-1BBL can be regulated extrinsically

Since 4-1BB and 4-1BBL can be expressed on the same cell type, it is possible that 4-1BB on the

same cell or on an adjacent cell regulates the expression/detection of 4-1BBL. To address

potential extrinsic regulation, immature CD45.2 4-1BB-/-

DCs were mixed with CD45.1 WT DCs

and stimulated with LPS for 24 hours. In this scenario, 4-1BBL expression on 4-1BB-/-

DCs can

only be regulated cell-extrinsically by 4-1BB on WT DCs. We found that 4-1BBL was readily

detectable on mature 4-1BB-/-

DC cultures in isolation (Fig. 2.3A, left panel), however, in the

mixed WT/4-1BB-/-

DC cultures, 4-1BBL was no longer detectable on the 4-1BB-/-

DCs (Fig.

2.3A, right panel). These data demonstrate that expression of 4-1BBL can be regulated or

masked in trans by 4-1BB on another cell. To confirm extrinsic regulation, we also added a

soluble fusion protein of 4-1BB (4-1BB-alkaline phosphatase, 4-1BB-AP) (439) to the mature 4-

1BBL-expressing 4-1BB-/-

DC cultures for 24 hours. Following addition of 4-1BB-AP, but not

AP alone, to the mature 4-1BB-/-

DC cultures, 4-1BBL was no longer detectable (Fig. 2.3B),

thereby providing further evidence that 4-1BB downregulates 4-1BBL or masks its detection.

Page 80: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

57

Page 81: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

58

Page 82: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

59

2.4.4 Evidence of constitutive interaction between 4-1BB and 4-1BBL on DCs

In order to investigate the trafficking of 4-1BBL on DCs, we generated fluorescent fusion

proteins of 4-1BBL that were transduced into cell lines and DCs. Microscopy studies identified

mainly surface expression of 4-1BBL fusion proteins, even in the presence of 4-1BB (Fig. 2.4),

prompting us to further investigate whether 4-1BBL surface detection was masked in the

presence of 4-1BB. To test this, we conducted blocking experiments in both cell lines and DCs.

293T cells overexpressing 4-1BBL were pre-treated with either 4-1BB-AP, anti-4-1BBL (clone

TKS-1, used in figures 2.2-2.3), anti-4-1BBL (clone 19H3), Rat IgG, or PBS for 10 minutes on

ice to prevent potential signal-induced downregulation of 4-1BBL. The cells were then

separately stained with the 19H3 and TKS-1 anti-4-1BBL antibodies. When cells had been pre-

treated with either PBS or Rat IgG, 4-1BBL was similarly detectable by the two antibodies (Fig.

2.5A). However, in the presence of 4-1BB-AP (which binds to 4-1BBL), the level of 4-1BBL

detected by TKS-1 was significantly reduced, whereas 19H3 binding to 4-1BBL was

independent of the presence of receptor (Fig. 2.5A). These findings demonstrate that the TKS-1

binding site is masked by receptor binding, arguing that they bind at similar or overlapping sites.

On the other hand, the 19H3 epitope on 4-1BBL is at a distinct site from the binding site of 4-

1BB or TKS-1. Consistently, pre-treatment with 19H3 affected 19H3 binding, but not TKS-1

binding. However, pre-treatment with TKS-1 slightly affected 19H3 binding (Fig. 2.5A). Similar

results were obtained when LPS-treated 4-1BBL-expressing 4-1BB-/-

DCs were used. Once

again, in the presence of 4-1BB-AP, ligand was detectable by 19H3 but not by TKS-1 (Fig.

2.5B). The finding that the presence of 4-1BB prevents TKS-1 binding, but still allows 19H3

binding, argues that 4-1BBL is present on LPS-activated DCs in the presence of 4-1BB, but its

binding site for TKS-1 is masked. This leads to the conclusion that 4-1BB and 4-1BBL are

constitutively interacting on the DCs.

To further test this hypothesis, we examined 4-1BBL expression on LPS-activated WT, 4-1BB-/-

,

4-1BBL-/-

, and DKO DCs using both TKS-1 and 19H3 antibodies. 4-1BBL expression was

detected by 19H3 but not by TKS-1 on LPS-activated WT DCs (Fig. 2.5C), consistent with the

hypothesis that 4-1BBL is expressed on the surface of LPS-activated WT DCs, but undetectable

by TKS-1, because it is bound to 4-1BB. Taken together, these data provide evidence of

constitutive interaction between 4-1BB and 4-1BBL on LPS-activated DCs in vitro.

Page 83: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

60

Page 84: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

61

Page 85: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

62

Page 86: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

63

2.4.5 Indistinguishable in vitro survival of WT and DKO DCs

Given the finding of constitutive interaction between 4-1BB and 4-1BBL on LPS-activated DCs,

we next investigated the biological relevance of this interaction. 4-1BB plays a key survival role

on T cells (432, 433, 500) and there is also evidence that it plays a similar role on DCs (460,

464). To test the effect of constitutive 4-1BB/4-1BBL on the survival of LPS-activated DCs in

vitro, we monitored cell viability by live/dead stain, as well as by intracellular expression of the

pro-survival molecules Bcl-xL and Bcl-2 over a period of 5 days. There was no statistically

significant difference in the survival rate of WT and DKO DCs (Fig. 2.6A), arguing against a

key role for 4-1BB/4-1BBL in the in vitro survival of DCs following LPS stimulation. There was

a trend toward lower MFI of Bcl-xL in the DKO compared to WT DCs at each time point (Fig.

2.6B), as well as for Bcl-2 between 48 and 72 hours (Fig. 2.6C). However, these changes in MFI

did not affect overall DC survival, as WT and DKO DCs died at a similar rate.

2.4.6 4-1BB and/or 4-1BBL do not impact the level of the cytokines IL-2, IL-6, IL-12, IL-18,

and TNF, nor the chemokines MCP-1 and RANTES in LPS-activated DCs

Following stimulation with foreign antigen, DCs produce cytokines and chemokines that

orchestrate the inflammatory response. To ask whether 4-1BB and 4-1BBL contribute to pro-

inflammatory cytokine/chemokine production by DCs, we measured LPS-induced production of

five pro-inflammatory cytokines and two chemokines in WT and DKO/4-1BBL-/-

DCs. The

production of IL-2, IL-6, TNF, MCP-1, and RANTES were similarly upregulated with LPS

stimulation between WT and KO DCs (Fig. 2.7A, B). There was more variability in the

production of IL-12 and IL-18 between WT and DKO DCs, but no significant differences at any

of the time points assessed (Fig. 2.7A). Overall, these data demonstrate that 4-1BB and/or 4-

1BBL are dispensable for IL-2, IL-6, IL-12, IL-18, MCP-1, RANTES and TNF production by

LPS-activated DCs.

2.4.7 4-1BB and 4-1BBL are dispensable for antigen presentation by DCs in vitro

Arguably, one of the defining features of DCs is their ability to present antigen and stimulate

naive T cells, thus we sought to determine whether this key function of DCs is modulated by 4-

1BB/4-1BBL interaction. To ensure a high efficiency of antigen delivery, we delivered antigen

Page 87: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

64

Page 88: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

65

Page 89: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

66

to the DCs using replication defective recombinant adenoviruses. To determine if 4-1BB/4-

1BBL could affect antigen uptake, WT and DKO DCs were transduced with different doses of

Ad-SIINFEKL-GFP and stimulated with LPS for 24 and 48 hours. The level of GFP expression

between the two groups was similar (Fig. 2.8A), suggesting that any observed differences in

antigen presentation were not due to differences in adenovirus-mediated antigen delivery. In

order to assay the capacity of the DCs to both process and present antigen, we next transduced

WT and DKO DCs with different doses of Ad-OVA for 24 and 48 hours in the presence of LPS,

and then looked at the expression of SIINFEKL peptide (OVA257-264) bound to MHC-I using an

MHC-restricted antibody. We did not find a statistically significant difference in the percentage

or MFI of SIINFEKL-MHC-I molecules as detected by anti-H-2Kb-SIINFEKL staining (Fig.

2.8B), suggesting that 4-1BB and 4-1BBL are dispensable for antigen processing by the DCs.

Next, we tested the ability of these cells to stimulate T cell responses. LPS-activated Ad-OVA

transduced DCs or SIINFEKL peptide-loaded DCs were mixed with CFSE-labelled OT-I CD8 T

cells at a 1:10 ratio of DCs to T cells. We tested two doses of both Ad-OVA and SIINFEKL

peptide at two time points (days 3 and 4), and found no striking difference in the CFSE dilution

profile of the T cells (Fig. 2.8C). We conducted similar experiments with OT-II cells to look at

CD4 T cell proliferation induced by DCs, and found no statistically significant difference

between WT and DKO DCs (data not shown). These findings demonstrate that the ability of DCs

to stimulate T cell proliferation in vitro is unaffected by the absence of 4-1BB/4-1BBL

interactions.

2.4.8 DC-intrinsic 4-1BB and 4-1BBL are largely dispensable for T cell priming against a

viral antigen in vivo

Although 4-1BB and 4-1BBL were dispensable on DCs for their in vitro activation of TCR

transgenic cells, it was conceivable that 4-1BB/4-1BBL had other effects in vivo and/or that

endogenous T cell responses would be more sensitive to the effects of 4-1BB/4-1BBL on DCs.

Therefore, we next investigated whether 4-1BB and 4-1BBL modulated DC immunogenicity in

vivo. To test this, in vitro generated WT, 4-1BB-/-

, and 4-1BBL-/-

DCs generated from bone

marrow of 4-1BB-/-

, and WT and 4-1BBL-/-

littermates, were modified with influenza NP (Ad-

NP) and used to prime mice (Fig. 2.9A). Of note, we titrated the dose of DCs, such that the

Page 90: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

67

Page 91: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

68

Page 92: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

69

Page 93: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

70

Page 94: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

71

induced response to WT DCs was around 0.5% NP-specific CD8 T cells by day 7. As the natural

influenza infection produces a more than 10x higher response, this means that we are in a range

of response that is submaximal and should be sensitive to changes in the quality of the DCs. CD8

T cell responses were monitored, based on the previous evidence that the 4-1BB/4-1BBL

costimulatory pathway modulates the CD8 T cell response following influenza infection (436,

443). Moreover, previous studies have shown that addition of 4-1BB/4-1BBL signals during

priming enhances the NP-specific CD8 T cell response to influenza virus (502, 506, 507). In

order to deduce the DC-intrinsic effect of 4-1BB/4-1BBL, we used DKO hosts. At 7 days post

DC vaccination, we found no statistically significant difference in the proportions or numbers of

NP-tetramer-specific CD8 T cells (Fig. 2.9B), and we observed similar production of IFNγ (Fig.

2.9C) and TNF (data not shown), and expression of CD107a (data not shown) following re-

stimulation with NP peptide ex vivo. Taken together, these data show that 4-1BB and 4-1BBL

interact constitutively on DCs, but this interaction is dispensable for primary clonal expansion of

influenza-specific CD8 T cells.

Page 95: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

72

2.5 Discussion

The activity of 4-1BB and 4-1BBL on DCs is poorly understood. Here we show that LPS-

activated DCs express both 4-1BB and 4-1BBL, and that these molecules constitutively interact

on this cell type. However, this interaction is dispensable for upregulation of key surface

molecules involved in APC function (MHC-I, CD80, CD86, CD40), survival,

cytokine/chemokine production (IL-2, IL-6, IL-12, IL-18, TNF, MCP-1, RANTES), in vitro

antigen processing and presentation to T cells, as well as in vivo priming against a viral antigen.

In the present study 4-1BBL was not readily detectable on LPS-activated WT DCs when using

the antibody TKS-1. 4-1BBL, however, was detected at high levels on LPS-activated 4-1BB-/-

DCs, suggesting that 4-1BB regulates the expression or detection of surface 4-1BBL. 4-1BBL

can be cleaved from the cell surface of leukocytes, and this cleavage is blocked by

metalloproteinase inhibitors in vitro (508). Enhanced levels of soluble 4-1BBL are found in the

sera of patients with leukemia and multiple sclerosis when compared to healthy controls (508,

509). Thus it was conceivable that 4-1BB could regulate 4-1BBL cleavage or could lead to

receptor-induced downregulation. Indeed, CD27 binding to CD70 results in reverse signaling

leading to downregulation of CD70 protein and mRNA levels (510). However, in the present

study, the major effect of 4-1BB on 4-1BBL was to block access of the antibody TKS-1. 4-1BBL

was still detected by the 19H3 antibody, arguing that 4-1BBL is present on the cell surface, but

that the TKS-1 epitope on 4-1BBL is masked by binding of its receptor.

The finding that 4-1BB and 4-1BBL interact constitutively on DCs does not distinguish whether

this interaction is between receptor and ligand on the same cells (in cis) or neighbouring cells (in

trans). 4-1BBL could be detected on 4-1BB-/-

cells but not WT cells within 3 hours following

LPS stimulation (data not shown) - suggesting a rapid interaction. Thus it is possible that 4-

1BB/4-1BBL interact within the same cell prior to surface exposure. Several studies have shown

that 4-1BB on T cells receives costimulatory signals (414), although the cell types that provide 4-

1BBL for these interactions have not been clearly delineated. If 4-1BB/4-1BBL interactions are

in trans, as was shown to occur in mixed DC cultures in the present study, then it is likely that

during DC-T cell interaction, that 4-1BBL on DCs could interact predominantly with 4-1BB on

T cells. However, if the interaction were cell-intrinsic, this might function to limit the 4-1BBL

signal available to the T cells. A previous study showed that 4-1BB and 4-1BBL transduced into

Page 96: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

73

Jurkat T cells can interact in cis to activate NF-κB in a single cell assay (511). Using coculture

systems, the same study also showed that these molecules can interact in trans (511). Thus, 4-

1BB/4-1BBL activity on DCs could be mediated both within the same cell and between cells.

A key question raised by the finding of constitutive 4-1BB/4-1BBL interaction on DCs is how

this interaction modulates DC function. A previous study that used 4-1BB-/-

DCs stimulated with

LPS reported a pro-survival function of 4-1BB on DCs. Choi et al. showed that in the absence of

4-1BB, DCs had lower expression of Bcl-2 and Bcl-xL and a lower survival rate in vitro.

However, the differences in the survival rate between WT and 4-1BB-/-

DCs were quite marginal

(half-life of 36 vs. 24 hours), and only significantly increased with agonist anti-4-1BB

stimulation (460). In contrast, we find no striking effects of deficiency of 4-1BB or 4-1BBL on

the survival rate of DCs in vitro. Supraphysiological stimulation of 4-1BB on DCs, either by

anti-4-1BB antibody or in the presence of 4-1BBL overexpressing cell lines, has also shown a

role for this receptor in modulating CD80/CD86 expression, production of the cytokines IL-6,

IL-12 and TNF, and the ability to enhance in vitro T cell proliferation, independent of effects of

T cells (441, 462, 463). We show in this report that in contrast to their activity in response to

supraphysiological stimulation, endogenous 4-1BB and 4-1BBL do not modulate the ability of

DCs to mediate these functions, at least in the context of LPS-activated DCs. It is unlikely that

the concentrations of LPS used to stimulate DCs in the present study (0.01-1 μg/ml) were

suboptimal to see an effect of 4-1BB/4-1BBL on the DCs as previous studies that found an effect

of 4-1BB on DC survival and cytokine production used higher concentrations of LPS to

stimulate DCs [1-10μg/ml (462) and 5μg/ml (460)].

As antigen presentation and the ability to stimulate naive T cell expansion are hallmarks of DC

function, we further investigated whether DC-expressed 4-1BB and 4-1BBL contribute to this

function by using an in vivo model. We designed a DC vaccination experiment, in which

influenza NP was expressed in WT or 4-1BB/4-1BBL-deficient DCs, which were then used to

vaccinate mice lacking both 4-1BB and 4-1BBL. DKO mice were chosen as recipients in order

to assess the effects of 4-1BB/4-1BBL interaction only on the transferred DCs. We selected a

dose of antigen and DCs that induced an NP-specific CD8 T cell population that constituted

about 0.5% of CD8 T cells by day 7 following DC vaccination. This immunization dose was

chosen deliberately to be suboptimal such that it would be sensitive to effects of 4-1BB/4-1BBL

Page 97: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

74

on outcome. However, under these conditions we found no significant role for 4-1BB/4-1BBL

interaction on the DCs in modulating the clonal expansion of influenza-specific CD8 T cells.

The influenza model was of interest as we have previously shown that incorporation of 4-1BBL

into an adenovirus vector containing influenza NP greatly augments the CD8 T cell response to

influenza (502). Bone marrow chimeras were used to show that these effects were largely due to

effects of 4-1BBL on T cells, although there was a small effect of 4-1BBL on non-T cells in this

model (502). The finding that we do not see an effect of 4-1BBL in the DC vaccination model

may again be due to use of endogenous 4-1BBL in the present study. Another caveat to this

vaccination model is that DCs used for vaccination may die rapidly and the contents cross-

presented via secondary DCs, in which case, 4-1BBL may not be relevant on the immunizing

DCs.

It should be noted that there is evidence in the literature that 4-1BB/4-1BBL interaction on

developing myeloid progenitor cells limits steady state myelopoiesis and differentiation into

CD11c+ DC lineages (470). The present study compared the numbers of CD11c-expressing cells

generated from WT and 4-1BBL-/-

littermate bone marrow derived DC cultures (data not shown),

but did not identify a role for these molecules in limiting DC differentiation. It is possible that

our DC culture conditions were not sensitive enough to observe these differences, as we

developed progenitor cells in the presence of 40 ng/ml of GM-CSF, whereas the aforementioned

study used 10 ng/ml of GM-CSF (470). However, it is not clear in (470) whether appropriate

littermate controls were used to rule out effects of incomplete backcrossing and the microflora,

which we did control for in the present study. Anti-4-1BB stimulation of DCs has also been

suggested to suppress T cell immunity. In an oral tolerance model, anti-4-1BB stimulation of

GALT DCs was suggested to promote the development of iTreg (461). Furthermore, early

administration of anti-4-1BB in the context of LCMV Armstrong infection was shown to target

both DCs and T cells, and result in AICD of CD8 T cells (463). It is possible that the 4-1BB

pathway in DCs augments the ability of DCs to induce Tregs or mediate T cell apoptosis;

however, we did not see an effect of endogenous DC-intrinsic 4-1BB in modulating CD8 T cell

responses in a very sensitive in vivo model. These studies (461, 463) investigated

supraphysiological stimulation of 4-1BB on DCs, which we have shown here does not

necessarily compare with the endogenous function of this molecule.

Page 98: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

75

Overall, this study provides evidence of constitutive interaction between 4-1BB and 4-1BBL on

LPS-activated DCs. Although we have not identified a physiological role for this interaction, the

finding that this interaction is so complete in DC cultures as to fully mask 4-1BBL when probed

with antibodies that target the receptor binding site, makes it hard to believe it does not play a

role in DC biology. These data highlight the need for further investigation into the role of

endogenous 4-1BB/4-1BBL expression and interaction on DC biology. As the 4-1BB/4-1BBL

pathway is a target in various anti-tumor and anti-viral therapeutic strategies, it is important that

we understand how these molecules impact not only T cell biology, but the biology of other cell

types that express these molecules, including DCs.

Page 99: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

76

Chapter 3

Natural killer cells contribute to splenic atrophy observed months following

LCMV clone 13 infection

Mbanwi AN, Wang C, Geddes K, Philpott DJ, Watts TH. Natural killer cells contribute to

splenic atrophy observed months following LCMV clone 13 infection.

Manuscript in preparation.

Author Contributions

Mbanwi AN and Watts TH designed the experiments and wrote the paper.

Wang C made the observation that spleens are small following LCMV clone 13 infection and

contributed data points to Fig 3.1A and E.

Geddes K prepared overnight cultures of Salmonella and infected mice with Salmonella.

Philpott DJ provided Salmonella and Salmonella-infected mice were maintained under Dr.

Philpott’s permit.

Mbanwi AN conducted all other experiments.

Page 100: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

77

3.1 Abstract

Lymphocytic choriomeningitis virus (LCMV) clone 13 infection of mice is a widely used model

for investigating the mechanisms driving persistent viral infection in humans. Early during

LCMV clone 13 infection, there is a disruption of splenic architecture, which then returns to

normal within a few weeks. However, the long-term effects of clone 13 infection on splenic

structure have not been reported. Here, we report that persistent infection with LCMV clone 13

results in sustained splenic atrophy that persists for up to 10 months following infection, whereas

infection with the acutely infecting LCMV Armstrong is associated with a return to pre-infection

spleen weights. The pathophysiological mechanisms underlying splenic atrophy often found in

autoimmune conditions such as celiac disease are not known. By blocking NK cells at the onset

of splenic atrophy (19-21 dpi), we show that NK cells contribute to the dramatic late-stage loss

in spleen size and cell number. Compared to LCMV clone 13-infected isotype control antibody

treated mice, treatment of LCMV clone 13-infected mice with a type I interferon receptor 1

(IFNAR1) blocking antibody at the onset of splenic atrophy significantly increased the

proportion of NK cells in the spleen and exacerbated splenic contraction around 2 months

following initial infection. LCMV clone 13 infection and associated atrophy of the spleen were

shown to compromise subsequent immunity to a bacterial pathogen. These findings implicate

NK cells in the pathophysiological mechanisms underlying splenic atrophy following viral

infection, raising the possibility that NK cells contribute to splenic atrophy in other diseases.

3.2 Introduction

LCMV is a non-cytopathic rodent pathogen. Infection with LCMV Armstrong leads to a robust

CD8 T cell response that results in rapid clearance of this acutely infecting strain of the virus.

LCMV clone 13 is a variant of LCMV Armstrong, isolated by serial passage in mice (258), that

differs by only 3 amino acids (267), resulting in a persistent viral infection, that is ultimately

cleared from most organs by 60-90 dpi. LCMV clone 13 infection of mice is a widely used

model to study the mechanisms driving persistent viral infection and has provided valuable

insights into how such mechanisms could be manipulated to treat chronic human viral disease

and other chronic conditions, such as cancer (437, 512-517). Despite the lack of cell lysis

Page 101: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

78

induced by the virus itself, chronic infection with LCMV is associated with specific and

generalized immunosuppression (518) due to induction of immunoregulatory mechanisms as a

consequence of persistent immune stimulation. This immune regulation leads to functional

exhaustion of the immune system (295, 519). Included in these outcomes is the loss of function

as well as early depletion of anti-viral T cells, culminating in poor viral control.

Previous studies have shown that infection with various strains of LCMV, including WE and

clone 13, leads to disruption of lymphoid architecture, a process that is reversed after viral

clearance (363, 366). However, the long term effect of LCMV clone 13 on splenic structure has

not been reported. Here we note a remarkable difference in the size of the spleen and the number

of immune cells following clone 13 as compared to Armstrong infection of mice. In contrast to

Armstrong infection, which induces transient splenomegaly, with a return to baseline spleen size

after viral clearance, clone 13-infected mice exhibit extensive and sustained splenic atrophy and

lymphopenia persisting up to 10 months following initial infection, well after virus can no longer

be detected in the spleen, kidney or liver. Here, we show that the reduced spleen size following

chronic viral infection is also associated with delayed control of a subsequent bacterial infection.

Splenic atrophy and the resulting perturbation in anti-bacterial immunity is a major issue in

autoimmune conditions such as celiac disease (229). The mechanisms causing splenic atrophy in

autoimmunity or following infection are ill-defined.

NK cells are cytotoxic lymphocytes that recognize their targets via an array of both activating

and inhibitory receptors, and mediate their effector functions by release of perforin and

granzymes and cytokines such as IFNγ and TNF (91, 520). In the context of LCMV infection,

NK cells were recently shown to play a regulatory role in the first few days following infection,

as NK cell depletion prior to infection increased anti-viral T cell responses and improved viral

control (322-324). However, the role of NK cells late in the infection is not clear. Here, we

implicate NK cells in modulating late-stage splenic atrophy following LCMV clone 13 infection.

NK cell depletion at 3 weeks pi, the time when splenic atrophy is first noted, significantly

ameliorated splenic contraction and loss of cell numbers. IFNAR1 blocking antibody treatment

at the onset of splenic atrophy dramatically increased the proportion of NK cells compared to

isotype control antibody treatment, which correlated with exacerbated splenic contraction and

Page 102: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

79

decreased cell recovery from the spleen. These findings implicate NK cells as contributing to

splenic atrophy following viral infection.

3.3 Materials and Methods

3.3.1 Mice and infections

5-6 week old C57BL/6 (B6) mice purchased from Charles River Laboratories (Saint-Constant,

QC, Canada) were infected intravenously with either 2 × 106 PFU of LCMV clone 13 or 5 × 10

3

PFU of LCMV Armstrong. Armstrong and clone 13 strains were prepared and quantitated as

previously described (258). Naive mice were either assessed on day 0 at 5-6 weeks old, or aged

to correspond with late-stage LCMV-infected mice. Mice were euthanized, and spleen weights

and number of splenocytes (as determined by trypan blue exclusion counting) were tracked from

days 0-300 for LCMV clone 13 infections, and from days 0-80 for LCMV Armstrong infections.

LCMV titers in the spleen, liver, and kidney were determined by a plaque-forming assay as

previously described (521).

Overnight cultures of streptomycin-resistant Salmonella enterica serovar Typhimurium (SL1344

ΔaroA) were washed two times with PBS, and the concentration of bacteria was measured by

determining the optical density at 600 nm. 14-15 week old naive B6 mice and B6 mice that had

been infected with 2 × 106 PFU of LCMV clone 13 2 months prior were infected

intraperitoneally with 105 CFU of Salmonella. 7-14 days post Salmonella infection, mice were

euthanized, and spleens and livers were homogenized in PBS containing 1% Triton X-100 with a

rotor homogenizer. Spleen and liver homogenates were serially diluted in PBS and plated on

Luria-Bertani agar plates containing 50 μg/ml streptomycin. Colonies were counted after

overnight incubation at 37°C.

All mice were maintained under specific pathogen-free conditions in sterile microisolators at the

University of Toronto. All animal studies were approved by the University of Toronto animal

care committee in accordance with the regulations of the Canadian Council on Animal Care.

3.3.2 In vivo antibody treatments

Page 103: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

80

On days 19 and 21 post LCMV clone 13 infection, B6 mice were injected intravenously with 0.2

mg/mouse of Rat IgG control (Sigma-Aldrich) or anti-NK1.1 [clone PK136, hybridoma kindly

provided by Dr. Pamela Ohashi (Ontario Cancer Institute, University Health Network, Toronto,

ON, Canada)] in PBS.

