phospholipase d signaling in t cells

209
Loyola University Chicago Loyola University Chicago Loyola eCommons Loyola eCommons Dissertations Theses and Dissertations 2010 Phospholipase D Signaling in T Cells Phospholipase D Signaling in T Cells Uma Chandrasekaran Loyola University Chicago Follow this and additional works at: https://ecommons.luc.edu/luc_diss Part of the Immunology and Infectious Disease Commons Recommended Citation Recommended Citation Chandrasekaran, Uma, "Phospholipase D Signaling in T Cells" (2010). Dissertations. 162. https://ecommons.luc.edu/luc_diss/162 This Dissertation is brought to you for free and open access by the Theses and Dissertations at Loyola eCommons. It has been accepted for inclusion in Dissertations by an authorized administrator of Loyola eCommons. For more information, please contact [email protected]. This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 3.0 License. Copyright © 2010 Uma Chandrasekaran

Upload: others

Post on 06-Feb-2022

8 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Phospholipase D Signaling in T Cells

Loyola University Chicago Loyola University Chicago

Loyola eCommons Loyola eCommons

Dissertations Theses and Dissertations

2010

Phospholipase D Signaling in T Cells Phospholipase D Signaling in T Cells

Uma Chandrasekaran Loyola University Chicago

Follow this and additional works at: https://ecommons.luc.edu/luc_diss

Part of the Immunology and Infectious Disease Commons

Recommended Citation Recommended Citation Chandrasekaran, Uma, "Phospholipase D Signaling in T Cells" (2010). Dissertations. 162. https://ecommons.luc.edu/luc_diss/162

This Dissertation is brought to you for free and open access by the Theses and Dissertations at Loyola eCommons. It has been accepted for inclusion in Dissertations by an authorized administrator of Loyola eCommons. For more information, please contact [email protected].

This work is licensed under a Creative Commons Attribution-Noncommercial-No Derivative Works 3.0 License. Copyright © 2010 Uma Chandrasekaran

Page 2: Phospholipase D Signaling in T Cells

LOYOLA UNIVERSITY CHICAGO

PHOSPHOLIPASE D SIGNALING IN T CELLS

A DISSERTATION SUBMITTED TO

THE FACULTY OF THE GRADUATE SCHOOL

IN CANDIDACY FOR THE DEGREE OF

DOCTOR OF PHILOSOPHY

PROGRAM IN MICROBIOLOGY AND

IMMUNOLOGY

BY

UMA CHANDRASEKARAN

CHICAGO, IL

DECEMBER 2010

Page 3: Phospholipase D Signaling in T Cells

Copyright by UMA CHANDRASEKARAN, 2010 All rights reserved

Page 4: Phospholipase D Signaling in T Cells

iii

ACKNOWLEDGEMENTS

First and foremost, I would like to thank my mentor, Dr. Makio Iwashima

for providing an excellent learning environment in the laboratory that has

facilitated my development as a scientist. I am forever grateful to Dr. Knight for

her mentoring and encouragement throughout my graduate school career.

I would also like to thank my dissertation committee, Dr. Christopher

Wiethoff, Dr. Herbert Mathews and Dr. Adriano Marchese for their helpful

suggestions. I am especially grateful to Medical College of Georgia for providing

me a solid foundation during the start of my graduate school career.

I would like to thank all present and past members of the Iwashima Lab for

their technical help and suggestions. Special thanks to Dr. Nagendra Singh,

Mayuko Takezaki, Dr. Yoichi Seki and Dr. Mutsumi Yamamoto; without their

technical expertise, my project would not have been possible. I would also like to

thank my friends Oana Maier, Mariko Takami and Kathleen Mishler for their

friendship and unwavering support.

Finally, I would like to thank my parents, my brother, my husband and the

rest of my family for their continued support, love and encouragement.

Page 5: Phospholipase D Signaling in T Cells

iv

TABLE OF CONTENTS ACKNOWLEDGEMENTS iii LIST OF TABLES vii LIST OF FIGURES viii LIST OF ABBREVIATIONS xi ABSTRACT xv INTRODUCTION 1 CHAPTER ONE: LITERATURE REVIEW 5 The T lymphocyte in Immunology 5 Introduction 5 Establishing the role and function of Thymus 7 Delineation of two major lymphocyte subsets 8 Subdivision of T lymphocytes: CD4 and CD8 10 Heterogeneity of helper T cells 12 Introduction 12 Identification of Th1 and Th2 cells 12 Development and regulation of Th1 and Th2 cells 14 Effector functions and disease implications 16 Th17 cells: new member of effector T cells 16 Heterogeneity of Regulatory T cells 20 Introduction 20 Identification of distinct subsets of Treg cells 21 Phenotypic and functional characteristics of Treg cells 23 Immune homeostasis: population balance between 29 effector and regulatory T cell subsets Role of TCR mediated signaling pathways in altering 32 the immune balance Balancing act of Phospholipase D signaling in T cells 34 PLD: structure, function and expression 34 Regulation of catalytic activity of PLD 38 TCR signaling and PLD 40 Modulation of Phospholipase D signaling by 42 extra-cellular stimuli

Page 6: Phospholipase D Signaling in T Cells

v

Adenosine and PLD signaling 45 Conclusion 47 CHAPTER TWO: MATERIALS AND EXPERIMENTAL 48 METHODS Mice 48 Southern blot 49 Adoptive transfer in mice 49 Cell isolation and culture 50 Flow cytometry 53 T cell proliferation assay 54 ELISA 55 Phospholipase D (PLD) assay 56 cAMP measurement 57 Western blot 57 Generation of PLD YFP fusion constructs 58 Confocal microscopy 59 CHAPTER THREE: EXPERIMENTAL RESULTS 65 Regulation of Phospholipase D signaling by exogenous factors 65 Ethanol and CD4 T cell population dynamics 66 Effect of alcohol ingestion on the balance between 80 Foxp3+ and Foxp3- CD4 T cells Clostridium difficile toxin: Hijacking the immune system 82 through PLD signaling Regulation of phospholipase D signaling by endogenous factors 90 Adenosine and CD4 T cell population dynamics 97 Cross talk between adenosine receptor and 101 Phospholipase D signaling Adenosine A3 receptor signaling and cAMP 105 PLD signaling: synergy between ethanol and adenosine 111 Structure-function analysis of Phospholipase D 115 Determinants of PLD1 peri-nuclear localization 116 Determinants of PLD2 plasma membrane localization 122 Effect of PLD2 gene deletion on T cell response 125 CHAPTER FOUR: DISCUSSION 136 Introduction 136 Immune balance: effect of exogenous factors 137

Page 7: Phospholipase D Signaling in T Cells

vi

Immune balance: effect of endogenous factors 140 AB-MECA and Ethanol: Potential use as a 146 Tolerogenic adjuvant PLD localization: interaction between multiple structures 147 PLD2 and T cell functioning 149 Concluding remarks 151 REFERENCES 152 VITA 191

Page 8: Phospholipase D Signaling in T Cells

vii

LIST OF TABLES

Figure Page

1. PCR primers 64

Page 9: Phospholipase D Signaling in T Cells

viii

LIST OF FIGURES

Figure Page

1. A schematic presentation of the hypothesis proposed 4

2. Heterogeneity of CD4 T cells 19

3. Different subsets of regulatory T cells 28

4. Schematic representation of catalytic reaction of PLD 36 and domain structure of PLD isoforms found in mice 5. Western blot analysis of PLD1 deletion mutant constructs 60

6. Western blot analysis of PLD2 deletion mutant constructs 61

7. Confocal microscopic analysis of YFP fusion constructs 63

8. Effect of ethanol on CD4 T cells in vitro 67

9. Effect of ethanol on CD4 T cells in the presence of 69 antigenic stimulation in vitro

10. Proliferation kinetics of CFSE labeled CD4 T cells 70 in the presence of ethanol

11. Flow cytometry analysis and quantification of cell 73 numbers of CD4 T cells in mice injected with ethanol 12. Effect of ethanol on the proliferation kinetics of Foxp3 74 positive and Foxp3 negative CD4 T cells in vivo in the presence of antigen

Page 10: Phospholipase D Signaling in T Cells

ix

13. Effect of ethanol on homeostatic proliferation of 77 CD4 T cells 14. Effect of alcohol containing diet on mice CD4 T cells 81

15. Analysis of proliferation kinetics of Foxp3+ and Foxp3- 83 T cells in the presence of C.difficile toxin supernatants 16. Flow cytometric analysis of activation markers expressed 86 by CD4 T cells following antigenic stimulation in the presence of C.difficile supernatants 17. Flow cytometric analysis of Foxp3+ T cell percentage in 88 mice infected with C.difficile spores 18. Western blot analysis of adenosine A3 receptor 92 expression on CD4 T cells 19. Analysis of activation markers on T cells following 93 antigenic stimulation in the presence of A3 receptor signaling 20. Comparison of proliferation and cytokine secretion profile 95 of DMSO and AB-MECA treated CD4 T cells 21. Effect of adenosine A3 receptor signaling on CD4 T 98 cell population dynamics 22. Flow cytometric analysis of Foxp3 induction in CD4+CD25- 100 T cells following adenosine A3 receptor signaling

23. Effect of A3 receptor signaling on the proliferation kinetics of 102 Foxp3 positive and Foxp3 negative CD4 T cells in vivo 24. Assay of PLD activity in AB-MECA treated CD4 T cells 104 25. Assay for cAMP levels and activity in CD4 T cells 106 26. Western blot analysis of phosphorylation patterns of 110 PKA substrates in AB-MECA treated CD4 T cells 27. Combinatorial effect of ethanol and adenosine on CD4 112 T- cell population dynamics.

Page 11: Phospholipase D Signaling in T Cells

x

28. Western blot analysis of phosphorylation patterns of PKA 114 substrates in CD4 T cells treated with a combination of ethanol and AB-MECA 29. Confocal microscopic analysis of the sub-cellular localization 117 of PLD1 loop deletion mutant 30. Confocal microscopic analysis of the sub-cellular localization 118 of PLD1 loop alone or loop and full length PLD1 combinations 31. Schematic representation of the mutant PLD1 constructs 120 and confocal microscopy of their sub-cellular localization 32. Schematic representation of the mutant PLD2 constructs 123 and confocal microscopy of their sub-cellular localization 33. Phenotypic analysis of conditional PLD2 deficient mice 126 34. Characterization of CD4 T cell function in PLD2 128 deficient T cells 35. Cytokine profiles of PLD2 deficient CD4 T cells 130 36. Differentiation of PLD2 CD4 T cells into Th1, Th2 132 and Th17 effector cells 37. Effect of A3 receptor agonist on PLD2 deficient 133 CD4 T cells 38. Effect of ethanol on PLD2 deficient CD4 T cells 134

39. Schematic representation of A3 receptor signaling 145 in CD4 T cells

Page 12: Phospholipase D Signaling in T Cells

xi

LIST OF ABBREVIATIONS

AB-MECA N6-(4-Aminobenzyl)-9-[5-(methylcarbonyl) -b-D-ribofuranosyl]adenine

Ado Adenosine

Ag antigen

AHR aryl hydrocarbon receptor

APC Antigen presenting cells

ARF ADP ribosylation factor

CAMP Cyclic adenosine monophosphate

CD Cluster of differentiation antigen

CDAD Clostridium Difficile - associated diarrhoea

CFSE Carboxyfluorescein succinimidyl ester

CFU Colony forming unit

ConA Concanavalin A

CTLA-4 Cytotoxic T lymphocyte antigen-4

DAG Diacylglycerol

DC Dendritic cells

DEX dexamethasone

DMSO Dimethyl sulfoxide

Page 13: Phospholipase D Signaling in T Cells

xii

DNA Deoxyribonuclease

EAE Experimental autoimmune encephalomyelitis

ELISA Enzyme-linked immunosorbent assay

ERK Extracellular signal-regulated protein kinase

FACS Fluorescence activated cell sorting

FCS Fetal Calf Serum

Foxp3 Forkhead box protein 3

FICZ Formylindolo (3,2-b) carbazole

GITR Glucocorticoid-induced TNF receptor family related protein

HIV Human immunodeficiency virus

IBD Inflammatory Bowel disease

IFN-γ Interferon-γ

IL Interleukin

i.p intraperitoneal

i.v intravenous

ITAM Immunoreceptor tyrosine-based activation motif kDa kilodalton

LAT Linker of activated T cell

Lck leukocyte-specific protein tyrosine kinase

LP Lamina propria

MACS Magnetic cell sorting

MBP Myelin basic protein

Page 14: Phospholipase D Signaling in T Cells

xiii

MHC Major histocompatibility complex

mTOR Mammalian target of rapamycin

nTregs Naturally occurring regulatory T cells

PA Phosphatidic acid

PBS Phosphate buffered saline

PC Phosphatidyl Choline

PCR Polymerase chain reaction

PGE Prostaglandin

PH Pleckstrin homology

PKA Protein kinase A

PLC Phospholipase C

PLD Phospholipase D

PKC Protein kinase C

PX Phox homology domain

RA Rheumatoid arthritis

RAG-1 Recombination activating gene-1

ROR Retinoid-related orphan receptor

RPMI Roswell Park Memorial Institute

SDS-PAGE Sodium dodecylsulfate-polyacyrlamide gel electrophoresis STAT Signal Transducer and Activator of Transcription

T-bet T-box transcription factor

TCR T cell receptor

TcdB Toxin B

Page 15: Phospholipase D Signaling in T Cells

xiv

TGF Transforming growth factor

Th T helper

TNF Tumour necrosis factor

Tregs T regulatory cells

T1D Type-1 diabetes

Zap-70 Zeta-chain-associated protein 70

Page 16: Phospholipase D Signaling in T Cells

xv

ABSTRACT

Antigen stimulation of T lymphocytes induces the activation of

phospholipase D (PLD) signaling. Phospholipase D (PLD) is a

phosphodiesterase that catalyzes the conversion of phosphatidyl choline (PC) to

phosphatidic acid (PA). PA is an important lipid second messenger and is known

to mediate a variety of cellular functions. However, the specific role of PA in T

lymphocytes has not been established. Previous studies indicated differential

requirement for TCR induced PLD signaling in regulatory and non-regulatory T

cells. Inhibition of TCR induced PLD signal preferentially suppressed the growth

of non-regulatory T cells while allowing the proliferation of regulatory T cells in

the presence of exogenous IL-2. Based on this observation, we hypothesized

that PLD signaling is a critical factor that balances the population dynamics

between regulatory and non-regulatory T cells.

For my dissertation work, I focused on elucidating the functional and

molecular regulation of PLD signaling in T cells. In this study, we identified that

various exogenous (alcohol and Clostridium difficile toxin) and endogenous

factors (adenosine) modulate PLD signaling altering the population dynamics of

regulatory and non-regulatory T cells.

Page 17: Phospholipase D Signaling in T Cells

xvi

PLD has two isoforms namely PLD1 and PLD2 expressed in mammalian

cells. These isoforms are 50% identical and have distinct localization in the cell.

PLD1 has peri-nuclear localization while PLD2 localizes to the plasma

membrane. Molecular analysis of PLD1 and PLD2 using domain deletion

suggested that a region unique to PLD1 known as the ‘loop’ confers the distinct

peri-nuclear localization of PLD1.

Further, we addressed the specific function of PLD2 in CD4 T cells using

PLD2 knock out mice. We found that PLD2 is dispensable for T cell activation

and proliferation but might play an important role in effector T cell differentiation.

The results from this study delineate some of the functions of PLD signaling in T

cells and provide insight into immunological regulation during T cell activation.

Page 18: Phospholipase D Signaling in T Cells

1

INTRODUCTION

The mammalian immune system exists in a state of equilibrium that allows

for immune activation (reactivity) against pathogens (foreign antigens) while

maintaining tolerance (non-reactivity) towards self-antigens. This balance is

achieved primarily through interactions between different subsets of T

lymphocytes (Littman and Rudensky, 2010). T cell receptors (TCR) recognize

short peptides derived from both self and foreign antigens in the context of major

histocompatibility complex (MHC) thus underscoring their role in both host

defense and autoimmunity (Sette et al., 1993). The fate of a T cell following

antigen recognition is influenced by the environmental cues it receives from the

surrounding milieu like the presence of different cytokines. Through the activation

of specific transcription factors, various cytokines promote distinct effector T cell

lineage commitment (Th1, Th2, Th17) thus conferring protection against a wide

variety of pathogens (Bettelli et al., 2007; Delgoffe et al., 2009). To avoid

uncontrolled immune activation against foreign and host molecules causing harm

to the host, the immune system employs various tolerance mechanisms.

Regulatory T cell mediated immune suppression plays a major role in regulating

immune response by effector T cells and maintenance of tolerance towards self

(Sakaguchi, 2004).

Page 19: Phospholipase D Signaling in T Cells

2Hence, tightly controlled balance between effector and regulatory T cell

populations maintain immune homeostasis. But various ex vivo (alcohol, bacterial

toxins) and in vivo factors (anti-inflammatory agents) can shift the balance

between these two T cell populations with opposing effects. For example, alcohol

and adenosine have long been known to be immunosuppressive (Cook, 1998;

Hasko et al., 2008; Nelson and Kolls, 2002). However, the precise molecular

mechanism through which these molecules alter the immune balance was largely

unknown. Previous reports have suggested that immune balance is sensitive to

alterations in TCR signaling pathways (Dragone et al., 2006; Goodnow, 2001).

Also, differences in the signaling pathways downstream of TCR activation have

been reported in Treg cells versus effector T cells (Gavin et al., 2002). In

particular, our lab had previously reported differential requirement for TCR

induced phospholipase D (PLD) signaling between effector and regulatory T

cells. Inhibition of PLD signaling in effector T cells blocked their proliferation and

survival while it had no significant effect on Treg proliferation (Singh et al., 2006).

Taken together, these data indicated that PLD signaling in T cells is a critical

factor controlling the balance between Treg cells and non-Treg cells.

The goal of my dissertation project was to identify the mechanism of PLD

regulation in T cells. Based on previous data, we hypothesized that inhibition of

PLD signal in T cells may promote dominance of Treg cells while activation of

PLD signaling may promote expansion of effector T cells (Fig 1). We focused on

the possibility that external factors (alcohol and bacterial toxin) and an

endogenous factor (adenosine) might mediate immune-suppression by

Page 20: Phospholipase D Signaling in T Cells

3

negatively regulating PLD signaling. Also, the specific role of PLD2 isoform in

altering the T cell balance was determined using mice with T cell specific deletion

of PLD2. Further, we identified the critical region controlling the sub-cellular

localization of PLD isoforms.

In the sections to follow, the significance of PLD signaling in T cells and

rationale and discussion of the experiments performed to address the hypothesis

is described.

Page 21: Phospholipase D Signaling in T Cells

4

Ethanol, Adenosine, C.diff toxin

Fig 1. A schematic presentation of the hypothesis proposed. Active PLD

signaling following TCR stimulation leads to expansion of effector T cells (white

circles) accompanied by a modest proliferation of nTreg cells (grey circles). TCR

signaling in the presence of various exogenous factors (alcohol, bacterial toxins)

and endogenous factors (adenosine) suppresses PLD signaling thus inhibiting

the expansion of effector T cells. (Courtesy: Dr.Iwashima)

Page 22: Phospholipase D Signaling in T Cells

5

CHAPTER ONE

LITERATURE REVIEW

THE T LYMPHOCYTE IN IMMUNOLOGY

Introduction

The immune system can be perceived as an army of organs, tissues, cells

and molecules that protect the host against a wide variety of invading pathogens

(Eberl, 2010). The coordinated effort of these various immune components

provides the host with layered defenses of increasing specificity (Alberts, 2002) .

At first, the physical barrier like skin prevents bacteria and viruses from entering

the host. In case of entry into the host, the innate immune response provides an

immediate but non-specific immune response. If pathogens escape the innate

immune response, vertebrates are equipped with an adaptive immune system.

The adaptive immune system makes use of variable receptors to target specific

pathogen antigens. Also, adaptive immune responses provide faster and

heightened responses upon re-infection with the pathogen due to maintenance of

immunological memory.

Bone marrow derived (B cells) and Thymus derived lymphocytes (T cells)

are now considered the key mediators of adaptive immune responses. However,

Page 23: Phospholipase D Signaling in T Cells

6 this was not always the case. Until 1950, the only property known about

lymphocytes was that they were motile (Miller, 2002). Using tritiated thymidine

and cannulation of thoracic duct, James Gowans showed that small lymphocytes

continuously re-circulate from the thoracic duct to blood and secondary lymphoid

tissue and back to the thoracic duct (Gowans, 1996) (Masopust et al., 2007).

Miller et al., in 1961 were the first to demonstrate the existence of two major

interacting subsets of lymphocytes, T and B cells. Further research suggested

that T cells could be divided into two distinct subsets on the basis of their cell

surface markers: CD4 and CD8. In terms of their function, CD4 T cells were

thought to help antibody synthesis (thus, helper T cells) while CD8 T cells

mediated cytotoxic effect on pathogens. In 1986, Mossman and Coffman showed

that CD4 T cells were heterogeneous and based on their pattern of cytokine

production can be further divided into Th1 and Th2 cells. Recent investigations

have further expanded the helper T cell subsets into Th17, regulatory T cells and

follicular helper T cells. Thus, the central role of T cells in orchestrating the

immune response against a wide variety of pathogens and maintenance of self-

regulatory network has been firmly established.

Despite the tremendous increase in the knowledge of T cell subsets, their

specific contribution to disease progression is not well established. Researchers

are only now beginning to apply the knowledge gained from scientific

experiments to therapeutic interventions.

Page 24: Phospholipase D Signaling in T Cells

7

Establishing the role and function of Thymus

Ancient Greeks noted the presence of a large mass of tissue in the chest above

the heart and they concluded that it had to be the seat of the ‘Soul’ (Miller, 2002).

The wasting away of the soul after puberty was never addressed (Miller, 2002).

As early as 1777, William Hewson noted that the thymus was filled with particles

resembling those in lymph and blood. Also, he believed that the thymus exists

during the early periods of life only when those particles seem to be most wanted

(Miller, 2002). Another hematologist, John Beard also echoed the same views as

Hewson and suggested that thymus must be regarded as the parent source of all

lymphoid structures of the body and that leukocytes starting from their birth place

in the thymus penetrate into all parts of the body and perform functions in the

body (Miller, 2002). Although, both Hewson and Beard had the right views they

did not have the right tools and technical skills to demonstrate their idea.

In the late 1950s, Gross showed that filtered extracts of leukemic tissues

from high leukemic strain mice could induce the disease in a low leukemic strain

of mice if the extracts were injected at birth (Gross, 1951; Miller, 1999, 2002).

The role of thymus in leukemogenesis was known as it was previously

demonstrated that adult thymectomy prevented spontaneous mouse leukemia

developing in high leukemic strains of mice as well as leukemia induced by

chemical carcinogens. Miller hypothesized that virus given at birth multiplied in

the developing thymus and thus led to leukemogenic transformation. In order to

test this hypothesis, he neonatally thymectomized mice. If his hypothesis was

correct, he expected that neonatal mice lacking a thymus from birth should no

Page 25: Phospholipase D Signaling in T Cells

8

longer be susceptible to virus infection and would not develop leukemia when

grafted with thymic tissue later. However, he found that these neonatally

thymectomized mice were healthy at first but wasted away after weaning and

died irrespective of virus inoculation. On the other hand, adult thymectomized

mice survived and never showed any signs of weight loss or any other pathology.

This led him to conclude that “ the thymus at birth may be essential to life “(Miller,

1961).

Histological examination of the tissues of neonatally thymectomized mice

displayed a marked deficiency of lymphocytes in the lymphoid tissues and in the

circulation along with the presence of liver lesions suggesting infection by

hepatitis virus (Miller, 1999, 2002). Further these mice failed to reject skin grafts

derived from foreign mouse strains indicating immune deficiency. Conversely,

inoculation of lymph node/spleen cells or implantation of thymic tissue led to the

recovery of immunological functions in these mice as efficiently as normal mice

(Miller, 2002). In summary, these studies firmly established the crucial role of

thymus in normal immune system development and function.

Delineation of two major lymphocyte subsets

Based on the observations of defective cellular immunity and impaired antibody

producing capacity in neonatally thymectomized mice, it was accepted that

mammalian thymus produced lymphocytes that participated in both cellular and

humoral immunity. In 1962, Burnet and colleagues reported division of labor

among chicken lymphocytes as they found that early bursectomy was associated

Page 26: Phospholipase D Signaling in T Cells

9

with defects in antibody formation and early thymectomy was associated with

defects in cellular response (failure to reject homografts) (Warner et al., 1962).

Also, Parrott et al., observed that neonatally thymectomized mice had a

deficiency of lymphocytes limited to those areas of lymph node and spleen

associated with histological changes induced by cell mediated immune

responses but not in those areas where antibody producing cells appeared

(Parrott et al., 1966). All these emerging data intrigued Dr.Miller and he set out to

understand the contribution of thymus to the pool of small lymphocytes and to

antibody forming capacity. Using histocompatibility differences as markers, Miller

et al., proposed the existence of two major subsets of lymphocytes. They were

antibody forming cell precursors derived from lymphocytes in bone marrow (B

cells) and thymus-derived cells (T cells) essential to allow the former to respond

to antigen by producing antibody and mediating cellular immunity against foreign

grafts (Miller and Mitchell, 1967) (Mitchell and Miller, 1968).