On days 19 and 21 post LCMV clone 13 infection, B6 mice were injected intraperitoneally with

500 μg of a mouse IgG1 isotype control (anti-TNP; clone 1B711) or IFNAR1 blocking antibody

(clone MAR1-5A3; Bio X Cell, West Lebanon, NH, USA). On day 23 post clone 13 infection,

mice received a final intraperitoneal injection of 250 μg of control or IFNAR1 blocking

antibody.

3.3.3 Flow cytometry and antibodies

Single cell suspensions of splenocytes were surface stained with antibodies, including anti-

TCRβ-PE-Cy5 (clone H57-597), B220-eFluor 450 (clone RA3-6B2), NK1.1-FITC (clone

PK136), NKp46-PE (clone 29A1.4), biotinylated NKG2D (clone A10) and streptavidin-PE,

MULT1-PE (clone 5D10), CD4-PE-Cy5 (clone RM4-5; BD Biosciences), and CD3-PE-Cy7

(clone 145-2C11). Intracellular staining was performed using Cytofix/Cytoperm solution (BD

Biosciences) and anti-RORγt-PE (clone B2D). All antibodies were purchased from eBioscience,

unless otherwise noted. Cell viability was determined using a LIVE/DEAD Violet

Viability/Vitality Kit (Life Technologies). Fluorescence minus one controls were used as

negative controls for antibody staining. Samples were analyzed using FACSCanto II (BD

Biosciences) with FACSDiva acquisition software. Data analysis was performed using FlowJo

software (Tree Star, Inc.).

For detection of LTi, spleens were cut into small pieces and treated with collagenase IV for 45

minutes at 37°C in a bacterial shaker. Spleen samples were then pipetted up and down to create a

single cell suspension. Splenocytes were washed with PBS, counted, and stained with the

appropriate antibodies.

3.3.4 Immunofluorescence microscopy

Spleens from naive and LCMV clone 13-infected B6 mice were removed from the animals and

frozen in Optimal Cutting Temperature (OCT) compound (Sakura Finetek). Spleen tissue

Page 104: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

81

sections were then cut at 6-8 microns using a Leica CM3050 cryostat (Leica Microsystems),

mounted on glass microscope slides and fixed in acetone. Sections were blocked with TBS-T

[TBS containing 0.05% Tween-20 (Sigma-Aldrich)] supplemented with 10% normal mouse

serum (The Jackson Laboratory) and 2 μg ml-1

anti-CD16/CD32 Fc block (clone 93;

eBioscience) for 30 minutes. Sections were stained using the following antibodies: anti-Thy1.2-

PE (clone 30-H12), B220-FITC (clone RA3-6B2), CD31-APC (clone 390), VCAM-1-Alexa

Fluor 647 (clone 429), biotinylated CD21/CD35 (clone 8D9) followed by streptavidin-PE in a

second step, and isotype control antibodies. Stains were performed in the dark for at least 1 hour.

All antibodies were purchased from eBioscience, unless otherwise noted. Slides were washed 3

times with TBS-T, followed by a final wash in PBS. DAPI nucleic acid stain (Invitrogen) was

then applied to the slides for 30 seconds, and washed off 3 times with PBS. Slides were then

mounted with Gel/Mount (Biomeda Corp.) and images were acquired with a Leica DMRA2

microscope (Leica Microsystems) equipped with a Retiga EXi digital camera (Q Imaging) using

OpenLab software (Improvision).

3.3.5 Statistical analysis

All statistical analyses were performed using Graphpad software (Prism). For comparison of two

groups, p values were obtained using the Student’s t test (unpaired, two tailed, 95% confidence

interval). Statistically significant differences are indicated as *, p < 0.05, **, p < 0.01 and ***, p

< 0.001.

3.4 Results

3.4.1 Persistent splenic atrophy following infection with LCMV clone 13

In the course of studying the resolution phase of the immune response in mice infected with

LCMV clone 13, we noted dramatic splenic atrophy at 60 days following infection, when

compared to pre-infection spleen (Fig. 3.1A left). Atrophic spleens at this time point were 64%

of their pre-infection weight (median of 93.7 mg on day 0 compared to a median of 59.8 on days

60-70 following infection, p < 0.0001). To ascertain the kinetics of this atrophy, spleen weight

and number of splenocytes were tracked from days 0-300 following infection with LCMV clone

Page 105: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

82

Page 106: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

83

13 (Fig. 3.1A center and right). Following infection with LCMV clone 13, spleen size increased

to a median weight of 203.7 mg by day 8. After reaching a peak size at day 8, spleen weight

decreased between days 8 and 21 to just below baseline and continued to decrease between days

35 and 50, thereafter maintaining a median weight of around 60 mg, and remained significantly

below baseline weight out to day 300 (Fig. 3.1A center). In addition, at 300 dpi, inguinal LNs in

LCMV-infected mice were about half the size of the inguinal LNs in naive animals (data not

shown). The size of the spleen was mirrored by the total number of splenocytes recovered at

each time point (Fig. 3.1A right). Over this time period, the overall weight of the mice increased

as expected, therefore splenic atrophy was not due to an overall cachexia in the animal (Fig.

3.1B). These data show that mice undergo splenic atrophy at days 21-50 pi and that the spleens

fail to recover to normal pre-infection weights up to 10 months pi.

To rule out the possibility that aging of the infected mice, compared to their 5-6 week naive

counterparts was responsible for splenic atrophy, we repeated this analysis with age-matched

uninfected controls (Fig. 3.1C). Naive WT age-matched mice had slightly bigger spleens than

their naive 5-6 week old counterparts (medians of 106.2 compared to 93.7 mg, p = 0.0809),

indicating that splenic atrophy is not a normal process associated with aging, rather that healthy

aging normally results in an increase in spleen size, and that splenic atrophy is a result of the

persistent infection.

We next asked whether splenic atrophy also occurred with an acutely infecting LCMV variant,

LCMV Armstrong, or whether it was a feature of more prolonged infection. We found a similar

increase in spleen weight by 8 dpi between clone 13 and Armstrong, however with Armstrong

infection, spleens contracted back to pre-infection weight around 30 dpi, and maintained this

weight up to 80 dpi (Fig. 3.1D). These findings suggest that persistent, but not acute infection,

results in late-stage splenic atrophy.

It was possible that splenic atrophy observed following LCMV clone 13 infection was due to the

persistence of the virus at late time points. To address this, we assessed viral load at time points

when sustained splenic atrophy is observed using plaque assays on the spleen, kidney and liver

of infected mice. Virus was detectable at 8 dpi in the spleen, but largely cleared by day 60, and

undetectable in the spleen thereafter (Fig. 3.1E), consistent with previous reports. At late time

points (80 dpi +), virus was no longer detectable in the kidney and liver (data not shown). These

Page 107: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

84

data suggest that the continued presence of virus is not required to sustain splenic atrophy, as the

spleen size fails to recover after virus is no longer detectable in spleen, liver or kidney.

3.4.2 Atrophic spleens are lymphopenic, but show T cell/B cell segregation within the white

pulp

To assess the impact of splenic atrophy on the cellular composition of the spleen, we compared

the proportion and number of immune cells in age-matched naive and 60 days post LCMV-

infected mice, as well as in naive 5-6 week old mice (Fig. 3.2A). We observed a significant

increase in the proportion of T cells in the late-stage LCMV-infected mice compared to young

and age-matched groups of naive mice, and a concomitant decrease in the proportion of B cells.

The proportion of non-B non-T cells was not significantly different between naive and infected

(Fig. 3.2A). However, when converted to total numbers, it was clear that, there were significantly

less T, B, and non-B non-T cells in the atrophic spleens. We further characterized the non-B non-

T cell population with antibodies to CD11b, CD11c, MHC-II (antigen presenting cells), F4/80

(macrophages), NK1.1 (NK cells), and Gr-1 (granulocytes), and similarly found a significant

reduction in the number of these cells in atrophic spleens (data not shown). Overall, these

findings show that splenic atrophy is due to an overall decrease in the numbers of T, B and non-

B non-T cells in the spleen. However, based on frequency, the B cell compartment seems to be

disproportionately affected compared to the T cell compartment.

Persistent LCMV infection has been shown to disrupt splenic architecture early following

infection, with recovery between days 14-20 pi (363). However, at these time points, maximal

splenic atrophy has not yet occurred. Thus, we investigated the organization of the spleen at late

time points when there is maximal and sustained splenic atrophy. Thy1.2, B220, and CD31 were

used to define T cells, B cells, and endothelial cells, respectively. CD35, a marker of follicular

dendritic cells (FDCs) and B cells, and VCAM-1 were used to look at stromal cells in the spleen.

We found T cell/B cell segregation, and similar stromal cell networks between day 60 LCMV-

infected and naive mice (Fig. 3.2B). Thus splenic atrophy persists despite the appearance of

organization within the white pulp at late time points following clone 13 infection.

Page 108: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

85

Page 109: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

86

3.4.3 NK cells contribute to splenic atrophy following LCMV clone 13 infection

NK cells have been implicated in the resolution phase of the immune response (522), and have

been identified as critical players both early and late following LCMV clone 13 infection, where

they were suggested to eliminate virus-specific T cells and promote viral persistence (196, 322-

324). Thus, to evaluate whether NK cells were participating in the drastic loss of spleen cells, we

evaluated the effect of NK cell depletion on splenic atrophy following clone 13 infection. Mice

were infected with LCMV clone 13 on day 0, and then on days 19 and 21 pi, the time point when

spleen size returns to baseline just before undergoing further atrophy, we treated mice with either

an NK cell-depleting antibody (anti-NK1.1) or an isotype control antibody (Fig. 3.3A). The

extent of NK cell depletion was measured in the blood at 25 dpi (69% depletion), and in the

spleen on days 35 (80% depletion) and 50 (47% depletion) following infection (Fig. 3.3B). NK

cells were largely depleted up to day 35 following infection, but their numbers started to recover

by day 50. Spleen weight and cell numbers were assessed on day 50 following infection, which

corresponded to around 1 month following NK cell depletion (Fig. 3.3C). On day 50 pi, mice

that had been treated with NK cell-depleting antibody on days 19 and 21 pi showed a 22%

increase in spleen weight compared to mice treated with control antibody (median of 62.3 mg for

control mice, and median of 75.7 mg for NK cell-depleted mice, p = 0.0001), as well as a 70%

increase in the number of cells in the spleen (median of 54 × 106 cells in control mice and 92 ×

106 cells in NK cell-depleted mice, p < 0.0001) (Fig. 3.3C). Compared to the median spleen

weight on day 21 pi (81.9 mg), control treated mice experienced a 24% reduction in spleen

weight (median of 62.3 mg), whereas NK cell-depleted mice only experienced a 7.6% reduction

in spleen weight (75.7 mg). NK cell depletion did not change the proportion of cells in the

spleen, but did increase the number of T, B, and non-B non-T cells (data not shown). Thus, these

findings suggest that NK cells contribute to splenic atrophy following persistent LCMV

infection.

We next asked how NK cells could be contributing to splenic atrophy. NKG2D is an activating

receptor on NK cells that was recently implicated in the regulatory functions of NK cells

following LCMV infection (322). As the expression of NKG2D ligands renders cells susceptible

to NK cell-mediated lysis, we looked at the expression of the NKG2D ligands, MULT1 and Rae-

1δ, 50 days-post LCMV clone 13 infection, however, we did not detect expression in the spleen

Page 110: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

87

Page 111: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

88

Page 112: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

89

at this time point (data not shown). We speculated that by day 50, NKG2D ligand-expressing

cells would have been depleted, and thus looked on day 21, a time point at which the spleen has

started to contract below baseline, but has not yet reached the maximal contraction weight. On

day 21 pi, NK cells, as defined by NKp46 expression, expressed NKG2D (Fig. 3.3D), and

MULT1 expression was found predominantly on B cells (Fig. 3.3E). Rae-1δ expression was not

detectable (data not shown). These data point to a possible role for NKG2D-mediated lysis of

MULT1-expressing B cells, in splenic atrophy following LCMV clone 13 infection.

3.4.4 IFNAR1 blocking antibody treatment increases the proportion of NK cells, and

exacerbates splenic atrophy

Chronic infection results in high levels of type I IFNs. As NK cells can be activated by type I

IFNs (344), and chronic IFN signaling plays a regulatory role following LCMV clone 13

infection (320, 353), we asked whether IFN signaling contributes to the process of splenic

atrophy following LCMV clone 13 infection. To this end, mice were infected with LCMV clone

13 on day 0, and then treated with either IFNAR1 blocking (320, 353) or isotype control

antibody on days 19, 21, and 23 (Fig. 3.4A). Spleen weights, number of splenocytes, and cell

populations were assessed about a month later on day 50. Surprisingly, spleens from IFNAR1

blocking antibody treated mice had an 18% decrease in spleen weight compared to isotype

control antibody treated mice (medians of 68.5 mg and 56.5 mg in isotype control treated and

IFNAR1 blocking antibody treated mice, respectively, p < 0.0001), as well as a 44% decrease in

cell numbers (medians of 110 × 106 cells and 62 × 10

6 cells in isotype control antibody treated

and IFNAR1 blocking antibody treated mice, respectively, p = 0.0002) (Fig. 3.4B). Compared to

the median spleen weight on day 21 pi (81.9 mg), isotype control antibody treated mice

experienced a 16% decrease in spleen weight (median of 68.5), while IFNAR1 blocking

antibody treated mice experienced a 31% decrease in spleen weight (median of 56.5 mg). In

terms of proportion of cells on day 50 pi, there was a significant decrease in the proportion of B

cells, and a concomitant increase in the proportion of T cells in the IFNAR1 blocking antibody

treated mice compared to isotype control antibody treated mice (Fig. 3.4C), whereas there was

no statistically significant difference in the proportion of non-B non-T cells (Fig. 3.4C). When

converted to absolute numbers, there were significantly fewer T, B, and non-B non-T cells in the

IFNAR1 blocking antibody treated mice compared to isotype control antibody treated mice (Fig.

Page 113: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

90

Page 114: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

91

Page 115: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

92

3.4C). To understand these findings, we looked at the proportion of NK cells, and found an 87%

increase in the proportion of NK cells in IFNAR1 blocking antibody treated compared to isotype

control antibody treated mice (Fig. 3.4D, 3.4E). The cells in the IFNAR1 blocking antibody

treated mice also had a higher MFI of NKG2D, suggesting increased functionality (Fig. 3.4E).

Thus, compared to isotype control antibody treatment, IFNAR1 blocking antibody treatment

during persistent LCMV infection resulted in increased proportions of NK cells, with evidence of

increased activation, correlating with increased splenic atrophy. These data further support a role

for NK cells in splenic contraction at the late stages of LCMV clone 13 infection.

As the type I IFN response did not appear to account for splenic atrophy following LCMV clone

13 infection, in fact the opposite was observed, we asked if other cytokines might contribute. As

TNF is a cytokine associated with cell death and cachexia, we also measured the spleen weights

and counted the number of cells in TNF-/-

mice at 60 days post LCMV clone 13 infection. Mice

lacking 4-1BBL were also included as 4-1BBL has been shown to regulate TNF levels in

macrophages (440). However, similar to WT mice, TNF-/-

and 4-1BBL-/-

mice had small spleens

(medians of 46.1 and 60.5 mg, respectively), suggesting that neither TNF, nor 4-1BBL,

contribute significantly to late-stage splenic atrophy (data not shown).

3.4.5 Splenic atrophy delays clearance of a bacterial pathogen

Splenectomy, hyposplenism, and asplenia have been shown to greatly increase susceptibility to

certain bacterial infections in humans (248). Therefore we asked whether splenic atrophy

following persistent viral infection compromises subsequent anti-bacterial immunity. To

investigate the response to secondary bacterial challenge, age-matched naive and 60 days-post

LCMV-infected mice were infected with a high dose of an avirulent aroA mutant strain of

Salmonella typhimurium. 7 days post Salmonella infection, the bacterial burden in the spleen and

liver were assessed. An attenuated strain of Salmonella was chosen as the virulent strain of

Salmonella killed the mice too quickly to assess bacterial burden with or without prior LCMV

infection. We found that mice that had been previously infected with LCMV clone 13 had about

a 3-fold increase in the bacterial load in the spleen (Fig. 3.5A) and liver (Fig. 3.5B), compared to

age-matched naive mice, although by day 14, both naive and LCMV clone 13-infected mice

largely cleared the attenuated pathogen. These findings demonstrate that infection with LCMV

Page 116: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

93

Page 117: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

94

clone 13 with its associated splenic atrophy can delay subsequent control of Salmonella

infection.

3.4.6 The proportion of LTi is similar between aged naive and LCMV-infected mice

It is remarkable that months following LCMV clone 13 infection and viral clearance from the

spleen, liver, and kidney, atrophied spleens do not recover to pre-infection weight. Lymphoid

tissue inducer cells (LTi) fall into the category of group 3 innate lymphoid cells (ILCs), which

are capable of producing IL-17A and/or IL-22, and depend on the transcription factor RORγt for

their development and function (90). LTi are critical in LN and Peyer’s patch organogenesis in

the embryo and post-natal period (523, 524). Adult LTi were recently shown to participate in the

restoration of lymphoid tissue integrity following LCMV WE infection (370). To ask whether

the lack of spleen recovery following LCMV clone 13 infection is due to a potential depletion of

LTi, we infected mice with clone 13, and assessed both the proportion and number of LTi

(CD4+CD3

-CD11c

-B220

-RORγt

+) in the spleen 300 days later (Fig. 3.6). LCMV-infected mice

were compared to naive mice aged between 3 and 9 months. Our findings show that the

proportion of LTi is similar between naive and LCMV-infected mice. When converted to total

number, there is a trend (p = 0.1567) of lower numbers in the LCMV-infected group, likely due

to the overall loss in the number of splenocytes. It should be noted that our analysis of LTi could

also include the recently identified subset of group 3 ILCs, NCR+ILC3s, which also express

RORγt and can express CD4, but unlike LTi, express NKp46 (90). Taken together, it appears

that late-stage LCMV infection does not affect the proportion of LTi or LTi-like cells present in

the spleen.

Page 118: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

95

Page 119: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

96

3.5 Discussion

The mechanisms governing the resolution of inflammation following LCMV infection are ill-

defined. Both LCMV Armstrong and LCMV clone 13 infections result in a substantial immune

response that initially doubles the size of the spleen. Following LCMV clone 13 infection, the

spleen contracts up to 50% of its pre-infection weight months following infection, whereas

spleens from LCMV Armstrong-infected mice return to their pre-infection weights. We observed

sustained splenic atrophy following LCMV clone 13 infection up to 10 months following

infection, a time point several months after virus is no longer detectable in the spleen, kidney and

liver. Atrophic spleens were lymphopenic, but did not show gross abnormalities in architecture.

Using an NK cell-depleting antibody at the onset of splenic atrophy, we provide evidence that

NK cells contribute to splenic atrophy. In keeping with the highest expression of the NKG2D

ligand, MULT1, on the B cells 21 dpi, B cells showed the biggest detriment in numbers over the

naive spleen. Thus it appears that NK cells contribute to splenic atrophy by impacting T cells, B

cells and non-B non-T cells, but B cells may be most susceptible. IFNAR1 blocking antibody

treatment at the onset of splenic atrophy increased the proportion of NK cells in the spleen,

which correlated with exacerbated splenic atrophy. Moreover, LCMV clone 13 infection and

associated splenic atrophy resulted in compromised immunity when mice were challenged with

an attenuated strain of Salmonella.

We found that mice undergo sustained splenic atrophy months following persistent, but not

acute, infection with LCMV. Persistent viral replication appears to initiate a program that results

in splenic atrophy at late stages following initial infection – a program that is distinct from that

induced by acute viral replication. It is well known that persistent as opposed to acute LCMV

infection, induces a state of generalized and specific immune suppression that is associated with

increased immune regulation, virus-specific T cell dysfunction/exhaustion and deletion, and

subsequent poor viral control (2). This poor control of virus following clone 13, but not

Armstrong infection, could lead to the excessive bystander activation of cells in the spleen,

thereby making these cells susceptible to either intrinsic or extrinsic forms of cell death. It is

unlikely that direct infection of T and B cells, followed by lysis of infected cells, mediated their

removal as these cells have very low expression of the cellular receptor for LCMV, α-

dystroglycan, and clone 13 has been shown to preferentially infect dendritic cells (257, 270).

Page 120: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

97

The mechanisms underlying late-stage atrophy observed with LCMV clone 13 have not been

previously reported. The NK cell depletion studies reported here indicate that NK cells are either

directly or indirectly contributing to splenic atrophy late following LCMV clone 13 infection.

NK cells were recently implicated in the resolution phase of a self-limited asthma model in mice.

Either depleting NK cells or blocking their migration to target tissues delayed clearance of

eosinophils and antigen-specific CD4 T cells (522). In the context of LCMV clone 13 and/or WE

infection, several groups have identified an immunoregulatory role for NK cells both early (322-

324) and later (196) in the infection. Depleting NK cells prior to infection (322-324) and

delaying NK cell depletion from 1 day to 4 weeks pi (196) increased virus-specific T cell

responses and improved viral control. Waggoner et al. point to a direct effect of NK cells on

CD4 T cells (196, 323), which then impact the CD8 T cell response, whereas Lang et al. point to

a direct effect of NK cells on CD8 T cells (322). However, these studies (196, 322-324) do not

report on the effects of NK cell depletion on spleen size, or on the long term outcome of these

effects.

The receptor-ligand interactions underlying NK cell recognition and depletion of T cells

following LCMV clone 13 infection are not clear, but might involve the activating NK cell

receptor, NKG2D. In the context of LCMV WE infection, Lang et al. point to a role for NKG2D

in the recognition and early removal of virus-specific CD8 T cells (322). Ligands for the

activating receptor NCR1, and the inhibitory ligands MHC-I, Qa-1b, and CD48 have also been

implicated in recognition of T cells by NK cells (325, 327, 360). The present study identifies an

effect of NK cells on not only T cells, but on several other immune cell types during the

resolution phase of persistent LCMV infection. Notably, our finding that NK cells express

NKG2D and that B cells express the NKG2D ligand, MULT1, at the onset of splenic atrophy,

identifies B cells as a potential target for NK cell-mediated killing. These data likely explain why

late-stage LCMV clone 13-infected mice have a lower proportion of B cells in the spleen. On

day 21 pi, MULT1 expression was not readily detectable on T cells and non-B non-T cells. This

could have been due to these cell types having lower levels of the ligand on day 21 pi, acquiring

ligand expression and being depleted prior to day 21, or acquiring ligand expression after day 21

and being depleted at a later time point. It is also possible that the receptor-ligand interactions

Page 121: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

98

that mediate B cell depletion are distinct from those that mediate T cell and non-B non-T cell

depletion.

Our finding that IFNAR1 blocking antibody treatment (compared to isotype control antibody

treatment) at the onset of splenic atrophy increases the proportion of NK cells in the spleen is

consistent with a role for type I IFNs in limiting the accumulation of NK cells at later stages of

LCMV clone 13 infection. Type I IFNs are known activators of NK cell effector function during

viral infection (344), but have been shown to exert anti-proliferative effects on mouse and human

NK cells in vitro (347, 525, 526). Although there is evidence that type I IFNs augment mouse

NK cell proliferation in vivo (347), this was later attributed to a role for these cytokines in

inducing IL-15 (527). It is possible that persistent IFN signaling induced by LCMV clone 13

infection (320, 353) coupled with potentially differential induction of IL-15 or other cytokines at

different stages of infection, normally limits the number of splenic NK cells present at later

stages of infection. Regardless of mechanism, the finding that IFNAR1 blocking antibody

treatment results in an increased proportion of NK cells and exacerbates splenic contraction at

late time points, adds further evidence for a role of NK cells in splenic atrophy.

The finding that spleens from LCMV clone 13-infected mice do not return to pre-infection

weights up to 10 months following infection raised the question of whether these mice have

compromised immunity. As humans with splenic atrophy or those that have undergone

splenectomy are more susceptible to certain bacterial infections (248), we tested the effect of

prior LCMV clone 13 infection and splenic atrophy on control of a bacterial infection. Previous

studies have shown that infection of mice with LCMV WE led to significantly higher bacterial

loads in the spleen and liver following challenge with either Listeria monocytogenes, S. aureus,

or Salmonella typhimurium when compared to LCMV-naive mice. However, in this study, mice

were challenged 2 days following LCMV infection (528), a time point when virus is still present.

Here, we investigated challenge during late stages of LCMV infection, at time points when virus

is largely cleared from the spleen and undetectable in the liver in our model, to look not at the

effect of viral replication per se, but at the effect of spleen size. We found that when mice were

challenged with Salmonella at 60 days following LCMV clone 13 infection, they had a higher

bacterial burden than Salmonella-challenged LCMV-naive mice, suggesting that LCMV clone

13-induced splenic atrophy compromises subsequent immunity.

Page 122: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

99

A key question that our study raises is why the spleen fails to recover to pre-infection weight

after NK cells have removed the activated/stressed cells and virus has been cleared. Is it possible

that LCMV infection and subsequent cell loss leads to the depletion of a cell type required for

lymphoid tissue homeostasis? A previous study (362) has shown that a lymphotoxin (LT) and B

cell dependent pathway is responsible for peripheral LN expansion following LCMV WE

infection. Similarly, LT-expressing LTi were shown to participate in the restoration of lymphoid

organ structure following LCMV infection (370). These cells were of interest because of their

critical role during LN and spleen organogenesis in the embryo and post-natal period (523, 524).

However, as our data suggest that LTi are still present 300 days after clone 13 infection, lack of

LTi are unlikely to explain the failure of spleens to recover following LCMV clearance.

Moreover, despite their small size, the atrophied spleens have largely regained their normal

architecture. As chemokine gradients required for T cell/B cell segregation are dependent on

LTi, it appears unlikely that loss of these cells is the cause of splenic atrophy.

Lack of spleen recovery might also be due to defective chemokine/cytokine expression by

splenic stromal cells, aberrant generation of immune cells in the bone marrow/thymus, altered

homing of lymphocytes to non-lymphoid organs or possibly sustained autoreactive phenomena

induced by persistent viral infection. Indeed, splenic atrophy in humans is associated with celiac

disease and other autoimmune conditions such as SLE and Sjögren’s syndrome (226-231, 233).

It is not entirely clear whether splenic atrophy predisposes to autoimmunity, or vice versa, and

the mechanisms underlying splenic atrophy are not defined.