Following the reports of two families of lymphocytes, several physical and

biochemical techniques were developed to distinguish these cell populations on

the basis of their surface markers, response to a number of mitogens etc (Hirst et

al., 1975). By 1970, it was widely accepted that T cells reacted with Thy-1 (θ)

antisera, thus serving as a specific T cell marker (Masopust et al., 2007) (Raff,

2008).

Page 27: Phospholipase D Signaling in T Cells

10

Sub-division of T lymphocytes: CD4 and CD8

In 1960, Govaerts demonstrated that lymphocytes from immunized

animals rather than the serum, destroyed allogeneic targets in vitro (Govaerts,

1960). Interestingly, destruction of T cells with anti-θ antiserum eliminated this

capability of lymphocytes to destroy allogeneic targets in vitro (Cerottini et al.,

1970). Several researchers corroborated this finding of cytotoxicity mediated by

T cells (Golstein et al., 1972).

T lymphocytes were reported to participate in a variety of cell-mediated

immune reactions like alloreactivity, cell mediated cytotoxicity and amplification of

antibody production by B cells (Cantor and Boyse, 1975). However, it was not

known whether T cells acquire these diverse functions following antigenic

stimulation or whether functionally distinct T cell subclass were present in non-

immune animals. To address this question, Cantor et al., undertook experiments

to define cell surface components selectively expressed by T cells mediating

these distinct functions. Cantor et al., hypothesized that genes coding for such

components would be exclusively expressed in T cells. Previous work had

reported that Ly antigens are expressed exclusively on thymus-derived

lymphocytes, so Cantor determined if these functionally distinct T cell populations

could be classified on the basis of expression of different Ly alloantigens. He

reported that helper T cells exclusively expressed Ly-1 while cytotoxic T cells

expressed Ly-23. Also, these subclasses of T cells were found in non-immune

animals indicating that commitment of T cells to participate in either helper or

cytotoxic function is a differentiative process independent of antigen exposure.

Page 28: Phospholipase D Signaling in T Cells

11

Further, Cantor et al, demonstrated using adoptive transfer of Ly-1 and Ly-23

expressing T cells into lethally irradiated mice, the absence of inter/ sequential

conversion of Ly-1 and Ly-23 expressing T cells in the host mice even after

prolonged periods (Huber et al., 1976). Taken together, these studies

demonstrated the presence of two functionally different subsets of T-cells,

representing distinct lineages of thymus-derived lymphocytes.

Further, researchers extended the surface molecule analysis of T cells to

humans and found the human analog of Ly-1 as CD4 and Ly-23 as CD8 (Chess,

2006). Moreover, Rao et al., demonstrated that expression of Ly antigens

determined major histocompatibility complex (MHC) specificity of T cells in

addition to their function (Rao et al., 1983). Thus, CD4+ T cells were found to be

MHC class II restricted, whereas CD8+ T cells were found to be MHC class I

restricted (Chess, 2006). Many years of research culminated in the

understanding of the role of CD4 and CD8 as not just surface markers of T cell

subsets but rather functionally important molecules involved in the recognition of

antigen along with the Ag-specific α,β TCR (Chess, 2006).

Page 29: Phospholipase D Signaling in T Cells

12

HETEROGENEITY OF HELPER T CELLS Introduction

Following the discovery of B and T lymphocytes, Miller performed

additional experiments using neonatally thymectomized mice and suggested

possible interactions between B and T cells in antibody responses (Miller, 1999).

This idea was further strengthened by work of Claman et al., and led to the

proposal of the “two cell” theory of antibody production (Claman and Chaperon,

1969). The discovery of two distinct lymphocyte populations and their synergism

in antibody production posed many interesting questions to researchers like the

nature of the contact between these two cells, timing, dosage of antigen, method

of information transfer etc. The process of answering these questions and much

technical advancement led to the identification of heterogeneity among helper T

cells (Fig 2).

Identification of Th1 and Th2 cells

By 1970, it was generally accepted that a homogeneous population of

thymus derived T cells participated in both cell mediated immunity (delayed–type

hypersensitivity reactions) and humoral immunity (helper cells in antibody

response (Liew and Parish, 1974). In 1971, Parish et al, found an inverse

relationship between humoral and cell mediated immunity and proposed the

possibility of different sub-populations of T cells mediating delayed type

hypersensitivity and helper functions (Liew, 2002; Liew and Parish, 1974).

Page 30: Phospholipase D Signaling in T Cells

13

Marrack et al further confirmed the existence of different sub-populations of CD4

T cells. They demonstrated the presence of antigen specific and non-specific Th

cells (Marrack and Kappler, 1975). Furthermore, Tada et al., demonstrated the

presence of two types of helper T cells that act either independently or

synergistically to help the B-cell response to an antigen (Tada et al., 1978). He

further distinguished these subsets of CD4 T cells on the basis of their

adherence to nylon wool column and the expression of Ia antigen. Accordingly,

Th1 cells were nylon non-adherent, did not express the Ia antigen and led to

modest increase in B cell responses, while Th2 cells were nylon adherent,

expressed the Ia antigen and led to maximal stimulation of B cell responses.

All of the above-mentioned experiments were conducted by stimulating

whole T cell fractions isolated from spleen with an antigen, thus depicting

polyclonal activation of T cells. However, by using the recently established

method for deriving alloreactive T cell clones (Fathman and Hengartner, 1978),

Mossman and Coffman established a panel of antigen specific helper T cell

clones. They categorized these helper T cell clones on the basis of their pattern

of cytokine production (Liew, 2002). They tested the effect of these cytokines

(derived by stimulating individual Th cell clones with ConA) on the proliferative

ability of 3 main cell lines namely HT2, NFS60 and MM3 cell lines (Mosmann et

al., 1986). This led to the identification of two distinct types of CD4 T cells. Type

I cells produced IL-2, IFN-γ, GM-CSF and IL-3 in response to antigenic

stimulation while type II cells produced IL-3, IL-4 and IL-5 (Liew, 2002). In line

with previous observations of Tada et al., type I helper T cell clones enhanced

Page 31: Phospholipase D Signaling in T Cells

14

IgG1 production by 10 fold and did not induce Ia expression on B cells while type

II T cell clones enhanced IgE production by 100 fold and induced Ia expression

on B cells. Hence, Mossman et al., used the same nomenclature proposed

earlier and categorized these CD4 T cells as Th1 and Th2 (Liew, 2002).

Following this discovery using T cell lines, attempts were made to define

CD4 T cell subsets in mice under physiological conditions. This led to the

proposal of four different CD4 T cell subsets: cells producing Th1 cytokines, Th2

cytokines, both Th1 and Th2 cytokines and neither Th1 /Th2 cytokine (Hayakawa

and Hardy, 1988). Additional results on the same cell lines led to the proposal of

a Th0 subset, which produced both Th1 and Th2 cytokines and were thought to

be uncommitted precursors of Th1 and Th2 subsets of CD4 T cells (Firestein et

al., 1989).

Development and regulation of Th1 and Th2 cells

Confirmation of the existence of Th1/Th2 subsets of CD4 T cells led to

further investigations into their development and mechanisms of regulation.

Gajewski and Fernandez-Botran et al., reported that the products of Th1 and Th2

cells could act as autocrine growth factors for further expansion of these cells

and also reciprocally inhibit the opposite cell type (Liew, 2002). IFN- γ enhanced

Th1 cell growth and inhibited the proliferation of Th2 cells while IL-4 promoted

Th2 cell expansion and limited the proliferation of Th1 cell (Fernandez-Botran et

al., 1988; Gajewski and Fitch, 1988).

Page 32: Phospholipase D Signaling in T Cells

15

These findings posed many important questions: What initiates the

development of Th1 versus Th2 cells? Are these cells derived from a common

precursor that are instructed to differentiate into various lineages or do they arise

from different precursors that are pre-programmed to develop into different

lineages? Researchers answered these questions using cells from TCR

transgenic mice. Hsieh et al, were the first to demonstrate that naïve T cells from

TCR transgenic DO11.10 mice can develop into both Th1 and Th2 phenotypes

under appropriate cytokine and stimulation conditions (Hsieh et al., 1992).

Similar findings by others established that Th1 and Th2 cells have a common

precursor and that the cytokine microenvironment is the primary determining

factor of the helper T cell subset fates (Liew, 2002). Over the years, many other

factors like antigen dose (Hosken et al., 1995), major histocompatibility complex

haplotypes (Constant et al., 1995) have also been shown to influence the

differentiation of Th1 and Th2 subsets. Also, studies were conducted to

elucidate the mechanism through which cytokines determine the lineage

decisions of helper T cells. Evidence so far indicates that IL-12 drives Th1

differentiation through activation of the transcription factor T-bet in T cells through

STAT4 signaling (Jacobson et al., 1995) (Szabo et al., 2000). Conversely, IL-4

induces Th2 cell differentiation through activation of another transcription factor,

GATA3 via STAT6 signaling (Takeda et al., 1996; Zheng and Flavell, 1997). It is

also proposed that the mutually exclusive expression of T-bet and GATA3 might

reflect the ability of these factors to antagonize each other (Liew, 2002).

Page 33: Phospholipase D Signaling in T Cells

16

Effector functions and disease implications

One of the earliest evidence for the role of helper T cell lineages in

disease pathogenesis was suggested in 1981 based on studies of Leishmania

major infections. Howard et al reported that BALB/c mice are highly susceptible

to Leishmania tropica infection compared to C57BL/6 mice (Howard et al., 1980;

Howard et al., 1981). Further using adoptive cell transfer it was shown that the

protection of C57BL/6 mice (resistant strain) correlated with an appropriate

increase in IFN-γ mediated Th1 response and susceptibility of Balb/c mice

correlated with an inappropriate IL-4 mediated Th2 response (Heinzel et al.,

1989; Scott et al., 1988). Th1 response and IFN-γ were also found to be critical to

protect hosts from a variety of infections mediated by intracellular pathogens,

viruses and tumors (Wan and Flavell, 2009). Also, excessive Th1 response has

been related to incidence of inflammatory diseases like IBD and autoimmune

diseases like rheumatoid arthritis (Wan and Flavell, 2009). On the other hand,

Th2 response protects hosts against infections from a variety of extra cellular

pathogens like helminthes. Th2 responses also participate in mucosal immunity

and in allergic diseases (Wan and Flavell, 2009).

Th17 cells: new member of effector T cells

Mossman used the ability of T cell clones to induce footpad swelling as a marker

for delayed type hypersensitivity reactions (Mosmann et al., 1986). Based on

these experiments, he proposed that DTH was mediated solely by Th1 cells. The

lymphocytic infiltrates found in the patients with autoimmune diseases like

Page 34: Phospholipase D Signaling in T Cells

17

rheumatoid arthritis (RA) and multiple sclerosis (MS) bear the hallmarks of DTH

(Steinman, 2007). So based on Mossman’s findings it was hypothesized that

administration of IFN-γ would worsen EAE (animal model of MS) while blocking

IFN-γ would ameliorate EAE. Further, one would have also predicted that EAE

would be attenuated or absent in IFN-γ deficient mice.

Contrary to these predictions, the results were just the opposite. For

example, paralysis in the EAE model was worsened in IFN-γ mice. In 2003,

multiple studies demonstrated that IL-23 knock out mice were resistant to the

induction of various autoimmune diseases like EAE, IBD and arthritis (Cua et al.,

2003) (Murphy et al., 2003). The cytokine IL-23 is a heterodimeric molecule,

sharing the p40 subunit with the Th1 cytokine IL-12 but differing from IL-12

because of its p19 subunit (Steinman, 2007). IL-23 does not promote the

development of Th1 cells, but instead it is one of the essential factors driving the

expansion of CD4 T cells producing IL17, IL17F, IL6 and TNF (Langrish et al.,

2005). Using passive transfer studies, authors demonstrated that IL-17 producing

T cells induced EAE while IFN-γ producing T cells did not (Steinman, 2007). Cua

and colleagues showed that EAE, driven by IL-23 and IL-17, was worsened with

administration of neutralizing antibody to γ-IFN suggesting that Th1 and Th17

were reciprocal in terms of their function (Cua et al., 2003).

In an attempt to identify factors that inhibited the TGF-β mediated

conversion of naïve T cells into Foxp3 expressing T cells, Bettelli et al,

discovered that TGF-β along with IL-6 led to differentiation of naïve T cells into

IL-17 producing T cells (Korn et al., 2009). Not surprisingly, IL-6 knock out mice

Page 35: Phospholipase D Signaling in T Cells

18

are resistant to EAE and anti-IL6 improves disease condition in EAE (Steinman,

2007). IL-6 or IL-21 in the presence of low amounts of TGF-β result in the

induction of the steroid nuclear receptor, RORα and RORγt in T cells through

STAT3 signaling (Korn et al., 2009). However, the mechanism through which

RORγt regulates Th17 cell differentiation is not yet determined.

In addition to inducing tissue inflammation in autoimmune diseases like

EAE and RA, Th17 cells also participate in host defense against variety of

microbes. Korn et al propose that Th17 cells function in the clearance of specific

types of pathogens that require massive inflammatory responses, which cannot

be dealt with by Th1 and Th2 immunity (Korn et al., 2009). Pathogens as diverse

as Citrobacter rodentium and Candida albicans can trigger Th17 responses

(Huang et al., 2004; Korn et al., 2009). Interestingly, Th17 cells are abundant in

the lamina propria (LP) of the small intestine. Ivanov et al, showed that Th17

cells are induced in the LP in response to specific components of the commensal

microbiota, underscoring their importance in maintaining intestinal immunity

(Ivanov et al., 2008).

Page 36: Phospholipase D Signaling in T Cells

19

Figure 2. Heterogeneity of CD4 T cells. Differentiation into different effector

CD4+ T cell lineages, T helper (Th) 1, Th2, Th17, and T regulatory (Treg) cells is

initiated following antigenic stimulation of uncommitted (naïve) CD4+ T helper

cells (Thp). The effector cell types are characterized by their synthesis of specific

cytokines and their immuno-regulatory functions, as indicated on the right. The

differentiation along different lineages involves different cytokines and the

activation of distinct signaling cascades and transcription factors that result in the

induction of additional cyto/chemokines and cyto/chemokine receptors, which

may be part of positive and negative feedback loops. Modified from Jetten et al

((Jetten, 2009)

Page 37: Phospholipase D Signaling in T Cells

20

HETEROGENEITY OF REGULATORY T CELLS

Introduction

Immunologists have appreciated the concept of self versus non-self

discrimination by the immune system since the late 19th century. Ray Owen was

the first to observe the phenomenon of immunological tolerance in vivo when he

documented the coexistence of two types of erythrocytes in the blood of dizygotic

cattle twins and the lack of immune reactivity against each other (Owen, 1945;

Wood et al., 2010). This led Burnet to propose the concept of “Neonatal

tolerance” wherein he stated that exposure to an antigen early in life would

induce specific tolerance to that antigen. Burnet further postulated in his clonal

selection theory that during ontogeny, antibodies against self-antigens would be

deleted (Martini and Burgio, 1999; Wood et al., 2010).

In 1970,Gershon and Kondo reported the finding that T cells not only

augmented but also dampened immune responses (Gershon and Kondo, 1970;

Sakaguchi et al., 2007). This suppressive function was found to be mediated by

a subset of T cells called suppressor T cells. It was soon determined that I-J was

the key suppressor molecule expressed by this subset of T cells. However, no

such gene could be found within MHC that encoded this molecule (Steinman,

2007) (Ishii et al., 1982) (Kronenberg et al., 1983).

In the 80s, using newly developed tools (monoclonal antibodies and TCR

transgenic mice) Marrack and von Boehmer’s group reported clonal deletion and

anergy as key mechanisms of immunological tolerance (Marrack and Kappler,

1987; Sakaguchi et al., 2007) (Kisielow et al., 1988). Most of these results were

Page 38: Phospholipase D Signaling in T Cells

21

inferred from experiments wherein tolerance was induced towards an exogenous

antigen.

The question of tolerance was addressed from a diametrically opposite

spectrum by investigators like Sakaguchi and Penhale. They studied the effect of

loss of tolerance towards self-antigens and thus the etiology of autoimmune

diseases. This led to the identification of a subset of T cell called regulatory T cell

(Treg cells) that prevented the onset of autoimmunity and is central to the

maintenance of tolerance to self-antigens. An improved understanding of the

mechanisms of tolerance has led to new possibilities for the treatment of

autoimmune diseases (Martini and Burgio, 1999).

Identification of regulatory T cell subset

Ehlrich postulated “Horror autotoxicus” in 1900 suggesting the

unwillingness of the organism to endanger itself by the formation of toxic

autoantibodies (Silverstein, 2001). However, Donath and Landsteiner

demonstrated that the cause of Paroxysmal cold hemoglobinuria is due to the

formation of autoantibodies against patient’s own erythrocytes (Silverstein,

2001). Many other diseases of autoimmune origin were discovered as well but

these results faded in the wake of biochemical pursuits by scientists to uncover

antibody–antigen interactions.

In the late 1960s many studies reported that neonatal thymectomy of

normal mice/rats (day 3 after birth) led to the development of inflammatory tissue

damage along with the appearance of tissue-specific autoantibodies in circulation

Page 39: Phospholipase D Signaling in T Cells

22

(Penhale et al., 1973; Sakaguchi et al., 2007). Also, adoptive transfer of CD4 T

cells from these mice with autoimmune disease was sufficient to induce disease

in T cell deficient recipients (McKeever et al., 1990). Interestingly, adoptive

transfer of CD4 T cells from normal syngeneic animals into thymectomized mice

inhibited the development of these diseases. Based on these studies, it was

inferred that there might exist two types of CD4 T cells in the periphery of normal

untreated mice/rats: one capable of mediating autoimmune disease and the other

capable of dominantly suppressing them (Sakaguchi et al., 1985).

Following this, attempts were made to separate these two putative CD4+

T cells on the basis of cell surface markers. In 1985, Sakaguchi demonstrated

that transfer of the CD5low CD4+ T cell subset into nude mice led to the

development of autoimmunity similar to neonatally thymectomized mice while co-

transfer with normal untreated CD4+ T cells inhibited autoimmunity (Sakaguchi et

al., 1985). Similar findings by Powrie and Morrissey, demonstrated the role of

CD45RB CD4+T cells in mediating autoimmune diseases (Powrie et al., 1993)

(Morrissey et al., 1993). In 1995, Sakaguchi et al., identified CD25 as a definitive

marker of CD4 T cells capable of suppressing autoimmune diseases based on

adoptive transfer studies (Sakaguchi et al., 1995). As one would expect based

on previous findings, CD25+ T cells were confined in the CD5 and CD45RB

fraction of CD4 T cells (Sakaguchi et al., 2007). Furthermore, removal of CD25+

T cells not only elicited autoimmune disease but also led to enhanced immune

response against non-self antigens (Sakaguchi et al., 1995). Together, all these

results collectively led to the identification of thymus derived CD4+ CD25+ T cells

high

high low

Page 40: Phospholipase D Signaling in T Cells

23

that mediates the maintenance of tolerance to self and controls the magnitude of

immune response to non-self. These CD4+CD25+ T cells were termed as

naturally occurring regulatory T cells (nTreg cells).

Around the same time as the discovery of nTreg cells, two other reports of

CD4 T cells with regulatory properties surfaced. Groux et al., showed that naive

T cells from OVA TCR-transgenic mice, repeatedly stimulated with OVA and IL-

10, differentiated into T cells with a unique cytokine profile distinct from Th1 or

Th2 cells (Groux et al., 1997). They produced IL-10, some IL-5, and IFN- , with

or without TGF- , but showed only marginal or no IL-2 and IL-4 production.

These cells prevented the development of T cell mediated autoimmune

responses. Consequently, these cells were designated as type 1 regulatory T

cells (Tr1) (Jonuleit and Schmitt, 2003) (Beissert et al., 2006) (Fig 3).

Also, Chen et al., described a subset of T cells in mice that suppressed

the onset of myelin basic protein (MBP)-specific experimental autoimmune

encephalitis following induction of oral tolerance to MBP (Jonuleit and Schmitt,

2003). The suppression of these T cells was primarily mediated through TGF

(Transforming growth factor)-β and was called T helper type 3 (Th3) cells

(Beissert et al., 2006)(Fig 3).

Phenotypic and functional characteristics of Treg cells

+ +Naturally arising Treg cells are mostly CD4 CD25 and constitute 5–10% of

peripheral T cells in normal mice (Fehervari and Sakaguchi, 2004). These cells

suppress cytokine production (e.g. IL-2, IL-4, IFN-γ) and proliferation of antigen-

Page 41: Phospholipase D Signaling in T Cells

24

receptor stimulated non-regulatory CD4 and CD8 T cells. Although their mode of

suppression is not clearly understood, emerging studies have put forth many

interestingly mechanisms that can be broadly classified into three classes:

contact dependent, soluble factors and killing of target cells. The contribution of

cell contact dependent mechanisms have been suggested based on the inability

of Treg cells to suppress effector T cell proliferation when the two populations

were separated using a semi-permeable membrane (Thornton and Shevach,

1998). Following cell contact, Treg cells might deliver negative signals to

responding T cells through increased cAMP levels (Bopp et al., 2007), which is

inhibitory for T cell proliferation and IL-2 production. Also, production of the

immunosuppressive nucleoside, adenosine through the action of CD39 and

CD73 expressed on the surface of Treg cells has been suggested (Deaglio et al.,

2007). Also, Treg cells have been proposed to suppress immune responses

through cytokine deprivation induced apoptosis of effector T cells (Pandiyan et

al., 2007). IL-2 is a critical growth/survival factor for Treg cells (Fontenot et al.,

2005). At the same time, the presence of exogenous IL-2 abrogates the

suppressive property of Treg cells.

Although surface phenotype of CD4+CD25+ was originally used to isolate

these populations, it is not a definitive marker of natural regulatory T cells since

activated effector T cells also express CD25. Many other cell surface markers

expressed by Treg cells are CD38 (Martins and Aguas, 1999; Read et al., 1998),

CD62L (Ermann et al., 2005; Mudter et al., 2002; You et al., 2004), CD103 (Banz

et al., 2003), glucocorticoid-induced tumor necrosis factor (TNF) receptor

Page 42: Phospholipase D Signaling in T Cells

25

(GITR)(Shimizu et al., 2002), or low levels of cell-surface CD45RB (Powrie et al.,

1996; Powrie et al., 1994; Read et al., 2000; Taams et al., 2001), though none of

these are exclusive to Treg cells. Currently, the most reliable marker that

exclusively distinguishes Treg cells from non-Treg cells is a transcriptional

repressor Foxp3 (forkhead box P3)(Fontenot et al., 2003; Gambineri et al., 2003;

Hori et al., 2003; Khattri et al., 2003; Schubert et al., 2001). Mutations of foxp3

gene result in severe autoimmune disorders both in human and mouse (Bennett

et al., 2001; Brunkow et al., 2001; Chatila et al., 2000; Wildin et al., 2001). Foxp3

gene is required for the production and maintenance of regulatory T cells

(Fontenot et al., 2003; Kim et al., 2007) and most importantly Foxp3 is not

induced upon activation of non-suppressive murine CD4+CD25- T cells (Fontenot

et al., 2003; Hori et al., 2003; Khattri et al., 2003).

Accumulating evidence suggests that the function of Treg cells is not

limited to suppression of autoimmunity and they play significant roles impairing

the immune response to chronic viral infections (Mills, 2004; Mills and McGuirk,

2004; Rouse and Suvas, 2004; Suvas et al., 2004; Suvas et al., 2003; Toka et

al., 2004). This was first reported using persistent Friend leukemia virus infection

of mice (Iwashiro et al., 2001). Mice infected with this virus have suppressed

CD8 T cell-mediated immunity against virus-infected cells. When CD4+CD25+ T

cells are removed from peripheral T cells of infected mice, CD8 T cells markedly

reduced viral levels during persistence (Dittmer et al., 2004). Increase of Treg

cells was also reported in cases of infection with HIV (Weiss et al., 2004).

Page 43: Phospholipase D Signaling in T Cells

26

Other studies have demonstrated that the frequency of Treg cells

increases under tumor bearing conditions (Waldmann, 2006), as well as during

pregnancy (Saito et al., 2005; Zenclussen, 2005). Conversely, decrease of Treg

cells was reported in cases of autoimmune disease such as multiple sclerosis,

type I diabetes, and rheumatoid arthritis (Lan et al., 2005).

As mentioned previously, regulatory T cells (CD4+Foxp3+) can also be

induced in the periphery and hence are called “adaptive” or “induced” Treg cells.

IL-10 producing Tr1 cells proliferate poorly after polyclonal or Ag-specific

activation in vitro and functional studies revealed that these cells have

immunosuppressive properties and can prevent the development of T cell-

mediated autoimmune responses (Levings et al., 2002) (Jonuleit and Schmitt,

2003). Tr1 can control the activation of naive and memory T cells both in vitro and

in vivo and suppress Th1- and Th2-mediated immune responses to pathogens,

tumors, and alloantigens (Groux, 2003). Furthermore, supernatants of activated

Tr1 cells strongly reduce the capacity of dendritic cells (DC) to induce

alloantigen-specific T cell proliferation (Levings et al., 2002) (Roncarolo et al.,

2001). The suppressive effects of Tr1 cells are reversed by blocking Abs against

IL-10, showing that the inhibitory capacity of Tr1 cells is mainly mediated through

production of immunosuppressive IL-10 (Groux et al., 1997; Levings et al., 2002).