An association between splenic atrophy and viral infection has been shown for several viruses,

including SARS and H5N1 infection in humans (234, 235), as well as for coxsackievirus B3

(236), YN strain of parainfluenza virus (237), MHV68 (239), and TBEV (240) infections in

mice. In the mouse models, atrophic spleens were shown to be approximately 2-3 fold smaller

than uninfected controls in size (236, 237) and number of immune cells (236, 240) and the

kinetics of atrophy ranged from 5-14 days following infection (236, 237, 239, 240). In those

studies where the numbers of specific cell types in the spleen were assessed, it was shown that T

cell, B cell and non-B non-T cell subsets were affected (236, 239, 240). The exact mechanisms

underlying splenic atrophy or its causes were not clear in these studies, but were attributed to the

cytopathicity of the virus (234, 235), the effector functions of T cells (239) and cytokine

Page 123: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

100

production (240). These studies did not investigate a potential role for NK cells in mediating

splenic atrophy, thus it is possible that similar to LCMV clone 13 infection, splenic atrophy

following these viral infections also depended in part on the presence of NK cells. In the

aforementioned studies, atrophy was irreversible as these viral infections ultimately killed their

hosts (236, 237, 240). MHV68 infection of IFNγR-/-

mice was an exception, in that similar to

LCMV clone 13 infection in our model, infection in these mice was not lethal, and mediated

irreversible splenic atrophy out to day 70 pi (239).

There is evidence that the spleen can regenerate following splenectomy and autotransplantantion

of spleen tissue in the peritoneum in mice and humans (251, 529, 530). The amount of spleen

tissue regenerated varies, but does not exceed that of the recipient’s own spleen (251, 531).

Regenerated splenic tissue appears histologically normal and can function in blood filtration,

however, the extent to which regenerated spleens protect from subsequent infection is not clear

(251, 532). The mechanisms regulating spleen tissue neogenesis have been investigated in

animal models, and although these are poorly defined, they are thought to involve the activity of

LT-educated endothelial organizer cells in the grafted spleen (533). Factors such as the age of

splenic tissue at the time of transplantation affects its ability to regenerate (533). Tan et al.

showed that grafting of whole-spleen capsules from D3 donors resulted in 83% (20/24) of the

grafts regenerating spleen tissue in recipient mice 4 weeks following transfer whereas only 38%

(6/16) of 8 week old adult spleen capsule grafts regenerated normal spleen tissue (533). It is

therefore possible that in our study, the atrophied spleens were too old to properly regenerate

following viral clearance. Other studies have shown that splenic tissue transplanted into

splenectomized mice regenerate to a larger size and contain more lymphoid tissue than spleen

grafts in sham-operated mice (534, 535). These studies suggest that during steady-state

conditions, there are factors secreted by the spleen or elsewhere that normally prevent

regeneration (251). This might also explain why the atrophied spleens induced by clone 13

infection had not returned to pre-infection weight by 300 dpi.

Taken together, these findings implicate NK cells in the mechanisms underlying pathological

splenic atrophy, and support a therapeutic role for NK cell depletion in the context of persistent

viral infection.

Page 124: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

101

Chapter 4

Discussion and Future Directions

Page 125: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

102

4.1 Overview

In this thesis, I have explored two aspects of innate immunity. In chapter 2, I demonstrated that

4-1BB and 4-1BBL interact consitutively on DCs and in chapter 3, I demonstrated a role for NK

cells in splenic atrophy in chronic LCMV infection. Here I discuss the implications of these

findings and possible future directions for the projects.

4.2 Role of 4-1BB/4-1BBL on DCs: What unique transcripts are induced by 4-1BB/4-1BBL

on DCs and does this impact secondary immune responses?

The finding that 4-1BB/4-1BBL interact on LPS-activated DCs (Fig. 4.1) raises the question of

the reason for such an interaction. What is the biological outcome of 4-1BB/4-1BBL interaction

for the DCs, and how does this interaction impact the interaction between DCs and other cell

types during an ongoing immune response in vivo? It is possible that 4-1BB/4-1BBL interaction

on DCs shuts down their ability to activate T cell responses by limiting the available 4-1BBL

signal for 4-1BB-expressing T cells, or by actively inhibiting pro-inflammatory signaling in the

DCs. Alternatively, this constitutive interaction could further enhance the stimulatory capabilities

of DCs before they encounter a T cell or during the DC-T cell interaction.

My studies in chapter 2 showed that 4-1BB/4-1BBL interaction on DCs was dispensable for the

primary response to influenza NP. As the conditions of immunization were chosen such that the

response was suboptimal, it seems unlikely that this negative result is due to the saturation of the

response. Although I tested the impact of 4-1BB/4-1BBL on DCs in the primary expansion of

influenza NP-specific CD8 T cells, it is conceivable that this interaction could be important in

programming a secondary response. The precedent for this type of experiment is the work of

Schoenberger et al. showing that T cells primed in the absence of CD4 T cell help, show

relatively normal primary expansion, but impaired secondary expansion (536), later attributed to

a role for CD4 T help during priming in inducing autocrine IL-2 production by the CD8 T cells

(537), which in turn prevented TRAIL upregulation during secondary antigen exposure, which

otherwise led to CD8 T cell death (538). To rule out this kind of scenario, one could check the

secondary response to influenza infection in mice immunized with DCs expressing influenza NP,

but lacking 4-1BB/4-1BBL. However, this scenario seems unlikely as two studies (436, 506)

showed that T cells primed in 4-1BBL-/-

mice by i.p. infection with influenza A/X31 and then

Page 126: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

103

Page 127: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

104

transferred to 4-1BBL-sufficient mice show normal secondary T cell expansion to a second

influenza infection with influenza A/PR8, suggesting that 4-1BBL is dispensable for

programming of CD8 T cell memory. Conflicting data on this point were obtained by Hendriks

et al. (448) using a different influenza strain, A/NT/60, which suggested that with a weak

primary response, 4-1BBL could be important in programming T cell memory. Thus an

outstanding question for the studies in chapter 2, is whether T cells primed with 4-1BB or 4-

1BBL-deficient DCs can mount an effective secondary T cell response. If not, this raises the

question of what 4-1BB/4-1BBL do on DCs during priming to allow full programming of CD8 T

cell memory.

Although we did not find a role for 4-1BB/4-1BBL interaction on DCs in priming influenza NP-

specific T cell responses, it is possible, indeed likely, that 4-1BB/4-1BBL interactions within the

DC compartment has a biological impact in some contexts. Thus, in order to take an unbiased

approach to decipher how 4-1BB and 4-1BBL regulate DC biology, future projects should

consider a high-throughput whole-genome screening approach, such as microarray technology

for gene expression studies in WT and deficient DCs. In this regard, it is worth considering

whether this should be done with LPS-activated BM-derived DCs ex vivo, or whether conditional

knockout mice lacking 4-1BB or 4-1BBL only on DCs, should be generated, so that the

consequences of this signal can be studied in vivo. Within the TNF family, conditional knockout

mice exist for TNF and CD70, and in the case of TNF, have been useful for determining which

TNF expressing cell types are mediators of inflammation in several models (539-541). However,

to date, 4-1BBL conditional knockout mice have not been generated.

4.3 4-1BB signaling in DCs

The 4-1BB signaling pathway in T cells has been well characterized and was briefly summarized

in the introduction section of this thesis (1.7.6). One of the outstanding questions is whether

TRAF recruitment and signaling have different consequences in different cell types. For

example, how do 4-1BB signals in a T cell compare to 4-1BB signals in a DC, and are these

important considerations for understanding the differential activity of 4-1BB on T cells versus

DCs. As 4-1BB regulates transcription factors such as NF-κB and MAPK-induced

transcritiptional regulators, it is likely that the genes induced by 4-1BB in different cell types

Page 128: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

105

depend on the context. The molecular pathway of 4-1BB signaling in DCs has not been

investigated and is a topic for future studies.

4.4 Potential bidirectional signaling by 4-1BB and 4-1BBL on DCs

An interesting feature of the TNF superfamily is bidirectional signaling, that is, for some family

members, both receptor and ligand can signal (432, 542-546). When signals are transmitted

through the ligand, this is referred to as “reverse signaling”. Since DCs express both 4-1BB and

4-1BBL, it is not clear in our model whether regulation of DC biology will be mediated by 4-

1BB and/or 4-1BBL signaling.

4-1BBL reverse signaling has been described in T cells, monocytes, macrophages, B cells,

osteoclasts, and DCs (432, 544). For the most part, reverse signals have been shown to enhance

the activation and pro-inflammatory activity of these cell types, although in some cases, reverse

signaling mediates an inibitory signal. One caveat to these experiments is that it is often difficult

to interpret whether an antibody is mediating its effects by inducing reverse signals, blocking

receptor-ligand interactions, or both. Overall, studies evaluating the biological contribution of

reverse signaling during ongoing immune responses are lacking, as most studies have been

conducted in vitro using stimulating agents in cell lines and some primary cells. Another issue

here is the reagents, as well as their respective controls, used to stimulate 4-1BBL. A 4-1BB-Fc

protein that could also target Fc receptors is commonly used without the inclusion of 4-1BBL-

deficient control cells. This is an issue, because the oligomerization state of 4-1BB-Fc may be

different than that of the human IgG1 control that is often used (497, 547). Moving forward,

knockin-mutations in the cytoplasmic tail of 4-1BBL would aid in assessing the true ability of a

ligand to transmit signals.

Another area that requires further investigation is the precise mechanisms used by 4-1BBL to

transmit signals, and the molecules involved in the 4-1BBL signaling pathway. The cytoplasmic

tails of six TNF family members, 4-1BBL, CD70, TNF, FasL, CD40L and CD30L, contain one

or more consensus sequences for phosphorylation by the ubiquitously expressed kinase, casein

kinase I (CKI) (545). Recombinant CKI was shown to phosphorylate 4-1BBL (548), TNF (549)

and FasL (550), and mutagenesis experiments involving CKI inhibitors revealed that the CKI

motif is important for NFAT activation in FasL-mediated T cell costimulation (550).

Page 129: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

106

Interestingly, 4-1BBL has been suggested to interact with molecules other than its traditional

receptor, 4-1BB, in mediating signals, including TLRs (440, 551), TNFR1 (552) and

TMEM126A (553). Taken together, evidence exists for bidirectional signaling during 4-1BB/4-

1BBL interactions, however, a demonstration that these signals have in vivo biological

significance is lacking.

4.5 Splenic atrophy following LCMV clone 13 infection

Chapter 3 identified NK cells as contributing to splenic atrophy following LCMV clone 13

infection, however, the mechanisms whereby NK cells mediate cell death were not delineated

(Fig. 4.2). My studies identified a potential role for the activating NK cell receptor, NKG2D, but

it is also possible that other activating NK cell receptors such as NCR1, are involved.

Alternatively, NK cell inhibitory ligands, such as MHC-I and Qa-1b which bind to the NK cell

inhibitory Ly49 and CD94/NKG2 receptors on NK cells, could be downregulated on these cells,

thereby making them susceptible to NK cell-mediated lysis. The upregulation of activating

ligands for NCR1 and downregulation of inhibitory ligands MHC-I and Qa-1b were recently

identified as mechanisms that render anti-viral T cells susceptible to NK cell killing in the

absence of early type I IFN signaling during LCMV WE infection (326, 327). The inhibitory

receptor 2B4 on NK cells has also been shown to be involved in NK cell killing of activated T

cells during LCMV clone 13 infection (325). Future studies should investigate further the NK

cell receptor-ligand interactions mediating splenic atrophy during LCMV clone 13 infection.

To look at a potential involvement of the activating receptor NCR1, future studies can utilize an

NCR1-IgG fusion protein, which detects NCR1 ligands (139), to assess the expression of NCR1

ligands at later time points following Cl13 infection. An NCR1 blocking antibody and NCR1-

deficient mice (554) are also available to test the impact of NCR1/NCR1 ligand interactions in

vivo. The expression of inhibitory ligands such as MHC-I, Qa-1b, and CD48 following Cl13

infection can be detected by using specifc antibodies for flow cytometry. β-2 microglobulin-

deficient (β-2m-/-

) mice (555), which lack surface MHC class I expression, and CD48-/-

mice

(556) are also available. Splenocytes from these mice could be used in ex vivo or in vivo NK cell

cytotoxicity assays to assess whether NK cells require detection of these ligands for killing. Ex

vivo, one can measure the apoptosis of cells cocultured with NK cells isolated from LCMV-

infected mice (19-21+ dpi) by flow cytometry using reagents specific for apoptotic cells

Page 130: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

107

Page 131: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

108

such as an antibody to Annexin-V and 7-AAD viability staining solution (326). In vivo, CFSE-

labelled target cells can be transferred into LCMV-infected mice (19-21+ dpi) and tracked for

short periods following transfer by CFSE expression. To further look at the expression of

NKG2D ligands, an NKG2D-human IgG fusion protein (557), which detects MULT1, the

isoforms of Rae-1, and H60, can be used. The involvement of NKG2D can also be verified by

using an NKG2D blocking antibody (558, 559) in vivo or Klrk1-/-

mice, which are deficient in

NKG2D.

The caveat to using mice that have a deficit in NK cell numbers, e.g. Nfil3-/-

(E4bp4-/-

) mice

(560), in the Cl13 splenic atrophy model is that Cl13 infection of these mice is predicted to not

cause a persistent infection due to an increased number of functional CD8 T cells mediating viral

clearance. Indeed, infection of Nfil3-/-

mice with LCMV WE resulted in double the frequency of

IFNγ-producing CD8 T cells 6 dpi when compared to WT mice (322). Thus, Cl13 infection of

NK cell-deficient mice could resemble an acute LCMV infection, which I showed in this model

does not result in splenic atrophy.

The findings in chapter 3 of this thesis do not definitively identify NK cells as the only

mechanism causing splenic atrophy following LCMV clone 13 infection as NK cell depletion

did not fully abrogate splenic contraction below pre-infection spleen weight. It is not clear,

however, whether the lack of complete reversal of splenic atrophy by NK cell depletion is

because NK cells are not the only mechanism involved or whether the depletion is incomplete. It

is possible that other cell types such as cytotoxic CD8 T cells or macrophages contribute to the

dramatic loss of cells. Indeed, in a model of splenic atrophy following MHV68 infection of

IFNγR-/-

mice, CD8 T cell depletion abrogated splenic atrophy (239). CD8 T cells also express

NKG2D and are involved in the killing of NKG2D ligand-expressing cells. Depleting CD8 T

cells or possibly macrophages at the onset of splenic atrophy would allow us to evaluate the

potential contribution of these cell types.

It should be noted that NKT cells also express NK1.1 and could be depleted by the high dose of

anti-NK1.1 (clone PK136) antibody used in this study. NKT cells are non-conventional αβ T

cells that express molecules associated with the NK cell lineage and are restricted by the

monomorphic MHC class-I-like molecule CD1d that presents self and exogenous glycolipids

(561). Type I NKT cells or invariant NKT cells (iNKT) cells express an invariant TCRα chain

Page 132: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

109

encoded by Vα14-Jα18 genes in mice and Vα24-Jα18 genes in humans and a restricted set of

TCRβ chains (Vβ2, Vβ7 and Vβ8 in mice and Vβ11 in humans), whereas type II NKT cells

express more diverse TCR Vα chains (561). Following activation, NKT cells produce large

amounts of IFNγ and IL-4, which influence a wide array of immune responses, including tumor

surveillance, maintenance of self-tolerance and anti-microbial defenses (561). LCMV Armstrong

infection of Vα14 transgenic mice, which contain an elevated frequency of iNKT cells, and

treatment of C57BL/6 mice with the iNKT cell agonist α-galactoside ceramide (α-GalCer) during

Armstrong infection revealed a role for NKT cells in controlling viral load in the liver and

pancreas, but not at early time points (up to 4 dpi) in the spleen (562). Compared to control mice,

viral load was about 1 log lower in Armstrong-infected Vα14 transgenic mice 6-8 dpi, suggesting

a possible delayed role for NKT cells in controlling virus in the spleen (562). NKT cells were

shown to be dispensable for NK cell immunoregulatory function during LCMV Cl13 infection as

NK cell depletion of Cl13-infected CD1d-/-

mice, which lack NKT cells, enhanced LCMV-

specific T cell responses and reduced viral load, similar to that in WT mice (323).

To evaluate a potential role of NKT cells in the LCMV clone 13 splenic atrophy model, the

frequency of NKT cells before and after anti-NK1.1 treatment should first be determined using

NKT tetramers. The frequency of other NK1.1-expressing cell types such as activated CD8 T

cells and γδ T cells following anti-NK1.1 depletion should also be assessed. A role for NKT cells

and γδ T cells can be further evaluated using CD1d-/-

and TCRδ-/-

mice.

4.6 Can the spleen regenerate following splenic atrophy?

One question that this study raises is whether during steady state conditions, the spleen fully

regenerates following splenic atrophy, and if so, how. If the spleen does not regenerate, then why

not?

Splenic regeneration or splenosis (autotransplantation of splenic tissue) is a normal process that

can occur following splenectomy due to trauma or pathology of the spleen. This occurs when

cells from a damaged spleen seed elsewhere on the peritoneal surface and grow into nodules of

differentiated splenic tissue with varying size. They are supplied by newly formed arteries that

penetrate the capsule (563). Patients that undergo splenectomy due to trauma rather than

hematological disorders are more likely to experience splenic regeneration due to the increased

Page 133: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

110

likelihood of dissipation of splenic tissue, which favours autotransplantation, and therefore these

patients have an overall lower risk of subsequent infection and death. The rate of splenic

regeneration following traumatic splenectomy is as high as 66% (564). Another factor reducing

the risk of infection following splenectomy is the hypertrophy of functional splenuncli, which are

accessory spleens resulting from the incomplete fusion of separated fetal spleen tissue

originating from the dorsal mesogastrium (565). Accessory spleens are reported to have an

incidence of over 10% (566).

Splenic regeneration also occurs following manual autotransplantation of splenic tissue.

Following splenectomy in humans, the removed spleen is cut into thin slices and re-implanted

into omental pouches, in order for it to regenerate (530). The specific cells and molecules

regulating spleen tissue neogenesis have been investigated in animal models, although these are

poorly defined. It is known that following spleen transplantation, the process of tissue

regeneration involves tissue necrosis within 4 days of autotransplantation, regeneration of outer

tissue layer and differentiation of connective tissue into splenic reticular cells, vascular re-growth

within 8 days with lymphocytes around primitive vessels, and white pulp differentiation within 5

weeks. Remarkably, the newly generated spleen appears histologically normal, and red and white

pulp areas are clearly distinct (251).

A recent study showed that mouse splenic tissue regeneration following transplantation of

neonatal spleen capsule tissue into the renal subcapsular space depended only on the presence of

stromal cells in the transplant and on the LT pathway. RAG-/-

spleen capsules were capable of

regenerating following transplantation, whereas LTα-/-

spleen capsules failed to induce tissue

regeneration. These investigators point to the activity of LT-educated endothelial cell organizers

in orchestrating splenic regeneration (533).

A major question in this field is whether the function of these newly generated spleens is

comparable to a normal spleen. Reported post-operative findings include a reduction in

thrombocytosis, increased levels of IgM, and loss of Howell-Jolly bodies and red-pitted cells

(532). Whether these regenerated spleens are able to protect from subsequent bacterial infection

is less clear-cut, but the data does suggest that the volume of regenerated splenic tissue seems to

correlate with protection against post-splenectomy infection (567, 568). It is noteworthy that

factors including the age of the transplanted splenic tissue, the location to which it is

Page 134: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

111

transplanted, and the time between removal of tissue and transplantation are all factors

determining the ability of the splenic tissue to regenerate, whereas age of the donor seems not to

be an important factor.

Although it is clear that the spleen can regenerate following disorder/trauma and splenectomy

and autotransplantation, it is not clear whether it recovers from splenic atrophy. Splenic atrophy

was shown to be irreversible in a study of celiac disease patients. Necropsy studies in adult celiac

disease show the spleen to become thickly encapsulated with fibrous tissue, and the red pulp to

be replaced with fibrous tissue, processes which are not easily reversible (231). Of interest,

transplantation studies showed that splenic fragments do not grow as well if the spleen is also

present, and that there is a limit to the size to which splenic fragments will grow, a size not

exceeding that of the original spleen. This suggests that the spleen or an intermediary system

produces negative feedback inhibitors of growth that normally prevent regeneration (251, 531,

534, 569). This might explain why the spleen may not normally regenerate following atrophy.

In the LCMV clone 13-induced splenic atrophy model discussed in chapter 3 of this thesis,

spleens did not recover to pre-infection weight out to day 300 pi, which was the last time point

assessed. The reason for this is not defined, but might involve the age of the mice when atrophy

occurs and when virus is cleared. Tan et al. showed that grafting of whole-spleen capsules from

D3 donors resulted in 83% (20/24) of the grafts regenerating spleen tissue in recipient mice 4

weeks following transfer whereas only 38% (6/16) of adult spleen capsule grafts regenerated

normal spleen tissue (533). Therefore, as briefly mentioned above, the age of splenic tissue does

factor into its ability to regenerate. It is possible that the spleens from Cl13-infected mice are too

old to properly regenerate following viral clearance. To determine whether the spleen ever fully

regenerates to pre-infection weight following atrophy in this model, LCMV clone 13-infected

mice should be housed in specific pathogen-free conditions for at least 2 years.

4.7 Conclusion

My studies in chapter 2 have shown that 4-1BB and 4-1BBL constitutively interact on LPS-

activated DCs, adding to the growing body of evidence that receptors and ligands of the TNF

family have the ability to interact on multiple cell types, including interacting within a single cell

type. More work is required to understand the functional role of this interaction. In chapter 3, I

Page 135: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

112

revealed remarkably long-term splenic atrophy following LCMV clone 13 infection, months

after virus can no longer be detected. I also showed a role for NK cells in mediating splenic

atrophy at late stages following clone 13 infection. This raises the issue of whether NK cells are

prominent in human conditions with splenic atrophy, including chronic infections such as HIV.

Page 136: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

113

Chapter 5

References

Page 137: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

114

References

1. Oldstone MB. 2013. Lessons learned and concepts formed from study of the pathogenesis

of the two negative-strand viruses lymphocytic choriomeningitis and influenza. Proc Natl

Acad Sci U S A 110: 4180-3

2. Virgin HW, Wherry EJ, Ahmed R. 2009. Redefining chronic viral infection. Cell 138:

30-50

3. Akira S, Uematsu S, Takeuchi O. 2006. Pathogen recognition and innate immunity. Cell

124: 783-801

4. Takeuchi O, Akira S. 2009. Innate immunity to virus infection. Immunol Rev 227: 75-86

5. Barton GM, Kagan JC. 2009. A cell biological view of Toll-like receptor function:

regulation through compartmentalization. Nat Rev Immunol 9: 535-42

6. Kawai T, Akira S. 2010. The role of pattern-recognition receptors in innate immunity:

update on Toll-like receptors. Nat Immunol 11: 373-84

7. Kawai T, Akira S. 2009. The roles of TLRs, RLRs and NLRs in pathogen recognition. Int

Immunol 21: 317-37

8. Loo YM, Gale M, Jr. 2011. Immune signaling by RIG-I-like receptors. Immunity 34: 680-

92

9. Paz S, Sun Q, Nakhaei P, Romieu-Mourez R, Goubau D, Julkunen I, Lin R, Hiscott J.

2006. Induction of IRF-3 and IRF-7 phosphorylation following activation of the RIG-I

pathway. Cell Mol Biol (Noisy-le-grand) 52: 17-28

10. Dinarello CA. 2009. Immunological and inflammatory functions of the interleukin-1

family. Annu Rev Immunol 27: 519-50

11. Schroder K, Tschopp J. 2010. The inflammasomes. Cell 140: 821-32

12. Fogg DK, Sibon C, Miled C, Jung S, Aucouturier P, Littman DR, Cumano A, Geissmann

F. 2006. A clonogenic bone marrow progenitor specific for macrophages and dendritic

cells. Science 311: 83-7

13. Jakubzick C, Bogunovic M, Bonito AJ, Kuan EL, Merad M, Randolph GJ. 2008. Lymph-

migrating, tissue-derived dendritic cells are minor constituents within steady-state lymph

nodes. J Exp Med 205: 2839-50

14. Naik SH, Metcalf D, van Nieuwenhuijze A, Wicks I, Wu L, O'Keeffe M, Shortman K.

2006. Intrasplenic steady-state dendritic cell precursors that are distinct from monocytes.

Nat Immunol 7: 663-71

15. Varol C, Landsman L, Fogg DK, Greenshtein L, Gildor B, Margalit R, Kalchenko V,

Geissmann F, Jung S. 2007. Monocytes give rise to mucosal, but not splenic,

conventional dendritic cells. J Exp Med 204: 171-80

16. Liu K, Nussenzweig MC. 2010. Origin and development of dendritic cells. Immunol Rev

234: 45-54

17. Ginhoux F, Liu K, Helft J, Bogunovic M, Greter M, Hashimoto D, Price J, Yin N,

Bromberg J, Lira SA, Stanley ER, Nussenzweig M, Merad M. 2009. The origin and

development of nonlymphoid tissue CD103+ DCs. J Exp Med 206: 3115-30

18. Kabashima K, Banks TA, Ansel KM, Lu TT, Ware CF, Cyster JG. 2005. Intrinsic

lymphotoxin-beta receptor requirement for homeostasis of lymphoid tissue dendritic

cells. Immunity 22: 439-50

19. Liu K, Waskow C, Liu X, Yao K, Hoh J, Nussenzweig M. 2007. Origin of dendritic cells

in peripheral lymphoid organs of mice. Nat Immunol 8: 578-83

Page 138: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

115

20. Maraskovsky E, Brasel K, Teepe M, Roux ER, Lyman SD, Shortman K, McKenna HJ.

1996. Dramatic increase in the numbers of functionally mature dendritic cells in Flt3

ligand-treated mice: multiple dendritic cell subpopulations identified. J Exp Med 184:

1953-62

21. Waskow C, Liu K, Darrasse-Jeze G, Guermonprez P, Ginhoux F, Merad M, Shengelia T,

Yao K, Nussenzweig M. 2008. The receptor tyrosine kinase Flt3 is required for dendritic

cell development in peripheral lymphoid tissues. Nat Immunol 9: 676-83

22. Kamath AT, Henri S, Battye F, Tough DF, Shortman K. 2002. Developmental kinetics

and lifespan of dendritic cells in mouse lymphoid organs. Blood 100: 1734-41

23. Idoyaga J, Suda N, Suda K, Park CG, Steinman RM. 2009. Antibody to Langerin/CD207

localizes large numbers of CD8alpha+ dendritic cells to the marginal zone of mouse

spleen. Proc Natl Acad Sci U S A 106: 1524-9

24. Belz GT, Behrens GM, Smith CM, Miller JF, Jones C, Lejon K, Fathman CG, Mueller

SN, Shortman K, Carbone FR, Heath WR. 2002. The CD8alpha(+) dendritic cell is

responsible for inducing peripheral self-tolerance to tissue-associated antigens. J Exp

Med 196: 1099-104

25. den Haan JM, Lehar SM, Bevan MJ. 2000. CD8(+) but not CD8(-) dendritic cells cross-

prime cytotoxic T cells in vivo. J Exp Med 192: 1685-96

26. Iyoda T, Shimoyama S, Liu K, Omatsu Y, Akiyama Y, Maeda Y, Takahara K, Steinman

RM, Inaba K. 2002. The CD8+ dendritic cell subset selectively endocytoses dying cells

in culture and in vivo. J Exp Med 195: 1289-302

27. Shortman K, Heath WR. 2010. The CD8+ dendritic cell subset. Immunol Rev 234: 18-31

28. Dudziak D, Kamphorst AO, Heidkamp GF, Buchholz VR, Trumpfheller C, Yamazaki S,

Cheong C, Liu K, Lee HW, Park CG, Steinman RM, Nussenzweig MC. 2007.