Regulatory Th3 cells are a unique T cell subset induced by orally

administered Ag in vivo and triggered in an Ag-specific fashion (Jonuleit and

Schmitt, 2003). They provide help for IgA production and have suppressive

properties for Th1 and Th2 cells (Jonuleit and Schmitt, 2003; Weiner, 2001a).

Page 44: Phospholipase D Signaling in T Cells

27

However, the suppression of immune responses mediated by Th3 cells are Ag

nonspecific and can occur as bystander suppression through secretion of TGF-β.

Because TGF-β is broadly expressed and influences the functional activity of

multiple cell types, TGF-β secreting Th3 cells probably have a major role in many

aspects of immune regulation and T cell homeostasis (Weiner, 2001b).

Page 45: Phospholipase D Signaling in T Cells

28

Fig 3. Different subsets of regulatory T cells. Naturally occurring regulatory T

cells mature and migrate from the thymus and constitute 5–10% of peripheral T

cells in normal mice. The inducible populations of regulatory T cells include

distinct subtypes of CD4+ T cell: TR1 cells, which secrete high levels of IL-10 and

TH3 cells, which secrete high levels of TGF-β. TGF-β can also induce Foxp3

expression CD25- T cells and thus confer regulatory properties to T cells in the

periphery. Modified from Milojevic et al (Milojevic et al., 2008).

Page 46: Phospholipase D Signaling in T Cells

29

IMMUNE HOMEOSTASIS: POPULATION BALANCE BETWEEN EFFECTOR

AND REGULATORY T CELL SUBSETS

The immune system faces the daunting task of protecting the host against

a wide variety of pathogens at the same time maintaining peace towards self.

CD4 T cells play key roles in orchestrating both the functions of the immune

response. As mentioned previously, naïve CD4 T cells specialize to become

distinct subsets and produce restricted patterns of cytokines that are tailored to

combat various pathogens. The range of infections afflicting HIV infected

individuals following decline in their CD4 T cell count underscores the importance

of CD4 T cells in host defense (O'Shea and Paul, 2010). On the other hand,

heterogenous populations of CD4+ regulatory T cells suppress the immune

response to both self and non-self antigens. Adoptive transfer of Treg depleted

CD4 T cells leads to development of spontaneous autoimmunity, illustrating the

dominant role of Treg cells in mediating tolerance and autoimmunity.

Homeostasis of the immune response depends on the balance of

population dynamics and functionality between these effector and regulatory T

cell subsets (Mills, 2004). Perturbations in this balance have been reported in

many diseases. Tang et al, reported that type I diabetes (T1D) progression in the

NOD mice was associated with a progressive loss of functional Treg cells in the

inflamed islets (Tang et al., 2008). Conversely, expansion of Treg cells in

inflammatory mouse models like EAE has been reported to be associated with

disease resolution (Knoechel et al., 2005) (Korn et al., 2007).

Page 47: Phospholipase D Signaling in T Cells

30

Various mechanisms have been proposed to explain the alterations in the

balance between Treg cells and effector T cells in vivo. The importance of IL-2

levels in the maintenance of homeostasis is evident from different studies. For

instance, Tang et al observed that low dose injections of IL-2 supported the

growth and survival of Treg cells over effector T cells (Teff) whereas a high dose

of IL-2 led to rapid expansion of effector T cells (Tang et al., 2008). Based on

these observations they further proposed that Treg cells versus effector T cells

regulate each other through a feedback loop. IL-2 production by activated Teff

cells expands and sustains Treg cells, which in turn feeds back to suppress the

Teff cell response and maintain normal immune homeostasis. Disruption of this

crosstalk can lead to the dysregulation of the Treg cell: Teff balance thus

contributing to the development of disease (Tang et al., 2008).

Recent emerging studies suggest that plasticity and interconvertibility

among different subset of T cells might also alter the immune balance (Wan and

Flavell, 2009). Foxp3 expressing T cells from wild type mice lose Foxp3

expression and exhibit activated-memory phenotype and produce inflammatory

cytokines (Zhou et al., 2009). Also, fully differentiated Th17 cells were shown to

convert into Th1 cells in the absence of TGF-β (Lee et al., 2009). However, the

mechanism and the proportion of T cells undergoing such inter-conversion are

not known. This newly defined property of CD4 T cells can have far reaching

consequences in the pathogenesis and treatment of variety of diseases ranging

from autoimmunity to cancer.

Page 48: Phospholipase D Signaling in T Cells

31

Studies of immune evasion by various pathogens also provide clues to the

mechanisms of alteration of immune balance. One of the common immune

evasion strategy employed by many pathogens involves increasing the

production of various immunosuppressive or anti-inflammatory molecules to

skew the immune response towards regulatory phenotype. Many intracellular

pathogens and viruses either induce cellular IL-10 or encode their own IL-10

homolog to delay or suppress the host immune response (Redpath et al., 2001).

Various environmental toxins have also been shown to tip the immune

balance between the effector and regulatory T cells. Quintana and Veldhoen et al

demonstrated that dioxin signaling through aryl hydrocarbon receptor (AHR)

increased Treg activity and proliferation while activation of AHR by FICZ led to

increased Th17 activity (Quintana et al., 2008) (Veldhoen et al., 2008).

Manipulation of the immune balance serves as an attractive target for

therapy for a range of human diseases. The ideal treatment for autoimmune

disease would be to decrease the frequency of effector T cells and increase the

frequency of Treg cells. Several investigators have devised many therapeutic

strategies to tip the Treg: effector T cell balance. For example, administration of

monoclonal antibodies to CD3, CD4 or CD40L in the presence of antigen causes

selective expansion of Foxp3+ Treg cells, producing the dominancy of Treg cells

over effector T cells (Wing and Sakaguchi, 2010). Rapamycin, an inhibitor of the

Akt-mTOR pathway, increases the number of Foxp3+ T cells in part by enhancing

Foxp3 transcription and by rendering Treg cells resistant to apoptosis (Basu et al.,

2008; Strauss et al., 2007c; Wing and Sakaguchi, 2010). Administration of IL-2–

Page 49: Phospholipase D Signaling in T Cells

32

anti-IL-2 complexes selectively expands Foxp3+ Treg cell populations (Webster et

al., 2009; Wing and Sakaguchi, 2010). On the other hand, the ideal treatment in

the case of tumours would be to increase the frequency of effector T cells and

reduce the numbers of Treg cells. Administration of anti-CD25 mAb (PC61) has

been reported to cause reduction in the number of CD4+CD25+ cells thus

enabling tumour rejection (Onizuka et al., 1999).

Role of TCR mediated signaling pathwyas in altering the immune balance

The underlying theme in the above mentioned strategies for selective

expansion/inhibition of Treg cells/Teff is the manipulation of IL-2 and/or IL-2R

signaling in CD4 T cells. The potential caveat in these strategies is that IL-2 is a

T cell growth factor for both effector and regulatory T cells and this might lead to

high variability in the in vivo effect in a heterogeneous patient population.

An alternative strategy would be to identify specific factors/signaling

cascades present exclusively in one subset versus the other and to exploit those

molecules for therapeutic applications. Consistent with this notion several studies

have reported the role of specific TCR associated signaling molecules in

controlling the direction of helper T cell subset differentiation.

Recognition of MHC–antigen peptide complex by T cell receptor triggers a

cascade of downstream signaling events. The most proximal biochemical event

is the tyrosine phosphorylation of the immunoreceptor tyrosine-based activation

motifs (ITAMs) of the TCR/CD3 complex (Guy and Vignali, 2009; Iwashima,

2003; Nakayama and Yamashita, 2010). Lck, an Src family tyrosine kinase plays

Page 50: Phospholipase D Signaling in T Cells

33

an important role in the tyrosine phosphorylation of ITAMs. Then, the Zap70 Syk

family kinase is recruited to the phosphorylated ITAMs through the SH2 domains

of ZAP70. The recruited ZAP70 is then phosphorylated and activated by

surrounding Lck tyrosine kinase, thus leading to the phosphorylation of LAT

molecules (Nakayama and Yamashita, 2010). Following these proximal events,

the activation of various distinct signaling pathways is initiated, including (i) the

Ras/ERK MAPK cascade, (ii) the Ca/calcineurin/NF-AT pathway, and (iii) the

PKC/NF-κB pathway (Nakayama and Yamashita, 2010). Interestingly, the

differential requirement for various signaling molecules involved in both proximal

and distal events of TCR signaling has been reported in different subsets of CD4

T cells. For instance, Lck activity is required for the generation of Th2 cells as

dominant negative (DN) Lck transgenic mice shows impaired Th2 cell generation

as against intact Th1 cells (Yamashita et al., 1998). In the same lines, Tan et al.,

showed that PKC is necessary for IL-17 production in the mice (Tan et al., 2006).

Further, inhibition of ERK/MAPK cascade using pharmacological inhibitor or DN

Ras results in increased Th1 cell differentiation (Yamashita et al., 1999). Recent

studies from our lab group reported the differential requirement for phospholipase

D (PLD) signaling between regulatory versus non-regulatory T cells. Suppression

of TCR induced increases in PLD activity led to selective inhibition in the

proliferation and survival of effector T cells. As a consequence, the frequency of

Treg cells increased under these conditions. Thus, manipulation of PLD activity

may serve as a tool to alter the immune balance. Hence, identification of

Page 51: Phospholipase D Signaling in T Cells

34

factors/molecules that modulate PLD activity will be of significant therapeutic

importance.

BALANCING ACT OF PHOSPHOLIPASE D (PLD) IN T CELLS

PLD: structure, function and expression

PLD belongs to the phosphodiesterase family of proteins, found in

bacteria, fungi and animals (Melendez and Allen, 2002). Mammalian PLDs

catalyze the hydrolysis of phosphatidylcholine (PC) to phosphatidic acid (PA) and

choline (Exton, 2002). Phosphatidic acid is an important lipid second messenger,

which mediates the downstream effects of PLD signaling. PA is an abundant

phospholipid that can be also synthesized from pathways other than PLD

signaling (Athenstaedt and Daum, 1999) .

A unique characteristic of PLD is that primary (1–) alcohol serves as a

more competitive substrate compared to water (> 1000 fold), and the presence of

1–alcohol effectively blocks PLD signaling (Morris et al., 1997) (Fig 4a). In the

presence of 1–alcohol (such as ethanol or 1–butanol), PLD prefers

transphosphatidylation to hydrolysis and produces phosphatidyl–alcohol, which is

poorly metabolized. As a result, the production of PA is significantly reduced.

Thus, 1-alcohol is a potent suppressive regulator of PLD signaling.

Two isoforms of PLDs are expressed in mammalian tissues namely PLD1

and PLD2, which share 55% identity (Frohman et al., 1999). The domain

structure of PLDs contains phox homology (PX) and pleckstrin homology (PH)

Page 52: Phospholipase D Signaling in T Cells

35

domains at the N-terminal followed by four conserved sequences (I-IV) (Sung et

al., 1999)(Fig 4b). PX and PH domains are implicated in phospholipid and protein

binding. Results from deletional analysis indicate that PIP2 binds at PH domain

and at conserved sequences located between catalytic sites II and III (Hodgkin et

al., 1999) (Sciorra et al., 1999). The catalytic sites II and III contain the

characteristic ‘HxKxxxxD’ motifs (Frohman et al., 1999). In addition, PLD1

contains an 116 amino acid ‘loop’ region inserted between the II and III catalytic

sites. The C-terminal four amino acids of mammalian PLDs are evolutionarily

conserved and modification of these residues (mutation/deletion) causes loss of

catalytic activity (Xie et al., 2000) (Liu et al., 2001). PLD1 exists as two splice

variants, PLD1a and PLD1b that differ in the length of the loop region. This

region may function as a negative regulator of PLD1 since deletion of the loop

has been shown to cause modest up regulation of basal catalytic activity of PLD1

(Frohman et al., 1999).

Although PLD1 and PLD2 have the same substrate specificity, the

activities of PLD1 and PLD2 are regulated differently. PLD1 has low basal

activity in vitro and it can be stimulated in vitro using agonists as it is coupled to

receptor signaling via the small GTPases, ARF1/6, Rho, CDC42, and RalA. PKC

mediated phosphorylation also up regulates PLD1 activity. In contrast, PLD2 is

less responsive to small GTPases and may be constitutively active and regulated

by inhibition (Colley et al., 1997).

Page 53: Phospholipase D Signaling in T Cells

36

A)

B)

Fig 4. Schematic representation of catalytic reaction of PLD and domain

structure of PLD isoforms found in mice. (A) In aqueous solution, PLD

catalyzes hydrolysis of phospholipid to yield Phosphatidic acid (PA). But in the

presence of small amounts of ethanol, PLD yields Phosphatidyl ethanol (PEt).

R1, R2 and R3 are fatty acid side chains. B) Domain structure of PLD isoforms in

mice. PX, phox homology; PH, pleckstrin homology domain. Motifs I~IV are

conserved regions and II and IV contain HKD sequences. Modified from

(Frohman et al., 1999).

Page 54: Phospholipase D Signaling in T Cells

37

PLD1 is mainly found in perinuclear regions, whereas PLD2 localizes

predominantly in the plasma membrane (Colley et al., 1997; Sung et al., 1997).

Both PLD1 and PLD2 are palmitoylated in the pleckstrin homology (PH) domain

and found associated with membrane raft fractions (Sugars et al., 1999; Xie et

al., 2001). The site of generation of PA and the availability of effector targets

determines its downstream signaling. Hence, the sub–cellular distribution of both

the isoforms of PLD is an important factor affecting its function and regulation.

Functions of PA

PA is now recognized as a vital second messenger for many cell types

(Andresen et al., 2002; Chen, 2004; Chen and Fang, 2002; Cummings et al.,

2002; English, 1996; Ghosh and Bell, 1997; Houslay and Adams, 2003; Rizzo

and Romero, 2002). For instance, the direct interaction of PA has been

demonstrated for several signaling proteins, namely cRaf-1 (Ghosh et al., 1996;

Rizzo et al., 1999; Rizzo et al., 2000; Wang et al., 2002), mTOR (Chen and

Fang, 2002; Fang et al., 2003; Fang et al., 2001; Lim et al., 2003),

phosphodiesterase 4D3 (El Bawab et al., 1997; Grange et al., 1998; Grange et

al., 2000; Savany et al., 1996), PKC ξ (Bandyopadhyay et al., 2001; Limatola et

al., 1994),SHP-1(Frank et al., 1999; Tomic et al., 1995) and sphingosine kinase-1

(Delon et al., 2004). PA plays a significant role in the regulation of the function of

these proteins. The binding of PA to SHP-1, for instance, activates the

phosphates in vitro (Zhao et al., 1993). PA also functions in protein recruitment.

Page 55: Phospholipase D Signaling in T Cells

38

Interactions with PA are essential for the recruitment of cRaf-1 to membranes

(Rizzo et al., 2000).

PA is also a precursor for a number of alternative signaling molecules.

For example, PA can be converted to diacylglycerol (DAG), by phosphatidic acid

phosphohydrolases (Balboa et al., 1995; Billah et al., 1989; Lassegue et al.,

1993; Mullmann et al., 1990). DAG is an activator of conventional and novel

PKC isoforms and other signaling molecules (Cummings et al., 2002;

Gudermann et al., 2004; Haeffner, 1993; Kikkawa et al., 1989; Yang and

Kazanietz, 2003). DAG is also generated by the hydrolysis of

phosphatidylinositol (4,5)-bisphosphate by phospholipase C (Haeffner, 1993;

Nishizuka, 1992). The nature of the DAG species generated by the action of

phospholipase C differs from that derived from PA through the action of

phospholipid phosphohydrolases (Liu et al., 1999; Plo et al., 2000). The

consequences of this difference are currently unknown.

Hence, PA mediates a variety of cell functions including cell survival,

proliferation, cytoskeletal rearrangement and vesicular trafficking (Fang et al.,

2001; Ghosh et al., 1996; Lim et al., 2003; Rizzo et al., 1999). However, the

precise role of PA in modulating T cell function and survival is not clear.

Regulation of catalytic activity of PLD

Various in vitro studies have elucidated the role of certain co-factors in the

catalytic activation/inactivation of PLD. They can be classified into 3 classes.

Page 56: Phospholipase D Signaling in T Cells

39

i. Phospholipids

PIP2 was determined to be essential for catalytic activity of PLDs (Brown et al.,

1993). PIP3 is also effective in up regulation of PLD activity. A PIP2 binding site

is located between conserved regions II and III of the catalytic domain and

mutation in this region abrogates PLD activity without altering their sub cellular

localization (Frohman et al., 1999). Another PIP2 binding site has been reported

to be present within the PH domain. This binding site affects PLD activity by

altering their sub cellular localization (Oude Weernink et al., 2007).

ii. Small GTPase

Several small GTPase’s can activate PLD. Activation of PLD1 by ARF is GTP

dependent and requires the N-terminal region of PLD. However, the detailed

mechanism of PLD activation by ARF is not known (Frohman et al., 1999).

Several Rho family proteins, namely RhoA, Rac and CDC42 are also involved in

PLD activation. Invitro, GTPγs activated forms of these proteins have been

shown to activate PLD (Hammond et al., 1997). Botulinum C3 toxin has been

reported to block PLD activation by inactivating Rho (Malcolm et al., 1996;

Schmidt et al., 1996). C-terminus of PLD1 is reported to be the interaction site for

Rho (Du et al., 2000).

Iii PKC

Treatment of cells with phorbol esters effectively activated PLD, suggesting that

Protein Kinase C (PKC) is upstream of PLD activation (Exton, 2002). Several

Page 57: Phospholipase D Signaling in T Cells

40

phosphorylation dependent and independent modes phosphorylation by PKC has

been reported (Hammond et al., 1997).

TCR signaling and PLD

Attachment of the T cell receptor complex by its cognate antigen triggers a

cascade of downstream signaling events including phosphorylation of ITAM

motifs, phosphoinositide hydrolysis and calcium flux. Emerging results indicate

that antigen receptors that transduce signals via immuno–receptor tyrosine

based activation motifs (ITAMs) are coupled to PLD activation (Melendez and

Allen, 2002). Fcγ and ε receptors activate PLD by the antigen-antibody complex

(Cockcroft et al., 2002; Gewirtz and Simons, 1997; Gruchalla et al., 1990; Jose

Lopez-Andreo et al., 2003; Kusner et al., 1999; Loegering and Lennartz, 2004).

TCR stimulation was also shown to upregulate PLD activity by 2~300% (Reid et

al., 1997). Inhibition of PLD activity in T cell line caused loss of AP-1 activation,

cytokine production and proliferation (Mollinedo et al., 1994). BCR was also

shown to activate PLD (Forssell et al., 2000; Hitomi et al., 1999).

Previously our lab group had demonstrated that PLD1 and PLD2 signaling

are involved in different stages of TCR signaling (Singh et al., 2006). Over

expression of both PLD1 and PLD2 potentiated NF-AT activation by TCR in

Jurkat cells. CD2/ZAP-70, a fusion protein between the extracellular and

transmembrane domains of CD2 with Zap-70 activates NFAT when expressed in

Jurkat cells. When co-expressed with CD2/Zap-70, PLD1 induced marked

Page 58: Phospholipase D Signaling in T Cells

41

upregulation of NFAT activity whereas PLD2 induced only a slight upregulation.

Conversely, lipase inactive PLD2 but not PLD1, blocked anti-TCR antibody

induced tyrosine phosphorylation of TCR ζ. Together, the data indicated that

PLD2 is required for TCR signaling events proximal to the recruitment of Zap-70

to ITAM while PLD1 is involved in events distal to tyrosine phosphorylation of ζ.

It is not yet clear how PLDs are coupled to TCR. Several groups have

reported the presence of PLD enzymes in caveolae, which are specialized

domains of the plasma membrane, related to sphingolipid and cholesterol-rich

micro domains or rafts (Gidwani et al., 2003; Hekman et al., 2002; Zheng et al.,

2003). In view of the fact that molecules with a key role in T cell signaling like

Lck also localize to these domains, it is tempting to suggest that lipid rafts might

provide the perfect platform for PLD and TCR coupling.

In our recent study, we also demonstrated that the increase in PLD activity

after TCR stimulation was two fold in CD4+CD25- cells and rather unchanged in

CD4+CD25+ T cells. Moreover, PLD1 expression was substantially lower in

CD4+CD25+ cells than in CD4+CD25- cells indicating differences in the

requirement for PLD signaling between regulatory and non-regulatory T cells.

Inhibition of the TCR induced increase in PLD activity in CD4+CD25- T cells

substantially affected their activation; proliferation and cytokine secretion while it

had no effect on CD4+CD25+ T cells. Consequently, culture of heterogeneous

population of naïve CD4 T cells in the presence of TCR stimulation and

exogenous IL-2 led to preferential expansion of Foxp3+CD4+ T cells. Moreover,

these expanded Treg cells exhibited potent immune suppression in a mouse

Page 59: Phospholipase D Signaling in T Cells

42

model of inflammatory bowel disease (IBD) indicative of their functional

capabilities. These data also illustrated the biological significance of PLD

signaling in T cells. Investigations to elucidate the anti-proliferative effect of

inhibition of PLD signaling on CD4+CD25- T cells suggested that blockade of

high affinity IL-2 receptor (CD25) expression may be a factor. However, the

precise molecular mechanism of regulation of CD25 expression by PLD is still

unknown. Also, the specific function of the PLD isoforms in mediating this

differential effect on CD4 T cells is unknown.

Modulation of PLD activity by extracellular stimuli

The above mentioned data suggested to us that PLD signal regulation

could play a pivotal role in controlling the balance between regulatory and non-

regulatory T cells. In vivo suppression of PLD signaling may promote dominance

of naturally arising Treg cells under physiological conditions. Conversely,

augmenting PLD signaling may promote growth of effector T cells and break the

immune suppression by Treg cells. This led us to ask: Is it possible for

pathogens/tumours to inhibit PLD activity (directly / indirectly through secretion of

factors) to subvert the protective immune responses of the host? Alternatively,

does elevated PLD activity contribute to autoimmune responses?

There have been a number of studies that indicate the modulation of PLD

activity by various extracellular stimuli. These stimuli in turn mediate their effect

on PLD through the help of above-mentioned cofactors of PLD signaling. Alcohol

Page 60: Phospholipase D Signaling in T Cells

43

is an exogenous factor that alters the immune phenotype of the host (Nelson and

Kolls, 2002). T cells from individuals who were exposed to alcohol both

chronically and acutely showed a higher frequency of T cells expressing CD25

(Cao et al., 1998; Laso et al., 1996a; Laso et al., 1996b; Sacanella et al., 1998;

Santos-Perez et al., 1996). One study showed that these T cells were

exclusively CD4+ (Laso et al., 1996b). Moreover, the frequency of CD25+ T cell

remained high even 9 months after alcohol withdrawal. In another study, clear

positive correlation was observed between total estimated life time dose of

ethanol consumed and the percentage of PBLs expressing CD25 (Sacanella et

al., 1998). Our studies suggested that these effects might be channeled through

suppression of PLD signaling and increased frequency of Treg cells.

Pathogens like Clostridium difficile might also alter the host immune

response through PLD signaling. The major virulence factors produced by

C.difficile are Toxin A (TcdA) and Toxin B (TcdB) (Genth et al., 2008). Inside the

host cell, these toxins mono-glucosylate and thereby inactivate small G proteins

of the Rho sub-family (Genth et al., 2008). Since PLD is one of the downstream

effector molecules of Rho, these toxins indirectly inhibit PLD activity (Schmidt et

al., 1996). We propose that the functional consequence of inhibiting PLD

signaling is enrichment of Treg cells. This in turn might facilitate the evasion of

host immune response by C.diff through elimination of the effector response by

non-Treg cells.

The receptor system that has been linked to PLD regulation includes G-

protein coupled receptors and tyrosine kinase coupled receptors (Frohman et al.,

Page 61: Phospholipase D Signaling in T Cells

44

1999). G-protein coupled receptors include chemokine receptors (RANTES, IL8)

(Bacon et al., 1995; Bacon et al., 1998), adenosine receptors (Selvatici et al.,

2006; Thibault et al., 2002), N-formyl peptide receptor (FPR) (Selvatici et al.,

2006), P2Z/P2X7 nucleotide receptor (Coutinho-Silva et al., 2003) and

prostaglandin (PGE2) receptors (Burelout et al., 2004). Tyrosine kinase coupled

receptors include TCR, BCR, FcReRI, and FcRI (Melendez and Allen, 2002) and

Insulin receptors (Farese, 1988).

These extracellular stimuli alter PLD activity in both a positive and

negative manner. Antigen and chemokine receptors elevate the activity of PLD

(Bacon et al., 1995; Bacon et al., 1998). FPR, which recognizes bacterial

peptides, also elevates PLD activity (Selvatici et al., 2006). On the other hand,

inflammation related receptors, including adenosine receptors (Selvatici et al.,

2006; Thibault et al., 2002), lipoxin A (4) receptor (Levy et al., 1999) and PGE2

receptors (Burelout et al., 2004) suppress PLD activation. Interestingly, a recent

report indicated that PGE2 induces enrichment of regulatory T cells (Baratelli et

al., 2005).

Most of the studies indicate that PLD1 is associated with receptor induced

PLD activity and this involves the role of small GTPases or PKCs whereas PLD2

may be constitutively active and is a target of negative regulation. In contrast to

the activation process of PLD, the process that inactivates PLD is not well

characterized. Adenosine (A2a) receptor signaling in neutrophils (Thibault et al.,

2002) and A3 receptor signaling in ischemic heart (Mozzicato et al., 2004) has

been shown to modulate PLD activity. PGE2 receptor has also been shown to

Page 62: Phospholipase D Signaling in T Cells

45

inhibit PLD activity. Both A2a and PGE2 stimulate adenylate cyclase and thereby

increase the intracellular cAMP levels. These data indicate that downstream

targets of cAMP, such as PKA may mediate suppression of PLD activity.

However, no clear connection between PKA and PLD has been made at this

point. Interestingly, a recent report showed that βγ subunits of G protein could

inhibit PLD1 activity by direct interactions (Preininger et al., 2006). Other

negative regulatory factors of PLD include ceramide (Nakamura et al., 1996),

presqualene diphosphate (PSDP) (Levy et al., 1999) and munc-18-1 (Lee et al.,

2004).

Adenosine and PLD signaling

Adenosine (Ado) is a purine nucleoside formed from its precursor ATP by

the action of CD39 [nucleoside triphosphate dephosphorylase (NTPD)] and

CD73 (5-ectonucleotidase)(Hasko et al., 2008). Adenosine is constitutively

present at low concentrations in extracellular space however metabolically

stressful conditions (hypoxia, cell death) increase its levels in extracellular space

drastically (Fredholm et al., 2001; Linden, 2001). Numerous studies suggest the

role of Ado as an immune modulator. First, high concentrations of Ado represent

a pre-eminent alarm signal indicating tissue injury to the surrounding cells.

Secondly, Ado occupies its cognate receptor present on a variety of immune

cells including dendritic cells (Panther et al., 2003; Schnurr et al., 2004),

neutrophils (Cronstein et al., 1983) and lymphocytes and mediates immune

Page 63: Phospholipase D Signaling in T Cells

46

suppression (Borsellino et al., 2007; Hasko et al., 2008; Kobie et al., 2006; Zarek

et al., 2008). A physiological response of Ado is elicited by engaging one or

more of the four G-protein coupled receptors (A1, A2a, A2b and A3) (Jacobson

and Gao, 2006).

Recent studies have elucidated an important role for adenosine in

mediating the immunosuppressive properties of Treg cells. It was demonstrated

that Treg cells express CD39/CD73 and thus hydrolyze extra cellular ADP and

ATP to adenosine (Deaglio et al., 2007; Kobie et al., 2006). Also, Treg cells

isolated from CD39 knockout mice do not suppress the proliferation of

CD4+CD25- cells, indicating the role of CD39 in modulating Treg function

(Deaglio et al., 2007). Similarly, Treg cells isolated from wild type mice failed to

suppress CD4+CD25- cells from A2a receptor knockout mice (Naganuma et al.,

2006; Zarek et al., 2008). Numerous studies have established the

immunosuppressive function of A2a receptor signaling. Indeed activation of A2a

receptor on Treg cells causes upregulation of Foxp3 expression (Zarek et al.,

2008). Interestingly, A2a receptor occupancy on neutrophils reduces PLD activity

by limiting the recruitment of Arf, RhoA, and PKC to membranes (Thibault et al.,

2000).

The combined effects of activated adenosine A2 and A3 receptors (A2AR,

A3AR) on mononuclear cells of the immune system are considered anti-

inflammatory (Hasko et al., 2008; Jacobson and Gao, 2006). Activation of A3

receptors on macrophages inhibits the release of tumor necrosis factor alpha

(TNF-α) and thus exerts beneficial effect in treating inflammatory disorders like

Page 64: Phospholipase D Signaling in T Cells

47

arthritis (Baharav et al., 2005). Conversely, A3 receptor activation on mast cells

triggers histamine release (Salvatore et al., 2000) and thus is an important player

in mediating asthma (Hasko et al., 2008). In addition, A3 receptors have been

demonstrated to utilize PLD and RhoA in dictating cell functions. However, the

precise function of A3 receptor in T cells has not been established.

Based on these studies, we set out to determine the effect of adenosine in

modulating PLD signaling in T cells. We also tested if PLD signaling plays any

role in mediating the immune suppression of adenosine.

CONCLUSION

The heterogeneity of T cell subsets has led us to appreciate the functional

importance of CD4 T cells in fighting pathogens, inducing autoimmunity as well

as maintaining tolerance to self. It is widely accepted that the population balance

between the various subsets of CD4 T cells is central to maintaining immune

homeostasis. Hence, identification of factors controlling this population balance

like PLD signaling will provide novel therapeutic strategies for controlling variety

of diseases ranging from autoimmunity to tumors.

Page 65: Phospholipase D Signaling in T Cells

48

CHAPTER TWO

MATERIAL AND EXPERIMENTAL METHODS

Mice

All mouse work was done in accordance with the Animal Care and Use

Committee guidelines of Medical College of Georgia and Loyola University.

Animals were housed in pathogen-free conditions prior to manipulations.

C57BL/6 mice and RAG-1 knock out mice were obtained from Jackson

laboratory. CD4 CRE mice were obtained from Taconic. P25 TCR transgenic

mice was generously gifted by Dr. T. Saito (Japan) (Tamura et al., 2004).

D011.10 TCR transgenic mice were housed in pathogen free facility in Medical

College of Georgia (Augusta, Georgia, USA).

Generation of PLD2 -/- mice

PLD2fl/fl mice were generated by selecting the PLDneo ES cell clone

(Mayuko Takezaki). The floxed mouse PLD2 (PLD2fl/fl) mice line was bred with

FLP transgenic mice to delete the neo transgene. Then PLD2fl/fl (minus neo)

mice were bred with mice carrying CD4 cre transgene. Mice were routinely

screened by genotyping of the tail DNA using PCR. Primers used for genotyping

Page 66: Phospholipase D Signaling in T Cells

49

are listed in Table I. Genomic DNA was isolated from CD4 T cells and analyzed

by southern genomic blot for targeted recombination.

Southern Blot

Genomic DNA was isolated from CD4 T cells using standard procedures

(Blin and Stafford, 1976). Ten micrograms of genomic DNA was digested with

the restriction enzyme Bgl II, electrophoresed on a 0.7% agarose gel, transferred

to Hybond-N and UV cross-linked according to the manufacturer’s suggestion

(Amersham Pharmacia Biotech). Filters containing the immobilized DNA were

incubated with 1-5*106 cpm/ml of radio labeled probe. 25ng cDNA-probe was

labeled with 32P using the Prime It® II Random Primer Labeling Kit (Stratagene

#300385). The PLD2 targeting probe corresponds to a 340bp XhoI-EcoRI

fragment located immediately upstream of the short arm of the targeting

construct. Filters were washed at 65 °C in a solution containing 0.1X SSC and

0.1% SDS and analyzed by autoradiography.

Adoptive transfer

CFSE labeled (0.5uM) (donor) p25 CD4 T cells (2.5-5×106 ) were injected

intravenously into Ly5.1 (recipient) mice on day 0. Recipient mice were injected

with 2.5ug of p25 peptide intravenously on day 1. Recipient mice were given

intraperitoneal injections of vehicle (DMSO in PBS), AB-MECA (200ug /kg),

Ethanol (2.9g/kg body weight in 20% vol/vol) in 200 ul from day 0-6. Mice were

sacrificed on day 8 and spleen, mesenteric lymph nodes and all peripheral lymph

Page 67: Phospholipase D Signaling in T Cells

50

nodes (axillary, brachial, cervical, inguinal and popliteal) were harvested and

intracellular staining for Foxp3 expression was performed. Proliferation function

in flowjo software was used to determine number of cells in each cell division.

RAG1 -/- mice

CFSE (0.5uM) labeled CD4 T cells (1- 5x106) from wild type B6 mice were

intravenously transferred into recipient RAG1-/- mice. Mice were injected with

ethanol or PBS intraperitoneally as described previously for 5 days. Mice were

sacrificed on day 6 and splenocytes were analyzed for Foxp3 expression by flow

cytometry.

Cell isolation and culture

Splenic CD4 T cells were isolated using either positive or negative

selection kit from BD Biosciences or MACS Miltenyi Biotec as per manufacturers

protocol. Cell purity was confirmed to be 95% by BDFACS Canto. Cells were

cultured in vitro in RPMI 1640 medium supplemented with 10% v/v heat-

inactivated FBS, 2mM glutamine, 5×10-5 M 2-ME, and 100 U/mL penicillin and

100 µg/mL streptomycin (all were obtained from Thermo Scientific Hyclone).

Treg enrichment

61×10 CD4 T cells were pre-incubated with DMSO, AB-MECA (A3R agonist,

SIGMA), 0.5 % ethanol (SIGMA, 200 proof) and 2 % ethanol (2ml in 24 well

plate). After 16 hrs, we replaced 1.5 ml of culture supernatant with fresh medium

containing 0.2ug/ml of anti-CD3 (e-bioscience), γ-irradiated splenocytes (2500

rad) (2-5×106 cells/well) and different treatment constituents like DMSO. We

Page 68: Phospholipase D Signaling in T Cells

51

replaced 1 ml of medium for the next 72 hrs with medium containing anti-CD3,

2ng/ml of mouse recombinant IL-2 and respective treatment constituents. We

washed the cells on day 4 and cultured them with IL-2 (2ng/ml) for next 3 days.

On day 7, cells were harvested, washed and counted. Cells were then stained

with different fluorescent antibodies and analyzed by flow cytometry.

Activation markers

1×106 CD4 T cells were activated with anti-CD3 (0.2ug/ml) and APCs (T

cell depleted γ-irradiated splenocytes (2500 rad), (2-5×106 cells/well) in the

presence of DMSO, AB-MECA, varying concentrations of ethanol and C.difficile

culture supernatants. 24 hrs later cells were harvested and analyzed by flow

cytometry. In some experiments, CD4 T cells were further sorted into CD4+

CD25+ and CD4+ CD25- T cells using BDFACSAria.

CFSE labeling

Purified CD4 T cells were labeled with 2.5μM CFSE (Invitrogen) for 10 min

at 37°C in PBS. The reaction was quenched by the addition of medium

containing 10% FCS and then washed twice in PBS. CFSE labeled cells were

cultured for 7 days as mentioned above. In some experiments, cells were labeled

with 2.5uM PKH-26 (a red fluorescent cell labeling dye, SIGMA). Cells were

harvested on Day 5 after stimulation.

Cytokine assay

To analyze cytokine production by ELISA, CD4 T cells were stimulated

with plate bound anti-CD3 (2 ug/ml) and soluble CD28 (1 ug/ml) in the presence

Page 69: Phospholipase D Signaling in T Cells

52

of DMSO or AB-MECA. Supernatants were collected at 24, 48 and 72 hrs and

enzyme linked immunoassays were performed as described previously.

To detect cytokine production by intracellular staining, CD4 T cells were

stimulated with plate bound anti-CD3 (2ug/ml) and soluble CD28 (1ug/ml) for 48-

72 hrs. Cells were then rested for 48 hrs in the presence of IL-2. On day 5, cells

were washed and 1×106 were restimulated with PMA (50ng/ml) and Ionomycin

(1ug/ml) for 6 hrs in the presence of Golgistop (Monesin, 2uM). Cells were then

harvested and stained with fluorescent antibodies and analyzed by flow

cytometry.

Th1, Th2 and Th17 skewing conditions

For Th1 skewing conditions, CD4 T cells were stimulated as before and

cultured in media supplemented with mIL-12 (10ng/ml) and anti-IL-4 (10ug/ml).

After 48 hrs, medium was supplemented with additional IL-2.

For Th2 skewing conditions, CD4 T cells were stimulated as before and

cultured in media supplemented with mIL-4 (10ng/ml), anti-IL-12 (10ug/ml) and

anti-IFNγ(10ug/ml). After 48 hrs, medium was supplemented with additional IL-4.

Cells skewed to Th17 were supplemented with TGF-β (2ng/ml), mIL-6

(10ng/ml), anti-IL-4 (2ug/ml) and anti-IFNγ (2ug/ml). Th17 skewed cells were

harvested on day3 or 4 washed and stimulated with PMA and Ionomycin and

analyzed by flow cytometry. Th1 and Th2 skewed cells were harvested on day5

after stimulation and restimulated with PMA and ionomycin.

Page 70: Phospholipase D Signaling in T Cells

53

Induction of Treg cells

To test for induction of Treg cells, CD4+CD25- T cells were stimulated

with APCs (whole splenocytes) and anti-CD3 (0.2-0.4ug/ml). Medium was

supplemented with AB-MECA or DMSO. TGF-β (5ng/ml) was added to positive

control wells. IL-2 (2ng/ml) was added to all wells on day 1 and day 2. Cells were

harvested on Day 5 or 6 and analyzed for Foxp3 expression by flow cytometry.

Flow cytometry

Cell surface staining

Cells in culture were harvested and washed once with FACS buffer (5%

FCS and 0.1% azide in 1X PBS). 1x106

cells were stained with various antibodies

for 1hr at 4 degree and cells were washed three times with FACS buffer. Before

staining with antibodies, cells were incubated for 5-10 mins with 2.4G2(FcR

block). Purified, fluorochrome and biotin-coupled monoclonal antibodies specific

for CD4 (RMA 4–4, GK1.5), CD8 (53–6.7), CD25 (PC61, 7D4), CD3 (145-2C11)

and FoxP3 (FJK-16s) were purchased from eBiosciences (San Diego, CA). Thy

1.2, Ly 5.2 and Vβ11were purchased from Biolegend (San Diego, CA).

Fluorescent antibodies against IL-2, IL-4, IFN-γ was purchased from e-bioscience

and IL-17 was purchased from Biolegend.

Intracellular cytokine staining

0.5-1x106

cells were surface stained and then fixed for 15 mins with 4%

Paraformaldehyde (Sigma) at room temperature. Cells were diluted with FACS

Page 71: Phospholipase D Signaling in T Cells

54

buffer and kept at 4 degrees overnight. The following day, cells were washed and

incubated with permeabilization buffer (50mM Nacl, 5mM EDTA, 0.02% Azide,

0.5 % Triton-X pH7.5) for 15 mins followed by blocking (3% BSA in PBS). Cells

were then incubated with fluorescent antibodies for 45 mins on ice. Finally, cells

were washed three times in FACS buffer and analyzed by flow cytometry.

Intracellular Foxp3 staining

1x106

cells were surface stained with anti-CD4 and anti-CD25, fixed

overnight with Fix/Perm buffer (eBioscience). After 12-16 hrs, cells were then

washed twice with IX permeabilization buffer (eBioscience) and stained with anti-

Foxp3 antibody in IX FACS buffer at 4°C for 1hr. Cells were washed thrice with

IX FACS buffer and analyzed by flow cytometry.

All the data was acquired using FACSCanto or FACSCanto II (Becton

Dickinson) at the LUMC FACS Core Facility, and data was analyzed using

FlowJo analysis software (©Tree Star, Inc).

T cell proliferation assay

CD4 T cells (2x104 – 3x104) were stimulated with anti-CD3 (0.2ug/ml) and

(T cell depleted splenocytes, γ-irradiated, 2500 rads) APCs in 96 well plates

(200ul). Depending on the experiment, medium was supplemented with DMSO,

AB-MECA, and 0.5%-2% ethanol and toxin or no-toxin supernatants from

C.difficile culture. 72 hrs after stimulation cells were assayed for proliferation

using CellTiter-Glo Luminescent cell viability assay (Promega). The kit uses

luciferase as the detection enzyme. Luciferase enzyme requires ATP to generate

Page 72: Phospholipase D Signaling in T Cells

55

light. Since metabolically active cells produce more ATP, the light signal detected

is proportional to the amount of ATP present that in turn correlates with the

number of viable cells. Luminescence is measured using plate reader (Turner

Biosystems) and analyzed using VERITAS software.

ELISA

96 well plates were coated with purified mIL-2, mIL-4 or mIFN-γ (capture

antibody, 1:500-1:1000) in 0.1M NaHCO3 buffer overnight at 4 deg. Next day,

wells were washed twice in ELISA wash buffer (0.05% Tween in 1xPBS) and

blocked with blocking buffer (3% BSA in PBS) at room temperature for 1 hr. Then

the wells were washed again and incubated with sample supernatants (serially

diluted in 1:3) overnight at 4 deg. The following day wells were washed and

incubated with biotin-conjugated antibodies (mIL-2, mIL-4 or mIFN-γ) for 1-2hrs.

Further, streptavidin-HRP (1:1000) was added and incubated for 30 mins at room

temperature. 100ul of 3,3', 5,5’-Tetramethylbenzidine (TMB) liquid substrate

(Sigma, St. Louis, MO) was added to each well after washing the plates five

times with wash buffer. Upon color development, the reaction was stopped by

adding 50ul of stop buffer (4 water: 1 HCl: 1 H2SO4) to the wells. The plates were

then read at 450 nm using plate reader (Biotek, El x 800) and the data was

analyzed using KC junior software.

Page 73: Phospholipase D Signaling in T Cells

56

Phospholipase D Assay

Lymphocytes were harvested from P25 TCR transgenic mice and

incubated overnight with (1-10ug/ml) P25 peptide. The following day cells were

harvested, washed with RPMI three times to remove any residual antigen

peptide. CD4 T cells were sorted and kept at room temperature until use. Cells

were then suspended in 10% RPMI containing 10mM HEPES (1-5x106/500ul)

and pretreated with DMSO or AB-MECA (50uM) for 30 mins in 37 deg water

bath. Following pretreatment, CD4 T cells were stimulated with soluble biotin

conjugated anti-CD3 and avidin for different time points (15-30 mins). Cells were

centrifuged and resuspended in RIPA lysis buffer (10mM phosphate buffer pH

7.2,150mM NaCl, 1% Triton-X, 0.5% sodium deoxycholate, 0.1% SDS)

containing Dnase. Cell lysates were assayed using the commercially available

Amplex Red PLD assay kit (Invitrogen (Molecular Probes)). In this assay, PLD

activity is monitored indirectly by using 10-acetyl-3, 7-dihydrophenoxasine, called

the Amplex Red reagent. PLD cleaves the L-α-phosphatidylcholine (PC) substrate

to yield choline and phosphatidic acid. Choline is oxidized by choline oxidase

(provided in the kit) (choline oxidase from an Alcaligenes sp.) to betaine and

H2O2. Finally, H2O2 in the presence of horseradish peroxidase reacts with the

Amplex Red reagent to generate the highly fluorescent product resorufin

(excitation and emission maxima, ~563 and 587 nm, respectively), an H2O2

fluorogenic sonde. As per manufacturers manual, this kit is specific for

membrane PLD enzymes, with optimal activity at nearly neutral pH (pH 8).

Purified PLD from Streptomyces chromofuscus and 10 μM H2O2 served as

Page 74: Phospholipase D Signaling in T Cells

57

positive controls. The PLD assay kit can detect PLD levels as low as 10 mU/ml.

One unit of PLD is defined as the amount of enzyme that liberates 1.0 μmol of

choline from PC per minute at pH 8.0 at 30°C. Cell lysates were incubated with

the reagents provided in the kit for 1hr at 37 deg and the fluorescence intensity

was read using fluorometer (BMG Labtech).

cAMP Assay

CD4 T cells from P25 transgenic mice were sorted and stimulated as previously

described in PLD assay. In the case of cAMP assay, CD4 T cells were stimulated

for 5 mins. Further, to assess cytosolic cAMP concentrations, T cells were

washed three times in ice-cold PBS, lysed in 0.1 N HCl (107/ml) for 30 mins.

Supernatants were assayed using the acetylated version of cAMP-specific ELISA

kit (Correlate EIA Direct Cyclic AMP Enzyme Immunoassay kit; Assay Design).

Western Blots

In general, cell pellets were suspended in 1xSDS sample buffer (0.5-1x106

cells in 25 ul) and boiled for 5-7 mins with intermediate vortexing. The proteins

were then separated on 8% polyacrylamide gels. Proteins were then transferred

to nitrocellulose membrane (Millipore) overnight in transfer buffer. The following

day membrane was blocked with 5% milk in TBST for 1 hr. Blots were then

washed three times in 1X TBST and incubated with the primary antibody

overnight at 4 deg. Next day, blots were washed and incubated with secondary

antibody for 1 hr at room temperature. ECL substrate (GE healthcare) was

Page 75: Phospholipase D Signaling in T Cells

58

added to the blot for 1 min before developing.

To determine adenosine A3 receptor expression, CD4+CD25+ and

CD4+CD25- T cells were sorted using MACS column. Cells were stimulated with

plate bound anti-CD3 (2ug/ml) and anti-CD28 (2ug/ml) for 72 hrs. Both

unstimulated and stimulated cells were harvested, washed in 1xPBS and

pelleted. Cells pellets were processed as described above. Adenosine A3

receptor antibody (Santa Cruz Biotechnology, 1:500) was diluted in antibody

dilution buffer (0.2% BSA, 0.1% azide in 1X TBST). Anti-rabbit HRP (Cell

Signaling, 1:5000) was used as secondary antibody. The membrane was

stripped with buffer containing 2-ME and probed with β-actin (Sigma) antibody.

For phospho PKA analysis, splenocytes from P25 TCR transgenic mice

were stimulated as described in PLD assay. Sorted CD4 T cells were incubated

with AB-MECA or DMSO for 30 mins before stimulation. T cells were stimulated

with biotin anti-CD3 (2C11, 10ug/ml) for 1 min followed by addition of 2.5ug/ml of

avidin for 5 mins at 37 deg. Cells were then pelleted and processed as described

above. Phospho-PKA substrate antibody (Cell Signaling, 1:1000) was diluted in

antibody dilution buffer. Anti-rabbit HRP (1:5000) was used as secondary

antibody.

Generation of PLD YFP fusion constructs

The plasmid encoding human PLD1 and mouse PLD2 were kindly provided by

Dr. Michael Frohman. DNA encoding full length PLD1 and PLD2 were sub-

cloned into pMEYFPzeo-C1 vector by Dr. Nagendra Singh. Deletion mutants of

Page 76: Phospholipase D Signaling in T Cells

59

PLD1 and PLD2 in pMEYFPzeo-C1 were constructed using convenient

restriction sites or polymerase chain reaction-based strategies and were

sequenced at all junctions to ensure that the reading frame was maintained.

Plasmids were sequenced to confirm the intended mutation. The mutant

constructs of both PLD1 and PLD2 were confirmed to be expressed at similar

levels and at the expected sizes (Fig 5 and 6). Primers used for the generation of

the constructs are listed in Table 1.

Confocal microscopy

Jurkat T antigen cells were transfected with the PLD YFP fusion constructs by

electroporation. Live cells were isolated (16-18 hrs after transfection,) using

lymphocyte separation medium (Mediatech, Inc). Approximately 105 cells in 50μl

medium were spread on 0.1% lysine coated glass slides. The cells were left to

adhere to the glass slide for 5 mins and the excess medium was drained. Cells

were fixed with 4% Paraformaldehyde for 10-20 mins and washed with 1x PBS

for three times. For nuclear staining, DAPI (Molecular probes) or Propidium

iodide was added to cells for 5 mins and washed with 1x PBS three times. One

drop of prolong antifade (Molecular probes) was added on top of the cells and

mounted with cover slip. Slides were left overnight at room temperature in the

dark and analyzed using Zeiss® LSM-510 laser scanning microscope at the

LUMC and MCG core imaging facility. LSM image examiner software was used

for image acquisition and analysis.

Page 77: Phospholipase D Signaling in T Cells

60

kDa

A) B)

Fig 5. Western blot analysis of PLD1 deletion mutant constructs. Full length

and deletion mutant constructs of hPLD1b were transfected into Jurkat T antigen

cells by electroporation. The following day live cells were isolated and lysed in

1xSDS sample buffer. A) Proteins were separated on 8% polyacrylamide gel and

probed with anti-GFP antibody. B) Proteins were separated on 12%

polyacrylamide gel and probed with anti-GFP antibody. Protein markers sizes

(kDa) are indicated on the left side of the gel. Protein of expected size was

obtained for the different constructs. Vector only: 30 kDa, Full length PLD1: 150

kDa, loop deleted PLD1: 130 kDa, PH+PX+C: 75 kDa, PH + PX only: 62 kDa, N-

terminal deletion: 100 kDa, C-terminal only: 40 kDa.

Page 78: Phospholipase D Signaling in T Cells

61

kDa

Fig 6. Western blot analysis of PLD2 deletion mutant constructs. Full length

and deletion mutant constructs of mPLD2 were transfected into Jurkat T antigen

cells by electroporation. The following day live cells were isolated and lysed in 1x

SDS sample buffer. Proteins were separated on 8% polyacrylamide gel and

probed with anti-GFP antibody. Protein markers sizes (kDa) are indicated on the

left side of the gel. Protein of expected size was obtained for the different

constructs. Full length PLD2: 125 kDa, PH+PX+C: 75 kDa, C-terminal only: 40

kDa.

Page 79: Phospholipase D Signaling in T Cells

62

In contrast to the diffuse cytoplasm and nuclear localization of YFP-vector

alone, PLD1YFP and PLD2YFP have peri-nuclear and plasma membrane

localization respectively (Fig 7).

Page 80: Phospholipase D Signaling in T Cells

63

Vector PLD1 PLD2

Fig 7. Confocal microscopic analysis of YFP fusion constructs. YFP fusion

(green) construct of vector alone, full length PLD1 and PLD2 were transfected in

Jurkat T antigen cell line. The sub cellular localization of these constructs was

determined using confocal microscopy.