Differential antigen processing by dendritic cell subsets in vivo. Science 315: 107-11

29. Kamphorst AO, Guermonprez P, Dudziak D, Nussenzweig MC. 2010. Route of antigen

uptake differentially impacts presentation by dendritic cells and activated monocytes. J

Immunol 185: 3426-35

30. Shortman K, Liu YJ. 2002. Mouse and human dendritic cell subtypes. Nat Rev Immunol

2: 151-61

31. Allan RS, Waithman J, Bedoui S, Jones CM, Villadangos JA, Zhan Y, Lew AM,

Shortman K, Heath WR, Carbone FR. 2006. Migratory dendritic cells transfer antigen to

a lymph node-resident dendritic cell population for efficient CTL priming. Immunity 25:

153-62

32. Ohl L, Mohaupt M, Czeloth N, Hintzen G, Kiafard Z, Zwirner J, Blankenstein T,

Henning G, Forster R. 2004. CCR7 governs skin dendritic cell migration under

inflammatory and steady-state conditions. Immunity 21: 279-88

33. Itano AA, Jenkins MK. 2003. Antigen presentation to naive CD4 T cells in the lymph

node. Nat Immunol 4: 733-9

34. Dzionek A, Fuchs A, Schmidt P, Cremer S, Zysk M, Miltenyi S, Buck DW, Schmitz J.

2000. BDCA-2, BDCA-3, and BDCA-4: three markers for distinct subsets of dendritic

cells in human peripheral blood. J Immunol 165: 6037-46

35. Bachem A, Guttler S, Hartung E, Ebstein F, Schaefer M, Tannert A, Salama A,

Movassaghi K, Opitz C, Mages HW, Henn V, Kloetzel PM, Gurka S, Kroczek RA. 2010.

Superior antigen cross-presentation and XCR1 expression define human

Page 139: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

116

CD11c+CD141+ cells as homologues of mouse CD8+ dendritic cells. J Exp Med 207:

1273-81

36. Crozat K, Guiton R, Contreras V, Feuillet V, Dutertre CA, Ventre E, Vu Manh TP,

Baranek T, Storset AK, Marvel J, Boudinot P, Hosmalin A, Schwartz-Cornil I, Dalod M.

2010. The XC chemokine receptor 1 is a conserved selective marker of mammalian cells

homologous to mouse CD8alpha+ dendritic cells. J Exp Med 207: 1283-92

37. Robbins SH, Walzer T, Dembele D, Thibault C, Defays A, Bessou G, Xu H, Vivier E,

Sellars M, Pierre P, Sharp FR, Chan S, Kastner P, Dalod M. 2008. Novel insights into the

relationships between dendritic cell subsets in human and mouse revealed by genome-

wide expression profiling. Genome Biol 9: R17

38. Crozat K, Guiton R, Guilliams M, Henri S, Baranek T, Schwartz-Cornil I, Malissen B,

Dalod M. 2010. Comparative genomics as a tool to reveal functional equivalences

between human and mouse dendritic cell subsets. Immunol Rev 234: 177-98

39. Perussia B, Fanning V, Trinchieri G. 1985. A leukocyte subset bearing HLA-DR antigens

is responsible for in vitro alpha interferon production in response to viruses. Nat Immun

Cell Growth Regul 4: 120-37

40. Cella M, Jarrossay D, Facchetti F, Alebardi O, Nakajima H, Lanzavecchia A, Colonna M.

1999. Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large

amounts of type I interferon. Nat Med 5: 919-23

41. Siegal FP, Kadowaki N, Shodell M, Fitzgerald-Bocarsly PA, Shah K, Ho S, Antonenko

S, Liu YJ. 1999. The nature of the principal type 1 interferon-producing cells in human

blood. Science 284: 1835-7

42. Colonna M, Trinchieri G, Liu YJ. 2004. Plasmacytoid dendritic cells in immunity. Nat

Immunol 5: 1219-26

43. O'Keeffe M, Hochrein H, Vremec D, Scott B, Hertzog P, Tatarczuch L, Shortman K.

2003. Dendritic cell precursor populations of mouse blood: identification of the murine

homologues of human blood plasmacytoid pre-DC2 and CD11c+ DC1 precursors. Blood

101: 1453-9

44. Crotty S. 2011. Follicular helper CD4 T cells (TFH). Annu Rev Immunol 29: 621-63

45. Granucci F, Foti M, Ricciardi-Castagnoli P. 2005. Dendritic cell biology. Adv Immunol

88: 193-233

46. Zygmunt B, Veldhoen M. 2011. T helper cell differentiation more than just cytokines.

Adv Immunol 109: 159-96

47. Maldonado-Lopez R, De Smedt T, Michel P, Godfroid J, Pajak B, Heirman C,

Thielemans K, Leo O, Urbain J, Moser M. 1999. CD8alpha+ and CD8alpha- subclasses

of dendritic cells direct the development of distinct T helper cells in vivo. J Exp Med 189:

587-92

48. Pulendran B, Smith JL, Caspary G, Brasel K, Pettit D, Maraskovsky E, Maliszewski CR.

1999. Distinct dendritic cell subsets differentially regulate the class of immune response

in vivo. Proc Natl Acad Sci U S A 96: 1036-41

49. Shen L, Rock KL. 2006. Priming of T cells by exogenous antigen cross-presented on

MHC class I molecules. Curr Opin Immunol 18: 85-91

50. Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K.

2000. Immunobiology of dendritic cells. Annu Rev Immunol 18: 767-811

51. Manicassamy S, Pulendran B. 2011. Dendritic cell control of tolerogenic responses.

Immunol Rev 241: 206-27

Page 140: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

117

52. Bevan MJ. 2004. Helping the CD8(+) T-cell response. Nat Rev Immunol 4: 595-602

53. Bennett SR, Carbone FR, Karamalis F, Flavell RA, Miller JF, Heath WR. 1998. Help for

cytotoxic-T-cell responses is mediated by CD40 signalling. Nature 393: 478-80

54. Ridge JP, Di Rosa F, Matzinger P. 1998. A conditioned dendritic cell can be a temporal

bridge between a CD4+ T-helper and a T-killer cell. Nature 393: 474-8

55. Schoenberger SP, Toes RE, van der Voort EI, Offringa R, Melief CJ. 1998. T-cell help

for cytotoxic T lymphocytes is mediated by CD40-CD40L interactions. Nature 393: 480-

3

56. Summers deLuca L, Ng D, Gao Y, Wortzman ME, Watts TH, Gommerman JL. 2011.

LTbetaR signaling in dendritic cells induces a type I IFN response that is required for

optimal clonal expansion of CD8+ T cells. Proc Natl Acad Sci U S A 108: 2046-51

57. Ng D, Gommerman JL. 2013. The Regulation of Immune Responses by DC Derived

Type I IFN. Front Immunol 4: 94

58. Bergtold A, Desai DD, Gavhane A, Clynes R. 2005. Cell surface recycling of internalized

antigen permits dendritic cell priming of B cells. Immunity 23: 503-14

59. Jego G, Pascual V, Palucka AK, Banchereau J. 2005. Dendritic cells control B cell

growth and differentiation. Curr Dir Autoimmun 8: 124-39

60. Qi H, Egen JG, Huang AY, Germain RN. 2006. Extrafollicular activation of lymph node

B cells by antigen-bearing dendritic cells. Science 312: 1672-6

61. Batista FD, Harwood NE. 2009. The who, how and where of antigen presentation to B

cells. Nat Rev Immunol 9: 15-27

62. Freer G, Matteucci D. 2009. Influence of dendritic cells on viral pathogenicity. PLoS

Pathog 5: e1000384

63. Geijtenbeek TB, van Kooyk Y. 2003. Pathogens target DC-SIGN to influence their fate

DC-SIGN functions as a pathogen receptor with broad specificity. APMIS 111: 698-714

64. de Witte L, Nabatov A, Geijtenbeek TB. 2008. Distinct roles for DC-SIGN+-dendritic

cells and Langerhans cells in HIV-1 transmission. Trends Mol Med 14: 12-9

65. Marzi A, Moller P, Hanna SL, Harrer T, Eisemann J, Steinkasserer A, Becker S,

Baribaud F, Pohlmann S. 2007. Analysis of the interaction of Ebola virus glycoprotein

with DC-SIGN (dendritic cell-specific intercellular adhesion molecule 3-grabbing

nonintegrin) and its homologue DC-SIGNR. J Infect Dis 196 Suppl 2: S237-46

66. Mahnke K, Guo M, Lee S, Sepulveda H, Swain SL, Nussenzweig M, Steinman RM.

2000. The dendritic cell receptor for endocytosis, DEC-205, can recycle and enhance

antigen presentation via major histocompatibility complex class II-positive lysosomal

compartments. J Cell Biol 151: 673-84

67. Bozzacco L, Trumpfheller C, Siegal FP, Mehandru S, Markowitz M, Carrington M,

Nussenzweig MC, Piperno AG, Steinman RM. 2007. DEC-205 receptor on dendritic

cells mediates presentation of HIV gag protein to CD8+ T cells in a spectrum of human

MHC I haplotypes. Proc Natl Acad Sci U S A 104: 1289-94

68. Lahoud MH, Ahmet F, Zhang JG, Meuter S, Policheni AN, Kitsoulis S, Lee CN,

O'Keeffe M, Sullivan LC, Brooks AG, Berry R, Rossjohn J, Mintern JD, Vega-Ramos J,

Villadangos JA, Nicola NA, Nussenzweig MC, Stacey KJ, Shortman K, Heath WR,

Caminschi I. 2012. DEC-205 is a cell surface receptor for CpG oligonucleotides. Proc

Natl Acad Sci U S A 109: 16270-5

69. Shrimpton RE, Butler M, Morel AS, Eren E, Hue SS, Ritter MA. 2009. CD205 (DEC-

205): a recognition receptor for apoptotic and necrotic self. Mol Immunol 46: 1229-39

Page 141: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

118

70. Zhang SS, Park CG, Zhang P, Bartra SS, Plano GV, Klena JD, Skurnik M, Hinnebusch

BJ, Chen T. 2008. Plasminogen activator Pla of Yersinia pestis utilizes murine DEC-205

(CD205) as a receptor to promote dissemination. J Biol Chem 283: 31511-21

71. Bousarghin L, Hubert P, Franzen E, Jacobs N, Boniver J, Delvenne P. 2005. Human

papillomavirus 16 virus-like particles use heparan sulfates to bind dendritic cells and

colocalize with langerin in Langerhans cells. J Gen Virol 86: 1297-305

72. Da Silva DM, Fausch SC, Verbeek JS, Kast WM. 2007. Uptake of human papillomavirus

virus-like particles by dendritic cells is mediated by Fcgamma receptors and contributes

to acquisition of T cell immunity. J Immunol 178: 7587-97

73. de Witte L, Nabatov A, Pion M, Fluitsma D, de Jong MA, de Gruijl T, Piguet V, van

Kooyk Y, Geijtenbeek TB. 2007. Langerin is a natural barrier to HIV-1 transmission by

Langerhans cells. Nat Med 13: 367-71

74. Di Pucchio T, Chatterjee B, Smed-Sorensen A, Clayton S, Palazzo A, Montes M, Xue Y,

Mellman I, Banchereau J, Connolly JE. 2008. Direct proteasome-independent cross-

presentation of viral antigen by plasmacytoid dendritic cells on major histocompatibility

complex class I. Nat Immunol 9: 551-7

75. Smit JJ, Lindell DM, Boon L, Kool M, Lambrecht BN, Lukacs NW. 2008. The balance

between plasmacytoid DC versus conventional DC determines pulmonary immunity to

virus infections. PLoS One 3: e1720

76. Sevilla N, McGavern DB, Teng C, Kunz S, Oldstone MB. 2004. Viral targeting of

hematopoietic progenitors and inhibition of DC maturation as a dual strategy for immune

subversion. J Clin Invest 113: 737-45

77. Lockridge KM, Zhou SS, Kravitz RH, Johnson JL, Sawai ET, Blewett EL, Barry PA.

2000. Primate cytomegaloviruses encode and express an IL-10-like protein. Virology

268: 272-80

78. Barends M, de Rond LG, Dormans J, van Oosten M, Boelen A, Neijens HJ, Osterhaus

AD, Kimman TG. 2004. Respiratory syncytial virus, pneumonia virus of mice, and

influenza A virus differently affect respiratory allergy in mice. Clin Exp Allergy 34: 488-

96

79. Bueno SM, Gonzalez PA, Pacheco R, Leiva ED, Cautivo KM, Tobar HE, Mora JE, Prado

CE, Zuniga JP, Jimenez J, Riedel CA, Kalergis AM. 2008. Host immunity during RSV

pathogenesis. Int Immunopharmacol 8: 1320-9

80. Frank I, Piatak M, Jr., Stoessel H, Romani N, Bonnyay D, Lifson JD, Pope M. 2002.

Infectious and whole inactivated simian immunodeficiency viruses interact similarly with

primate dendritic cells (DCs): differential intracellular fate of virions in mature and

immature DCs. J Virol 76: 2936-51

81. Freer G, Matteucci D, Mazzetti P, Tarabella F, Catalucci V, Bendinelli M. 2007. Effects

of feline immunodeficiency virus on feline monocyte-derived dendritic cells infected by

spinoculation. J Gen Virol 88: 2574-82

82. Izquierdo-Useros N, Blanco J, Erkizia I, Fernandez-Figueras MT, Borras FE, Naranjo-

Gomez M, Bofill M, Ruiz L, Clotet B, Martinez-Picado J. 2007. Maturation of blood-

derived dendritic cells enhances human immunodeficiency virus type 1 capture and

transmission. J Virol 81: 7559-70

83. Palucka K, Banchereau J. 2012. Cancer immunotherapy via dendritic cells. Nat Rev

Cancer 12: 265-77

Page 142: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

119

84. Palucka K, Banchereau J. 2013. Dendritic-cell-based therapeutic cancer vaccines.

Immunity 39: 38-48

85. Lapenta C, Santini SM, Logozzi M, Spada M, Andreotti M, Di Pucchio T, Parlato S,

Belardelli F. 2003. Potent immune response against HIV-1 and protection from virus

challenge in hu-PBL-SCID mice immunized with inactivated virus-pulsed dendritic cells

generated in the presence of IFN-alpha. J Exp Med 198: 361-7

86. Lu W, Wu X, Lu Y, Guo W, Andrieu JM. 2003. Therapeutic dendritic-cell vaccine for

simian AIDS. Nat Med 9: 27-32

87. Yoshida A, Tanaka R, Murakami T, Takahashi Y, Koyanagi Y, Nakamura M, Ito M,

Yamamoto N, Tanaka Y. 2003. Induction of protective immune responses against R5

human immunodeficiency virus type 1 (HIV-1) infection in hu-PBL-SCID mice by

intrasplenic immunization with HIV-1-pulsed dendritic cells: possible involvement of a

novel factor of human CD4(+) T-cell origin. J Virol 77: 8719-28

88. Aline F, Brand D, Bout D, Pierre J, Fouquenet D, Verrier B, Dimier-Poisson I. 2007.

Generation of specific Th1 and CD8+ T-cell responses by immunization with mouse

CD8+ dendritic cells loaded with HIV-1 viral lysate or envelope glycoproteins. Microbes

Infect 9: 536-43

89. Garcia F, Climent N, Guardo AC, Gil C, Leon A, Autran B, Lifson JD, Martinez-Picado

J, Dalmau J, Clotet B, Gatell JM, Plana M, Gallart T. 2013. A dendritic cell-based

vaccine elicits T cell responses associated with control of HIV-1 replication. Sci Transl

Med 5: 166ra2

90. Spits H, Artis D, Colonna M, Diefenbach A, Di Santo JP, Eberl G, Koyasu S, Locksley

RM, McKenzie AN, Mebius RE, Powrie F, Vivier E. 2013. Innate lymphoid cells--a

proposal for uniform nomenclature. Nat Rev Immunol 13: 145-9

91. Vivier E, Tomasello E, Baratin M, Walzer T, Ugolini S. 2008. Functions of natural killer

cells. Nat Immunol 9: 503-10

92. Yokoyama WM, Kim S, French AR. 2004. The dynamic life of natural killer cells. Annu

Rev Immunol 22: 405-29

93. Di Santo JP. 2006. Natural killer cell developmental pathways: a question of balance.

Annu Rev Immunol 24: 257-86

94. Williams NS, Klem J, Puzanov IJ, Sivakumar PV, Bennett M, Kumar V. 1999.

Differentiation of NK1.1+, Ly49+ NK cells from flt3+ multipotent marrow progenitor

cells. J Immunol 163: 2648-56

95. Schmitt TM, Zuniga-Pflucker JC. 2002. Induction of T cell development from

hematopoietic progenitor cells by delta-like-1 in vitro. Immunity 17: 749-56

96. Lehar SM, Dooley J, Farr AG, Bevan MJ. 2005. Notch ligands Delta 1 and Jagged1

transmit distinct signals to T-cell precursors. Blood 105: 1440-7

97. Kim S, Iizuka K, Kang HS, Dokun A, French AR, Greco S, Yokoyama WM. 2002. In

vivo developmental stages in murine natural killer cell maturation. Nat Immunol 3: 523-8

98. Anfossi N, Andre P, Guia S, Falk CS, Roetynck S, Stewart CA, Breso V, Frassati C,

Reviron D, Middleton D, Romagne F, Ugolini S, Vivier E. 2006. Human NK cell

education by inhibitory receptors for MHC class I. Immunity 25: 331-42

99. Chalifour A, Scarpellino L, Back J, Brodin P, Devevre E, Gros F, Levy F, Leclercq G,

Hoglund P, Beermann F, Held W. 2009. A Role for cis Interaction between the Inhibitory

Ly49A receptor and MHC class I for natural killer cell education. Immunity 30: 337-47

Page 143: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

120

100. Fernandez NC, Treiner E, Vance RE, Jamieson AM, Lemieux S, Raulet DH. 2005. A

subset of natural killer cells achieves self-tolerance without expressing inhibitory

receptors specific for self-MHC molecules. Blood 105: 4416-23

101. Johansson S, Johansson M, Rosmaraki E, Vahlne G, Mehr R, Salmon-Divon M,

Lemonnier F, Karre K, Hoglund P. 2005. Natural killer cell education in mice with single

or multiple major histocompatibility complex class I molecules. J Exp Med 201: 1145-55

102. Kim S, Poursine-Laurent J, Truscott SM, Lybarger L, Song YJ, Yang L, French AR,

Sunwoo JB, Lemieux S, Hansen TH, Yokoyama WM. 2005. Licensing of natural killer

cells by host major histocompatibility complex class I molecules. Nature 436: 709-13

103. Sun JC, Lanier LL. 2011. NK cell development, homeostasis and function: parallels with

CD8(+) T cells. Nat Rev Immunol 11: 645-57

104. Brady J, Carotta S, Thong RP, Chan CJ, Hayakawa Y, Smyth MJ, Nutt SL. 2010. The

interactions of multiple cytokines control NK cell maturation. J Immunol 185: 6679-88

105. Loza MJ, Peters SP, Zangrilli JG, Perussia B. 2002. Distinction between IL-13+ and IFN-

gamma+ natural killer cells and regulation of their pool size by IL-4. Eur J Immunol 32:

413-23

106. Loza MJ, Zamai L, Azzoni L, Rosati E, Perussia B. 2002. Expression of type 1

(interferon gamma) and type 2 (interleukin-13, interleukin-5) cytokines at distinct stages

of natural killer cell differentiation from progenitor cells. Blood 99: 1273-81

107. Perussia B, Chen Y, Loza MJ. 2005. Peripheral NK cell phenotypes: multiple changing

of faces of an adapting, developing cell. Mol Immunol 42: 385-95

108. Di Santo JP, Vosshenrich CA. 2006. Bone marrow versus thymic pathways of natural

killer cell development. Immunol Rev 214: 35-46

109. Ma A, Koka R, Burkett P. 2006. Diverse functions of IL-2, IL-15, and IL-7 in lymphoid

homeostasis. Annu Rev Immunol 24: 657-79

110. Dubois S, Mariner J, Waldmann TA, Tagaya Y. 2002. IL-15Ralpha recycles and presents

IL-15 In trans to neighboring cells. Immunity 17: 537-47

111. Lucas M, Schachterle W, Oberle K, Aichele P, Diefenbach A. 2007. Dendritic cells

prime natural killer cells by trans-presenting interleukin 15. Immunity 26: 503-17

112. Mortier E, Woo T, Advincula R, Gozalo S, Ma A. 2008. IL-15Ralpha chaperones IL-15

to stable dendritic cell membrane complexes that activate NK cells via trans presentation.

J Exp Med 205: 1213-25

113. Cooper MA, Bush JE, Fehniger TA, VanDeusen JB, Waite RE, Liu Y, Aguila HL,

Caligiuri MA. 2002. In vivo evidence for a dependence on interleukin 15 for survival of

natural killer cells. Blood 100: 3633-8

114. Huntington ND, Puthalakath H, Gunn P, Naik E, Michalak EM, Smyth MJ, Tabarias H,

Degli-Esposti MA, Dewson G, Willis SN, Motoyama N, Huang DC, Nutt SL, Tarlinton

DM, Strasser A. 2007. Interleukin 15-mediated survival of natural killer cells is

determined by interactions among Bim, Noxa and Mcl-1. Nat Immunol 8: 856-63

115. Ranson T, Vosshenrich CA, Corcuff E, Richard O, Muller W, Di Santo JP. 2003. IL-15 is

an essential mediator of peripheral NK-cell homeostasis. Blood 101: 4887-93

116. Gregoire C, Chasson L, Luci C, Tomasello E, Geissmann F, Vivier E, Walzer T. 2007.

The trafficking of natural killer cells. Immunol Rev 220: 169-82

117. Jamieson AM, Isnard P, Dorfman JR, Coles MC, Raulet DH. 2004. Turnover and

proliferation of NK cells in steady state and lymphopenic conditions. J Immunol 172:

864-70

Page 144: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

121

118. Walzer T, Blery M, Chaix J, Fuseri N, Chasson L, Robbins SH, Jaeger S, Andre P,

Gauthier L, Daniel L, Chemin K, Morel Y, Dalod M, Imbert J, Pierres M, Moretta A,

Romagne F, Vivier E. 2007. Identification, activation, and selective in vivo ablation of

mouse NK cells via NKp46. Proc Natl Acad Sci U S A 104: 3384-9

119. Zhang Y, Wallace DL, de Lara CM, Ghattas H, Asquith B, Worth A, Griffin GE, Taylor

GP, Tough DF, Beverley PC, Macallan DC. 2007. In vivo kinetics of human natural

killer cells: the effects of ageing and acute and chronic viral infection. Immunology 121:

258-65

120. Prlic M, Blazar BR, Farrar MA, Jameson SC. 2003. In vivo survival and homeostatic

proliferation of natural killer cells. J Exp Med 197: 967-76

121. Sun JC, Beilke JN, Bezman NA, Lanier LL. 2011. Homeostatic proliferation generates

long-lived natural killer cells that respond against viral infection. J Exp Med 208: 357-68

122. Chiossone L, Chaix J, Fuseri N, Roth C, Vivier E, Walzer T. 2009. Maturation of mouse

NK cells is a 4-stage developmental program. Blood 113: 5488-96

123. Hayakawa Y, Smyth MJ. 2006. CD27 dissects mature NK cells into two subsets with

distinct responsiveness and migratory capacity. J Immunol 176: 1517-24

124. Cooper MA, Fehniger TA, Caligiuri MA. 2001. The biology of human natural killer-cell

subsets. Trends Immunol 22: 633-40

125. Raulet DH, Vance RE. 2006. Self-tolerance of natural killer cells. Nat Rev Immunol 6:

520-31

126. Lanier LL. 2005. NK cell recognition. Annu Rev Immunol 23: 225-74

127. Diefenbach A, Tomasello E, Lucas M, Jamieson AM, Hsia JK, Vivier E, Raulet DH.

2002. Selective associations with signaling proteins determine stimulatory versus

costimulatory activity of NKG2D. Nat Immunol 3: 1142-9

128. Gilfillan S, Ho EL, Cella M, Yokoyama WM, Colonna M. 2002. NKG2D recruits two

distinct adapters to trigger NK cell activation and costimulation. Nat Immunol 3: 1150-5

129. Billadeau DD, Upshaw JL, Schoon RA, Dick CJ, Leibson PJ. 2003. NKG2D-DAP10

triggers human NK cell-mediated killing via a Syk-independent regulatory pathway. Nat

Immunol 4: 557-64

130. Jost S, Altfeld M. 2013. Control of human viral infections by natural killer cells. Annu

Rev Immunol 31: 163-94

131. Lanier LL, Corliss BC, Wu J, Leong C, Phillips JH. 1998. Immunoreceptor DAP12

bearing a tyrosine-based activation motif is involved in activating NK cells. Nature 391:

703-7

132. Wu J, Song Y, Bakker AB, Bauer S, Spies T, Lanier LL, Phillips JH. 1999. An activating

immunoreceptor complex formed by NKG2D and DAP10. Science 285: 730-2

133. Raulet DH, Gasser S, Gowen BG, Deng W, Jung H. 2013. Regulation of ligands for the

NKG2D activating receptor. Annu Rev Immunol 31: 413-41

134. Carayannopoulos LN, Naidenko OV, Fremont DH, Yokoyama WM. 2002. Cutting edge:

murine UL16-binding protein-like transcript 1: a newly described transcript encoding a

high-affinity ligand for murine NKG2D. J Immunol 169: 4079-83

135. Diefenbach A, Hsia JK, Hsiung MY, Raulet DH. 2003. A novel ligand for the NKG2D

receptor activates NK cells and macrophages and induces tumor immunity. Eur J

Immunol 33: 381-91

136. Nice TJ, Coscoy L, Raulet DH. 2009. Posttranslational regulation of the NKG2D ligand

Mult1 in response to cell stress. J Exp Med 206: 287-98

Page 145: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

122

137. Nice TJ, Deng W, Coscoy L, Raulet DH. 2010. Stress-regulated targeting of the NKG2D

ligand Mult1 by a membrane-associated RING-CH family E3 ligase. J Immunol 185:

5369-76

138. Yokoyama WM, Riley JK. 2008. NK cells and their receptors. Reprod Biomed Online 16:

173-91

139. Mandelboim O, Lieberman N, Lev M, Paul L, Arnon TI, Bushkin Y, Davis DM,

Strominger JL, Yewdell JW, Porgador A. 2001. Recognition of haemagglutinins on

virus-infected cells by NKp46 activates lysis by human NK cells. Nature 409: 1055-60

140. Bloushtain N, Qimron U, Bar-Ilan A, Hershkovitz O, Gazit R, Fima E, Korc M,

Vlodavsky I, Bovin NV, Porgador A. 2004. Membrane-associated heparan sulfate

proteoglycans are involved in the recognition of cellular targets by NKp30 and NKp46. J

Immunol 173: 2392-401

141. Pogge von Strandmann E, Simhadri VR, von Tresckow B, Sasse S, Reiners KS, Hansen

HP, Rothe A, Boll B, Simhadri VL, Borchmann P, McKinnon PJ, Hallek M, Engert A.