Page 81: Phospholipase D Signaling in T Cells

64

G TGG GCT CAC CAT GAG - 3'6 5'- TAT CAC ATG GAC GTA AGC GGC GT -3'

7 hPLD1b (PX+PH+C) 5'- CTC AAA GAT CAT CGA TTT GGG TCA GTT GTC CTT GGC TAT CTT GAT GAC - 3'

Table 1. PCR primers

T TA

Number Primer Name Sequence

1 hPLD1b ( PX+PH only) 5'- GGC CGC GAT ATC TCA CTA A - 3'2 5'- TA

T TT

G TGA GAT ATC GC- 3'

3 hPLD1b ( loop deletion) 5'- TAA ACT ACG GAT GGA CCC GGT AGT GAC CCG CTT CAC ACT - 3'

4 5'- AGT GTG AAG CGG GTC ACT ACC GGG TCC ATC CGT AGT TTA - 3'

5 5'- TA

8 5'- TTC AAA ATT AAG GTG GGG AAG - 3'

9 hPLD1b (C terminal only) 5'- GGA TAT TCA TGC GGC CGC TCA TTA AGT CCA AAC CTC CAT GGG - 3'

10 5'- TTT AGG CTC GAG CTG TTG TCC TTG GCT ATC TTG AT - 3'

11 mPLD2 (PX+PH+C) 5'- CTA CAG CTA CAT CAG CAT GAC AGC GTG ATT CTT GGG GCA AAT ACC TGG - 3'

12 5'- TCC CTG ATT CTC AAA TGC AGC AGC -3'13 5'- TTC ACC TTC TGT GGC CGA GAC C - 3'14 5'- CCA GGT ATT TGC CCC AAG AAT CAC

GCT GTC ATG CTG ATG TAG CTG TAG - 3'

15 mPLD2 (C terminal only) 5'- AAG GAC TCC TTC CTG CTG TAC ATG - 3'16 5'- TTC AGT CTC GAG CTG TGA TTC TTG

GGG CAA ATA CC- 3'

PLD2 (- / -) mice genotyping primers17 P274 5'- GCC ATC TCA TTT GTT CAG CTT T - 3'18 P275 5'- GCT GTC ATG CTG ATG TAG CTG T - 3'

19 CD4 CRE FWD 5'- CGA TGC AAC GAG TGA TGA GG - 3'20 CD4 CRE REV 5'- GCA TTG CTG TCA CTT GGT CGT - 3'

21 FLP FWD 5'- CAC TGA TAT TGT AAG TAG TTT GC- 3'22 FLP REV 5'- CTA GTG CGA AGT AGT GAT CAG G - 3'

Page 82: Phospholipase D Signaling in T Cells

CHAPTER THREE

EXPERIMENTAL RESULTS

REGULATION OF PHOSPHOLIPASE D SIGNALING BY EXOGENOUS

FACTORS

Antigen stimulation of lymphocytes results in increased PLD activity.

Primary alcohol is a well-known inhibitor of PLD signaling. Previous studies have

documented the inhibition of T cell activation in the presence of primary alcohols

in vitro. Singh et al (Singh et al., 2006) analyzed the effect of inhibiting PLD

signaling in different subsets of CD4 T cells using 0.3% 1-butanol. Inhibition of

PLD signaling selectively blocked the proliferation of CD4+CD25- (effector) T

cells and led to the enrichment of Foxp3+ regulatory T cells. This study

highlighted the differential requirement for PLD signaling among effector versus

regulatory T cell subsets.

We then sought to determine the role of PLD signaling in dictating the

immune response of the host. We hypothesized that in vivo suppression of PLD

signaling may promote immune suppression through dominance of Treg cells

under physiological/pathological conditions. Conversely, enhancement of PLD

signaling under certain conditions might promote dominance of effector T cells

65

Page 83: Phospholipase D Signaling in T Cells

66

and thus promote immune reactivity. For my studies, I focused on identifying

various exogenous and endogenous factors that alter the immune status of the

host through modulating PLD signaling.

Ethanol and CD4 T cell population dynamics

Based on the effect of 1-butanol, we predicted that another 1-alcohol,

ethanol would also inhibit PLD signaling. Ethanol is widely consumed for

recreational purposes and is considered as an immunosuppressive agent (Cook,

1998; Nelson and Kolls, 2002; Zhang and Meadows, 2005). Excessive alcohol

intake has been associated with increased incidence of infection, cancer and

autoimmunity (Song et al., 2002). Previous studies in mice indicate that alcohol

administration for short duration causes reduced cell numbers of immune cells

like T cell, B cell and NK cells and concomitant reduction in the cellularity of

spleen and thymus (Chadha et al., 1991). However, the effect of alcohol on CD4

T cells in specific and the mode of action were not delineated.

To determine if ethanol can enrich for Treg cells, we treated murine CD4 T

cells with varying concentrations of ethanol in the presence of anti-CD3

stimulation in vitro for 4 days. After 72 hrs of stimulation, cells were washed and

cultured for 3 days in the presence of IL-2. Cells were harvested and stained for

regulatory T cell markers, Foxp3 and CTLA4. When T cells were stimulated by

anti-CD3 antibody in the presence of low concentrations of alcohol (0.5%), no

enrichment of Foxp3+ Treg cells was observed and the frequency of Foxp3+

Page 84: Phospholipase D Signaling in T Cells

67

0.5% Ethanol Medium

1% Ethanol 2% Ethanol

0.86

4.91 16.56

Foxp3

CTL

A4

0.78

Figure 8. Effect of ethanol on CD4 T cells in vitro. Total CD4+ T cells were

cultured with anti-CD3 plus antigen presenting cells in medium containing

increasing concentrations (0.5% - 2%) ethanol for 3 days. After 3 days, cells

were placed in medium containing IL-2 (20ng/ml) for 4 days. Seven days after

the start of culture, cells were harvested and analyzed by flow cytometry for

Foxp3 and CTLA4 expression. Representative data from 3 independent

experiments is shown.

Page 85: Phospholipase D Signaling in T Cells

68

cells was similar to medium-treated samples. When higher doses of ethanol

were used (1.0%, 2.0%), however, we observed a clear dose dependent

increase in Treg frequency (Fig 8) among CD4 T cells and observed 4.9 and

16.5% of CD4 T cells becoming Foxp3+, respectively. Similar to 1-butanol, CD4

T cells expanded in the presence of 2% ethanol contain higher percentages of

Foxp3+ cells.

The experiment shown above used stimulation of T cells by anti-CD3

antibodies. To determine if ethanol can enrich for antigen specific Treg cells, we

next tested the effect of ethanol on antigen-induced T cell proliferation using CD4

T cells from P25 TCR transgenic mice. The P25 mouse carries a transgene

encoding the TCR specific for peptide-25 of Mycobaterium tuberculosis (Tamura

et al., 2004). T cells were stimulated with the antigen peptide in the presence of

irradiated antigen presenting cells for 3 days followed by culture in the presence

of IL-2 for 4 days. Similar to anti-CD3 stimulated cultures, the frequency of

Foxp3+ cells in 2% ethanol treated cultures were 22% as against 0.6% in both

medium and 0.5% ethanol treated samples (Fig 9).

The increase in percentage of Foxp3+ cells could be due to increased

proliferation of Foxp3+ cells in 2% ethanol treated cultures or due to decreased

proliferation of Foxp3- T cells. To test for these possibilities, we labeled CD4 T

cells with CFSE and measured the dilution of CFSE to monitor proliferation.

Compared to 0.5% ethanol, very few Foxp3- T cells proliferated in 2% ethanol

treated cultures as seen from the CFSE dilution plots (Fig 10).

Page 86: Phospholipase D Signaling in T Cells

69

Medium 0.5 % ethanol

2% ethanol

Foxp

3

CD25

Figure 9. Effect of ethanol on CD4 T cells in the presence of antigenic

stimulation in vitro. Total CD4+ T cells from P25 TCR transgenic mice were

cultured with P25 peptide (0.1ug/ml) plus antigen presenting cells in medium

containing different percentages of ethanol for 3 days. After 3 days, cells were

placed in medium containing IL-2 (20ng/ml) for 4 days. Seven days after the start

of culture, cells were harvested and analyzed by flow cytometry for Foxp3 and

CD25 expression. Representative data from 2 independent experiments is

shown.

Page 87: Phospholipase D Signaling in T Cells

70

Tota

l cel

l num

bers

Figure 10. Proliferation kinetics of CFSE labeled CD4 T cells in the

presence of ethanol. Total CD4+ T cells were labeled with CFSE (2.5ug/ml) and

cultured with anti-CD3 (0.2ug/ml) and APCs in medium containing 0.5% ethanol

(solid line) or 2% ethanol (dotted line) for 3 days. Cells were placed in medium

containing IL-2 (20ng/ml) for next 4 days. On day 7 after stimulation, cells were

harvested and analyzed by flow cytometry for Foxp3 and CFSE expression. A)

Line graph depicting number of Foxp3- T cells in different generations B) Line

graph depicting number of Foxp3+ T cells in different generations.

0

0

0

0

0

5000

0

0 1 2 3 4 5 6 7

100

200

300

400

6000.5 % ethanol2 % ethanol

Tota

l cel

l num

bers

0

0.05

0.1

0.15

2

0.25

3

0.35

0 1 2 3 4 5 6 7 8

0.

0.0.5 % ethanol2 % ethanol

(106) A)

B)

Generations

Page 88: Phospholipase D Signaling in T Cells

71

As a result, the total Foxp3- T cell numbers recovered from 0.5% ethanol treated

cultures were 8 times more than that recovered from 2% ethanol cultures. In

contrast, Foxp3+ cells from samples treated with 2.0% ethanol showed no

difference in CFSE levels between 0.5% and 2% ethanol treated samples.

Together, the data demonstrate that ethanol causes increased frequency of

Foxp3+ cells by decreasing the proliferation of Foxp3- T cells.

Epidemiological studies showed that alcohol abusers are predisposed to

more frequent and more severe infections (Cook, 1998). In particular, increased

incidence of infections and enhanced severity of disease such as pneumonia and

tuberculosis were associated with chronic consumption of alcohol (Cortese et al.,

1992; Esposito, 1984; Fernandez-Sola et al., 1995; Friedman et al., 1987; Moss

et al., 1996; Ruiz et al., 1999; Winterbauer et al., 1969). The incidence of chronic

Hepatitis C Virus (HCV) infection is also increased in alcohol abusers (Degos,

1999; Harris et al., 2001; Khan and Yatsuhashi, 2000; Lima et al., 2000; Schiff,

1999). Also, studies on rodents show that animals are more vulnerable to

infection after chronic or acute exposure to alcohol. These observations can be

interpreted based on our in vitro studies in the following manner: Consistent

intake of alcohol might lead to selective elimination of effector T cells and

consequent enrichment of regulatory T cells. This in turn might increase the

susceptibility of the host to various infections. Also, antigenic exposure in the

presence of alcohol might inhibit the proliferation of antigen specific effector T

cells thus compromising the overall health of the host.

Page 89: Phospholipase D Signaling in T Cells

72

We tested for these possibilities in vivo in mice using two different

experimental set-ups. First, the effect of ethanol on CD4 T cell subsets in mice in

the absence of antigen was determined. Wild type C57BL/6 mice were injected

with either PBS or 20% ethanol intraperitoneally (i.p) for 7 days. Mice were then

sacrificed and their spleen and mesenteric lymph nodes were harvested and

stained for regulatory T cell markers. As expected, spleen of mice treated with

ethanol contained 23.56% of Foxp3+ cells compared to 17.5% in PBS treated

mice (Fig 11a). This increase in frequency of Foxp3+ cells was due to significant

decrease in the total Foxp3- CD4 T cell numbers; no significant change in

Foxp3+ T cell numbers was observed (Fig 11b). These data confirmed our

prediction that ethanol selectively eliminates Foxp3- T cells even in the absence

of antigenic proliferation.

Secondly, we determined if ethanol inhibits antigen-induced effector T cell

proliferation while allowing Treg cells to proliferate in vivo, as we observed in

vitro. To test the effect of ethanol in vivo, we used the protocol that has been

previously described by others. Briefly, mice were injected with 2.9g/kg body

weight ethanol in 20% vol/vol saline intraperitoneally (i.p) (Meadows et al., 1989;

Obradovic and Meadows, 2002; Song et al., 2002). This dose of ethanol injection

leads to blood ethanol level of 300mg/dl in 30 minutes. We adoptively transferred

CFSE labeled CD4 T cells from P25 TCR transgenic mice into C57.BL/6 CD45.1

recipient mice intravenously. These recipient mice express CD45.1 isoform,

which is distinguishable from the CD45.2 isoform expressed by the donor mice.

Page 90: Phospholipase D Signaling in T Cells

73

EthanolPBS A)

CTL

A4

Foxp3

Figure 11. Flow cytometry analysis and quantification of cell numbers of

CD4 T cells in mice injected with ethanol. Wild type mice were injected

intraperitoneally with either PBS or 20% ethanol for 7 days. On day 7, mice were

sacrificed and their spleen cells were stained for Foxp3, CD4 and CTLA4. A)

Representative FACS plots of spleen CD4 T cells from PBS and ethanol treated

mice. B) Bar graphs of total Foxp3- T cell numbers in spleen of PBS (white bars)

and ethanol (black bars) treated mice. C) Bar graphs total Foxp3+ T cell numbers

in spleen of PBS (white bars) and ethanol (black bars) treated mice.

0

5

10

15

20

25

30

1

PBSEtoH

B) C)

0

1

2

3

4

5

6

1

(103) (103)

Page 91: Phospholipase D Signaling in T Cells

74

CFSE

Tota

l cel

l num

bers

To

tal c

ell n

umbe

rs

PBSNo antigen Ethanol Fo

xp3

(103)

B)

C)

A)

0

1

2

3

4

5

6

7

0 1 2 3 4 5 6 7

GENERATIONS

CONTROLETHANOL

0

50

100

150

200

250

300

0 1 2 3 4 5 6 7

GENERATIONS

CTRLETHANOL

Page 92: Phospholipase D Signaling in T Cells

75

Figure 12. Effect of ethanol on the proliferation kinetics of Foxp3 positive

and Foxp3 negative CD4 T cells in vivo in the presence of antigen. CFSE

labeled CD4 T cells from P25 TCR transgenic (Ly5.2) mice were intravenously

transferred to Ly5.1 recipient mice. 24 hrs later, the recipients were challenged

with 2.5ug of P25 peptide along with either PBS (control) or 20% ethanol (i.p) for

a period of 7 days. On day 8, mice were sacrificed and all their peripheral lymph

nodes (axial, cervical, inguinal, popliteal) were isolated and pooled for analysis

by flow cytometry. A) Representative FACS plots showing CFSE dilution in

lymphocytes of control and ethanol treated mice. B) Line graphs depicting the

number of foxp3- T cells in each generation of control (dotted lines) and ethanol

treated (solid lines) mice. C) Line graphs depicting the number of foxp3+ T cells

in each generation of control (dotted lines) and ethanol treated (solid lines) mice.

Data is representative of 2 independent experiments.

Page 93: Phospholipase D Signaling in T Cells

76

One day after injection of T cells the recipient mice were injected with the antigen

peptide. Immediately after injection of peptide, mice received ethanol injection as

described above. Mice were administered ethanol daily for 7 days. On day 8,

mice were euthanized and spleen, mesentric and peripheral lymph nodes

(inguinal, brachial, axial, popliteal, cervical) were harvested. The state of

proliferation of adoptively transferred T cells was determined by analyzing the

CFSE level of CD45.2+ T cells (Fig. 12a). T cells of mice that received no antigen

showed no proliferation after 7 days while T cells from mice that received antigen

peptide in PBS showed significant level of proliferation in both Foxp3+ and

Foxp3- populations. The frequency of Foxp3+ T cells in PBS treated mice was

3.5%. Mice injected with ethanol showed a marked reduction in proliferation of

Foxp3- T cells and the frequency of Foxp3+ T cells was 9.75%. The majority of

Foxp3- T cells in control mice were in between generation 3 to generation 6 (i.e.

most of these cells underwent 3 cell divisions)(Fig 12b). However, there was no

clear peak in the cell division of Foxp3- T cells of ethanol treated mice suggesting

that majority of cells did not proliferate effectively. In stark contrast, the level of

cell division of Foxp3+ cells was comparable between ethanol treated and PBS

treated mice (Fig 12c). As a consequence, the total cell numbers that were

obtained from ethanol treated and untreated mice were comparable for Foxp3+ T

cells but the cell numbers was significantly lower in ethanol treated animals for

Foxp3- cells. Together, the data demonstrate that ethanol does block antigen

induced Foxp3- effector T cell proliferation while allowing Treg cells to expand in

vivo.

Page 94: Phospholipase D Signaling in T Cells

77

PBS ETHANOL ETHANOL

CFSE

FOXP

3

A)

B)

0

5 0

1 0 0

1 5 0

2 0 0

2 5 0

T O T A L C D 4

CEL

L C

OU

NT

P B SE T H A N O L # 1E T H A N O L # 2

C) D)

(103)

(103

0

50

100

150

200

250

1

CEL

L C

OU

NT

0

5

10

15

20

25

30

FOXP3 POS

) (103

PBS)

ETHANOL #1ETHANOL #2

Page 95: Phospholipase D Signaling in T Cells

78

Figure 13. Effect of ethanol on homeostatic proliferation of CD4 T cells.

CD4 T cells from wild type mice were intravenously transferred into RAG1 KO

mice along with intraperitoneal injections of either PBS or 20% ethanol for 5

days. On day 6, mice were sacrificed and their spleen cells were stained for

Foxp3 and CD4 T cells. A) Representative FACS plots of PBS and two ethanol

treated mice showing CFSE dilution in spleen CD4 T cells. B) Bar graphs of total

cell numbers of CD4 T cells in the spleen. C) Bar graphs depicting the number of

foxp3- T cells in each of control (white bars) and ethanol treated mice (filled

bars). D) Bar graphs depicting the number of foxp3+ T cells in each of control

(white bars) and ethanol treated mice (filled bars).

Page 96: Phospholipase D Signaling in T Cells

79

Zhang et al reported that chronic alcohol consumption (2 weeks-6 months)

results in T cell lymphopenia, which in turn induces homeostatic proliferation

(Zhang and Meadows, 2005). Since we also observed similar reduction in cell

numbers in mice injected with ethanol for 1 week (Fig 11b), we investigated

whether ethanol affects homeostatic proliferation. We tested the primary effect of

ethanol on the proliferation of CD4 T cells under well-established homeostatic

condition like RAG1 KO mice. We adoptively transferred CFSE labeled CD4 T

cells from wild type mice into RAG-1 knock out mice intravenously followed by

ethanol injection for 5 days. Under lymphopenic conditions, T cells are stimulated

by self-peptide and self-MHC (Zhang and Meadows, 2005). We monitored

proliferation of T cells based on dilution of CFSE. As seen in (Fig 13b), the total

CD4 T cell counts in ethanol injected mice were 5 times lower than PBS injected

mice. Unlike antigen-induced proliferation of CD4 T cells, we did not observe

selective decrease in Foxp3- T cells alone. Both Foxp3- and Foxp3+ T cells

counts were reduced by injection of ethanol (Fig 13 c). Thus, the data show that

in acute phase of homeostatic expansion, ethanol inhibits proliferation of both

Foxp3- and Foxp3+ CD4 T cells. Further experiments could be aimed at looking

for specific markers of homeostatic proliferation such as expression of CD44 and

lack of CD69.

Page 97: Phospholipase D Signaling in T Cells

80

Effect of alcohol ingestion on the balance between Foxp3+ and Foxp3- CD4 T

cells

The data indicate that continuous and high dose exposure of T cells to alcohol

may induce a relative increase in the frequency of Foxp3+ T cells. Despite the

minute presence of endogenous ethanol, alcohol consumption by humans is the

major source of ethanol in the human body (Antoshechkin, 2001). To study the

effects of in vivo ethanol exposure on CD4 T cells, we fed mice with liquid diet

containing alcohol. DO11.10 TCR (T cell receptor) transgenic mice (receptor

specific for ovalbumin (OVA) plus I-Ad) were fed an alcohol-containing nutrient-

supplemented liquid diet (Bio-Serv, NJ) (ethanol concentration was gradually

increased from 1% to 6.3% w/v every 2 days) or calorie matched control diet for

10 days (sex-matched 12-week-old male mice, 6 mice per group). Mice were

euthanized on day 10, and mucosal CD4 T cells were analyzed for expression of

Foxp3 by flow cytometry. CD4 T cells from the mesenteric lymph nodes of

alcohol-fed mice showed a moderate but clear increase in Foxp3+ CTLA4+ T

cells compared to the control group (Fig 14a). Average frequency of Foxp3+

CD4 T cells increased significantly compared to control diet fed animals (Fig

14b). These results show that continuous presence of ethanol in the diet causes

relative increase of Foxp3+ T cell in mucosal associated lymphoid organs and

suggest that continuous alcohol consumption by humans might lead to selective

enrichment of Treg cells and immune suppression leading to heightened

susceptibility to opportunistic infections.

Page 98: Phospholipase D Signaling in T Cells

81

A)

B)

Figure 14. Effect of alcohol containing diet on mice CD4 T cells. Mice were

fed on liquid diet containing water or varying concentration of ethanol for 10 days.

Mice were sacrificed on day 10 and their mesenteric lymph node was stained for

Foxp3 and CTLA4. A) Representative FACS plots of mesenteric lymph node of

two control and two alcohol fed mice. B) Data from 2 different experiments were

pooled and the percentage of Foxp3 positive cells in control versus alcohol fed

mice is shown as a graph.

Page 99: Phospholipase D Signaling in T Cells

82

Clostridium difficile toxin: Hijacking the immune system through PLD

Clostridium difficile (C.diff) is a spore forming, anaerobic, gram-positive

bacterium (Genth et al., 2008). Its role in the pathogenesis of

pseudomembranous colitis is well established. Recently, it has emerged as the

leading cause of nosocomial diarrhea in patients (Genth et al., 2008). Bacterial

pathogens interact with the mammalian cells through the production of protein

toxins (Fiorentini et al., 2003). These toxins serve as virulence factors and aid in

establishing their niche in the host. The major virulence factors produced by

C.difficile are Toxin A and Toxin B. Following cell entry, these toxins mono-

glucosylate and thereby inactivate lower molecular mass GTP binding proteins of

the Rho sub family (Genth et al., 2008). As mentioned previously, PLD is one of

the downstream effector molecules of Rho. As a consequence, C.difficile toxins

are considered inhibitors of PLD signaling.

Thus, we hypothesized that C.difficile might evade the host T cell effector

response by inhibiting PLD signaling. If these toxins inhibit PLD signaling in T

cells as well, then we expect to see inhibition of effector T cell proliferation in the

presence of these toxins. To test the effect of C.difficile toxins on T cell

proliferation, PKH-26 (analog of CFSE) labeled CD4 T cells were stimulated with

anti-CD3 in the presence of varying amounts of supernatants from toxigenic and

non-toxigenic strains of C.difficile (C.difficile supernatants were kindly provided

by Michelle Merrigan). As seen in (Fig 15a,b), following TCR stimulation 50% of

both Foxp3+ and Foxp3- T cells had undergone one cell division.

Page 100: Phospholipase D Signaling in T Cells

83

A)

B)

0 10 2 10 3 104 105

PE-A 0 20 40 60 80

100 % of Max

US / S

8.37 / 50.2

0 102 103 104 105

PE-A

0

20

40

60

80

100

% o

f Max

6.79 / 55

0 102 103 104 105

PE-A

0

20

40

60

80

100

NT / TO.15%

50 / 31.8

% o

f Max

0 102 103 104 105

PE-A

0

20

40

60

80

100

% o

f Max

56.6 / 47.7

Foxp3 negative

PKH

Foxp3 positive

PKH

Page 101: Phospholipase D Signaling in T Cells

84

C)

Toxin B concentration (

μg/ml)

Fig 15. Analysis of proliferation kinetics of Foxp3+ and Foxp3- T cells in the

presence of C.difficile toxin supernatants. CD4 T cells were labeled with 5μM

PKH-26 and stimulated with anti-CD3 (0.2ug/ml) and APCs. Varying amounts of

supernatants from toxigenic (Tcd) or non-toxigenic (non-Tcd) (0.05%-0.25%)

strains of C.difficile were added to the culture medium. 72 hrs after stimulation,

cell were harvested and stained with anti-Foxp3 antibody. A) Histograms

comparing the proliferation of Foxp3- T cells (top row) and Foxp3+ T cells

(bottom row) between medium (left side) and 0.15% of C.difficile supernatants

(right side) treated samples. Left panels: blue line (stimulated sample) and red

line (unstimulated sample). Right side: blue line (supernatant from toxigenic C.diff

strain) and red line (supernatant from non-toxigenic C.difficile strain) B) Graph

showing percentage of growth inhibition between samples containing increasing

amounts of C.difficile culture supernatants. C) Bar graph showing percentage of

growth inhibition in Foxp3+ and Foxp3- T cells in the presence of purified TcdB

(μg/ml).

Page 102: Phospholipase D Signaling in T Cells

85

In comparison, the Foxp3- T cell proliferation was decreased substantially

when the concentration of toxin containing supernatants was increased. Also, we

observed some inhibition in the proliferation of Foxp3+ T cells at higher

concentrations of the toxin supernatants (Fig 15b). Since we used supernatants

from C.difficile cultures as the source of toxin, it is possible that components

other than toxins might exhibit non-specific effects at high concentrations.

Further, to confirm that toxins specifically mediate the effect on T cell

proliferation, we performed this experiment with purified Toxin B. Purified toxin B

(1ug/ml) inhibited the proliferation of Foxp3- T cells by 60% while inhibition of

proliferation of Foxp3+ T cells was 10% (Fig 15c). These results suggest that

C.difficile toxin B (TcdB) through inhibiting PLD signaling blocks effector T cell

proliferation significantly.

Why does inhibiting PLD signaling selectively affect the proliferation of

Foxp3- (effector) T cells? In our studies with 1-butanol, we showed that PLD

signal inhibition abrogates activation induced expression of CD25 on Foxp3-

(effector) T cells. In contrast, Foxp3+ Treg cells express CD25 constitutively.

CD25 is IL-2 receptor alpha chain, up regulated on effector T cells following their

activation. IL-2 is an important survival/proliferation cytokine for activated T cells.

To test the effect of C.difficile toxin on the expression of CD25, CD4 T cells were

stimulated with anti-CD3 both in the presence and absence of toxin containing

supernatants. CD25 expression on these samples was determined 24 hrs after

stimulation. Stimulation of T cells resulted in significant up regulation of CD25

and CD69 in medium or non-toxigenic supernatant containing samples (Fig 16).

Page 103: Phospholipase D Signaling in T Cells

86

Figure 16. Flow cytometric analysis of activation markers expressed by

CD4 T cells following antigenic stimulation in the presence of C.difficile

supernatants. CD4 T cells were stimulated with anti-CD3 and APCs. Equal

amounts (0.15%) of culture supernatants from either toxigenic or non-toxigenic

C.difficile strains were added. 24 hrs after stimulation, cells were harvested,

stained for activation markers, CD25 and CD69 and analyzed by flow cytometry.

Page 104: Phospholipase D Signaling in T Cells

87

In contrast, the expression of CD25 was not increased in samples treated with

toxin containing supernatants. This result is in agreement with our previous

finding that inhibition of PLD signal blocks CD25 up regulation. Future

experiments can be performed to determine if C.difficile toxin B specifically

inhibits CD25 expression on Foxp3- T cells.

Based on our data on alcohol diet fed mice, we predicted that mice

exposed to C. difficile toxin/spores would have increased frequency of Treg cells.

To test this possibility, we analyzed the frequency of Foxp3+ T cells in mice

infected with C. difficile spores. Treatment with antibiotics disrupts the normal

commensal microbiota in the host intestine and is often associated with

increased incidence of Clostridium Difficile- associated diarrhoea (CDAD) (Giel et

al., 2010; Vollaard and Clasener, 1994). Based on these studies, mice were

treated with various antibiotics prior to introduction of C. difficile spores to induce

CDAD. Mice were treated with antibiotics and infected with C. difficile spores by

Pehga Mohseni. Compared to wild type mice, mesenteric lymph nodes of mice

colonized with C. difficile had higher percentages of Foxp3+ T cells (Fig 17 a). As

expected, this increase in Foxp3+ percentage was due to concomitant decrease

in Foxp3- T cell numbers (Fig 17b). However, a potential caveat of this

experiment is the absence of mice injected with spores derived from non-

toxigenic strain of C. difficile as a negative control. It is possible that the stringent

treatment with antibiotics might have contributed to the increased frequency of

Treg cells. Future experiments could be aimed at comparing mice colonized with

spores from both toxigenic and non-toxigenic strains.

Page 105: Phospholipase D Signaling in T Cells

88

Wild type C. Difficile spores # 1

Fig 17. Flow cytometric analysis of Foxp3+ T cell percentage in mice

infected with C. difficile spores. C57BL/6 mice were treated with broad-

spectrum antibiotics for 72 hrs. For the next 48 hrs, these mice were fed normal

drinking water and injected intraperitonealy with clindamycin. 24 hrs later, these

mice were given (50,000 CFU) C. difficile spores by oral gavage. 72 hrs later

these mice were sacrificed and their mesenteric lymph nodes were harvested

and stained using anti-CD4 and Foxp3 antibodies. A) FACS plots from wild type

0 1 2 1 3 1 4 1 5<APC

01 2

1 3

1 4

1 5

<P

0. 4.

12820 1 2 1 3 1 4 1 5

<APC-

01 2

1 3

1 4

1 5

<P

17. 31.

9.141.0 1 2 1 3 1 4 1 5

<APC

01 2

1 3

1 4

1 5

<P

19 26

350

CTL

A4

Foxp3

A)

B)

0

10

20

30

40

50

60

70

1

Tota

l cel

l num

bers

C) (10

WTC.diff # 1C.diff # 2

0

5

10

15

1

WTC.diff # 1C.diff # 2

3) (103)

C. Difficile spores # 2

Page 106: Phospholipase D Signaling in T Cells

89

mice (WT) and two different mice infected with C. difficile spores showing the

percentage of CD4 T cells expressing Foxp3. B) Bar graph of Foxp3- T cell

numbers in wild type and C. difficile spores-infected mice. C) Bar graph of

Foxp3+ T cell numbers in wild type and C. difficile spores-infected mice.

Page 107: Phospholipase D Signaling in T Cells

90

Nevertheless, based on evidence from other models of PLD signal inhibition, it is

tempting to speculate that exposure to C. difficile toxins might lead to preferential

enrichment of Treg cells in vivo.

REGULATION OF PHOSPHOLIPASE D SIGNALING BY NATURALLY

PRESENT ENDOGENOUS FACTORS

Adenosine (Ado) is an endogenous molecule that mediates a wide

spectrum of biologic effects by engaging 4 kinds of cell surface receptors: A1,

A2a, A2b and A3. Ado is found in very high concentrations under hypoxic

conditions like tissues surrounding inflammatory sites and tumors. Notably, these

Ado rich environments have an immune suppressive phenotype. For example,

increased frequency of Treg cells has been reported in most human solid tumors

(Beyer and Schultze, 2006). Several reports highlight the immune modulatory

effects of Ado acting through A2a receptor on T cells. However, there is little

experimental evidence for a role of A3 receptors mediating effects of Ado in T

cells. Interestingly, both A2a and A3 receptors have been previously reported to

modulate cell functions through regulating PLD activity. We hypothesized that

one mechanism by which adenosine can decrease immune responsiveness is

through inhibiting PLD signaling.

TCR stimulation has been shown to modulate the expression of adenosine

receptors in mice (Lappas et al., 2005) and humans (Gessi et al., 2004b). To

understand the role of adenosine A3 receptor signaling on T cell activation, we

investigated the kinetics of A3 receptor expression on CD4+CD25+ regulatory T

Page 108: Phospholipase D Signaling in T Cells

91

cells (Treg cells) and CD4+CD25- T cells (non-Treg cells) under resting and

activated conditions. We purified CD4+CD25- and CD4+CD25+ T cells from

C57.BL/6 mouse splenocytes and stimulated them with anti-CD3 and anti-CD28

antibodies. Total cell lysates from each group of T cells were separated by SDS-

PAGE and analyzed by Western blot for expression of the A3 receptor. Under

resting conditions, freshly isolated CD4+CD25- T cells did not express detectable

level of adenosine A3 receptor while CD4+CD25+ T cells expressed barely

detectable levels of the receptor. However, following stimulation both

CD4+CD25+ and CD4+CD25- T cells express comparable levels of adenosine A3

receptor (Fig 18).

We then examined the functional relevance of A3 receptor expression on

CD4 T cells using adenosine A3 receptor selective agonist, AB-MECA. We

stimulated splenic CD4 T cells with anti-CD3 antibody plus irradiated T-depleted

splenocytes (APCs) in the presence or absence of AB-MECA. We first examined

the effect of AB-MECA on the expression of early activation markers on T cells

namely CD69 and CD25 (IL-2 receptor α chain) following 24 hrs of stimulation.

Under these conditions, 65% of stimulated T cells expressed both CD25 and

CD69 whereas CD69 and CD25 alone were expressed by 5% and 13%

respectively (Fig 19a). In contrast, in cultures treated with AB-MECA, the

frequency of CD69+CD25+ T cells decreased to 40% and CD25 alone increased

to 21% and we observed little effect on CD25+CD69- T cells as the frequency

remained at 5%. Since naturally occurring regulatory T cells (nTreg cells)

constitutively express CD25 and non-Treg (CD4+CD425-) T cells express CD25

Page 109: Phospholipase D Signaling in T Cells

92

- + - + CD25 CD 25

Freshly isolated Invitro expanded

75 kD

50 kD

66 kD

Fig 18. Western blot analysis of adenosine A3 receptor expression on CD4

T cells. Treg cells (CD4+CD25+) and non-Treg (CD4+CD25-) T cells were

sorted from wild type mice by flow cytometry. Lysates of unstimulated versus

stimulated T cells were separated on 8% polyacrylamide gel, transferred onto

nitrocellulose membrane and blotted with antibody against A3 receptor

(66kDa)(Top blot). Molecular weights are indicated on the left side of the blot.

The membrane was stripped and probed with antibody against β-actin as loading

control (bottom row).

Page 110: Phospholipase D Signaling in T Cells

93

Unstimulated DMSO AB-MECA

Fig 19. Analysis of activation markers on T cells following antigenic

stimulation in the presence of A3 receptor signaling. CD4 T cells or

CD4+CD25- T cells were stimulated with anti-CD3 (0.2ug/ml) and T cell depleted

splenocytes (APCs) for 24 hrs in the presence of DMSO or AB-MECA. T cells

were stained with anti-CD25, anti-CD69 and analyzed by flow cytometry. A) Dot

plots of CD4 T cells expressing CD25 and CD69. B) Histograms of CD4+ CD25-

T cells expressing CD25 after TCR stimulation. Shaded peaks represent

unstimulated controls and solid black lines denote activation induced up -

regulation of CD25.

CD

25

CD 69

0 102 103 104 105

<PE- 0

20

40

60

80

100

% of Max

53 46

AB-MECA

0 102 103 104 105

<PE- 0

20

40

60

80

100

% of Max

30 70

DMSO

CD 25

A)

B)

Page 111: Phospholipase D Signaling in T Cells

94

only upon activation, we sorted Treg cells and non-Treg cells and determined the

expression of CD25. 70% of non-Treg cells in control cultures were CD25

positive in comparison to 45% in AB-MECA treated cultures (Fig 19 b). These

data collectively indicate that AB-MECA inhibits the expression of CD25 on non-

Treg cells and has no effect on Treg cells as they constitutively express CD25.

Next, we examined the effect of AB-MECA on CD4 T cell proliferation at

72 hrs post stimulation (Fig 20a). In the presence of AB-MECA, there was

approximately 3-fold reduction in the proliferation of CD4 T cells. A similar

inhibitory effect of AB-MECA was observed even in the presence of exogenous

IL-2 consistent with the reduced expression of CD25 (IL-2 receptor α chain) (Fig

20b). Also, we measured cytokine secretion by CD4 T cells both in the presence

and absence of AB-MECA. For this experiment, purified CD4 T cells were

stimulated with plate-bound anti-CD3 antibody plus soluble anti-CD28 antibody.

After 24~72 hours of stimulation, culture supernatant was collected and analyzed

by ELISA (Fig 20c). Stimulation of CD4 T cells under these conditions induced

substantial amount of IL-2, IFN-γ, and IL-4 and the level of cytokines in the

supernatant continued to increase for 72 hours. When AB-MECA was present,

the initial 24 hours of cytokine production was not significantly affected.

However, the level of cytokine production in AB-MECA treated samples showed

profound reduction compared to the control samples at later time points (48 and

72 hrs). Among the cytokines examined, IFNγ and IL-4 production were severely

reduced, potentially due to inhibition of proliferation of activated T cells.

Page 112: Phospholipase D Signaling in T Cells

95

(10

3)

0

50

100

150

200

250

CD3 + IL2

DMSOA3

p= 0.04

DMSO AB-MECA

A)

(103)

0

10

20

30

40

50

60

CD3

p = 0.02265

O.D

B)

IL- 2

0

0.2

0.4

0.6

0.8

1.0

1.2

1.4

1.6

0 24 48 72 HRS

IFN- γ

0

0.1

0.2

0.3

0.4

0.5

0.6

0 24 48 72 HRS

DMSO AB-MECA

O.D

Page 113: Phospholipase D Signaling in T Cells

96

DMSO

AB-MECA IL- 4

0

0.1

0.2

0.3

0.4

0.5

0.6

0 24 48 72 HRS

Fig 20. Comparison of proliferation and cytokine secretion profile of DMSO

and AB-MECA treated CD4 T cells. A) CD4 T cells were stimulated anti-CD3

(0.2ug/ml) and APCs in the presence of DMSO or AB-MECA (50uM) for 72 hrs.

Cells were harvested and proliferation was measured using cell titer glo kit

(Molecular probes). (A) in the absence of IL-2 (B) in the presence of IL-2

(20ng/ml) C) CD4 T cells were stimulated with plate bound anti-CD3 (2ug/ml) and

anti-CD28 (1ug/ml). Supernatants were collected at different time points and

analyzed using ELISA.

Page 114: Phospholipase D Signaling in T Cells

97

Adenosine and CD4 T cell population dynamics

In addition to determining the effect of A3 receptor signaling on the effector

functions of CD4 T cells, we investigated the effect of A3 receptor signaling on

CD4 T population dynamics. Based on the previous observation of inhibition of

CD25 expression on non-Treg cells by AB-MECA, we predicted that culture of

CD4 T cells with exogenous IL-2 and AB-MECA would cause preferential

enrichment of Treg cells. To test this hypothesis, CD4 T cells were stimulated

with anti-CD3 in the presence of DMSO or AB-MECA for 3 days followed by

expansion in IL-2 containing medium for 3 days. The culture medium was

supplemented with fresh AB-MECA and anti-CD3 every 24 hrs for the first 3

days. As predicted, flow cytometry showed 22% of CD4 T cells were Foxp3 and

CTLA4+ in AB-MECA treated samples in contrast to <2% in DMSO treated

samples (Fig 21 a).

The increased percentage of Foxp3+ CD4 T cells in AB-MECA treated

cultures could result from either expansion of Foxp3+ T cells or from induction of

Foxp3 expression in CD4+CD25- T cells. To test for the possibility of increased

expansion of Foxp3+ cells, we tested the effect of AB-MECA on the proliferation

of CD4+CD25- and CD4+CD25+ T cells separately. AB-MECA substantially

reduced the proliferation of CD4+CD25- T cells (50% reduction) and had no

significant effect on the proliferation of CD4+CD25+ T cells (Fig 21b). This result

of inhibition of CD4+CD25- T cell proliferation was confirmed using CFSE

proliferation assay as well.

Page 115: Phospholipase D Signaling in T Cells

98

AB-MECA DMSO

22.01.61

FOXP3

C

TLA

4

p < 0.001

0

20

40

60

80

100

CD4+CD25- CD4+CD25+

Rel

ativ

e lig

ht u

nit

DMSO

A3

A)

B)

AB-MECA

Fig 21. Effect of adenosine A3 receptor signaling on CD4 T cell population

dynamics. A) Total CD4 T cells were cultured with anti-CD3 antibody plus APCs

in medium containing DMSO or adenosine A3 receptor specific agonist (AB-

MECA, 50uM) for 72 hrs. After 3 days, cells were placed in medium containing

IL-2 (20ng/ml) for 4 days and analyzed for Foxp3 and CTLA4 expression. B)

CD4+CD25- and CD4+ CD25+ T cells were sorted from mice splenocytes and

cultured with anti-CD3 and APCs along with DMSO or AB-MECA for 72 hrs.

Proliferation was assessed using cell titer glo kit (Molecular probes).

Page 116: Phospholipase D Signaling in T Cells

99

Relative increase in Foxp3+ T cells with AB-MECA could also be caused

by de novo induction of Foxp3+ cells from naïve T cells (Chen et al., 2003). To

address this possibility, CD4+CD25- T cells were stimulated with anti-CD3 in the

presence or absence of AB-MECA. TGF-beta has been shown previously to

induce Foxp3 expression in CD4+ CD25- T cells and thus served as positive

control for this experiment. However, similar treatment of naïve T cells with AB-

MECA did not lead to induction of Foxp3+ expression in CD4 T cells (Fig 22, top

row). Interestingly, co-culture of AB-MECA with TGF-β led to significant reduction

in the percentages of Foxp3+ CD4 T cells (8%) compared to 20% of TGF-β alone

treated cultures (Fig 22, bottom row). This result could mean that A3 receptor

signaling antagonizes TGF-β signaling in naïve T cells. Taken together, these

data rule out the possibility of Foxp3 induction by AB-MECA and rather strongly

suggest preferential enrichment of Foxp3+ T cells.

Next, we asked whether adenosine A3 receptor signaling could inhibit

effector T cell proliferation in vivo in mice. To address this question, CFSE

labeled transgenic mouse derived CD4 T cells were intravenously transferred

into congenic Ly5.1 mice and challenged in vivo with the cognate antigenic

peptide with or without AB-MECA. As the source of CD4 T cells, we used p25

TCR transgenic mouse T cells, specific to Mycobacterium Tuberculosis antigen

p25 peptide (Tamura et al., 2004). The antigen challenged mice also received

AB-MECA or vehicle control through intraperitoneal (i.p) injection daily for 1

week. Expression levels of Foxp3 and CFSE were measured on day 8 after

antigen administration.

Page 117: Phospholipase D Signaling in T Cells

100

Foxp3

0 102 103 104 105

<APC-A>

0

102

103

104

105

<PE-

A>

62.6 2.17

3.7631.40 102 103 104 105

<APC-A>

0

102

103

104

105

<PE-

A>

49.4 0.58

149.1

CTL

A4

DMSO AB-MECA

0 102 103 104 105

<APC-A>

0

102

103

104

105

<PE-

A>

43.7 8.64

2.1245.60 102 103 104 105

<APC-A>

0

102

103

104

105

<PE-

A>

42.7 21.6

4.431.4

Fig 22. Flow cytometric analysis of Foxp3 induction in CD4+CD25- T cells

following adenosine A3 receptor signaling. CD4+CD25- T cells were cultured

with anti-CD3 stimulation and APCs in medium containing DMSO or adenosine

A3 receptor agonist (AB-MECA) for 72 hrs. After day 3, cells were placed in

medium containing IL-2 (20ng/ml) for 4 days and analyzed for Foxp3 and CTLA4

expression (Top row). Exogenous TGF-β (5ng/ml) was added to the medium

(bottom row).

Page 118: Phospholipase D Signaling in T Cells

101

Intravenous injection of 2.5μg antigen peptide in the recipient mice caused

effective proliferation of CD4 T cells as seen from the progressive dilution of

CFSE in dividing cells (Fig 23a). As expected from in vitro data, CFSE labeled

CD4 T cells proliferated considerably less in AB-MECA treated mice compared

vehicle treated control mice. Also, we observed increased frequency of Foxp3+

CD4 T cells in AB-MECA treated mice. Further, we compared the total cell

numbers in each cell division of the Foxp3- and Foxp3+ T cell fraction (Fig 23b).

Consistent with the in vitro data, we found reduced cell numbers in Foxp3- T cell

fraction alone of AB-MECA treated mice. This result led us to suggest that A3

receptor (A3R) signaling selectively inhibits effector T cell proliferation in vivo as

well. Thus, under adenosine rich conditions (eg: excessive inflammation, hypoxia

induced cell death in solid tumors), A3R activation might inhibit effector T cell

proliferation with a concomitant increase in the frequency of Treg cells and

change the balance towards immune suppression.

Cross talk between Adenosine receptor and Phospholipase D signaling

Ado receptors, A2a and A3 have been reported to utilize PLD in mediating

cell functions (Thibault et al., 2000) (Mozzicato et al., 2004). In light of our

findings with A3 receptor signaling in CD4 T cells, we next determined the effect

of A3R signaling on PLD activity in CD4 T cells. TCR induced PLD activity was

measured in AB-MECA treated CD4 T cells using a commercially available PLD

assay kit (Molecular Probes). First, whole splenocytes from P25 TCR transgenic

mice were stimulated with the cognate P25 peptide overnight to induce the

Page 119: Phospholipase D Signaling in T Cells

102

CFSE

FOXP

3 No Antigen

Ag + DMSO Ag + AB-MECA

0

1000

2000

3000

4000

5000

6000

7000

0 1 2 3 4 5 6 7

GENERATIONS

TOTA

L C

ELL

NU

MB

ERS

DMSOA3

0

50

100

150

200

250

300

0 1 2 3 4 5 6 7GEN ER A T ION S

Tota

l cel

l num

bers

DMSOA3

A)

B)

C)

Page 120: Phospholipase D Signaling in T Cells

103

Figure 23. Effect of A3 receptor signaling on the proliferation kinetics of

Foxp3 positive and Foxp3 negative CD4 T cells in vivo. CFSE labeled CD4 T

cells from P25 TCR transgenic (Ly5.2) mice were intravenously transferred to

Ly5.1 recipient mice. 24 hrs later, the recipients were challenged with 2.5ug of

P25 peptide along with either DMSO (control) or AB-MECA (200ug/Kg) (i.p) for a

period of 7 days. On Day 8, mice were sacrificed and all their peripheral lymph

nodes (axial, cervical, inguinal, popliteal) were isolated and pooled for analysis

by flow cytometry. A) Representative FACS plots showing CFSE dilution of

adoptively transferred T cells in DMSO and AB-MECA treated mice. B) Line

graphs depicting the number of foxp3- T cells in each generation of DMSO

(dotted lines) and AB-MECA treated (solid lines) mice. C) Line graphs depicting

the number of foxp3+ T cells in each generation of DMSO (dotted lines) and

ethanol treated (solid lines) mice. Data is representative of 2 independent

experiments.

Page 121: Phospholipase D Signaling in T Cells

104

84

86

88

90

92

94

96

98

100

102

104

1

FLU

OR

SCEN

CE

INTE

NSI

TY

DMSO USp = 0.000592DMSO STIM A3 USA3 STIM

Fig 24. Assay of PLD activity in AB-MECA treated CD4 T cells. Total

splenocytes from P25 TCR transgenic mice were incubated overnight with the

P25 peptide (1ug/ml). Following day, CD4 T cells were sorted and pre-treated

with DMSO or AB-MECA for 30 mins. Cells were stimulated with soluble biotin

conjugated anti-CD3 (10ug/ml) and avidin for 30 mins. Lysates were then probed

for PLD activity as per manufacturers instruction. Filled bars are DMSO treated

samples and open bars are AB-MECA treated samples.

Page 122: Phospholipase D Signaling in T Cells

105

expression of A3 receptor. The following day CD4 T cells were sorted and

pretreated with AB-MECA for 30 min before restimulation with soluble anti-CD3

for 30 min. The total cell lysate from these samples were then used for PLD

assay as per manufacturers instruction. As reported previously, we observed an

increase in PLD activity in CD4 T cells following stimulation and this increase is

not seen in samples treated with AB-MECA (Fig 24). Thus, A3 receptor signaling

in CD4 T cells blocks TCR induced PLD activity. This indicates that the selective

inhibition of Foxp3- T cell proliferation in AB-MECA treated cells is caused in part

by blockade of PLD signaling.

Adenosine A3 receptor signaling and cAMP

A3 receptor activation is linked to Gi-mediated inhibition of adenylyl cyclase

(Gessi et al., 2008). Recent reports indicate that A3 receptors dictate cellular

responses such as mast cell degranulation in part by decreasing intracellular

cAMP concentration (Hasko et al., 2008). To determine the effect of A3 receptor

activation on cAMP levels in CD4 T cells, splenocytes from P25 TCR transgenic

mice were stimulated with P25 peptide overnight to induce expression of A3

receptor. On the following day, CD4 T cells were sorted by MACS column and

incubated with DMSO or AB-MECA for 30 min. CD4 T cells were then stimulated

with biotin conjugated anti-CD3 and avidin for 5 mins. Cells were then lysed in

0.1N HCL and the supernatants were assayed using cAMP-specific ELISA (Fig

25a).

Page 123: Phospholipase D Signaling in T Cells

106

02468

101214161820

1

cAM

P (p

mol

/ml)

DMSOUnstimulatedDMSO Stimulated

AB-MECAUnstimulatedAB-MECAStimulated

FOXP3

CTL

A4

cAMP (1uM) cAMP (10uM)1.4

cAMP (100uM) cAMP (1mM)

21.2 30.0

A)

B)

#

*

Page 124: Phospholipase D Signaling in T Cells

107

Fig 25. Assay for cAMP levels and activity in CD4 T cells. A) Total

splenocytes from P25 TCR transgenic mice were incubated overnight with the

P25 peptide (1ug/ml). On the following day, CD4 T cell were sorted and pre-

treated with DMSO or AB-MECA for 30 mins. Cells were stimulated with soluble

biotin conjugated anti-CD3 (10ug/ml) and avidin for 5 mins and lysed in 0.1N

HCL. A) The supernatants were assayed for cAMP levels using cAMP specific

ELISA. (* p<0.07, # p= 0.065) B) CD4 T cells were cultured with anti-CD3 and

APCs along with varying doses of cAMP (1μM to 1mM) for 3 days. Cells were

further cultured in IL-2 containing medium for 4 days and analyzed by flow

cytometry. C) Total Foxp3- T cell numbers (left) and Foxp3+ T cell numbers

(right) in exogenous cAMP treated cultures on day 7.