2007. Human leukocyte antigen-B-associated transcript 3 is released from tumor cells

and engages the NKp30 receptor on natural killer cells. Immunity 27: 965-74

142. Halfteck GG, Elboim M, Gur C, Achdout H, Ghadially H, Mandelboim O. 2009.

Enhanced in vivo growth of lymphoma tumors in the absence of the NK-activating

receptor NKp46/NCR1. J Immunol 182: 2221-30

143. Pessino A, Sivori S, Bottino C, Malaspina A, Morelli L, Moretta L, Biassoni R, Moretta

A. 1998. Molecular cloning of NKp46: a novel member of the immunoglobulin

superfamily involved in triggering of natural cytotoxicity. J Exp Med 188: 953-60

144. Karre K, Ljunggren HG, Piontek G, Kiessling R. 1986. Selective rejection of H-2-

deficient lymphoma variants suggests alternative immune defence strategy. Nature 319:

675-8

145. Ljunggren HG, Karre K. 1990. In search of the 'missing self': MHC molecules and NK

cell recognition. Immunol Today 11: 237-44

146. Correa I, Raulet DH. 1995. Binding of diverse peptides to MHC class I molecules

inhibits target cell lysis by activated natural killer cells. Immunity 2: 61-71

147. Hanke T, Takizawa H, McMahon CW, Busch DH, Pamer EG, Miller JD, Altman JD, Liu

Y, Cado D, Lemonnier FA, Bjorkman PJ, Raulet DH. 1999. Direct assessment of MHC

class I binding by seven Ly49 inhibitory NK cell receptors. Immunity 11: 67-77

148. Orihuela M, Margulies DH, Yokoyama WM. 1996. The natural killer cell receptor Ly-

49A recognizes a peptide-induced conformational determinant on its major

histocompatibility complex class I ligand. Proc Natl Acad Sci U S A 93: 11792-7

149. Veinotte LL, Wilhelm BT, Mager DL, Takei F. 2003. Acquisition of MHC-specific

receptors on murine natural killer cells. Crit Rev Immunol 23: 251-66

150. Raulet DH, Held W, Correa I, Dorfman JR, Wu MF, Corral L. 1997. Specificity,

tolerance and developmental regulation of natural killer cells defined by expression of

class I-specific Ly49 receptors. Immunol Rev 155: 41-52

151. Doucey MA, Scarpellino L, Zimmer J, Guillaume P, Luescher IF, Bron C, Held W. 2004.

Cis association of Ly49A with MHC class I restricts natural killer cell inhibition. Nat

Immunol 5: 328-36

152. Scarpellino L, Oeschger F, Guillaume P, Coudert JD, Levy F, Leclercq G, Held W. 2007.

Interactions of Ly49 family receptors with MHC class I ligands in trans and cis. J

Immunol 178: 1277-84

Page 146: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

123

153. Arase H, Mocarski ES, Campbell AE, Hill AB, Lanier LL. 2002. Direct recognition of

cytomegalovirus by activating and inhibitory NK cell receptors. Science 296: 1323-6

154. Smith HR, Heusel JW, Mehta IK, Kim S, Dorner BG, Naidenko OV, Iizuka K, Furukawa

H, Beckman DL, Pingel JT, Scalzo AA, Fremont DH, Yokoyama WM. 2002.

Recognition of a virus-encoded ligand by a natural killer cell activation receptor. Proc

Natl Acad Sci U S A 99: 8826-31

155. Barten R, Torkar M, Haude A, Trowsdale J, Wilson MJ. 2001. Divergent and convergent

evolution of NK-cell receptors. Trends Immunol 22: 52-7

156. Colonna M, Samaridis J. 1995. Cloning of immunoglobulin-superfamily members

associated with HLA-C and HLA-B recognition by human natural killer cells. Science

268: 405-8

157. Wagtmann N, Biassoni R, Cantoni C, Verdiani S, Malnati MS, Vitale M, Bottino C,

Moretta L, Moretta A, Long EO. 1995. Molecular clones of the p58 NK cell receptor

reveal immunoglobulin-related molecules with diversity in both the extra- and

intracellular domains. Immunity 2: 439-49

158. Pegram HJ, Andrews DM, Smyth MJ, Darcy PK, Kershaw MH. 2011. Activating and

inhibitory receptors of natural killer cells. Immunol Cell Biol 89: 216-24

159. Held W, Roland J, Raulet DH. 1995. Allelic exclusion of Ly49-family genes encoding

class I MHC-specific receptors on NK cells. Nature 376: 355-8

160. Held W, Raulet DH. 1997. Expression of the Ly49A gene in murine natural killer cell

clones is predominantly but not exclusively mono-allelic. Eur J Immunol 27: 2876-84

161. Saleh A, Davies GE, Pascal V, Wright PW, Hodge DL, Cho EH, Lockett SJ, Abshari M,

Anderson SK. 2004. Identification of probabilistic transcriptional switches in the Ly49

gene cluster: a eukaryotic mechanism for selective gene activation. Immunity 21: 55-66

162. Borrego F, Ulbrecht M, Weiss EH, Coligan JE, Brooks AG. 1998. Recognition of human

histocompatibility leukocyte antigen (HLA)-E complexed with HLA class I signal

sequence-derived peptides by CD94/NKG2 confers protection from natural killer cell-

mediated lysis. J Exp Med 187: 813-8

163. Lee N, Goodlett DR, Ishitani A, Marquardt H, Geraghty DE. 1998. HLA-E surface

expression depends on binding of TAP-dependent peptides derived from certain HLA

class I signal sequences. J Immunol 160: 4951-60

164. Mingari MC, Ponte M, Bertone S, Schiavetti F, Vitale C, Bellomo R, Moretta A, Moretta

L. 1998. HLA class I-specific inhibitory receptors in human T lymphocytes: interleukin

15-induced expression of CD94/NKG2A in superantigen- or alloantigen-activated CD8+

T cells. Proc Natl Acad Sci U S A 95: 1172-7

165. Kirkham CL, Carlyle JR. 2014. Complexity and Diversity of the NKR-P1:Clr

(Klrb1:Clec2) Recognition Systems. Front Immunol 5: 214

166. Aldemir H, Prod'homme V, Dumaurier MJ, Retiere C, Poupon G, Cazareth J, Bihl F,

Braud VM. 2005. Cutting edge: lectin-like transcript 1 is a ligand for the CD161 receptor.

J Immunol 175: 7791-5

167. Rosen DB, Bettadapura J, Alsharifi M, Mathew PA, Warren HS, Lanier LL. 2005.

Cutting edge: lectin-like transcript-1 is a ligand for the inhibitory human NKR-P1A

receptor. J Immunol 175: 7796-9

168. Tassi I, Colonna M. 2005. The cytotoxicity receptor CRACC (CS-1) recruits EAT-2 and

activates the PI3K and phospholipase Cgamma signaling pathways in human NK cells. J

Immunol 175: 7996-8002

Page 147: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

124

169. Veillette A. 2006. NK cell regulation by SLAM family receptors and SAP-related

adapters. Immunol Rev 214: 22-34

170. Degli-Esposti MA, Smyth MJ. 2005. Close encounters of different kinds: dendritic cells

and NK cells take centre stage. Nat Rev Immunol 5: 112-24

171. Moretta L, Ferlazzo G, Bottino C, Vitale M, Pende D, Mingari MC, Moretta A. 2006.

Effector and regulatory events during natural killer-dendritic cell interactions. Immunol

Rev 214: 219-28

172. Walzer T, Dalod M, Robbins SH, Zitvogel L, Vivier E. 2005. Natural-killer cells and

dendritic cells: "l'union fait la force". Blood 106: 2252-8

173. Piccioli D, Sbrana S, Melandri E, Valiante NM. 2002. Contact-dependent stimulation and

inhibition of dendritic cells by natural killer cells. J Exp Med 195: 335-41

174. Martin-Fontecha A, Thomsen LL, Brett S, Gerard C, Lipp M, Lanzavecchia A, Sallusto

F. 2004. Induced recruitment of NK cells to lymph nodes provides IFN-gamma for T(H)1

priming. Nat Immunol 5: 1260-5

175. Morandi B, Bougras G, Muller WA, Ferlazzo G, Munz C. 2006. NK cells of human

secondary lymphoid tissues enhance T cell polarization via IFN-gamma secretion. Eur J

Immunol 36: 2394-400

176. Nakayama M, Takeda K, Kawano M, Takai T, Ishii N, Ogasawara K. 2011. Natural killer

(NK)-dendritic cell interactions generate MHC class II-dressed NK cells that regulate

CD4+ T cells. Proc Natl Acad Sci U S A 108: 18360-5

177. Takeda K, Dennert G. 1993. The development of autoimmunity in C57BL/6 lpr mice

correlates with the disappearance of natural killer type 1-positive cells: evidence for their

suppressive action on bone marrow stem cell proliferation, B cell immunoglobulin

secretion, and autoimmune symptoms. J Exp Med 177: 155-64

178. van Dommelen SL, Sumaria N, Schreiber RD, Scalzo AA, Smyth MJ, Degli-Esposti MA.

2006. Perforin and granzymes have distinct roles in defensive immunity and

immunopathology. Immunity 25: 835-48

179. Sun JC, Beilke JN, Lanier LL. 2009. Adaptive immune features of natural killer cells.

Nature 457: 557-61

180. Welsh RM, Waggoner SN. 2013. NK cells controlling virus-specific T cells: Rheostats

for acute vs. persistent infections. Virology 435: 37-45

181. Burshtyn DN. 2013. NK cells and poxvirus infection. Front Immunol 4: 7

182. Lee SH, Miyagi T, Biron CA. 2007. Keeping NK cells in highly regulated antiviral

warfare. Trends Immunol 28: 252-9

183. Scalzo AA, Corbett AJ, Rawlinson WD, Scott GM, Degli-Esposti MA. 2007. The

interplay between host and viral factors in shaping the outcome of cytomegalovirus

infection. Immunol Cell Biol 85: 46-54

184. Gazit R, Gruda R, Elboim M, Arnon TI, Katz G, Achdout H, Hanna J, Qimron U, Landau

G, Greenbaum E, Zakay-Rones Z, Porgador A, Mandelboim O. 2006. Lethal influenza

infection in the absence of the natural killer cell receptor gene Ncr1. Nat Immunol 7: 517-

23

185. Stein-Streilein J, Guffee J. 1986. In vivo treatment of mice and hamsters with antibodies

to asialo GM1 increases morbidity and mortality to pulmonary influenza infection. J

Immunol 136: 1435-41

186. Asmal M, Sun Y, Lane S, Yeh W, Schmidt SD, Mascola JR, Letvin NL. 2011. Antibody-

dependent cell-mediated viral inhibition emerges after simian immunodeficiency virus

Page 148: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

125

SIVmac251 infection of rhesus monkeys coincident with gp140-binding antibodies and is

effective against neutralization-resistant viruses. J Virol 85: 5465-75

187. Hellmann I, Lim SY, Gelman RS, Letvin NL. 2011. Association of activating KIR copy

number variation of NK cells with containment of SIV replication in rhesus monkeys.

PLoS Pathog 7: e1002436

188. Sun Y, Asmal M, Lane S, Permar SR, Schmidt SD, Mascola JR, Letvin NL. 2011.

Antibody-dependent cell-mediated cytotoxicity in simian immunodeficiency virus-

infected rhesus monkeys. J Virol 85: 6906-12

189. Flores-Villanueva PO, Yunis EJ, Delgado JC, Vittinghoff E, Buchbinder S, Leung JY,

Uglialoro AM, Clavijo OP, Rosenberg ES, Kalams SA, Braun JD, Boswell SL, Walker

BD, Goldfeld AE. 2001. Control of HIV-1 viremia and protection from AIDS are

associated with HLA-Bw4 homozygosity. Proc Natl Acad Sci U S A 98: 5140-5

190. Martin MP, Gao X, Lee JH, Nelson GW, Detels R, Goedert JJ, Buchbinder S, Hoots K,

Vlahov D, Trowsdale J, Wilson M, O'Brien SJ, Carrington M. 2002. Epistatic interaction

between KIR3DS1 and HLA-B delays the progression to AIDS. Nat Genet 31: 429-34

191. Vidal SM, Lanier LL. 2006. NK cell recognition of mouse cytomegalovirus-infected

cells. Curr Top Microbiol Immunol 298: 183-206

192. Voigt S, Mesci A, Ettinger J, Fine JH, Chen P, Chou W, Carlyle JR. 2007.

Cytomegalovirus evasion of innate immunity by subversion of the NKR-P1B:Clr-b

missing-self axis. Immunity 26: 617-27

193. Williams KJ, Wilson E, Davidson CL, Aguilar OA, Fu L, Carlyle JR, Burshtyn DN.

2012. Poxvirus infection-associated downregulation of C-type lectin-related-b prevents

NK cell inhibition by NK receptor protein-1B. J Immunol 188: 4980-91

194. Fine JH, Chen P, Mesci A, Allan DS, Gasser S, Raulet DH, Carlyle JR. 2010.

Chemotherapy-induced genotoxic stress promotes sensitivity to natural killer cell

cytotoxicity by enabling missing-self recognition. Cancer Res 70: 7102-13

195. Norris BA, Uebelhoer LS, Nakaya HI, Price AA, Grakoui A, Pulendran B. 2013. Chronic

but not acute virus infection induces sustained expansion of myeloid suppressor cell

numbers that inhibit viral-specific T cell immunity. Immunity 38: 309-21

196. Waggoner SN, Daniels KA, Welsh RM. 2014. Therapeutic depletion of natural killer

cells controls persistent infection. J Virol 88: 1953-60

197. Cesta MF. 2006. Normal structure, function, and histology of the spleen. Toxicol Pathol

34: 455-65

198. Mebius RE, Kraal G. 2005. Structure and function of the spleen. Nat Rev Immunol 5:

606-16

199. Green MC. 1967. A defect of the splanchnic mesoderm caused by the mutant gene

dominant hemimelia in the mouse. Dev Biol 15: 62-89

200. Hecksher-Sorensen J, Watson RP, Lettice LA, Serup P, Eley L, De Angelis C, Ahlgren

U, Hill RE. 2004. The splanchnic mesodermal plate directs spleen and pancreatic

laterality, and is regulated by Bapx1/Nkx3.2. Development 131: 4665-75

201. Herzer U, Crocoll A, Barton D, Howells N, Englert C. 1999. The Wilms tumor

suppressor gene wt1 is required for development of the spleen. Curr Biol 9: 837-40

202. Lu J, Chang P, Richardson JA, Gan L, Weiler H, Olson EN. 2000. The basic helix-loop-

helix transcription factor capsulin controls spleen organogenesis. Proc Natl Acad Sci U S

A 97: 9525-30

Page 149: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

126

203. Roberts CW, Shutter JR, Korsmeyer SJ. 1994. Hox11 controls the genesis of the spleen.

Nature 368: 747-9

204. Brendolan A, Ferretti E, Salsi V, Moses K, Quaggin S, Blasi F, Cleary ML, Selleri L.

2005. A Pbx1-dependent genetic and transcriptional network regulates spleen ontogeny.

Development 132: 3113-26

205. Mebius RE, Rennert P, Weissman IL. 1997. Developing lymph nodes collect CD4+CD3-

LTbeta+ cells that can differentiate to APC, NK cells, and follicular cells but not T or B

cells. Immunity 7: 493-504

206. Seifert MF, Marks SC, Jr. 1985. The regulation of hemopoiesis in the spleen. Experientia

41: 192-9

207. Ansel KM, Ngo VN, Hyman PL, Luther SA, Forster R, Sedgwick JD, Browning JL, Lipp

M, Cyster JG. 2000. A chemokine-driven positive feedback loop organizes lymphoid

follicles. Nature 406: 309-14

208. Dejardin E, Droin NM, Delhase M, Haas E, Cao Y, Makris C, Li ZW, Karin M, Ware

CF, Green DR. 2002. The lymphotoxin-beta receptor induces different patterns of gene

expression via two NF-kappaB pathways. Immunity 17: 525-35

209. Vondenhoff MF, Desanti GE, Cupedo T, Bertrand JY, Cumano A, Kraal G, Mebius RE,

Golub R. 2008. Separation of splenic red and white pulp occurs before birth in a

LTalphabeta-independent manner. J Leukoc Biol 84: 152-61

210. Katakai T, Suto H, Sugai M, Gonda H, Togawa A, Suematsu S, Ebisuno Y, Katagiri K,

Kinashi T, Shimizu A. 2008. Organizer-like reticular stromal cell layer common to adult

secondary lymphoid organs. J Immunol 181: 6189-200

211. Koning JJ, Mebius RE. 2012. Interdependence of stromal and immune cells for lymph

node function. Trends Immunol 33: 264-70

212. Mueller SN, Germain RN. 2009. Stromal cell contributions to the homeostasis and

functionality of the immune system. Nat Rev Immunol 9: 618-29

213. Castagnaro L, Lenti E, Maruzzelli S, Spinardi L, Migliori E, Farinello D, Sitia G,

Harrelson Z, Evans SM, Guidotti LG, Harvey RP, Brendolan A. 2013. Nkx2-

5(+)islet1(+) mesenchymal precursors generate distinct spleen stromal cell subsets and

participate in restoring stromal network integrity. Immunity 38: 782-91

214. Shinkura R, Kitada K, Matsuda F, Tashiro K, Ikuta K, Suzuki M, Kogishi K, Serikawa T,

Honjo T. 1999. Alymphoplasia is caused by a point mutation in the mouse gene encoding

Nf-kappa b-inducing kinase. Nat Genet 22: 74-7

215. De Togni P, Goellner J, Ruddle NH, Streeter PR, Fick A, Mariathasan S, Smith SC,

Carlson R, Shornick LP, Strauss-Schoenberger J, et al. 1994. Abnormal development of

peripheral lymphoid organs in mice deficient in lymphotoxin. Science 264: 703-7

216. Miyawaki S, Nakamura Y, Suzuka H, Koba M, Yasumizu R, Ikehara S, Shibata Y. 1994.

A new mutation, aly, that induces a generalized lack of lymph nodes accompanied by

immunodeficiency in mice. Eur J Immunol 24: 429-34

217. Bronte V, Pittet MJ. 2013. The spleen in local and systemic regulation of immunity.

Immunity 39: 806-18

218. Luther SA, Tang HL, Hyman PL, Farr AG, Cyster JG. 2000. Coexpression of the

chemokines ELC and SLC by T zone stromal cells and deletion of the ELC gene in the

plt/plt mouse. Proc Natl Acad Sci U S A 97: 12694-9

219. Ngo VN, Korner H, Gunn MD, Schmidt KN, Riminton DS, Cooper MD, Browning JL,

Sedgwick JD, Cyster JG. 1999. Lymphotoxin alpha/beta and tumor necrosis factor are

Page 150: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

127

required for stromal cell expression of homing chemokines in B and T cell areas of the

spleen. J Exp Med 189: 403-12

220. Turley SJ, Fletcher AL, Elpek KG. 2010. The stromal and haematopoietic antigen-

presenting cells that reside in secondary lymphoid organs. Nat Rev Immunol 10: 813-25

221. Karrer U, Althage A, Odermatt B, Roberts CW, Korsmeyer SJ, Miyawaki S, Hengartner

H, Zinkernagel RM. 1997. On the key role of secondary lymphoid organs in antiviral

immune responses studied in alymphoplastic (aly/aly) and spleenless (Hox11(-)/-) mutant

mice. J Exp Med 185: 2157-70

222. Ciavarra RP, Buhrer K, Van Rooijen N, Tedeschi B. 1997. T cell priming against

vesicular stomatitis virus analyzed in situ: red pulp macrophages, but neither marginal

metallophilic nor marginal zone macrophages, are required for priming CD4+ and CD8+

T cells. J Immunol 158: 1749-55

223. Seiler P, Aichele P, Odermatt B, Hengartner H, Zinkernagel RM, Schwendener RA.

1997. Crucial role of marginal zone macrophages and marginal zone metallophils in the

clearance of lymphocytic choriomeningitis virus infection. Eur J Immunol 27: 2626-33

224. Al-Salem AH. 2011. Splenic complications of sickle cell anemia and the role of

splenectomy. ISRN Hematol 2011: 864257

225. Lau FY. 1959. Pulmonary infarction and atrophy of the spleen associated with sickle-cell

hemoglobin C disease. N Engl J Med 260: 907-11

226. Wardrop CA, Dagg JH, Lee FD, Singh H, Dyet JF, Moffat A. 1975. Immunological

abnormalities in splenic atrophy. Lancet 2: 4-7

227. Coppo P, Saadoun D, Varet B. 2006. Autoimmune manifestations in acquired idiopathic

splenic atrophy: A puzzling association. Eur J Intern Med 17: 580-2

228. Di Sabatino A, Brunetti L, Carnevale Maffe G, Giuffrida P, Corazza GR. 2013. Is it

worth investigating splenic function in patients with celiac disease? World J

Gastroenterol 19: 2313-8

229. Di Sabatino A, Rosado MM, Cazzola P, Riboni R, Biagi F, Carsetti R, Corazza GR.

2006. Splenic hypofunction and the spectrum of autoimmune and malignant

complications in celiac disease. Clin Gastroenterol Hepatol 4: 179-86

230. Harmon GS, Lee JS. 2010. Splenic atrophy in celiac disease. Clin Gastroenterol Hepatol

8: A22

231. Trewby PN, Chipping PM, Palmer SJ, Roberts PD, Lewis SM, Stewart JS. 1981. Splenic

atrophy in adult coeliac disease: is it reversible? Gut 22: 628-32

232. Santilli D, Govoni M, Prandini N, Rizzo N, Trotta F. 2003. Autosplenectomy and

antiphospholipid antibodies in systemic lupus erythematosus: A pathogenetic

relationship? Semin Arthritis Rheum 33: 125-33

233. Santos N, Silva R, Rodrigues J, Torres-Costa J. 2014. Sjogren's syndrome and acquired

splenic atrophy with septic shock: a case report. J Med Case Rep 8: 10

234. Zhan J, Deng R, Tang J, Zhang B, Tang Y, Wang JK, Li F, Anderson VM, McNutt MA,

Gu J. 2006. The spleen as a target in severe acute respiratory syndrome. FASEB J 20:

2321-8

235. To KF, Chan PK, Chan KF, Lee WK, Lam WY, Wong KF, Tang NL, Tsang DN, Sung

RY, Buckley TA, Tam JS, Cheng AF. 2001. Pathology of fatal human infection

associated with avian influenza A H5N1 virus. J Med Virol 63: 242-6

Page 151: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

128

236. Matteucci D, Toniolo A, Conaldi PG, Basolo F, Gori Z, Bendinelli M. 1985. Systemic

lymphoid atrophy in coxsackievirus B3-infected mice: effects of virus and

immunopotentiating agents. J Infect Dis 151: 1100-8

237. Shibuta H, Adachi A, Kanda T, Matumoto M. 1982. Experimental parainfluenzavirus

infection in mice: fatal illness with atrophy of thymus and spleen in mice caused by a

variant of parainfluenza 3 virus. Infect Immun 35: 437-41

238. Bartholdy C, Hogh-Petersen M, Storm P, Holst PJ, Orskov C, Christensen JP, Thomsen

AR. 2014. IFNgamma and perforin cooperate to control infection and prevent fatal

pathology during persistent gammaherpesvirus infection in mice. Scand J Immunol 79:

395-403

239. Dutia BM, Clarke CJ, Allen DJ, Nash AA. 1997. Pathological changes in the spleens of

gamma interferon receptor-deficient mice infected with murine gammaherpesvirus: a role

for CD8 T cells. J Virol 71: 4278-83

240. Hayasaka D, Nagata N, Fujii Y, Hasegawa H, Sata T, Suzuki R, Gould EA, Takashima I,

Koike S. 2009. Mortality following peripheral infection with tick-borne encephalitis virus

results from a combination of central nervous system pathology, systemic inflammatory

and stress responses. Virology 390: 139-50

241. Di Sabatino A, Corazza GR. 2009. Coeliac disease. Lancet 373: 1480-93

242. Lewis NR, Holmes GK. 2010. Risk of morbidity in contemporary celiac disease. Expert

Rev Gastroenterol Hepatol 4: 767-80

243. Morelli AE, Larregina AT, Shufesky WJ, Zahorchak AF, Logar AJ, Papworth GD, Wang

Z, Watkins SC, Falo LD, Jr., Thomson AW. 2003. Internalization of circulating apoptotic

cells by splenic marginal zone dendritic cells: dependence on complement receptors and

effect on cytokine production. Blood 101: 611-20

244. Rathmell JC, Townsend SE, Xu JC, Flavell RA, Goodnow CC. 1996. Expansion or

elimination of B cells in vivo: dual roles for CD40- and Fas (CD95)-ligands modulated

by the B cell antigen receptor. Cell 87: 319-29

245. Steinman RM, Pack M, Inaba K. 1997. Dendritic cells in the T-cell areas of lymphoid

organs. Immunol Rev 156: 25-37

246. Offner H, Subramanian S, Parker SM, Wang C, Afentoulis ME, Lewis A, Vandenbark

AA, Hurn PD. 2006. Splenic atrophy in experimental stroke is accompanied by increased

regulatory T cells and circulating macrophages. J Immunol 176: 6523-31

247. Savino W. 2006. The thymus is a common target organ in infectious diseases. PLoS

Pathog 2: e62

248. Di Sabatino A, Carsetti R, Corazza GR. 2011. Post-splenectomy and hyposplenic states.

Lancet 378: 86-97

249. Foster PN, Hardy GJ, Losowsky MS. 1984. Fatal Salmonella septicaemia in a patient

with systemic lupus erythematosus and splenic atrophy. Br J Clin Pract 38: 434-5

250. Logan RF, Rifkind EA, Turner ID, Ferguson A. 1989. Mortality in celiac disease.

Gastroenterology 97: 265-71

251. Holdsworth RJ. 1991. Regeneration of the spleen and splenic autotransplantation. Br J

Surg 78: 270-8

252. Fenner F. 1976. Classification and nomenclature of viruses. Second report of the

International Committee on Taxonomy of Viruses. Intervirology 7: 1-115

Page 152: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

129

253. Perez M, Craven RC, de la Torre JC. 2003. The small RING finger protein Z drives

arenavirus budding: implications for antiviral strategies. Proc Natl Acad Sci U S A 100:

12978-83

254. Lee KJ, Novella IS, Teng MN, Oldstone MB, de La Torre JC. 2000. NP and L proteins of

lymphocytic choriomeningitis virus (LCMV) are sufficient for efficient transcription and

replication of LCMV genomic RNA analogs. J Virol 74: 3470-7

255. Borrow P, Oldstone MB. 1992. Characterization of lymphocytic choriomeningitis virus-

binding protein(s): a candidate cellular receptor for the virus. J Virol 66: 7270-81

256. Parekh BS, Buchmeier MJ. 1986. Proteins of lymphocytic choriomeningitis virus:

antigenic topography of the viral glycoproteins. Virology 153: 168-78

257. Cao W, Henry MD, Borrow P, Yamada H, Elder JH, Ravkov EV, Nichol ST, Compans

RW, Campbell KP, Oldstone MB. 1998. Identification of alpha-dystroglycan as a

receptor for lymphocytic choriomeningitis virus and Lassa fever virus. Science 282:

2079-81

258. Ahmed R, Salmi A, Butler LD, Chiller JM, Oldstone MB. 1984. Selection of genetic

variants of lymphocytic choriomeningitis virus in spleens of persistently infected mice.

Role in suppression of cytotoxic T lymphocyte response and viral persistence. J Exp Med

160: 521-40

259. Zinkernagel RM, Doherty PC. 1979. MHC-restricted cytotoxic T cells: studies on the

biological role of polymorphic major transplantation antigens determining T-cell

restriction-specificity, function, and responsiveness. Adv Immunol 27: 51-177

260. Pircher H, Burki K, Lang R, Hengartner H, Zinkernagel RM. 1989. Tolerance induction

in double specific T-cell receptor transgenic mice varies with antigen. Nature 342: 559-

61

261. Tishon A, Borrow P, Evans C, Oldstone MB. 1993. Virus-induced immunosuppression.

1. Age at infection relates to a selective or generalized defect. Virology 195: 397-405

262. von Herrath MG, Guerder S, Lewicki H, Flavell RA, Oldstone MB. 1995. Coexpression

of B7-1 and viral ("self") transgenes in pancreatic beta cells can break peripheral

ignorance and lead to spontaneous autoimmune diabetes. Immunity 3: 727-38

263. Borrow P, Lewicki H, Wei X, Horwitz MS, Peffer N, Meyers H, Nelson JA, Gairin JE,

Hahn BH, Oldstone MB, Shaw GM. 1997. Antiviral pressure exerted by HIV-1-specific

cytotoxic T lymphocytes (CTLs) during primary infection demonstrated by rapid

selection of CTL escape virus. Nat Med 3: 205-11

264. Ahmed R, Hahn CS, Somasundaram T, Villarete L, Matloubian M, Strauss JH. 1991.

Molecular basis of organ-specific selection of viral variants during chronic infection. J

Virol 65: 4242-7

265. Matloubian M, Kolhekar SR, Somasundaram T, Ahmed R. 1993. Molecular determinants

of macrophage tropism and viral persistence: importance of single amino acid changes in

the polymerase and glycoprotein of lymphocytic choriomeningitis virus. J Virol 67:

7340-9

266. Wherry EJ, Blattman JN, Murali-Krishna K, van der Most R, Ahmed R. 2003. Viral

persistence alters CD8 T-cell immunodominance and tissue distribution and results in

distinct stages of functional impairment. J Virol 77: 4911-27

267. Sullivan BM, Emonet SF, Welch MJ, Lee AM, Campbell KP, de la Torre JC, Oldstone

MB. 2011. Point mutation in the glycoprotein of lymphocytic choriomeningitis virus is

Page 153: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

130

necessary for receptor binding, dendritic cell infection, and long-term persistence. Proc

Natl Acad Sci U S A 108: 2969-74

268. Matloubian M, Somasundaram T, Kolhekar SR, Selvakumar R, Ahmed R. 1990. Genetic

basis of viral persistence: single amino acid change in the viral glycoprotein affects

ability of lymphocytic choriomeningitis virus to persist in adult mice. J Exp Med 172:

1043-8

269. Salvato M, Borrow P, Shimomaye E, Oldstone MB. 1991. Molecular basis of viral

persistence: a single amino acid change in the glycoprotein of lymphocytic

choriomeningitis virus is associated with suppression of the antiviral cytotoxic T-

lymphocyte response and establishment of persistence. J Virol 65: 1863-9

270. Sevilla N, Kunz S, Holz A, Lewicki H, Homann D, Yamada H, Campbell KP, de La

Torre JC, Oldstone MB. 2000. Immunosuppression and resultant viral persistence by

specific viral targeting of dendritic cells. J Exp Med 192: 1249-60

271. Kunz S, Sevilla N, McGavern DB, Campbell KP, Oldstone MB. 2001. Molecular

analysis of the interaction of LCMV with its cellular receptor [alpha]-dystroglycan. J Cell

Biol 155: 301-10

272. Smelt SC, Borrow P, Kunz S, Cao W, Tishon A, Lewicki H, Campbell KP, Oldstone

MB. 2001. Differences in affinity of binding of lymphocytic choriomeningitis virus

strains to the cellular receptor alpha-dystroglycan correlate with viral tropism and disease

kinetics. J Virol 75: 448-57

273. Borrow P, Evans CF, Oldstone MB. 1995. Virus-induced immunosuppression: immune

system-mediated destruction of virus-infected dendritic cells results in generalized

immune suppression. J Virol 69: 1059-70

274. Hahm B, Trifilo MJ, Zuniga EI, Oldstone MB. 2005. Viruses evade the immune system

through type I interferon-mediated STAT2-dependent, but STAT1-independent,

signaling. Immunity 22: 247-57

275. Wilson EB, Kidani Y, Elsaesser H, Barnard J, Raff L, Karp CL, Bensinger S, Brooks

DG. 2012. Emergence of distinct multiarmed immunoregulatory antigen-presenting cells

during persistent viral infection. Cell Host Microbe 11: 481-91

276. Sevilla N, Kunz S, McGavern D, Oldstone MB. 2003. Infection of dendritic cells by

lymphocytic choriomeningitis virus. Curr Top Microbiol Immunol 276: 125-44

277. Kunz S, Sevilla N, Rojek JM, Oldstone MB. 2004. Use of alternative receptors different

than alpha-dystroglycan by selected isolates of lymphocytic choriomeningitis virus.

Virology 325: 432-45

278. Shimojima M, Kawaoka Y. 2012. Cell surface molecules involved in infection mediated

by lymphocytic choriomeningitis virus glycoprotein. J Vet Med Sci 74: 1363-6

279. Shimojima M, Stroher U, Ebihara H, Feldmann H, Kawaoka Y. 2012. Identification of

cell surface molecules involved in dystroglycan-independent Lassa virus cell entry. J

Virol 86: 2067-78

280. Sullivan BM, Welch MJ, Lemke G, Oldstone MB. 2013. Is the TAM receptor Axl a

receptor for lymphocytic choriomeningitis virus? J Virol 87: 4071-4

281. Bachmann MF, Barner M, Viola A, Kopf M. 1999. Distinct kinetics of cytokine

production and cytolysis in effector and memory T cells after viral infection. Eur J

Immunol 29: 291-9

282. Grayson JM, Murali-Krishna K, Altman JD, Ahmed R. 2001. Gene expression in

antigen-specific CD8+ T cells during viral infection. J Immunol 166: 795-9

Page 154: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

131

283. Kaech SM, Ahmed R. 2001. Memory CD8+ T cell differentiation: initial antigen

encounter triggers a developmental program in naive cells. Nat Immunol 2: 415-22

284. Schulz M, Aichele P, Vollenweider M, Bobe FW, Cardinaux F, Hengartner H,

Zinkernagel RM. 1989. Major histocompatibility complex--dependent T cell epitopes of

lymphocytic choriomeningitis virus nucleoprotein and their protective capacity against

viral disease. Eur J Immunol 19: 1657-67

285. Klavinskis LS, Whitton JL, Joly E, Oldstone MB. 1990. Vaccination and protection from

a lethal viral infection: identification, incorporation, and use of a cytotoxic T lymphocyte

glycoprotein epitope. Virology 178: 393-400

286. Gairin JE, Mazarguil H, Hudrisier D, Oldstone MB. 1995. Optimal lymphocytic

choriomeningitis virus sequences restricted by H-2Db major histocompatibility complex

class I molecules and presented to cytotoxic T lymphocytes. J Virol 69: 2297-305

287. Oldstone MB, Lewicki H, Borrow P, Hudrisier D, Gairin JE. 1995. Discriminated

selection among viral peptides with the appropriate anchor residues: implications for the

size of the cytotoxic T-lymphocyte repertoire and control of viral infection. J Virol 69:

7423-9

288. Selin LK, Vergilis K, Welsh RM, Nahill SR. 1996. Reduction of otherwise remarkably

stable virus-specific cytotoxic T lymphocyte memory by heterologous viral infections. J

Exp Med 183: 2489-99

289. Butz EA, Bevan MJ. 1998. Massive expansion of antigen-specific CD8+ T cells during

an acute virus infection. Immunity 8: 167-75

290. Murali-Krishna K, Altman JD, Suresh M, Sourdive DJ, Zajac AJ, Miller JD, Slansky J,

Ahmed R. 1998. Counting antigen-specific CD8 T cells: a reevaluation of bystander

activation during viral infection. Immunity 8: 177-87

291. Wang XZ, Stepp SE, Brehm MA, Chen HD, Selin LK, Welsh RM. 2003. Virus-specific

CD8 T cells in peripheral tissues are more resistant to apoptosis than those in lymphoid

organs. Immunity 18: 631-42

292. Lau LL, Jamieson BD, Somasundaram T, Ahmed R. 1994. Cytotoxic T-cell memory

without antigen. Nature 369: 648-52

293. Asano MS, Ahmed R. 1996. CD8 T cell memory in B cell-deficient mice. J Exp Med

183: 2165-74

294. Gallimore A, Glithero A, Godkin A, Tissot AC, Pluckthun A, Elliott T, Hengartner H,

Zinkernagel R. 1998. Induction and exhaustion of lymphocytic choriomeningitis virus-

specific cytotoxic T lymphocytes visualized using soluble tetrameric major

histocompatibility complex class I-peptide complexes. J Exp Med 187: 1383-93

295. Zajac AJ, Blattman JN, Murali-Krishna K, Sourdive DJ, Suresh M, Altman JD, Ahmed

R. 1998. Viral immune evasion due to persistence of activated T cells without effector

function. J Exp Med 188: 2205-13

296. Kaech SM, Wherry EJ, Ahmed R. 2002. Effector and memory T-cell differentiation:

implications for vaccine development. Nat Rev Immunol 2: 251-62

297. Khanolkar A, Fuller MJ, Zajac AJ. 2002. T cell responses to viral infections: lessons

from lymphocytic choriomeningitis virus. Immunol Res 26: 309-21

298. Wherry EJ. 2011. T cell exhaustion. Nat Immunol 12: 492-9

299. Barber DL, Wherry EJ, Masopust D, Zhu B, Allison JP, Sharpe AH, Freeman GJ, Ahmed

R. 2006. Restoring function in exhausted CD8 T cells during chronic viral infection.

Nature 439: 682-7

Page 155: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

132

300. Brooks DG, Trifilo MJ, Edelmann KH, Teyton L, McGavern DB, Oldstone MB. 2006.

Interleukin-10 determines viral clearance or persistence in vivo. Nat Med 12: 1301-9

301. Ejrnaes M, Filippi CM, Martinic MM, Ling EM, Togher LM, Crotty S, von Herrath MG.

2006. Resolution of a chronic viral infection after interleukin-10 receptor blockade. J Exp

Med 203: 2461-72

302. Ng CT, Oldstone MB. 2012. Infected CD8alpha- dendritic cells are the predominant

source of IL-10 during establishment of persistent viral infection. Proc Natl Acad Sci U S

A 109: 14116-21

303. Borrow P, Tishon A, Oldstone MB. 1991. Infection of lymphocytes by a virus that aborts

cytotoxic T lymphocyte activity and establishes persistent infection. J Exp Med 174: 203-

12

304. Foulds KE, Zenewicz LA, Shedlock DJ, Jiang J, Troy AE, Shen H. 2002. Cutting edge:

CD4 and CD8 T cells are intrinsically different in their proliferative responses. J

Immunol 168: 1528-32

305. Homann D, Teyton L, Oldstone MB. 2001. Differential regulation of antiviral T-cell

immunity results in stable CD8+ but declining CD4+ T-cell memory. Nat Med 7: 913-9

306. Varga SM, Welsh RM. 1998. Detection of a high frequency of virus-specific CD4+ T

cells during acute infection with lymphocytic choriomeningitis virus. J Immunol 161:

3215-8

307. Varga SM, Welsh RM. 2000. High frequency of virus-specific interleukin-2-producing

CD4(+) T cells and Th1 dominance during lymphocytic choriomeningitis virus infection.

J Virol 74: 4429-32

308. Sun JC, Williams MA, Bevan MJ. 2004. CD4+ T cells are required for the maintenance,

not programming, of memory CD8+ T cells after acute infection. Nat Immunol 5: 927-33

309. von Herrath MG, Yokoyama M, Dockter J, Oldstone MB, Whitton JL. 1996. CD4-

deficient mice have reduced levels of memory cytotoxic T lymphocytes after

immunization and show diminished resistance to subsequent virus challenge. J Virol 70:

1072-9

310. Brooks DG, Teyton L, Oldstone MB, McGavern DB. 2005. Intrinsic functional

dysregulation of CD4 T cells occurs rapidly following persistent viral infection. J Virol

79: 10514-27

311. Fuller MJ, Zajac AJ. 2003. Ablation of CD8 and CD4 T cell responses by high viral

loads. J Immunol 170: 477-86

312. Mothe BR, Stewart BS, Oseroff C, Bui HH, Stogiera S, Garcia Z, Dow C, Rodriguez-

Carreno MP, Kotturi M, Pasquetto V, Botten J, Crotty S, Janssen E, Buchmeier MJ, Sette

A. 2007. Chronic lymphocytic choriomeningitis virus infection actively down-regulates

CD4+ T cell responses directed against a broad range of epitopes. J Immunol 179: 1058-

67

313. Matloubian M, Concepcion RJ, Ahmed R. 1994. CD4+ T cells are required to sustain

CD8+ cytotoxic T-cell responses during chronic viral infection. J Virol 68: 8056-63

314. Elsaesser H, Sauer K, Brooks DG. 2009. IL-21 is required to control chronic viral

infection. Science 324: 1569-72

315. Frohlich A, Kisielow J, Schmitz I, Freigang S, Shamshiev AT, Weber J, Marsland BJ,

Oxenius A, Kopf M. 2009. IL-21R on T cells is critical for sustained functionality and

control of chronic viral infection. Science 324: 1576-80

Page 156: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

133

316. Boettler T, Moeckel F, Cheng Y, Heeg M, Salek-Ardakani S, Crotty S, Croft M, von

Herrath MG. 2012. OX40 facilitates control of a persistent virus infection. PLoS Pathog

8: e1002913

317. Probst HC, van den Broek M. 2005. Priming of CTLs by lymphocytic choriomeningitis

virus depends on dendritic cells. J Immunol 174: 3920-4

318. Montoya M, Edwards MJ, Reid DM, Borrow P. 2005. Rapid activation of spleen

dendritic cell subsets following lymphocytic choriomeningitis virus infection of mice:

analysis of the involvement of type 1 IFN. J Immunol 174: 1851-61

319. Thompson LJ, Kolumam GA, Thomas S, Murali-Krishna K. 2006. Innate inflammatory

signals induced by various pathogens differentially dictate the IFN-I dependence of CD8

T cells for clonal expansion and memory formation. J Immunol 177: 1746-54

320. Wilson EB, Yamada DH, Elsaesser H, Herskovitz J, Deng J, Cheng G, Aronow BJ, Karp

CL, Brooks DG. 2013. Blockade of chronic type I interferon signaling to control

persistent LCMV infection. Science 340: 202-7

321. Mack EA, Kallal LE, Demers DA, Biron CA. 2011. Type 1 interferon induction of

natural killer cell gamma interferon production for defense during lymphocytic

choriomeningitis virus infection. MBio 2

322. Lang PA, Lang KS, Xu HC, Grusdat M, Parish IA, Recher M, Elford AR, Dhanji S,

Shaabani N, Tran CW, Dissanayake D, Rahbar R, Ghazarian M, Brustle A, Fine J, Chen

P, Weaver CT, Klose C, Diefenbach A, Haussinger D, Carlyle JR, Kaech SM, Mak TW,

Ohashi PS. 2012. Natural killer cell activation enhances immune pathology and promotes

chronic infection by limiting CD8+ T-cell immunity. Proc Natl Acad Sci U S A 109:

1210-5

323. Waggoner SN, Cornberg M, Selin LK, Welsh RM. 2012. Natural killer cells act as

rheostats modulating antiviral T cells. Nature 481: 394-8

324. Cook KD, Whitmire JK. 2013. The depletion of NK cells prevents T cell exhaustion to

efficiently control disseminating virus infection. J Immunol 190: 641-9

325. Waggoner SN, Taniguchi RT, Mathew PA, Kumar V, Welsh RM. 2010. Absence of

mouse 2B4 promotes NK cell-mediated killing of activated CD8+ T cells, leading to

prolonged viral persistence and altered pathogenesis. J Clin Invest 120: 1925-38

326. Crouse J, Bedenikovic G, Wiesel M, Ibberson M, Xenarios I, Von Laer D, Kalinke U,

Vivier E, Jonjic S, Oxenius A. 2014. Type I Interferons Protect T Cells against NK Cell

Attack Mediated by the Activating Receptor NCR1. Immunity 40: 961-73

327. Xu HC, Grusdat M, Pandyra AA, Polz R, Huang J, Sharma P, Deenen R, Kohrer K,

Rahbar R, Diefenbach A, Gibbert K, Lohning M, Hocker L, Waibler Z, Haussinger D,

Mak TW, Ohashi PS, Lang KS, Lang PA. 2014. Type I interferon protects antiviral

CD8(+) T cells from NK cell cytotoxicity. Immunity 40: 949-60

328. Andrews DM, Estcourt MJ, Andoniou CE, Wikstrom ME, Khong A, Voigt V, Fleming P,

Tabarias H, Hill GR, van der Most RG, Scalzo AA, Smyth MJ, Degli-Esposti MA. 2010.

Innate immunity defines the capacity of antiviral T cells to limit persistent infection. J

Exp Med 207: 1333-43

329. Su HC, Nguyen KB, Salazar-Mather TP, Ruzek MC, Dalod MY, Biron CA. 2001. NK

cell functions restrain T cell responses during viral infections. Eur J Immunol 31: 3048-

55

Page 157: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

134

330. Littwitz E, Francois S, Dittmer U, Gibbert K. 2013. Distinct roles of NK cells in viral

immunity during different phases of acute Friend retrovirus infection. Retrovirology 10:

127

331. Zhou G, Juang SW, Kane KP. 2013. NK cells exacerbate the pathology of influenza virus

infection in mice. Eur J Immunol 43: 929-38

332. Khakoo SI, Thio CL, Martin MP, Brooks CR, Gao X, Astemborski J, Cheng J, Goedert

JJ, Vlahov D, Hilgartner M, Cox S, Little AM, Alexander GJ, Cramp ME, O'Brien SJ,

Rosenberg WM, Thomas DL, Carrington M. 2004. HLA and NK cell inhibitory receptor

genes in resolving hepatitis C virus infection. Science 305: 872-4

333. Knapp S, Warshow U, Hegazy D, Brackenbury L, Guha IN, Fowell A, Little AM,

Alexander GJ, Rosenberg WM, Cramp ME, Khakoo SI. 2010. Consistent beneficial

effects of killer cell immunoglobulin-like receptor 2DL3 and group 1 human leukocyte

antigen-C following exposure to hepatitis C virus. Hepatology 51: 1168-75

334. Paladino N, Flores AC, Marcos CY, Fainboim H, Theiler G, Arruvito L, Williams F,

Middleton D, Fainboim L. 2007. Increased frequencies of activating natural killer

receptors are associated with liver injury in individuals who do not eliminate hepatitis C

virus. Tissue Antigens 69 Suppl 1: 109-11

335. Alter G, Rihn S, Walter K, Nolting A, Martin M, Rosenberg ES, Miller JS, Carrington M,

Altfeld M. 2009. HLA class I subtype-dependent expansion of KIR3DS1+ and

KIR3DL1+ NK cells during acute human immunodeficiency virus type 1 infection. J

Virol 83: 6798-805

336. Jennes W, Verheyden S, Demanet C, Adje-Toure CA, Vuylsteke B, Nkengasong JN,

Kestens L. 2006. Cutting edge: resistance to HIV-1 infection among African female sex

workers is associated with inhibitory KIR in the absence of their HLA ligands. J Immunol

177: 6588-92

337. Martin MP, Qi Y, Gao X, Yamada E, Martin JN, Pereyra F, Colombo S, Brown EE,

Shupert WL, Phair J, Goedert JJ, Buchbinder S, Kirk GD, Telenti A, Connors M, O'Brien

SJ, Walker BD, Parham P, Deeks SG, McVicar DW, Carrington M. 2007. Innate

partnership of HLA-B and KIR3DL1 subtypes against HIV-1. Nat Genet 39: 733-40

338. Romero V, Azocar J, Zuniga J, Clavijo OP, Terreros D, Gu X, Husain Z, Chung RT,

Amos C, Yunis EJ. 2008. Interaction of NK inhibitory receptor genes with HLA-C and

MHC class II alleles in Hepatitis C virus infection outcome. Mol Immunol 45: 2429-36

339. Yawata M, Yawata N, Draghi M, Little AM, Partheniou F, Parham P. 2006. Roles for

HLA and KIR polymorphisms in natural killer cell repertoire selection and modulation of

effector function. J Exp Med 203: 633-45

340. Parham P. 2004. Immunology. NK cells lose their inhibition. Science 305: 786-7

341. Moesta AK, Norman PJ, Yawata M, Yawata N, Gleimer M, Parham P. 2008. Synergistic

polymorphism at two positions distal to the ligand-binding site makes KIR2DL2 a

stronger receptor for HLA-C than KIR2DL3. J Immunol 180: 3969-79

342. Koziel MJ. 2005. Cellular immune responses against hepatitis C virus. Clin Infect Dis 41

Suppl 1: S25-31

343. Alter G, Martin MP, Teigen N, Carr WH, Suscovich TJ, Schneidewind A, Streeck H,

Waring M, Meier A, Brander C, Lifson JD, Allen TM, Carrington M, Altfeld M. 2007.