0

1

2

3

4

5

6

1

Tota

l cel

l num

bers

0

0.05

0.1

0.15

0.2

0.25

0.3

1

cAMP (1uM)cAMP (10uM)

cAMP (100uM)cAMP (1000uM)

(106) (106)

C)

Page 125: Phospholipase D Signaling in T Cells

108

We did not observe any increase in cAMP levels in either stimulated or non-

stimulated cells of control (DMSO) treated samples. In contrast, we detected high

concentration of cAMP in both unstimulated and stimulated samples treated with

AB-MECA. This observation is in stark contrast to what has been reported in

other immune cells regarding A3 receptors decreasing intracellular cAMP

concentrations (Jin et al., 1997).

The second messenger cAMP is shown to be a negative regulator of T cell

activation and proliferation (Bopp et al., 2007; Ramstad et al., 2000). A previous

study showed that cAMP-dependent signal transduction interferes with CD25

expression and IL-2 production (Ramstad et al., 2000). To test if A3 receptor

signaling blocks effector T cell activation/proliferation via cAMP, CD4 T cell

proliferation was determined following addition of exogenous cAMP (dibutyryl-

cAMP, Db-cAMP, a cell permeable analog of cAMP, Calbiochem). Total CD4 T

cells were stimulated with anti-CD3 antibody and APCs in the presence of

increasing concentrations of Db-cAMP for 3 days followed by culture in IL-2

containing medium for 4 days. Flow cytometric analysis of Foxp3 and CTLA4

expression show that low concentrations (1uM-10uM) of cAMP have no effect of

CD4 T cells population dynamics, whereas high concentration (100-1000μM)

cause increased frequency of Treg cells (Fig 25 b, c). Hence, increase in cAMP

level selectively causes inhibition of Foxp3- T cell proliferation similar to those

observed with AB-MECA. Taken together, these findings indicate that cAMP is

involved in mediating the effects of adenosine A3 receptor on CD4 T cells.

Page 126: Phospholipase D Signaling in T Cells

109

Various studies suggest that the effect of cAMP in T cells is mediated by

the cytosolic cAMP-dependent protein kinase A (PKA) type I (Ramstad et al.,

2000). It is reported that cAMP binds to the two regulatory subunits in the inactive

PKA tetrameric holoenzyme to release two active catalytic subunits. In turn,

these catalytic subunits phosphorylate diverse proteins to regulate their activity

(Yao et al., 2002). Based on these observations, it was hypothesized that the

increase in cAMP levels in A3R agonist treated samples would lead to activation

of PKA followed by increased phosphorylation of its downstream effector

molecules. To test this experimentally, western blot analysis was performed on

CD4 T cells stimulated with anti-CD3 in the presence of AB-MECA. The lysates

were assayed for phosphorylation patterns by western blot analysis using anti-

phospho-PKA substrate antibody. In comparison to both unstimulated and

stimulated control (DMSO) treated samples, stimulated AB-MECA treated

samples showed increased phosphorylation of PKA substrates (Fig 26a). cAMP

assay showed increased cAMP levels in both unstimulated and stimulated

samples treated with AB-MECA. We further determined the effect of A3 receptor

signaling on PKA activity in both stimulated and unstimulated T cells. As

predicted from the data on cAMP, AB-MECA increased PKA substrate

phosphorylation in unstimulated AB-MECA treated CD4 T cells (Fig 26b). A mild

increase in PKA substrate phosphorylation was observed in AB-MECA treated

CD4 T cells stimulated with anti-CD3.

Page 127: Phospholipase D Signaling in T Cells

110

DMSO

DM

SO

DM

SO

AB

-MEC

A

(50u

M)

AB

-MEC

A

(100

uM)

US S US S

AB-MECA (50uM)

US S S SA)

B)

Fig 26. Western blot analysis of phosphorylation patterns of PKA

substrates in AB-MECA treated CD4 T cells. Total splenocytes from P25 TCR

transgenic mice were incubated overnight with the P25 peptide (1ug/ml). The

following day, CD4 T cell were sorted and pre-treated with DMSO or AB-MECA

for 30 mins. Cells were stimulated with soluble biotin conjugated anti-CD3

(10ug/ml) and avidin for 5 mins and the lysate were separated on 8%

polyacrylamide gels, transferred on to nitrocellulose membrane and blotted with

antibody against phospho-PKA substrates. A and B) (Top blot) phosphoPKA blot,

(bottom) beta actin, loading control. US = Unstimulated sample, S = Stimulated

sample.

Page 128: Phospholipase D Signaling in T Cells

111

In conclusion, these data indicate that adenosine signaling through A3

receptor blocks T cell proliferation via an increase in cAMP and decrease in PLD

signaling.

PLD signaling: synergy between ethanol and adenosine

Since both ethanol and adenosine A3 receptor agonist inhibit PLD activity

and result in enrichment of Treg cells, we examined if there is synergy between

the A3R agonist (AB-MECA) and ethanol. CD4 T cells from mouse spleen were

stimulated with anti-CD3 antibody both in the presence and absence of ethanol

and AB-MECA. Lower concentrations of ethanol (0.5%) alone did not cause

increased frequency of Treg cells. However, when 0.5% ethanol was combined

with 50μM AB-MECA, substantial increase in the percentage of Foxp3+ T cells

was observed (Fig 27 a). Also, the comparison of Foxp3+ and Foxp3-T cell

numbers indicate decreased proliferation of Foxp3- T cells in cultures treated

with either AB-MECA alone or AB-MECA and 0.5% ethanol (Fig 27 b) indicating

enrichment of Treg cells in these culture conditions. The synergy between these

two factors may be due to increased suppression of PLD signaling and/or

inhibition of multiple signaling pathways that collectively block non-Treg

activation.

Previously, it was reported that ethanol increases adenosine receptor

stimulated cAMP levels in human lymphocytes (Hynie et al., 1980) (Gordon et al.,

1986). Increased cAMP levels in the presence of both ethanol and adenosine

could contribute to the observed synergy in Treg enrichment. However, we

Page 129: Phospholipase D Signaling in T Cells

112

DMSO

0.78

AB-MECA (50μM)

4.74

0.5% ethanol 0.86

A3 + 0.5% ethanol 11.74

1.0% ethanol 4.91

A3 + 1.0% ethanol 20.35

A)

Foxp3

CTL

A4

Page 130: Phospholipase D Signaling in T Cells

113

Fig 27. Combinatorial effect of ethanol and adenosine on CD4 T cell

population dynamics. Total CD4 T cells were cultured with anti-CD3 and APCs

along with ethanol alone or in combination with AB-MECA(50uM) for 3 days.

Cells were further cultured in IL-2 containing medium for 4 days and analyzed by

flow cytometry. A) Flow cytometric plots depicting Foxp3 in the x-axis and CTLA4

in the y-axis. B) Total Foxp3- T cell (left) and Foxp3+ (right) T cell numbers in

0.5% ethanol, AB-MECA and in AB-MECA + 0.5% ethanol treated cultures.

0

1

2

3

4

5

6

7

8

1

Tota

l cel

l num

bers

0

20

40

60

80

100

120

1

0.5 % ethanol

AB-MECA

AB-MECA +0.5% ethanol

(106) (103)

B)

Page 131: Phospholipase D Signaling in T Cells

114

1 2 3

1. AB-MECA (50μM)

2. 0.5 % ethanol

3. AB-MECA + 0.5 % ethanol

Fig 28. Western blot analysis of phosphorylation patterns of PKA

substrates in CD4 T cells treated with a combination of ethanol and AB-

MECA. Total splenocytes from P25 TCR transgenic mice were incubated

overnight with the P25 peptide (1μg/ml). Following day, CD4 T cell were sorted

and pre-treated with ethanol (0.5%) or AB-MECA or both for 30 mins. Cells were

stimulated with soluble biotin conjugated anti-CD3 (10μg/ml) and avidin for 5

mins and the lysate were separated on 8% polyacrylamide gels, transferred on to

nitrocellulose membrane and blotted with antibody against phospho-PKA

substrates. A) (Top blot) phospho PKA blot, (bottom) beta actin, loading control.

Page 132: Phospholipase D Signaling in T Cells

115

observed very little, if any, increase in phosphorylation of PKA substrates in

samples treated with the combination of AB-MECA (50μM) and 0.5% ethanol

compared to 0.5% ethanol alone (Fig 28). Since there are many potential bands

that are not separated by 1D gel analysis, further analysis is needed to determine

whether ethanol and AB-MECA have synergistic effect on PKA activity.

STRUCTURE-FUNCTION ANALYSIS OF PHOSPHOLIPASE D

The data presented above show that various exogenous and endogenous

factors influences the immune outcome by regulating PLD signaling. However,

the contribution of PLD to T cell signaling is not clearly understood. As described

previously, the catalytic action of PLD on phosphatidyl choline (PC) results in the

formation of phosphatidic acid (PA) and soluble choline. The formation of PA

constitutes the crucial contribution of PLD to cellular signaling and function. PA is

an important lipid second messenger that can also be synthesized by several

other routes and thus the contribution of PLD to the overall mass of PA in cells is

quite miniscule (Ktistakis et al., 2003). Therefore, PLD is thought to only

influence the local levels of PA. Thus, delineation of the spatial regulation of PLD

isoforms in T cells will help us understand its function in T cells.

As mentioned previously, the two isoforms of PLD localize at different

locations in the cell. Over expression and epitope tag studies reveal that PLD1 is

found mainly in punctate structures around perinuclear regions while PLD2 is

primarily found in the plasma membrane. To determine the region that controls

the localization of PLD1 and PLD2, a series of mutants of both PLD1 and PLD2

Page 133: Phospholipase D Signaling in T Cells

116

were generated. In order to assess the localization of these mutants in the cell,

these mutants were tagged with YFP in the N-terminal and transfected in Jurkat

T antigen cells, which express SV40 large T antigen to allow high level of

expression of transfected genes (Northrop et al., 1993) . After transfection (24

hrs), live cells were separated using Ficoll gradient, mounted on glass slides

coated with lysine, fixed and covered with cover slip. These slides were then

analyzed using confocal microscopy.

Determinants of PLD1 perinuclear localization

At first, we addressed the region responsible for the differential localization

of PLD1 and PLD2. The primary sequence of PLD1a revealed the presence of

116 amino acid ‘loop region’ inserted between catalytic sites II and III (Frohman

et al., 1999). Previous studies in which the loop region of PLD1 was deleted

have reported modest increase in the basal activity of PLD1 (Sung et al., 1999).

Based on this observation, we hypothesized that deletion of the loop region

redistributes PLD1 similar to PLD2 and this results in increased catalytic activity

of PLD1. To test this idea, mutant human PLD1b with loop (aa. 504-585) deletion

was constructed and its localization was assessed using confocal microscopy.

The loop deletion mutant of PLD1 lost its ability to localize in punctate structures

surrounding the nucleus and localized to the plasma membrane, similar to PLD2

(Fig 29). These data suggested that the loop region is a required component for

PLD1 localization in vesicular structures.

Page 134: Phospholipase D Signaling in T Cells

117

PLDc

5851 1036

Fig 29. Confocal microscopic analysis of the sub-cellular localization of

PLD1 loop deletion mutant. PLD1 and PLD2 full length and loop deletion

mutant are tagged with YFP. Green indicates the localization of the respective

construct and red in propidium iodide staining of the nucleus. These constructs

were transfected in jurkat T antigen cells. LIve cells were isolated 24 hours later

by ficoll gradient and plated on lysine coated glass slides. Cells were then

imaged using the confocal microscope and images were processed using LSM

image examiner software.

PX PH PLDc

loop

C-term

504

PLDc

5861 1036

PX PH C-ter

503

PLDc

PLDc

1 933

PLDcPX PH C-term

Full length PLD1b

Full length PLD2

Loop deleted PLD1b

Full length PLD1

Full length PLD2

Loop deleted

Page 135: Phospholipase D Signaling in T Cells

118

Full length PLD1

+ Full length PLD1

58loop

50FLAG

58loop

50FLAG

Fig 30. Confocal microscopic analysis of the sub-cellular localization of

PLD1 loop region alone or loop region and full length PLD1 combinations.

PLD1 full length is tagged with YFP. Green indicates the localization of the full-

length PLD1 construct. Loop alone construct was tagged with FLAG. Blue is the

DAPI staining of the nucleus. Loop alone construct was detected using anti-

FLAG conjugated with flurochrome. PLD1 and loop construct were transfected in

jurkat T antigen cells either alone or together. Live cells were isolated 24 hours

later by ficoll gradient and plated on to lysine coated glass slides. Cells were then

imaged using the confocal microscope and the images were processed using

LSM image examiner software.

Page 136: Phospholipase D Signaling in T Cells

119

Next, we determined if the loop region is sufficient to determine the

localization of PLD1 into vesicular structures. Furthermore, if loop interacts with

unknown target molecules in the cell that dictate the localization of PLD1 then

expression of the loop alone construct could interfere with PLD1-target molecule

interaction. Therefore, flag tagged ‘loop alone’ construct was co-transfected with

YFP fusion full length PLD1. Full length PLD1 localized in punctate structures

and when co-transfected with the ‘loop alone” construct, there were no

observable differences in its localization (Fig 30). The loop alone construct was

mainly detected in the plasma membrane. The data suggest that the loop region

is required for PLD1 sub-cellular localization in the vesicular structures but loop

alone (aa 504-585) is not sufficient to create the structure of PLD1 that interacts

with the cellular target that determines the localization of PLD1.

Also, we determined the contribution of PX, PH and C-terminal regions to

the peri-nuclear localization of PLD1 by deletion of the corresponding regions

from full length PLD1. These constructs were transfected into Jurkat T antigen

cells and their localization was determined by confocal microscopy. The

construct lacking PX and PH domain (a.a 1-333 deletion) localized entirely in the

cytoplasm (Fig 31). The mutant with only PX and PH domain (a.a 1-342) had

diffuse localization in both nucleus and cytoplasm, similar to vector alone.

Interestingly, the construct with only the C-terminal (aa. 933 -1036) localized

entirely in the nucleus. This suggests that the C-terminal (aa-933-1036) has

nuclear import property. Indeed, PLD1b localization in the nucleus has been

reported before (Freyberg et al., 2001).

Page 137: Phospholipase D Signaling in T Cells

120

PLDc

5851 1036

PLDcPX PH loop

C-term

504

PLD1b Full length

C-term

PLDc

585334 1036

PLDc loop

504

N-terminal deletion of PLD1

1

PX PH

342PX+ PH only

C-terminal only

1036 933

C-termina

Full length PLD1b

N-terminal deletion PX+PH only C-terminal only

Merged

YFP

DAPI

Page 138: Phospholipase D Signaling in T Cells

121

Fig 31. Schematic representation of the mutant PLD1 constructs and

confocal microscopy of their sub-cellular localization. All the constructs are

tagged with YFP. Green indicates the localization of these constructs. Blue is the

DAPI staining of the nucleus. These constructs were transfected in jurkat T

antigen cells. Live cells were isolated 24 hours later by ficoll gradient and plated

on lysine coated glass slides. Cells were then imaged using the confocal

microscope and images were processed using LSM image examiner software.

Page 139: Phospholipase D Signaling in T Cells

122

Together, these data suggest that the interaction of PX, PH domain with the rest

of PLD1 domains determines the perinuclear localization of PLD1. It is evident

that interactions between different domains rather than a single domain are key

to the sub cellular localization of PLD1.

Determinants of PLD2 plasma membrane localization

PLD2 has strikingly restricted distribution to the plasma membrane. Thus,

YFP fusion constructs of PLD2 deletion mutants were made to determine the

domain necessary for its plasma membrane localization. The construct

containing PX+PH+ C-terminal localized to the cytoplasm (Fig 32). Similar to

PLD1, 'C-terminal only’ construct of PLD2 localized to the nucleus. This

observation is in agreement with previous studies indicating highly homologous

C-termini of both PLD isoforms (Liu et al., 2001). Together, these observations

suggest that the interaction of PX, PH and C-terminal with the catalytic region

determines the plasma membrane localization of PLD2.

Future studies can be aimed at assaying the catalytic activities of both

PLD1 and PLD2 mutant constructs and its effect of TCR signaling to pinpoint with

certainty the contribution of PLDs to the functioning of the immune system.

Page 140: Phospholipase D Signaling in T Cells

123

PLDc

1 933

PLDcPX PH C-term

Full length PLD2

C term

1 933

PX PHPH+PX+C

C

termC terminal only

Full length PLD2

PH+PX+C C terminal only

Merged

YFP

PI

Page 141: Phospholipase D Signaling in T Cells

124

Fig 32. Schematic representation of the mutant PLD2 constructs and

confocal microscopy of their sub-cellular localization. All the constructs are

tagged with YFP. Green indicates the localization of these constructs. Red is the

propidium iodide staining of the nucleus. These constructs were transfected in

jurkat T antigen cells. Live cells were isolated 24 hours later by ficoll gradient and

plated on lysine coated glass slides. Cells were then imaged using the confocal

microscope and the images were processed using LSM image examiner

software.

Page 142: Phospholipase D Signaling in T Cells

125

EFFECT OF PLD2 GENE DELETION ON T CELL RESPONSES

Delineating the isoform specific contribution of PLD1 and PLD2 would be

critical to address their role in regulatory T cell enrichment. In order to delineate

the potential role of PLD2 in regulating the balance between effector and

regulatory T cell subsets, we generated mice deficient in PLD2 specifically in T

cells. Mice carrying floxed PLD2 genes were crossed with CD4 CRE mice in

order to delete the gene in double positive (CD4+CD8+) and CD4 and CD8

single positive T lymphocytes. PLD2 gene was successfully deleted in CD4 T

cells as detected by the differential size of DNA segments (WT mice = 6 kb,

PLD2 +/- = 6 and 5.2kb, PLD2 -/- = 5.2Kb) on southern blots (Fig 33a).

At first, we characterized PLD2flox/floxCD4cre mice to determine the effect

of deletion of PLD2 on thymocyte differentiation and peripheral T cell activation.

The absence of PLD2 had minimal effect on T cell development as determined

by flow cytometric analysis of thymocytes from WT and PLD2 (+/- and -/-) mice

(Fig 33B). Also, CD4:CD8 ratios in the spleen were similar between PLD2 (+/-)

and PLD2 (-/-) mice (Fig 33 C). Based on our previous in vitro findings, we

expected increased frequency of regulatory T cells in PLD2 knock out mice.

Surprisingly, deletion of PLD2 did not alter the development of naturally occurring

CD4+ CD25+ Foxp3+ regulatory T cells (Fig 33D). These observations suggest

that deleting PLD2 at the double positive stage of T cell development does not

affect the development of CD4, CD8 and regulatory T cells.

Page 143: Phospholipase D Signaling in T Cells

126

WT +/- -/-

WT +/- - / -

CD

8

CD 4 CD 4

Foxp3

A)

B)

C)

D)

CD

8

CD

25

Fig 33. Phenotypic analysis of conditional PLD2 deficient mice. A) Southern

blot analysis of CD4 T cells from WT or PLD2 (+/-), PLD2 (-/-) mice. B) Flow

cytometric analysis of thymocytes from PLD2 deficient mice. X-axis corresponds

to CD4 T cells and y-axis corresponds to CD8 T cells. C and D) Flow cytometric

analysis of splenocytes from PLD2 deficient mice. Foxp3 versus CD25 is gated

on CD4+ T cells.

Page 144: Phospholipase D Signaling in T Cells

127

Next, we determined the functional consequences of TCR induced

signaling in PLD2 deficient T cells. Sorted CD4 T cells were stimulated with anti-

CD3 and irradiated APCs overnight. Expression of CD69 (very early activation

antigen) and CD25 (IL-2Rα) was determined by flow cytometry. Both WT and

PLD2 (-/-) mice up regulated activation markers to the same extent (Fig 34A).

Concomitant with activation status, both WT and PLD2 (-/-) mice had similar

proliferation kinetics 72 hrs after activation (Fig 34B). Although, our previous in

vitro experiments suggested that inhibition of PLD signaling using 1-

alcohol/adenosine blocks proliferation of effector CD4 T cells, proliferation of CD4

T cells from PLD2 knock out mice did not exhibit any significant differences. This

result indicates that the ability of PLD2 (-/-) CD4 T cells to initially become

activated is not affected by their lack of PLD2.

In addition, we determined the cytokine secretion by CD4 T cells of PLD2

knock out mice. Sorted CD4 T cells were added to tissue culture plates coated

with anti-CD3 along with soluble anti-CD28 for 72 hrs. After 72 hrs of stimulation,

cells were rested for 48 hrs and then re-stimulated with PMA and ionomycin.

Intracellular cytokine staining on these cells indicate that in PLD2 (-/-) mice, the

CD4 T cells have 24% IL-2 positive cells compared to 5% (WT) and 12%

(PLD2+/-) mice (Fig 35). In contrast, the percentage of IL-4 secreting cells was

lower in PLD2 (-/-) mice (6%) compared to WT (18%) and PLD2 (+/-) mice

(12%). We did not observe any significant differences in the cytokine levels of IL-

17 and TNF-α between WT and PLD2 (-/-) mice. Thus, the data indicate that

Page 145: Phospholipase D Signaling in T Cells

128

A)

B)

αCD3

0.5

CD25

CD

69

Unstimulated μg/ml 0.2 μg/ml

PLD2 (+/+)

PLD2 (+/-)

PLD2 (-/-)

APC + CD3

0.E+00

1.E+02

2.E+02

3.E+02

4.E+02

5.E+02

6.E+02

0 0.1 0.3 0.5 1anti-CD3 concentration

O.D

APC + CD3 + IL2

0.E+00

5.E+05

1.E+06

2.E+06

2.E+06

3.E+06

0 0.1 0.3 0.5 1

ANTI-CD3 CONCENTRATION (ug/ml)

wtwt hetero d2hetero d2 homo d2homo d2

Page 146: Phospholipase D Signaling in T Cells

129

Fig 34. Characterization of CD4 T cell function in PLD2 deficient T cells. A)

Activation markers, CD69 (y-axis) and CD25 (x-axis) following stimulation. Cells

are gated on CD4 T cells after 24 hrs of stimulation with anti-CD3. B)

Proliferation analysis of PLD2 deficient CD4 T cells after 72 hrs of stimulation

with different concentrations of anti - CD3. WT, PLD2 (+/-) and PLD2 (-/-) mice

are littermates.

Page 147: Phospholipase D Signaling in T Cells

130

IL-2

IL-4

IL

-17

TNF-α

IFN- γ

PLD2 (-/-) PLD2 (+/-)PLD2 (+/+)

Fig 35. Cytokine profiles of PLD2 deficient CD4 T cells. A) CD4 T cells from

WT and PLD2 KO mice were stimulated with plate bound anti-CD3 and soluble

CD28 for 72 hrs, following which cells were rested for 2 days. Cells were re-

stimulated with PMA and ionomycin and intracellular FACS staining for IL-2,

IFN- γ, IL-4, IL-17 and TNF-α was performed.

Page 148: Phospholipase D Signaling in T Cells

131

PLD2 deficient CD4 T cells remain as IL-2 producers after three days of

stimulation and have impairment in differentiation into Th2 type T cells.

To test if PLD2 is necessary for effector T cell differentiation into different

subsets namely Th1, Th2 or Th17, CD4 T cells from WT and PLD2 knock out

mice were activated under Th1 (in the presence of IFN-γ, IL-12 and anti-IL-4),

Th2 (in the presence of IL-4 and anti-IFN-γ) or Th17 (in the presence of TGF-β

and IL-6) skewing conditions and FACS staining was performed to detect

intracellular cytokine expression. Under Th1 induction conditions, 78% of WT

CD4 T cells were IFN-γ positive and 11% were IL-2 positive while in PLD2 (-/-)

mice 21% were IL-2 positive and 66% were IFN-γ positive (Fig 36, top row)

indicating slight resistance to differentiate into Th1 phenotype. On the other

hand, under Th2 induction conditions 1.8% of WT CD4 T cells were IL-4 positive

in comparison to 0.8% of PLD2 (-/-) T cells (Fig 36, middle row). Under Th17

skewing conditions, ~8% of CD4 T cells in PLD2 (-/-) mice were IL-17 positive

compared to 6% of WT T cells (Fig 36, bottom row). Together, the data suggest

that PLD2 (-/-) mice have slight impairment in differentiation into Th1/Th2 cell

types and remain at IL-2 secreting stage. We can further assess effector T cell

differentiation in PLD2 (-/-) mice in vivo (eg: by using P25 peptide, a strong

inducer of Th1 response).

In light of our previous findings, we next determined if ethanol/adenosine

inhibits effector T cell proliferation through inhibiting PLD2. To test this, we

treated WT, PLD2 (+/-) and PLD2 (-/-) mice with DMSO, A3 agonist or A3 agonist

plus 0.5% ethanol. We predicted that if A3 agonist inhibited PLD2 signaling, then

Page 149: Phospholipase D Signaling in T Cells

132

PLD2 (+/+) PLD2 (+/ -) PLD2 (- / -) IL

-2IL

-4IL

-17

TH1 induction

TH2 induction

TH 17 induction

IFN- γ

Fig 36. Differentiation of PLD2 CD4 T cells into Th1, Th2 and Th17 Effector

cells. Spleen CD4 T cells were stimulated with anti-CD3 in vitro and rested 5

days in the presence of Th1, Th2 or Th17 skewing conditions. T cells were re-

stimulated with PMA and Ionomycin and intracellular cytokine staining was

performed.