Differential natural killer cell-mediated inhibition of HIV-1 replication based on distinct

KIR/HLA subtypes. J Exp Med 204: 3027-36

Page 158: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

135

344. Biron CA, Nguyen KB, Pien GC. 2002. Innate immune responses to LCMV infections:

natural killer cells and cytokines. Curr Top Microbiol Immunol 263: 7-27

345. Louten J, van Rooijen N, Biron CA. 2006. Type 1 IFN deficiency in the absence of

normal splenic architecture during lymphocytic choriomeningitis virus infection. J

Immunol 177: 3266-72

346. Aichele P, Unsoeld H, Koschella M, Schweier O, Kalinke U, Vucikuja S. 2006. CD8 T

cells specific for lymphocytic choriomeningitis virus require type I IFN receptor for

clonal expansion. J Immunol 176: 4525-9

347. Biron CA, Sonnenfeld G, Welsh RM. 1984. Interferon induces natural killer cell

blastogenesis in vivo. J Leukoc Biol 35: 31-7

348. Gidlund M, Orn A, Wigzell H, Senik A, Gresser I. 1978. Enhanced NK cell activity in

mice injected with interferon and interferon inducers. Nature 273: 759-61

349. Kolumam GA, Thomas S, Thompson LJ, Sprent J, Murali-Krishna K. 2005. Type I

interferons act directly on CD8 T cells to allow clonal expansion and memory formation

in response to viral infection. J Exp Med 202: 637-50

350. Wiesel M, Kratky W, Oxenius A. 2011. Type I IFN substitutes for T cell help during

viral infections. J Immunol 186: 754-63

351. Ou R, Zhou S, Huang L, Moskophidis D. 2001. Critical role for alpha/beta and gamma

interferons in persistence of lymphocytic choriomeningitis virus by clonal exhaustion of

cytotoxic T cells. J Virol 75: 8407-23

352. Binder D, Fehr J, Hengartner H, Zinkernagel RM. 1997. Virus-induced transient bone

marrow aplasia: major role of interferon-alpha/beta during acute infection with the

noncytopathic lymphocytic choriomeningitis virus. J Exp Med 185: 517-30

353. Teijaro JR, Ng C, Lee AM, Sullivan BM, Sheehan KC, Welch M, Schreiber RD, de la

Torre JC, Oldstone MB. 2013. Persistent LCMV infection is controlled by blockade of

type I interferon signaling. Science 340: 207-11

354. Chen L, Borozan I, Feld J, Sun J, Tannis LL, Coltescu C, Heathcote J, Edwards AM,

McGilvray ID. 2005. Hepatic gene expression discriminates responders and

nonresponders in treatment of chronic hepatitis C viral infection. Gastroenterology 128:

1437-44

355. Sarasin-Filipowicz M, Oakeley EJ, Duong FH, Christen V, Terracciano L, Filipowicz W,

Heim MH. 2008. Interferon signaling and treatment outcome in chronic hepatitis C. Proc

Natl Acad Sci U S A 105: 7034-9

356. Guidotti LG, Chisari FV. 2006. Immunobiology and pathogenesis of viral hepatitis. Annu

Rev Pathol 1: 23-61

357. Su AI, Pezacki JP, Wodicka L, Brideau AD, Supekova L, Thimme R, Wieland S, Bukh J,

Purcell RH, Schultz PG, Chisari FV. 2002. Genomic analysis of the host response to

hepatitis C virus infection. Proc Natl Acad Sci U S A 99: 15669-74

358. Bosinger SE, Li Q, Gordon SN, Klatt NR, Duan L, Xu L, Francella N, Sidahmed A,

Smith AJ, Cramer EM, Zeng M, Masopust D, Carlis JV, Ran L, Vanderford TH,

Paiardini M, Isett RB, Baldwin DA, Else JG, Staprans SI, Silvestri G, Haase AT, Kelvin

DJ. 2009. Global genomic analysis reveals rapid control of a robust innate response in

SIV-infected sooty mangabeys. J Clin Invest 119: 3556-72

359. Jacquelin B, Mayau V, Targat B, Liovat AS, Kunkel D, Petitjean G, Dillies MA, Roques

P, Butor C, Silvestri G, Giavedoni LD, Lebon P, Barre-Sinoussi F, Benecke A, Muller-

Page 159: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

136

Trutwin MC. 2009. Nonpathogenic SIV infection of African green monkeys induces a

strong but rapidly controlled type I IFN response. J Clin Invest 119: 3544-55

360. Sandler NG, Bosinger SE, Estes JD, Zhu RT, Tharp GK, Boritz E, Levin D,

Wijeyesinghe S, Makamdop KN, del Prete GQ, Hill BJ, Timmer JK, Reiss E, Yarden G,

Darko S, Contijoch E, Todd JP, Silvestri G, Nason M, Norgren RB, Jr., Keele BF, Rao S,

Langer JA, Lifson JD, Schreiber G, Douek DC. 2014. Type I interferon responses in

rhesus macaques prevent SIV infection and slow disease progression. Nature 511: 601-5

361. Sharpe J, Ahlgren U, Perry P, Hill B, Ross A, Hecksher-Sorensen J, Baldock R,

Davidson D. 2002. Optical projection tomography as a tool for 3D microscopy and gene

expression studies. Science 296: 541-5

362. Kumar V, Scandella E, Danuser R, Onder L, Nitschke M, Fukui Y, Halin C, Ludewig B,

Stein JV. 2010. Global lymphoid tissue remodeling during a viral infection is

orchestrated by a B cell-lymphotoxin-dependent pathway. Blood 115: 4725-33

363. Odermatt B, Eppler M, Leist TP, Hengartner H, Zinkernagel RM. 1991. Virus-triggered

acquired immunodeficiency by cytotoxic T-cell-dependent destruction of antigen-

presenting cells and lymph follicle structure. Proc Natl Acad Sci U S A 88: 8252-6

364. Matter M, Odermatt B, Yagita H, Nuoffer JM, Ochsenbein AF. 2006. Elimination of

chronic viral infection by blocking CD27 signaling. J Exp Med 203: 2145-55

365. Engwerda CR, Ato M, Cotterell SE, Mynott TL, Tschannerl A, Gorak-Stolinska PM,

Kaye PM. 2002. A role for tumor necrosis factor-alpha in remodeling the splenic

marginal zone during Leishmania donovani infection. Am J Pathol 161: 429-37

366. Mueller SN, Matloubian M, Clemens DM, Sharpe AH, Freeman GJ, Gangappa S, Larsen

CP, Ahmed R. 2007. Viral targeting of fibroblastic reticular cells contributes to

immunosuppression and persistence during chronic infection. Proc Natl Acad Sci U S A

104: 15430-5

367. Ng CT, Nayak BP, Schmedt C, Oldstone MB. 2012. Immortalized clones of fibroblastic

reticular cells activate virus-specific T cells during virus infection. Proc Natl Acad Sci U

S A 109: 7823-8

368. Mueller SN, Vanguri VK, Ha SJ, West EE, Keir ME, Glickman JN, Sharpe AH, Ahmed

R. 2010. PD-L1 has distinct functions in hematopoietic and nonhematopoietic cells in

regulating T cell responses during chronic infection in mice. J Clin Invest 120: 2508-15

369. Zeng M, Southern PJ, Reilly CS, Beilman GJ, Chipman JG, Schacker TW, Haase AT.

2012. Lymphoid tissue damage in HIV-1 infection depletes naive T cells and limits T cell

reconstitution after antiretroviral therapy. PLoS Pathog 8: e1002437

370. Scandella E, Bolinger B, Lattmann E, Miller S, Favre S, Littman DR, Finke D, Luther

SA, Junt T, Ludewig B. 2008. Restoration of lymphoid organ integrity through the

interaction of lymphoid tissue-inducer cells with stroma of the T cell zone. Nat Immunol

9: 667-75

371. Benedict CA, De Trez C, Schneider K, Ha S, Patterson G, Ware CF. 2006. Specific

remodeling of splenic architecture by cytomegalovirus. PLoS Pathog 2: e16

372. Alexander-Miller MA, Derby MA, Sarin A, Henkart PA, Berzofsky JA. 1998.

Supraoptimal peptide-major histocompatibility complex causes a decrease in bc1-2 levels

and allows tumor necrosis factor alpha receptor II-mediated apoptosis of cytotoxic T

lymphocytes. J Exp Med 188: 1391-9

373. Badovinac VP, Tvinnereim AR, Harty JT. 2000. Regulation of antigen-specific CD8+ T

cell homeostasis by perforin and interferon-gamma. Science 290: 1354-8

Page 160: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

137

374. Opferman JT, Ober BT, Narayanan R, Ashton-Rickardt PG. 2001. Suicide induced by

cytolytic activity controls the differentiation of memory CD8(+) T lymphocytes. Int

Immunol 13: 411-9

375. Zheng L, Fisher G, Miller RE, Peschon J, Lynch DH, Lenardo MJ. 1995. Induction of

apoptosis in mature T cells by tumour necrosis factor. Nature 377: 348-51

376. Badovinac VP, Porter BB, Harty JT. 2002. Programmed contraction of CD8(+) T cells

after infection. Nat Immunol 3: 619-26

377. De Boer RJ, Homann D, Perelson AS. 2003. Different dynamics of CD4+ and CD8+ T

cell responses during and after acute lymphocytic choriomeningitis virus infection. J

Immunol 171: 3928-35

378. Kamperschroer C, Quinn DG. 1999. Quantification of epitope-specific MHC class-II-

restricted T cells following lymphocytic choriomeningitis virus infection. Cell Immunol

193: 134-46

379. Misumi I, Alirezaei M, Eam B, Su MA, Whitton JL, Whitmire JK. 2013. Differential T

cell responses to residual viral antigen prolong CD4+ T cell contraction following the

resolution of infection. J Immunol 191: 5655-68

380. Varga SM, Welsh RM. 1998. Stability of virus-specific CD4+ T cell frequencies from

acute infection into long term memory. J Immunol 161: 367-74

381. Williams MA, Ravkov EV, Bevan MJ. 2008. Rapid culling of the CD4+ T cell repertoire

in the transition from effector to memory. Immunity 28: 533-45

382. Nguyen LT, McKall-Faienza K, Zakarian A, Speiser DE, Mak TW, Ohashi PS. 2000.

TNF receptor 1 (TNFR1) and CD95 are not required for T cell deletion after virus

infection but contribute to peptide-induced deletion under limited conditions. Eur J

Immunol 30: 683-8

383. Reich A, Korner H, Sedgwick JD, Pircher H. 2000. Immune down-regulation and

peripheral deletion of CD8 T cells does not require TNF receptor-ligand interactions nor

CD95 (Fas, APO-1). Eur J Immunol 30: 678-82

384. Suresh M, Singh A, Fischer C. 2005. Role of tumor necrosis factor receptors in regulating

CD8 T-cell responses during acute lymphocytic choriomeningitis virus infection. J Virol

79: 202-13

385. Singh A, Suresh M. 2007. A role for TNF in limiting the duration of CTL effector phase

and magnitude of CD8 T cell memory. J Leukoc Biol 82: 1201-11

386. Suresh M, Gao X, Fischer C, Miller NE, Tewari K. 2004. Dissection of antiviral and

immune regulatory functions of tumor necrosis factor receptors in a chronic lymphocytic

choriomeningitis virus infection. J Virol 78: 3906-18

387. Razvi ES, Jiang Z, Woda BA, Welsh RM. 1995. Lymphocyte apoptosis during the

silencing of the immune response to acute viral infections in normal, lpr, and Bcl-2-

transgenic mice. Am J Pathol 147: 79-91

388. Zimmermann C, Rawiel M, Blaser C, Kaufmann M, Pircher H. 1996. Homeostatic

regulation of CD8+ T cells after antigen challenge in the absence of Fas (CD95). Eur J

Immunol 26: 2903-10

389. Weant AE, Michalek RD, Khan IU, Holbrook BC, Willingham MC, Grayson JM. 2008.

Apoptosis regulators Bim and Fas function concurrently to control autoimmunity and

CD8+ T cell contraction. Immunity 28: 218-30

390. Grayson JM, Weant AE, Holbrook BC, Hildeman D. 2006. Role of Bim in regulating

CD8+ T-cell responses during chronic viral infection. J Virol 80: 8627-38

Page 161: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

138

391. La Gruta NL, Kedzierska K, Stambas J, Doherty PC. 2007. A question of self-

preservation: immunopathology in influenza virus infection. Immunol Cell Biol 85: 85-92

392. Bouvier NM, Palese P. 2008. The biology of influenza viruses. Vaccine 26 Suppl 4: D49-

53

393. Ennis FA, Verbonitz M, Reichelderfer P, Daniel S. 1976. Recombination of influenza A

virus strains: effect on pathogenicity. Dev Biol Stand 33: 220-5

394. Thomas PG, Keating R, Hulse-Post DJ, Doherty PC. 2006. Cell-mediated protection in

influenza infection. Emerg Infect Dis 12: 48-54

395. Iwasaki A, Pillai PS. 2014. Innate immunity to influenza virus infection. Nat Rev

Immunol 14: 315-28

396. Kohlmeier JE, Woodland DL. 2009. Immunity to respiratory viruses. Annu Rev Immunol

27: 61-82

397. Hashimoto Y, Moki T, Takizawa T, Shiratsuchi A, Nakanishi Y. 2007. Evidence for

phagocytosis of influenza virus-infected, apoptotic cells by neutrophils and macrophages

in mice. J Immunol 178: 2448-57

398. Hikono H, Kohlmeier JE, Ely KH, Scott I, Roberts AD, Blackman MA, Woodland DL.

2006. T-cell memory and recall responses to respiratory virus infections. Immunol Rev

211: 119-32

399. Legge KL, Braciale TJ. 2003. Accelerated migration of respiratory dendritic cells to the

regional lymph nodes is limited to the early phase of pulmonary infection. Immunity 18:

265-77

400. Mellman I, Steinman RM. 2001. Dendritic cells: specialized and regulated antigen

processing machines. Cell 106: 255-8

401. Moll H. 2003. Dendritic cells and host resistance to infection. Cell Microbiol 5: 493-500

402. Vermaelen KY, Carro-Muino I, Lambrecht BN, Pauwels RA. 2001. Specific migratory

dendritic cells rapidly transport antigen from the airways to the thoracic lymph nodes. J

Exp Med 193: 51-60

403. Kim TS, Braciale TJ. 2009. Respiratory dendritic cell subsets differ in their capacity to

support the induction of virus-specific cytotoxic CD8+ T cell responses. PLoS One 4:

e4204

404. Kim TS, Gorski SA, Hahn S, Murphy KM, Braciale TJ. 2014. Distinct dendritic cell

subsets dictate the fate decision between effector and memory CD8(+) T cell

differentiation by a CD24-dependent mechanism. Immunity 40: 400-13

405. McGill J, Van Rooijen N, Legge KL. 2010. IL-15 trans-presentation by pulmonary

dendritic cells promotes effector CD8 T cell survival during influenza virus infection. J

Exp Med 207: 521-34

406. Belz GT, Wodarz D, Diaz G, Nowak MA, Doherty PC. 2002. Compromised influenza

virus-specific CD8(+)-T-cell memory in CD4(+)-T-cell-deficient mice. J Virol 76:

12388-93

407. Brown DM, Dilzer AM, Meents DL, Swain SL. 2006. CD4 T cell-mediated protection

from lethal influenza: perforin and antibody-mediated mechanisms give a one-two punch.

J Immunol 177: 2888-98

408. Kim TS, Sun J, Braciale TJ. 2011. T cell responses during influenza infection: getting

and keeping control. Trends Immunol 32: 225-31

409. McKinstry KK, Strutt TM, Swain SL. 2011. Hallmarks of CD4 T cell immunity against

influenza. J Intern Med 269: 507-18

Page 162: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

139

410. Doherty PC, Topham DJ, Tripp RA, Cardin RD, Brooks JW, Stevenson PG. 1997.

Effector CD4+ and CD8+ T-cell mechanisms in the control of respiratory virus

infections. Immunol Rev 159: 105-17

411. Oda T, Akaike T, Hamamoto T, Suzuki F, Hirano T, Maeda H. 1989. Oxygen radicals in

influenza-induced pathogenesis and treatment with pyran polymer-conjugated SOD.

Science 244: 974-6

412. Salek-Ardakani S, Croft M. 2010. Tumor necrosis factor receptor/tumor necrosis factor

family members in antiviral CD8 T-cell immunity. J Interferon Cytokine Res 30: 205-18

413. Watts TH. 2005. TNF/TNFR family members in costimulation of T cell responses. Annu

Rev Immunol 23: 23-68

414. Wortzman ME, Clouthier DL, McPherson AJ, Lin GH, Watts TH. 2013. The contextual

role of TNFR family members in CD8(+) T-cell control of viral infections. Immunol Rev

255: 125-48

415. Aggarwal BB. 2003. Signalling pathways of the TNF superfamily: a double-edged sword.

Nat Rev Immunol 3: 745-56

416. Croft M. 2009. The role of TNF superfamily members in T-cell function and diseases.

Nat Rev Immunol 9: 271-85

417. Croft M, Benedict CA, Ware CF. 2013. Clinical targeting of the TNF and TNFR

superfamilies. Nat Rev Drug Discov 12: 147-68

418. Shuford WW, Klussman K, Tritchler DD, Loo DT, Chalupny J, Siadak AW, Brown TJ,

Emswiler J, Raecho H, Larsen CP, Pearson TC, Ledbetter JA, Aruffo A, Mittler RS.

1997. 4-1BB costimulatory signals preferentially induce CD8+ T cell proliferation and

lead to the amplification in vivo of cytotoxic T cell responses. J Exp Med 186: 47-55

419. Melero I, Shuford WW, Newby SA, Aruffo A, Ledbetter JA, Hellstrom KE, Mittler RS,

Chen L. 1997. Monoclonal antibodies against the 4-1BB T-cell activation molecule

eradicate established tumors. Nat Med 3: 682-5

420. So T, Lee SW, Croft M. 2008. Immune regulation and control of regulatory T cells by

OX40 and 4-1BB. Cytokine Growth Factor Rev 19: 253-62

421. Niu L, Strahotin S, Hewes B, Zhang B, Zhang Y, Archer D, Spencer T, Dillehay D,

Kwon B, Chen L, Vella AT, Mittler RS. 2007. Cytokine-mediated disruption of

lymphocyte trafficking, hemopoiesis, and induction of lymphopenia, anemia, and

thrombocytopenia in anti-CD137-treated mice. J Immunol 178: 4194-213

422. Lee SW, Salek-Ardakani S, Mittler RS, Croft M. 2009. Hypercostimulation through 4-

1BB distorts homeostasis of immune cells. J Immunol 182: 6753-62

423. Zhu Y, Zhu G, Luo L, Flies AS, Chen L. 2007. CD137 stimulation delivers an antigen-

independent growth signal for T lymphocytes with memory phenotype. Blood 109: 4882-

9

424. Ascierto PA, Simeone E, Sznol M, Fu YX, Melero I. 2010. Clinical experiences with

anti-CD137 and anti-PD1 therapeutic antibodies. Semin Oncol 37: 508-16

425. Vinay DS, Kwon BS. 2012. Immunotherapy of cancer with 4-1BB. Mol Cancer Ther 11:

1062-70

426. Kwon BS, Weissman SM. 1989. cDNA sequences of two inducible T-cell genes. Proc

Natl Acad Sci U S A 86: 1963-7

427. Goodwin RG, Din WS, Davis-Smith T, Anderson DM, Gimpel SD, Sato TA,

Maliszewski CR, Brannan CI, Copeland NG, Jenkins NA, et al. 1993. Molecular cloning

Page 163: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

140

of a ligand for the inducible T cell gene 4-1BB: a member of an emerging family of

cytokines with homology to tumor necrosis factor. Eur J Immunol 23: 2631-41

428. Alderson MR, Smith CA, Tough TW, Davis-Smith T, Armitage RJ, Falk B, Roux E,

Baker E, Sutherland GR, Din WS. 1994. Molecular and biological characterization of

human 4-1BB and its ligand. Eur J Immunol 24: 2219-27

429. Schwarz H, Valbracht J, Tuckwell J, von Kempis J, Lotz M. 1995. ILA, the human 4-

1BB homologue, is inducible in lymphoid and other cell lineages. Blood 85: 1043-52

430. Zhou Z, Kim S, Hurtado J, Lee ZH, Kim KK, Pollok KE, Kwon BS. 1995.

Characterization of human homologue of 4-1BB and its ligand. Immunol Lett 45: 67-73

431. Won EY, Cha K, Byun JS, Kim DU, Shin S, Ahn B, Kim YH, Rice AJ, Walz T, Kwon

BS, Cho HS. 2010. The structure of the trimer of human 4-1BB ligand is unique among

members of the tumor necrosis factor superfamily. J Biol Chem 285: 9202-10

432. Wang C, Lin GH, McPherson AJ, Watts TH. 2009. Immune regulation by 4-1BB and 4-

1BBL: complexities and challenges. Immunol Rev 229: 192-215

433. Lee HW, Park SJ, Choi BK, Kim HH, Nam KO, Kwon BS. 2002. 4-1BB promotes the

survival of CD8+ T lymphocytes by increasing expression of Bcl-xL and Bfl-1. J

Immunol 169: 4882-8

434. Tan JT, Ha J, Cho HR, Tucker-Burden C, Hendrix RC, Mittler RS, Pearson TC, Larsen

CP. 2000. Analysis of expression and function of the costimulatory molecule 4-1BB in

alloimmune responses. Transplantation 70: 175-83

435. Lee SW, Park Y, Song A, Cheroutre H, Kwon BS, Croft M. 2006. Functional dichotomy

between OX40 and 4-1BB in modulating effector CD8 T cell responses. J Immunol 177:

4464-72

436. Lin GH, Sedgmen BJ, Moraes TJ, Snell LM, Topham DJ, Watts TH. 2009. Endogenous

4-1BB ligand plays a critical role in protection from influenza-induced disease. J

Immunol 182: 934-47

437. Wang C, McPherson AJ, Jones RB, Kawamura KS, Lin GH, Lang PA, Ambagala T,

Pellegrini M, Calzascia T, Aidarus N, Elford AR, Yue FY, Kremmer E, Kovacs CM,

Benko E, Tremblay C, Routy JP, Bernard NF, Ostrowski MA, Ohashi PS, Watts TH.

2012. Loss of the signaling adaptor TRAF1 causes CD8+ T cell dysregulation during

human and murine chronic infection. J Exp Med 209: 77-91

438. Seo SK, Park HY, Choi JH, Kim WY, Kim YH, Jung HW, Kwon B, Lee HW, Kwon BS.

2003. Blocking 4-1BB/4-1BB ligand interactions prevents herpetic stromal keratitis. J

Immunol 171: 576-83

439. Pollok KE, Kim YJ, Hurtado J, Zhou Z, Kim KK, Kwon BS. 1994. 4-1BB T-cell antigen

binds to mature B cells and macrophages, and costimulates anti-mu-primed splenic B

cells. Eur J Immunol 24: 367-74

440. Kang YJ, Kim SO, Shimada S, Otsuka M, Seit-Nebi A, Kwon BS, Watts TH, Han J.

2007. Cell surface 4-1BBL mediates sequential signaling pathways 'downstream' of TLR

and is required for sustained TNF production in macrophages. Nat Immunol 8: 601-9

441. Futagawa T, Akiba H, Kodama T, Takeda K, Hosoda Y, Yagita H, Okumura K. 2002.

Expression and function of 4-1BB and 4-1BB ligand on murine dendritic cells. Int

Immunol 14: 275-86

442. Snell LM, Lin GH, McPherson AJ, Moraes TJ, Watts TH. 2011. T-cell intrinsic effects of

GITR and 4-1BB during viral infection and cancer immunotherapy. Immunol Rev 244:

197-217

Page 164: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

141

443. Bertram EM, Lau P, Watts TH. 2002. Temporal segregation of 4-1BB versus CD28-

mediated costimulation: 4-1BB ligand influences T cell numbers late in the primary

response and regulates the size of the T cell memory response following influenza

infection. J Immunol 168: 3777-85

444. Lin GH, Snell LM, Wortzman ME, Clouthier DL, Watts TH. 2013. GITR-dependent

regulation of 4-1BB expression: implications for T cell memory and anti-4-1BB-induced

pathology. J Immunol 190: 4627-39

445. Lin GH, Edele F, Mbanwi AN, Wortzman ME, Snell LM, Vidric M, Roth K, Hauser AE,

Watts TH. 2012. Contribution of 4-1BBL on radioresistant cells in providing survival

signals through 4-1BB expressed on CD8(+) memory T cells in the bone marrow. Eur J

Immunol 42: 2861-74

446. Kwon BS, Hurtado JC, Lee ZH, Kwack KB, Seo SK, Choi BK, Koller BH, Wolisi G,

Broxmeyer HE, Vinay DS. 2002. Immune responses in 4-1BB (CD137)-deficient mice. J

Immunol 168: 5483-90

447. Tan JT, Whitmire JK, Ahmed R, Pearson TC, Larsen CP. 1999. 4-1BB ligand, a member

of the TNF family, is important for the generation of antiviral CD8 T cell responses. J

Immunol 163: 4859-68

448. Hendriks J, Xiao Y, Rossen JW, van der Sluijs KF, Sugamura K, Ishii N, Borst J. 2005.

During viral infection of the respiratory tract, CD27, 4-1BB, and OX40 collectively

determine formation of CD8+ memory T cells and their capacity for secondary

expansion. J Immunol 175: 1665-76

449. Taraban VY, Rowley TF, O'Brien L, Chan HT, Haswell LE, Green MH, Tutt AL,

Glennie MJ, Al-Shamkhani A. 2002. Expression and costimulatory effects of the TNF

receptor superfamily members CD134 (OX40) and CD137 (4-1BB), and their role in the

generation of anti-tumor immune responses. Eur J Immunol 32: 3617-27

450. Wen T, Bukczynski J, Watts TH. 2002. 4-1BB ligand-mediated costimulation of human

T cells induces CD4 and CD8 T cell expansion, cytokine production, and the

development of cytolytic effector function. J Immunol 168: 4897-906

451. Pulle G, Vidric M, Watts TH. 2006. IL-15-dependent induction of 4-1BB promotes

antigen-independent CD8 memory T cell survival. J Immunol 176: 2739-48

452. Rouse BT, Sarangi PP, Suvas S. 2006. Regulatory T cells in virus infections. Immunol

Rev 212: 272-86

453. Keynan Y, Card CM, McLaren PJ, Dawood MR, Kasper K, Fowke KR. 2008. The role of

regulatory T cells in chronic and acute viral infections. Clin Infect Dis 46: 1046-52

454. Zheng G, Wang B, Chen A. 2004. The 4-1BB costimulation augments the proliferation of

CD4+CD25+ regulatory T cells. J Immunol 173: 2428-34

455. Choi BK, Bae JS, Choi EM, Kang WJ, Sakaguchi S, Vinay DS, Kwon BS. 2004. 4-1BB-

dependent inhibition of immunosuppression by activated CD4+CD25+ T cells. J Leukoc

Biol 75: 785-91

456. Elpek KG, Yolcu ES, Franke DD, Lacelle C, Schabowsky RH, Shirwan H. 2007. Ex vivo

expansion of CD4+CD25+FoxP3+ T regulatory cells based on synergy between IL-2 and

4-1BB signaling. J Immunol 179: 7295-304

457. Irie J, Wu Y, Kachapati K, Mittler RS, Ridgway WM. 2007. Modulating protective and

pathogenic CD4+ subsets via CD137 in type 1 diabetes. Diabetes 56: 186-96

Page 165: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

142

458. Robertson SJ, Messer RJ, Carmody AB, Mittler RS, Burlak C, Hasenkrug KJ. 2008.

CD137 costimulation of CD8+ T cells confers resistance to suppression by virus-induced

regulatory T cells. J Immunol 180: 5267-74

459. Hippen KL, Harker-Murray P, Porter SB, Merkel SC, Londer A, Taylor DK, Bina M,

Panoskaltsis-Mortari A, Rubinstein P, Van Rooijen N, Golovina TN, Suhoski MM,

Miller JS, Wagner JE, June CH, Riley JL, Blazar BR. 2008. Umbilical cord blood

regulatory T-cell expansion and functional effects of tumor necrosis factor receptor

family members OX40 and 4-1BB expressed on artificial antigen-presenting cells. Blood

112: 2847-57

460. Choi BK, Kim YH, Kwon PM, Lee SC, Kang SW, Kim MS, Lee MJ, Kwon BS. 2009. 4-

1BB functions as a survival factor in dendritic cells. J Immunol 182: 4107-15

461. Lee SW, Park Y, Eun SY, Madireddi S, Cheroutre H, Croft M. 2012. Cutting edge: 4-

1BB controls regulatory activity in dendritic cells through promoting optimal expression

of retinal dehydrogenase. J Immunol 189: 2697-701

462. Wilcox RA, Chapoval AI, Gorski KS, Otsuji M, Shin T, Flies DB, Tamada K, Mittler

RS, Tsuchiya H, Pardoll DM, Chen L. 2002. Cutting edge: Expression of functional

CD137 receptor by dendritic cells. J Immunol 168: 4262-7

463. Zhang B, Zhang Y, Niu L, Vella AT, Mittler RS. 2010. Dendritic cells and Stat3 are

essential for CD137-induced CD8 T cell activation-induced cell death. J Immunol 184:

4770-8

464. Kuang Y, Weng X, Liu X, Zhu H, Chen Z, Chen H. 2012. Effects of 4-1BB signaling on

the biological function of murine dendritic cells. Oncol Lett 3: 477-81

465. Zhang X, Voskens CJ, Sallin M, Maniar A, Montes CL, Zhang Y, Lin W, Li G, Burch E,

Tan M, Hertzano R, Chapoval AI, Tamada K, Gastman BR, Schulze DH, Strome SE.

2010. CD137 promotes proliferation and survival of human B cells. J Immunol 184: 787-

95

466. Cole SL, Benam KH, McMichael AJ, Ho LP. 2014. Involvement of the 4-1BB/4-1BBL

Pathway in Control of Monocyte Numbers by Invariant NKT Cells. J Immunol

467. Melero I, Johnston JV, Shufford WW, Mittler RS, Chen L. 1998. NK1.1 cells express 4-

1BB (CDw137) costimulatory molecule and are required for tumor immunity elicited by

anti-4-1BB monoclonal antibodies. Cell Immunol 190: 167-72

468. Wilcox RA, Tamada K, Strome SE, Chen L. 2002. Signaling through NK cell-associated

CD137 promotes both helper function for CD8+ cytolytic T cells and responsiveness to

IL-2 but not cytolytic activity. J Immunol 169: 4230-6

469. Kim DH, Chang WS, Lee YS, Lee KA, Kim YK, Kwon BS, Kang CY. 2008. 4-1BB

engagement costimulates NKT cell activation and exacerbates NKT cell ligand-induced

airway hyperresponsiveness and inflammation. J Immunol 180: 2062-8

470. Lee SW, Park Y, So T, Kwon BS, Cheroutre H, Mittler RS, Croft M. 2008. Identification

of regulatory functions for 4-1BB and 4-1BBL in myelopoiesis and the development of

dendritic cells. Nat Immunol 9: 917-26

471. Nishimoto H, Lee SW, Hong H, Potter KG, Maeda-Yamamoto M, Kinoshita T,

Kawakami Y, Mittler RS, Kwon BS, Ware CF, Croft M, Kawakami T. 2005.

Costimulation of mast cells by 4-1BB, a member of the tumor necrosis factor receptor

superfamily, with the high-affinity IgE receptor. Blood 106: 4241-8

Page 166: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

143

472. Lee SC, Ju SA, Pack HN, Heo SK, Suh JH, Park SM, Choi BK, Kwon BS, Kim BS.

2005. 4-1BB (CD137) is required for rapid clearance of Listeria monocytogenes

infection. Infect Immun 73: 5144-51

473. Heinisch IV, Daigle I, Knopfli B, Simon HU. 2000. CD137 activation abrogates

granulocyte-macrophage colony-stimulating factor-mediated anti-apoptosis in

neutrophils. Eur J Immunol 30: 3441-6

474. Zhang B, Maris CH, Foell J, Whitmire J, Niu L, Song J, Kwon BS, Vella AT, Ahmed R,

Jacob J, Mittler RS. 2007. Immune suppression or enhancement by CD137 T cell

costimulation during acute viral infection is time dependent. J Clin Invest 117: 3029-41

475. Chung JY, Park YC, Ye H, Wu H. 2002. All TRAFs are not created equal: common and

distinct molecular mechanisms of TRAF-mediated signal transduction. J Cell Sci 115:

679-88

476. Silke J, Brink R. 2010. Regulation of TNFRSF and innate immune signalling complexes

by TRAFs and cIAPs. Cell Death Differ 17: 35-45

477. Cabal-Hierro L, Lazo PS. 2012. Signal transduction by tumor necrosis factor receptors.

Cell Signal 24: 1297-305

478. Rothe M, Pan MG, Henzel WJ, Ayres TM, Goeddel DV. 1995. The TNFR2-TRAF

signaling complex contains two novel proteins related to baculoviral inhibitor of

apoptosis proteins. Cell 83: 1243-52

479. Xie P. 2013. TRAF molecules in cell signaling and in human diseases. J Mol Signal 8: 7

480. Ely KR, Kodandapani R, Wu S. 2007. Protein-protein interactions in TRAF3. Adv Exp

Med Biol 597: 114-21

481. Chung JY, Lu M, Yin Q, Lin SC, Wu H. 2007. Molecular basis for the unique specificity

of TRAF6. Adv Exp Med Biol 597: 122-30

482. Lee HW, Nam KO, Park SJ, Kwon BS. 2003. 4-1BB enhances CD8+ T cell expansion by

regulating cell cycle progression through changes in expression of cyclins D and E and

cyclin-dependent kinase inhibitor p27kip1. Eur J Immunol 33: 2133-41

483. Lee do Y, Choi BK, Lee DG, Kim YH, Kim CH, Lee SJ, Kwon BS. 2013. 4-1BB

signaling activates the t cell factor 1 effector/beta-catenin pathway with delayed kinetics

via ERK signaling and delayed PI3K/AKT activation to promote the proliferation of

CD8+ T Cells. PLoS One 8: e69677

484. Starck L, Scholz C, Dorken B, Daniel PT. 2005. Costimulation by CD137/4-1BB inhibits

T cell apoptosis and induces Bcl-xL and c-FLIP(short) via phosphatidylinositol 3-kinase

and AKT/protein kinase B. Eur J Immunol 35: 1257-66

485. McPherson AJ, Snell LM, Mak TW, Watts TH. 2012. Opposing roles for TRAF1 in the

alternative versus classical NF-kappaB pathway in T cells. J Biol Chem 287: 23010-9

486. Sabbagh L, Pulle G, Liu Y, Tsitsikov EN, Watts TH. 2008. ERK-dependent Bim

modulation downstream of the 4-1BB-TRAF1 signaling axis is a critical mediator of CD8

T cell survival in vivo. J Immunol 180: 8093-101

487. Sabbagh L, Andreeva D, Laramee GD, Oussa NA, Lew D, Bisson N, Soumounou Y,

Pawson T, Watts TH. 2013. Leukocyte-specific protein 1 links TNF receptor-associated

factor 1 to survival signaling downstream of 4-1BB in T cells. J Leukoc Biol 93: 713-21

488. Sabbagh L, Srokowski CC, Pulle G, Snell LM, Sedgmen BJ, Liu Y, Tsitsikov EN, Watts

TH. 2006. A critical role for TNF receptor-associated factor 1 and Bim down-regulation

in CD8 memory T cell survival. Proc Natl Acad Sci U S A 103: 18703-8

Page 167: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

144

489. Zarnegar BJ, Wang Y, Mahoney DJ, Dempsey PW, Cheung HH, He J, Shiba T, Yang X,

Yeh WC, Mak TW, Korneluk RG, Cheng G. 2008. Noncanonical NF-kappaB activation

requires coordinated assembly of a regulatory complex of the adaptors cIAP1, cIAP2,

TRAF2 and TRAF3 and the kinase NIK. Nat Immunol 9: 1371-8

490. Vallabhapurapu S, Matsuzawa A, Zhang W, Tseng PH, Keats JJ, Wang H, Vignali DA,

Bergsagel PL, Karin M. 2008. Nonredundant and complementary functions of TRAF2

and TRAF3 in a ubiquitination cascade that activates NIK-dependent alternative NF-

kappaB signaling. Nat Immunol 9: 1364-70

491. Zheng C, Kabaleeswaran V, Wang Y, Cheng G, Wu H. 2010. Crystal structures of the

TRAF2: cIAP2 and the TRAF1: TRAF2: cIAP2 complexes: affinity, specificity, and

regulation. Mol Cell 38: 101-13

492. Zapata JM, Krajewska M, Krajewski S, Kitada S, Welsh K, Monks A, McCloskey N,

Gordon J, Kipps TJ, Gascoyne RD, Shabaik A, Reed JC. 2000. TNFR-associated factor

family protein expression in normal tissues and lymphoid malignancies. J Immunol 165:

5084-96

493. Arron JR, Pewzner-Jung Y, Walsh MC, Kobayashi T, Choi Y. 2002. Regulation of the

subcellular localization of tumor necrosis factor receptor-associated factor (TRAF)2 by

TRAF1 reveals mechanisms of TRAF2 signaling. J Exp Med 196: 923-34

494. Wicovsky A, Henkler F, Salzmann S, Scheurich P, Kneitz C, Wajant H. 2009. Tumor

necrosis factor receptor-associated factor-1 enhances proinflammatory TNF receptor-2

signaling and modifies TNFR1-TNFR2 cooperation. Oncogene 28: 1769-81

495. Madireddi S, Eun SY, Lee SW, Nemcovicova I, Mehta AK, Zajonc DM, Nishi N, Niki T,

Hirashima M, Croft M. 2014. Galectin-9 controls the therapeutic activity of 4-1BB-

targeting antibodies. J Exp Med 211: 1433-48

496. Schwarz H, Tuckwell J, Lotz M. 1993. A receptor induced by lymphocyte activation

(ILA): a new member of the human nerve-growth-factor/tumor-necrosis-factor receptor

family. Gene 134: 295-8

497. Jiang D, Chen Y, Schwarz H. 2008. CD137 induces proliferation of murine

hematopoietic progenitor cells and differentiation to macrophages. J Immunol 181: 3923-

32

498. Jiang D, Yue PS, Drenkard D, Schwarz H. 2008. Induction of proliferation and

monocytic differentiation of human CD34+ cells by CD137 ligand signaling. Stem Cells

26: 2372-81

499. Cannons JL, Lau P, Ghumman B, DeBenedette MA, Yagita H, Okumura K, Watts TH.

2001. 4-1BB ligand induces cell division, sustains survival, and enhances effector

function of CD4 and CD8 T cells with similar efficacy. J Immunol 167: 1313-24

500. Takahashi C, Mittler RS, Vella AT. 1999. Cutting edge: 4-1BB is a bona fide CD8 T cell

survival signal. J Immunol 162: 5037-40

501. Lin GH, Liu Y, Ambagala T, Kwon BS, Ohashi PS, Watts TH. 2010. Evaluating the

cellular targets of anti-4-1BB agonist antibody during immunotherapy of a pre-

established tumor in mice. PLoS One 5: e11003

502. Moraes TJ, Lin GH, Wen T, Watts TH. 2011. Incorporation of 4-1BB ligand into an

adenovirus vaccine vector increases the number of functional antigen-specific CD8 T

cells and enhances the duration of protection against influenza-induced respiratory

disease. Vaccine 29: 6301-12

Page 168: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

145

503. Steinman RM. 2007. Dendritic cells: understanding immunogenicity. Eur J Immunol 37

Suppl 1: S53-60

504. Summers deLuca L, Gommerman JL. 2012. Fine-tuning of dendritic cell biology by the

TNF superfamily. Nat Rev Immunol 12: 339-51

505. Lutz MB, Kukutsch N, Ogilvie AL, Rossner S, Koch F, Romani N, Schuler G. 1999. An

advanced culture method for generating large quantities of highly pure dendritic cells

from mouse bone marrow. J Immunol Methods 223: 77-92

506. Bertram EM, Dawicki W, Sedgmen B, Bramson JL, Lynch DH, Watts TH. 2004. A

switch in costimulation from CD28 to 4-1BB during primary versus secondary CD8 T

cell response to influenza in vivo. J Immunol 172: 981-8

507. Halstead ES, Mueller YM, Altman JD, Katsikis PD. 2002. In vivo stimulation of CD137

broadens primary antiviral CD8+ T cell responses. Nat Immunol 3: 536-41

508. Salih HR, Schmetzer HM, Burke C, Starling GC, Dunn R, Pelka-Fleischer R, Nuessler V,

Kiener PA. 2001. Soluble CD137 (4-1BB) ligand is released following leukocyte

activation and is found in sera of patients with hematological malignancies. J Immunol

167: 4059-66

509. Liu GZ, Gomes AC, Putheti P, Karrenbauer V, Kostulas K, Press R, Hillert J,

Hjelmstrom P, Gao XG. 2006. Increased soluble 4-1BB ligand (4-1BBL) levels in

peripheral blood of patients with multiple sclerosis. Scand J Immunol 64: 412-9

510. Kuka M, Munitic I, Giardino Torchia ML, Ashwell JD. 2013. CD70 is downregulated by

interaction with CD27. J Immunol 191: 2282-9

511. Stephan MT, Ponomarev V, Brentjens RJ, Chang AH, Dobrenkov KV, Heller G,

Sadelain M. 2007. T cell-encoded CD80 and 4-1BBL induce auto- and

transcostimulation, resulting in potent tumor rejection. Nat Med 13: 1440-9

512. Day CL, Kaufmann DE, Kiepiela P, Brown JA, Moodley ES, Reddy S, Mackey EW,

Miller JD, Leslie AJ, DePierres C, Mncube Z, Duraiswamy J, Zhu B, Eichbaum Q,

Altfeld M, Wherry EJ, Coovadia HM, Goulder PJ, Klenerman P, Ahmed R, Freeman GJ,

Walker BD. 2006. PD-1 expression on HIV-specific T cells is associated with T-cell

exhaustion and disease progression. Nature 443: 350-4

513. Boni C, Fisicaro P, Valdatta C, Amadei B, Di Vincenzo P, Giuberti T, Laccabue D,

Zerbini A, Cavalli A, Missale G, Bertoletti A, Ferrari C. 2007. Characterization of

hepatitis B virus (HBV)-specific T-cell dysfunction in chronic HBV infection. J Virol 81:

4215-25

514. Radziewicz H, Ibegbu CC, Fernandez ML, Workowski KA, Obideen K, Wehbi M,

Hanson HL, Steinberg JP, Masopust D, Wherry EJ, Altman JD, Rouse BT, Freeman GJ,

Ahmed R, Grakoui A. 2007. Liver-infiltrating lymphocytes in chronic human hepatitis C

virus infection display an exhausted phenotype with high levels of PD-1 and low levels of

CD127 expression. J Virol 81: 2545-53

515. Freeman GJ, Wherry EJ, Ahmed R, Sharpe AH. 2006. Reinvigorating exhausted HIV-

specific T cells via PD-1-PD-1 ligand blockade. J Exp Med 203: 2223-7

516. Wilson EB, Brooks DG. 2011. The role of IL-10 in regulating immunity to persistent

viral infections. Curr Top Microbiol Immunol 350: 39-65

517. Kim PS, Ahmed R. 2010. Features of responding T cells in cancer and chronic infection.

Curr Opin Immunol 22: 223-30

Page 169: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

146

518. Zinkernagel RM, Planz O, Ehl S, Battegay M, Odermatt B, Klenerman P, Hengartner H.

1999. General and specific immunosuppression caused by antiviral T-cell responses.

Immunol Rev 168: 305-15

519. Moskophidis D, Lechner F, Pircher H, Zinkernagel RM. 1993. Virus persistence in

acutely infected immunocompetent mice by exhaustion of antiviral cytotoxic effector T

cells. Nature 362: 758-61

520. Cerwenka A, Lanier LL. 2001. Ligands for natural killer cell receptors: redundancy or

specificity. Immunol Rev 181: 158-69

521. Battegay M, Cooper S, Althage A, Banziger J, Hengartner H, Zinkernagel RM. 1991.

Quantification of lymphocytic choriomeningitis virus with an immunological focus assay

in 24- or 96-well plates. J Virol Methods 33: 191-8

522. Haworth O, Cernadas M, Levy BD. 2011. NK cells are effectors for resolvin E1 in the

timely resolution of allergic airway inflammation. J Immunol 186: 6129-35

523. Cupedo T, Mebius RE. 2005. Cellular interactions in lymph node development. J

Immunol 174: 21-5

524. Finke D. 2005. Fate and function of lymphoid tissue inducer cells. Curr Opin Immunol

17: 144-50

525. Jewett A, Bonavida B. 1995. Interferon-alpha activates cytotoxic function but inhibits

interleukin-2-mediated proliferation and tumor necrosis factor-alpha secretion by

immature human natural killer cells. J Clin Immunol 15: 35-44

526. Loza MJ, Perussia B. 2004. Differential regulation of NK cell proliferation by type I and

type II IFN. Int Immunol 16: 23-32

527. Nguyen KB, Salazar-Mather TP, Dalod MY, Van Deusen JB, Wei XQ, Liew FY,

Caligiuri MA, Durbin JE, Biron CA. 2002. Coordinated and distinct roles for IFN-alpha

beta, IL-12, and IL-15 regulation of NK cell responses to viral infection. J Immunol 169:

4279-87

528. Navarini AA, Recher M, Lang KS, Georgiev P, Meury S, Bergthaler A, Flatz L, Bille J,

Landmann R, Odermatt B, Hengartner H, Zinkernagel RM. 2006. Increased susceptibility

to bacterial superinfection as a consequence of innate antiviral responses. Proc Natl Acad

Sci U S A 103: 15535-9

529. Miko I, Brath E, Nemeth N, Furka A, Sipka S, Jr., Peto K, Serfozo J, Kovacs J, Imre S,

Benko I, Galuska L, Sipka S, Acs G, Furka I. 2007. Spleen autotransplantation.

Morphological and functional follow-up after spleen autotransplantation in mice: a

research summary. Microsurgery 27: 312-6

530. Yamataka A, Fujiwara T, Tsuchioka T, Kurosu Y, Sunagawa M. 1996. Heterotopic

splenic autotransplantation in a neonate with splenic rupture, leading to normal splenic

function. J Pediatr Surg 31: 239-40

531. Tavassoli M. 1975. Limitation of splenic growth as studied by heterotopic splenic

implants. Blood 46: 631-5

532. Riera M, Buczacki S, Khan ZA. 2009. Splenic regeneration following splenectomy and

impact on sepsis: a clinical review. J R Soc Med 102: 139-42

533. Tan JK, Watanabe T. 2014. Murine spleen tissue regeneration from neonatal spleen

capsule requires lymphotoxin priming of stromal cells. J Immunol 193: 1194-203

534. Metcalf D. 1964. RESTRICTED GROWTH CAPACITY OF MULTIPLE SPLEEN

GRAFTS. Transplantation 2: 387-92

Page 170: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

147

535. Metcalf D. 1963. Spleen graft growth in splenectomised mice. Aust J Exp Biol Med Sci

41: 51-60

536. Janssen EM, Lemmens EE, Wolfe T, Christen U, von Herrath MG, Schoenberger SP.

2003. CD4+ T cells are required for secondary expansion and memory in CD8+ T

lymphocytes. Nature 421: 852-6

537. Feau S, Arens R, Togher S, Schoenberger SP. 2011. Autocrine IL-2 is required for

secondary population expansion of CD8(+) memory T cells. Nat Immunol 12: 908-13

538. Janssen EM, Droin NM, Lemmens EE, Pinkoski MJ, Bensinger SJ, Ehst BD, Griffith TS,

Green DR, Schoenberger SP. 2005. CD4+ T-cell help controls CD8+ T-cell memory via

TRAIL-mediated activation-induced cell death. Nature 434: 88-93

539. Munitic I, Kuka M, Allam A, Scoville JP, Ashwell JD. 2013. CD70 deficiency impairs

effector CD8 T cell generation and viral clearance but is dispensable for the recall

response to lymphocytic choriomeningitis virus. J Immunol 190: 1169-79

540. Kruglov AA, Lampropoulou V, Fillatreau S, Nedospasov SA. 2011. Pathogenic and

protective functions of TNF in neuroinflammation are defined by its expression in T

lymphocytes and myeloid cells. J Immunol 187: 5660-70

541. Winsauer C, Kruglov AA, Chashchina AA, Drutskaya MS, Nedospasov SA. 2013.

Cellular sources of pathogenic and protective TNF and experimental strategies based on

utilization of TNF humanized mice. Cytokine Growth Factor Rev

542. Clouthier DL, Watts TH. 2014. Cell-specific and context-dependent effects of GITR in

cancer, autoimmunity, and infection. Cytokine Growth Factor Rev

543. Juhasz K, Buzas K, Duda E. 2013. Importance of reverse signaling of the TNF

superfamily in immune regulation. Expert Rev Clin Immunol 9: 335-48

544. Shao Z, Schwarz H. 2011. CD137 ligand, a member of the tumor necrosis factor family,

regulates immune responses via reverse signal transduction. J Leukoc Biol 89: 21-9

545. Sun M, Fink PJ. 2007. A new class of reverse signaling costimulators belongs to the TNF

family. J Immunol 179: 4307-12

546. Eissner G, Kolch W, Scheurich P. 2004. Ligands working as receptors: reverse signaling

by members of the TNF superfamily enhance the plasticity of the immune system.

Cytokine Growth Factor Rev 15: 353-66

547. Sollner L, Shaqireen DOKMM, Wu JT, Schwarz H. 2007. Signal transduction

mechanisms of CD137 ligand in human monocytes. Cell Signal 19: 1899-908

548. Saito K, Ohara N, Hotokezaka H, Fukumoto S, Yuasa K, Naito M, Fujiwara T,

Nakayama K. 2004. Infection-induced up-regulation of the costimulatory molecule 4-

1BB in osteoblastic cells and its inhibitory effect on M-CSF/RANKL-induced in vitro

osteoclastogenesis. J Biol Chem 279: 13555-63

549. Watts AD, Hunt NH, Wanigasekara Y, Bloomfield G, Wallach D, Roufogalis BD,

Chaudhri G. 1999. A casein kinase I motif present in the cytoplasmic domain of members

of the tumour necrosis factor ligand family is implicated in 'reverse signalling'. EMBO J

18: 2119-26

550. Sun M, Lee S, Karray S, Levi-Strauss M, Ames KT, Fink PJ. 2007. Cutting edge: two

distinct motifs within the Fas ligand tail regulate Fas ligand-mediated costimulation. J

Immunol 179: 5639-43

551. Ma J, Bang BR, Lu J, Eun SY, Otsuka M, Croft M, Tobias P, Han J, Takeuchi O, Akira

S, Karin M, Yagita H, Kang YJ. 2013. The TNF family member 4-1BBL sustains

Page 171: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

148

inflammation by interacting with TLR signaling components during late-phase activation.

Sci Signal 6: ra87

552. Moh MC, Lorenzini PA, Gullo C, Schwarz H. 2013. Tumor necrosis factor receptor 1

associates with CD137 ligand and mediates its reverse signaling. FASEB J 27: 2957-66

553. Bae JS, Choi JK, Moon JH, Kim EC, Croft M, Lee HW. 2012. Novel transmembrane

protein 126A (TMEM126A) couples with CD137L reverse signals in myeloid cells. Cell

Signal 24: 2227-36

554. Narni-Mancinelli E, Jaeger BN, Bernat C, Fenis A, Kung S, De Gassart A, Mahmood S,

Gut M, Heath SC, Estelle J, Bertosio E, Vely F, Gastinel LN, Beutler B, Malissen B,

Malissen M, Gut IG, Vivier E, Ugolini S. 2012. Tuning of natural killer cell reactivity by

NKp46 and Helios calibrates T cell responses. Science 335: 344-8

555. Koller BH, Marrack P, Kappler JW, Smithies O. 1990. Normal development of mice

deficient in beta 2M, MHC class I proteins, and CD8+ T cells. Science 248: 1227-30

556. Gonzalez-Cabrero J, Wise CJ, Latchman Y, Freeman GJ, Sharpe AH, Reiser H. 1999.

CD48-deficient mice have a pronounced defect in CD4(+) T cell activation. Proc Natl

Acad Sci U S A 96: 1019-23

557. Cerwenka A, Bakker AB, McClanahan T, Wagner J, Wu J, Phillips JH, Lanier LL. 2000.

Retinoic acid early inducible genes define a ligand family for the activating NKG2D

receptor in mice. Immunity 12: 721-7

558. Lodoen M, Ogasawara K, Hamerman JA, Arase H, Houchins JP, Mocarski ES, Lanier

LL. 2003. NKG2D-mediated natural killer cell protection against cytomegalovirus is

impaired by viral gp40 modulation of retinoic acid early inducible 1 gene molecules. J

Exp Med 197: 1245-53

559. Ogasawara K, Hamerman JA, Hsin H, Chikuma S, Bour-Jordan H, Chen T, Pertel T,

Carnaud C, Bluestone JA, Lanier LL. 2003. Impairment of NK cell function by NKG2D

modulation in NOD mice. Immunity 18: 41-51

560. Kamizono S, Duncan GS, Seidel MG, Morimoto A, Hamada K, Grosveld G, Akashi K,

Lind EF, Haight JP, Ohashi PS, Look AT, Mak TW. 2009. Nfil3/E4bp4 is required for

the development and maturation of NK cells in vivo. J Exp Med 206: 2977-86

561. Diana J, Lehuen A. 2009. NKT cells: friend or foe during viral infections? Eur J Immunol

39: 3283-91

562. Diana J, Griseri T, Lagaye S, Beaudoin L, Autrusseau E, Gautron AS, Tomkiewicz C,

Herbelin A, Barouki R, von Herrath M, Dalod M, Lehuen A. 2009. NKT cell-

plasmacytoid dendritic cell cooperation via OX40 controls viral infection in a tissue-

specific manner. Immunity 30: 289-99

563. Pearson HA, Johnston D, Smith KA, Touloukian RJ. 1978. The born-again spleen.

Return of splenic function after splenectomy for trauma. N Engl J Med 298: 1389-92

564. Ludtke FE, Mack SC, Schuff-Werner P, Voth E. 1989. Splenic function after

splenectomy for trauma. Role of autotransplantation and splenosis. Acta Chir Scand 155:

533-9

565. Dodds WJ, Taylor AJ, Erickson SJ, Stewart ET, Lawson TL. 1990. Radiologic imaging

of splenic anomalies. AJR Am J Roentgenol 155: 805-10

566. Curtis GM, Movitz D. 1946. The surgical significance of the accessory spleen. Ann Surg

123: 276-98

567. Khan ZA, Dikki PE. 2004. Return of a normal functioning spleen after traumatic

splenectomy. J R Soc Med 97: 391-2

Page 172: Regulation of anti-viral immunity by dendritic cells and ... · 1.5.2 Armstrong vs. clone 13: mechanisms underlying clearance vs. persistence 24 1.5.3 The innate immune response to

149

568. Zoli G, Corazza GR, D'Amato G, Bartoli R, Baldoni F, Gasbarrini G. 1994. Splenic

autotransplantation after splenectomy: tuftsin activity correlates with residual splenic

function. Br J Surg 81: 716-8

569. Marine D, Manley OT. 1920. HOMEOTRANSPLANTATION AND

AUTOTRANSPLANTATION OF THE SPLEEN IN RABBITS : III. FURTHER DATA

ON GROWTH, PERMANENCE, EFFECT OF AGE, AND PARTIAL OR COMPLETE

REMOVAL OF THE SPLEEN. J Exp Med 32: 113-33