Page 150: Phospholipase D Signaling in T Cells

133

WT

PLD2 (+/-)

PLD2 (-/-)

DMSO AB-MECA

CTL

A4

Foxp3

Fig 37. Effect of A3 receptor agonist on PLD2 deficient CD4 T cells. CD4 T

cells isolated from WT and PLD2 KO mice spleen were stimulated with APC and

anti-CD3 either in presence of A3 agonist (AB-MECA) or carrier control (DMSO)

for 3 days. On day 4, cells were washed and placed in medium containing IL-2

for 4 days. On day 7, cells were harvested and stained for detection of Foxp3

expression and analyzed using flow cytometry.

Page 151: Phospholipase D Signaling in T Cells

134

WT

PLD2 (+/-)

PLD2 (-/-)

2% ethanol 1% ethanol0.5% ethanol

Foxp3

CTL

A4

Fig 38. Effect of ethanol on PLD2 deficient CD4 T cells. CD4 T cells isolated

from WT and PLD2 KO mice spleen were stimulated with APC and anti-CD3 in

the presence of varying concentrations of ethanol for 3 days. On day 4, cells

were washed and placed in medium containing IL-2 for 4 days. On day 7, cells

were harvested and stained for detection of Foxp3 expression and analyzed

using flow cytometry.

Page 152: Phospholipase D Signaling in T Cells

135

treatment of PLD2 (+/-) with A3 agonist and ethanol would lead to increased

frequency of Foxp3+ cells. However, we found no differences in the frequency of

Foxp3+ cells between WT and PLD2 knock out mice under these treatment

conditions (Fig 37 and 38). This suggests that A3 agonist/ethanol mediated

enrichment of Treg cells might involve additional signaling pathways other than

PLD2. Future experiments can be conducted to determine the level of PLD

activity in PLD2 (-/-) T cells. Based on the experiments of Treg enrichment using

AB-MECA/ethanol it is very likely that PLD1 compensates for the lack of PLD2 in

PLD2 (-/-) mice thus exhibiting no difference in PLDactivity levels.

Future studies in PLD2 (-/-) mice can be conducted to determine its role in

T cell signaling and function. Previous data from the lab indicated that PLD2 is

involved in proximal TCR signaling events. We can determine if PLD2 (-/-) mice

exhibit altered TCR signaling pathway compared to WT mice.

Page 153: Phospholipase D Signaling in T Cells

CHAPTER FOUR

DISCUSSION

Introduction

The response to pathogenic attack and maintenance of immune

homeostasis requires precise coordination between different cell types of helper

T cell subsets. Population balance between different types of cells is key to

sustaining a balanced immune system. Many studies have reported altered

balance between effector versus regulatory T cell subsets as the prime cause of

disease pathogenesis. For instance, increased frequency and suppressor

function of Treg cells have been reported and proposed as potential mechanism

of immune suppression in patients with tumor and HIV-infection (Strauss et al.,

2007a) (Strauss et al., 2007b) (Sempere et al., 2007). On the other hand,

reduced number/function of Treg cells and increased effector T cell function have

been reported in patients with allergic/autoimmune diseases (Dejaco et al., 2006)

(Bacchetta et al., 2006).

Hence, ideal treatment for conditions of excessive immune suppression

would be to enhance the frequency/ function of effector T cells and vice–versa for

autoimmune diseases. Therapeutic potential of this strategy has spurred the

136

Page 154: Phospholipase D Signaling in T Cells

137

interests of researchers leading to the discovery of many mechanisms for

specific enrichment of either Treg cells or effector T cells.

Previously, our lab reported the differential requirement for PLD signaling

in Treg cells versus effector T cells. As a consequence, inhibition of PLD

signaling led to preferential enrichment of Treg cells by inhibiting effector T cell

proliferation. Interestingly, other reports suggest increased PLD expression and

signaling in autoimmune myocarditis and chronic inflammation (Ahn et al., 2004).

For my dissertation work, I focused on identification of mechanisms regulating

PLD signaling in T cells. The data demonstrated that various exogenous

(ethanol, bacterial toxins) and endogenous (adenosine) factors alter the immune

balance through regulating PLD signaling. PLD2 deficient mice showed that the

effect of these molecules on the Teff: Treg balance is not dependent on PLD2

alone. Further the structure that controls the sub cellular localization of PLD1 and

PLD2 was determined using mutant construct analysis.

IMMUNE BALANCE: EFFECT OF EXOGENOUS FACTORS

The human body is exposed to myriads of pathogens, pollutants, and

environmental and bacterial toxins in day-to-day life. The immune system has

evolved in many ways to combat these intruders and to maintain peace with self

through mechanisms like plasticity of helper T cell subsets. But some pathogens

and various exogenous molecules selectively manipulate the immune balance to

their advantage by shifting the equilibrium between these activities one way or

Page 155: Phospholipase D Signaling in T Cells

138

the other. For instance, environmental toxin like dioxin has been shown to cause

immune suppression by increasing Treg frequency (Quintana et al., 2008). Our

data demonstrate that by inhibiting PLD signaling, exogenous factors like ethanol

and C. difficile toxin tip the balance towards immune suppression.

Our results suggest that these external factors by means of inhibiting PLD

signaling, block the up regulation of activation induced CD25 expression on

naïve CD4 T cells. IL-2 is an important T cell growth factor and CD25 is required

for the proper function/signaling of IL-2. However, Treg cells constitutively

express CD25 and thus expand in the presence of exogenous IL-2. The

functional significance of ethanol/C. difficile toxin mediated inhibition of PLD

signaling is suggested by the decreased proliferation of antigen specific effector

T cells and increased frequency of antigen specific Treg cells in vivo.

There is extensive evidence that ethanol consumption leads to immune

suppression. Recently, both acute and chronic alcohol intake have been shown

to result in specific defects in innate and adaptive immunity (Nelson and Kolls,

2002). It is suggested that ethanol results in loss of splenic and circulating T and

B cells through apoptosis (Shao et al., 1995) (Sibley and Jerrells, 2000). Our

experiments using ethanol provide evidence to suggest that this loss of T cells

might be due to lack of PLD signaling and its effect on naïve T cells. In addition

to mediating its effect through CD4 T cells, alcohol has been shown to modulate

the functioning of other immune cells like macrophages, neutrophils, dendritic

cells with antigen presenting capabilities (Szabo et al., 1993) (Nelson and Kolls,

Page 156: Phospholipase D Signaling in T Cells

139

2002). The lack of activation induced marker expression on CD4 T cells might

also be mediated through lack of antigen presentation through these APCs.

Thus, in our experimental set up we cannot rule out the effect of ethanol on these

APCs and the consequent reduction in TCR activation and PLD signaling.

In contrast to mice injected with ethanol intraperitoneally, mice fed on

alcohol diet showed increased frequency of Treg cells only locally (mesenteric

lymph nodes). This result can be explained based on previous studies

suggesting that oral administration of ethanol results in increased availability and

metabolism of ethanol in the gut resulting in less entry of ethanol into the blood

stream while intraperitoneal injections result in rapid appearance of ethanol in the

blood stream (Livy et al., 2003). Thus oral exposure of alcohol reflects the

localized effect of ethanol on T cells while intraperitoneal injection reflects

systemic effect of ethanol on T cells. This phenomenon of local immune

suppression could in principle be exploited for treatment of gut associated

inflammatory diseases like IBD and colitis.

Our studies with C. difficile toxin demonstrate a new facet of immune

evasion strategy through PLD signaling. C. difficile toxins have long been known

to induce reorganization of actin filaments due to modification of Rho proteins

(Ottlinger and Lin, 1988). Since PLD is an effector molecule of Rho, C. difficile

was also reported to inhibit PLD signaling. However, functional relevance for the

lack of PLD signaling was not previously established. On the basis of our

experimental results, we propose that by inhibiting PLD signaling in naïve T cells

Page 157: Phospholipase D Signaling in T Cells

140

through toxin B, C. difficile eliminates effector T cell functions. The consequent

increase in the frequency of Treg cells aids the pathogen’s success in promoting

illness and furthering its own survival. Although we report the effect of Toxin B on

altering the immune balance, we have not tested the effect of blocking Toxin B

using anti-Toxin B antibodies. Based on our results, we predict that blocking of

Toxin B would mitigate the effects of C. difficile on the immune system and

ameliorate the disease progression by promoting effector and memory T cell

expansion.

IMMUNE BALANCE: EFFECT OF ENDOGENOUS FACTORS

One of the drawbacks of a powerful immune response is uncontrolled immune

response against pathogens and associated collateral damage to self. The

restraint of uncontrolled immune response is mediated through negative feed

back mechanisms. Adenosine (Ado) is an endogenous molecule well known for

its role in mediating the negative feed back mechanisms of immune responses.

Previous studies have reported inhibition of inflammation following activation of

adenosine receptors. Adenosine receptors are expressed by all immune cells

and A2a receptor signaling on macrophages, dendritic cells and T cells have

been shown to inhibit effector function (Zarek et al., 2008). Based on many

studies, A2a receptors are considered the dominant receptor dictating

lymphocyte responses in T cells.

Page 158: Phospholipase D Signaling in T Cells

141

Here we demonstrate the role of A3 receptor in inhibiting effector T cell

responses. The data indicate that stimulation of T cells in the presence of A3

receptor agonist inhibits TCR induced activation of PLD signaling. This inhibition

of PLD signaling abrogates CD25 expression, effector T cell proliferation, and

cytokine secretion. Although, signaling through A3 receptor in other immune cells

results in low cAMP concentrations, we found that signaling through A3 receptor

in CD4 T cells results in elevated cAMP levels and increased PKA activity.

Previous studies on A3 receptors report both pro and anti-inflammatory

effects depending on the system investigated (mast cells, macrophages) and

species examined (rat, humans) etc (Gessi et al., 2008). However, the specific

effect of activating A3 receptors on CD4 T cells has never been studied. In the

heart, A3 receptors mediate cardioprotection through modulating PLD signaling

(Mozzicato et al., 2004). This led us to determine the effect of A3 receptor

signaling on CD4 T cells. We found that A3 receptor signaling blocked TCR

induced PLD activation. At present, we do not know the exact mechanism of A3

receptor signaling leading to PLD signal inhibition. Inhibition of PLD by A2a

receptors is mediated by interference with the translocation of small GTPases,

Arf and Rho (Thibault et al., 2000). In future, we can test if A3 receptor inhibits

PLD through translocation of small GTPases as well. One recent study also

demonstrated inhibition of PLD1 activity directly by Gβγ subunits (Preininger et

al., 2006).

Page 159: Phospholipase D Signaling in T Cells

142

Since adenosine receptors are coupled to G-protein, signaling is thought

to occur either through the inhibition or stimulation of adenylyl cyclase resulting in

a decrease or increase of intracellular cAMP concentration respectively. The

classical signaling pathway associated with A3 receptor suggests Gi mediated

inhibition of adenylyl cyclase with a concomitant decrease in cAMP

concentrations. We experimentally determined the effect of A3 receptor

activation on cAMP concentrations. Contrary to previous reports, we found

increased cAMP levels in A3 receptor agonist treated CD4 Tcells. This increased

cAMP levels also resulted in increased PKA activity. The effect of A3 receptor

activation on inhibition of effector T cell proliferation and concomitant increase in

Treg frequency was indeed reproducible by the cell permeable analog of cAMP,

db-cAMP. Future experiments can be done to determine if the effect of A3

receptor is solely mediated by cAMP and PKA by co-culturing CD4 T cells with

A3R agonist and PKA inhibitor H-89.

A3 receptor is coupled to trimeric GTP binding proteins, Gαi and/or Gαo

and Gβγ dimers. The αi subunits directly inhibit adenylyl cyclase. However βγ

dimers from Gi/o are known to activate adenylyl cyclase (AC) isozymes II and IV

(Yao et al., 2002). We speculate that the increase in cAMP levels caused by A3R

activation is mediated through βγ dimers. We can test if this is indeed the case by

inhibiting βγ signaling. One way to inhibit βγ signaling is through over expression

of the carboxyl terminal of βARK1 retrovirally in CD4 T cells. Previous evidence

Page 160: Phospholipase D Signaling in T Cells

143

suggests that expression of carboxyl terminus of βARK1 binds free βγ dimers and

thus inhibits βγ signaling (Yao et al., 2002) (Koch et al., 1994).

Depending on the cell type studied, cAMP has been reported to inhibit (Le

Stunff et al., 2000; Thibault et al., 2002) or activate (Mamoon et al., 1999) PLD

activity through PKA signaling. In our experiments, we detected increased

cAMP levels in CD4 T cells treated with A3R agonist 5 mins following T cell

stimulation and observed a decrease in the PLD activity 15-30 mins after TCR

stimulation. Based on these results, it is very likely that increased cAMP levels

caused by A3 receptor signaling results in inhibition of PLD signaling. The

experiment using inhibitor of PKA (H-89) and/or constitutively active form of PKA

could also be used to determine PLD activity following PKA signaling.

Based on our various observations, we propose the following model

depicting A3 receptor signaling in CD4 T cells (Fig 39). Two different pathways

can mediate the effect of A3 receptor signaling in T cells. A central role for Gi/o

βγ is proposed. A3 receptor activation can lead to increased cAMP levels and

PKA signaling. PKA has been demonstrated to inhibit PLD activation. Another

possibility is βγ dimers can directly inhibit PLD activity (Preininger et al., 2006).

The ability of A3 receptor agonist to promote regulatory T cell enrichment

in vivo suggests that it could be used for treatment of autoimmune diseases.

Studies in mice and rats demonstrate that the anti-inflammatory effects of

methotrexate are in part mediated via A3 receptors (Montesinos et al., 2003).

Page 161: Phospholipase D Signaling in T Cells

144

Perhaps, methotrexate acting through A3 receptor on CD4 T cells blocks PLD

signaling and leads to Treg enrichment.

Interestingly, it has been demonstrated that A3 receptor is over expressed

in cancer tissues in comparison to normal tissues (Gessi et al., 2004a). It should

be noted that the concentration of adenosine in solid tumor reaches as high as

100μM due to high levels of hypoxia (Hasko and Cronstein, 2004). Thus, it is

highly likely that T cells activated by MHC class II positive APCs in these tissues

are selectively blocked via A3 receptor signaling. Moreover, since A3 receptor

agonists inhibit IFN-γ secretion, A3 receptor antagonists could be effective in

tumor immunotherapy.

Page 162: Phospholipase D Signaling in T Cells

145

A3 R

adenosine

α i/o - βγ

AC II and IV

cAMP

PKA

PLD

adenosine

PA PC

Fig 39. Schematic representation of A3 receptor signaling in CD4 T cells. A

central role for Gi/o βγ subunits is proposed. A3 receptor activation leads to

increased cAMP levels and activates PKA. PKA has been demonstrated to inhibit

PLD activation. Another possibility is βγ dimers directly inhibit PLD activity

(Preininger et al., 2006).

Page 163: Phospholipase D Signaling in T Cells

146

AB-MECA AND ETHANOL: POTENTIAL USE AS A TOLEROGENIC

ADJUVANT

Our data show that concurrent presence of PLD suppressive factors such

as adenosine and ethanol impose substantial suppression of immune response.

We observed that 0.5% ethanol had no effect of inhibition of effector T cell

proliferation or expansion of Treg cells. However, combination of 0.5% ethanol

with A3 receptor agonist led to substantial decrease in effector T cell

proliferation. This suggests that there exists a threshold for PLD activity in T cells

and the combination of ethanol and AB-MECA pushes PLD activity well below

this threshold. Under these conditions, we see a sizable effect of PLD inhibition

on effector T cell proliferation and subsequent enrichment of Treg cells.

A very high concentration of ethanol leads to cell death due to the toxic

effects of by products of ethanol (Lieber, 1997). However, we can avoid these

toxic effects by using a combination of low doses of ethanol and AB-MECA.

Induction/expansion of antigen-specific Treg cells is of great therapeutic

interest for the treatment of autoimmunity, allergy and organ transplantation. Our

studies using AB-MECA/ethanol show expansion of antigen specific Treg cells in

vivo. Therefore, AB-MECA alone or combination of AB-MECA and low dose

ethanol may be used in immunotherapy of various diseases.

A recent study proposed the use of immunosuppressants like

dexamethasone (DEX) as tolerogenic adjuvants (Kang et al., 2008). They

demonstrated that antigenic immunization combined with DEX led to block of

Page 164: Phospholipase D Signaling in T Cells

147

dendritic cell (DC) maturation and preferential expansion of antigen specific

Foxp3+ cells. Based on the similar effect observed with AB-MECA /ethanol, we

propose that agents inhibiting PLD signaling can be used as tolerogenic

adjuvants. However, we do not know the effect of AB-MECA and ethanol on the

status of DC maturation. Future experiments can be designed to evaluate the

effect of PLD signaling in antigen presenting cells like DC.

PLD LOCALIZATION: INTERACTION BETWEEN MULTIPLE STRUCTURES

PLD1 and PLD2 are 50% identical and display similar domain structures.

However, they localize to different regions in the cell. It is suggested that

localization determines intermolecular interaction with downstream target

molecules thus serving different functions in signal transduction. A previous

report shows that deletion of the loop region in PLD1 led to an increase in its

basal activity levels but the localization of the loop deletion mutant was not

determined. We over expressed loop deletion mutant in the Jurkat T cell line and

found that it had plasma membrane localization similar to PLD2. Since PLD2 is

characterized by high basal activity it is very likely that the increased activity of

the loop deletion mutant is in part due to its plasma membrane localization and

potential modification by membrane attached enzymes (eg: phosphorylation).

A puzzling observation was that the isolated loop domain had plasma

membrane localization as well. Since deletion of loop from PLD1 abrogates its

distribution from vesicular structures we hypothesize that the coordinated

Page 165: Phospholipase D Signaling in T Cells

148

interaction of loop within PLD1 and with unknown structure in the cell sequesters

it to vesicular structures. One way to test this hypothesis is to insert the isolated

loop domain into full length PLD2. If the interaction of loop with the other domains

is the factor governing its vesicular localization, then PLD2 will localize similar to

PLD1.

A previous report suggests that PH, PX and palmitoylation sites on the N-

terminal region of PLD1 serve as potential membrane binding determinants

(Sugars et al., 2002). In agreement with previous studies, in a deletion mutant

encompassing all 3 potential sites (N-terminal deletion mutant), the membrane

binding determinants redistributed to the cytoplasm. We also tested the cellular

distribution of the isolated ‘PH+PX’ domain of PLD1. The localization of this

mutant did not resemble that of full length PLD1. Rather it was found to have a

diffuse localization in both cytoplasm and nucleus similar to vector construct.

Based on this result, we suggest that ‘PH+PX’ domain cannot be studied in

isolation. Other studies suggest that an intact C-terminus of PLD1 is required for

its function (Liu et al., 2001). We made a deletion mutant containing the C-

terminus fused with YFP. Interestingly, this mutant localized only in the nucleus

of the cell. Taken together, we propose that a single domain does not determine

the localization of PLD1. Rather interactions between different domains of PLD1

and with other cellular components regulate localization of PLD1 in sub cellular

structures. The presence of C-terminal region alone determines its nuclear

localization while interaction of the C-terminal region with catalytic domains and

Page 166: Phospholipase D Signaling in T Cells

149

the loop region redistributes PLD1 to the cytoplasm. Further, the interaction of N-

terminal domains (PX+PH) with the rest of PLD1 determines its localization in

membranous vesicles like golgi.

We undertook experiments to determine the domains necessary for the

plasma membrane localization of PLD2. As proposed for PLD1, similar

interaction between domains determines its plasma membrane localization.

Isolated C-terminus mutant of PLD2 had nuclear localization while the mutant

with the presence of PH+PX along with the C-terminus redistributed to the

cytoplasm. Further, the interaction of the region containing the catalytic domains

with the rest of PLD2 results in its plasma membrane localization. Our studies

provide evidence for the role of different domains of PLD1 and PLD2 in

determining their respective localizations under basal condition. Future

experiments can be conducted to determine the cellular binding target of PLD

proteins particularly the loop and N-terminal half. It is possible that various

modifications of typical residues mediate the hierarchy of localization signals.

This can be addressed using a site directed mutagenesis approach. Also, we

have not examined the functional effect of these mutations. Functional studies in

future will provide a better understanding of the role of localization in determining

specific functions.

PLD2 AND T CELL FUNCTIONING

As a result of the critical role of PLD in development, PLD null mutants are

embryonic lethal (LaLonde et al., 2006). In PLD2flox/flox CD4 cre mice, PLD2 is

Page 167: Phospholipase D Signaling in T Cells

150

deleted relatively late (double positive stage). Our data demonstrate that PLD2

is dispensable for T cell development after the double positive stage of

thymocyte differentiation. Our data does not address the role of PLD2 in earlier

aspects of T cell development.

Also, deficiency of PLD2 did not affect CD4 T cell activation and

proliferation. On the other hand, we did observe differences in cytokine secretion

profiles of PLD2 deficient T cells. PLD2 deficient T cell cultures had more IL-2

positive cells than wild type T cells and showed resistance to differentiate in to

Th1 and Th2 lineage in vitro. This effect on effector T cell differentiation might be

caused by blockade of transcription factors like T-bet and GATA3. These

possibilities can be investigated in the future.

Surprisingly, PLD2 deletion had very little effect, if any, on the expansion

of regulatory T cells. One possible explanation is that PLD1 compensates for the

lack of PLD2. We can test for this possibility by inducibly deleting PLD1 alone in

PLD2 CD4 cre knock out mice. Another possibility is that PLD2 deficient T cells

are inherently different due to their thymocyte development and differentiation in

the absence of PLD2 signals. The other exciting possibility is perhaps that PLD2

is more important under conditions of physiological stress. This possibility can be

addressed by inducing autoimmunity or by observing the immunological

response to pathogens in PLD2 deficient mice in vivo. Nonetheless, these

experiments in PLD2 knock out mice have brought to light previously unknown

questions and answers.

Page 168: Phospholipase D Signaling in T Cells

151

CONCLUDING REMARKS

PLD signaling in T cells was discovered in 1991. However, until now the

specific role of PA and PLD in T cell functioning has not been clear. Our lab

group was the first to highlight the differential requirement of PLD signaling in

effector versus regulatory T cells. This led us to investigate the role of PLD

signaling in T cells. My dissertation work has focused on identifying the

regulation of PLD signaling in T cells. I found that physiological molecules like

adenosine maintain immune homeostasis by modulating PLD signaling. In

addition, I discovered that pathogens/ exogenous molecules could alter the host

immune response by virtue of their PLD inhibiting properties. Further, I

determined the structure that controls the sub-cellular localization of PLD

isoforms and their specific role in T cell function. These studies highlight the

complex array of functions mediated by PLD in T cells. In the process of studying

PLD, we have also discovered the role of adenosine A3 receptor signaling in T

cells. This study highlights the role of crosstalk between different signaling

pathways in human health and disease.

Page 169: Phospholipase D Signaling in T Cells

152

REFERENCES

Ahn, M., Lee, Y., Sim, K.B., Min, D.S., Matsumoto, Y., Wie, M.B., Shin, Y.G., and Shin, T. (2004). Increased expression of phospholipase D in the heart with experimental autoimmune myocarditis in Lewis rats. Immunological investigations 33, 95-105.

Alberts, B.A.J., Julian Lewis, Martin Raff, Keith Roberts, and Peter Walters, ed. (2002). Molecular Biology of the Cell; Fourth Edition.

Andresen, B.T., Rizzo, M.A., Shome, K., and Romero, G. (2002). The role of phosphatidic acid in the regulation of the Ras/MEK/Erk signaling cascade. FEBS Lett 531, 65-68.

Antoshechkin, A.G. (2001). On intracellular formation of ethanol and its possible role in energy metabolism. Alcohol and alcoholism (Oxford, Oxfordshire) 36, 608.

Athenstaedt, K., and Daum, G. (1999). Phosphatidic acid, a key intermediate in lipid metabolism. European journal of biochemistry / FEBS 266, 1-16.

Bacchetta, R., Passerini, L., Gambineri, E., Dai, M., Allan, S.E., Perroni, L., Dagna-Bricarelli, F., Sartirana, C., Matthes-Martin, S., Lawitschka, A., et al. (2006). Defective regulatory and effector T cell functions in patients with FOXP3 mutations. The Journal of clinical investigation 116, 1713-1722.

Bacon, K.B., Flores-Romo, L., Life, P.F., Taub, D.D., Premack, B.A., Arkinstall, S.J., Wells, T.N., Schall, T.J., and Power, C.A. (1995). IL-8-induced signal transduction in T lymphocytes involves receptor-mediated activation of phospholipases C and D. J Immunol 154, 3654-3666.

Page 170: Phospholipase D Signaling in T Cells

153

Bacon, K.B., Schall, T.J., and Dairaghi, D.J. (1998). RANTES activation of phospholipase D in Jurkat T cells: requirement of GTP-binding proteins ARF and RhoA. J Immunol 160, 1894-1900.

Baharav, E., Bar-Yehuda, S., Madi, L., Silberman, D., Rath-Wolfson, L., Halpren, M., Ochaion, A., Weinberger, A., and Fishman, P. (2005). Antiinflammatory effect of A3 adenosine receptor agonists in murine autoimmune arthritis models. The Journal of rheumatology 32, 469-476.

Balboa, M.A., Balsinde, J., Dennis, E.A., and Insel, P.A. (1995). A phospholipase D-mediated pathway for generating diacylglycerol in nuclei from Madin-Darby canine kidney cells. J Biol Chem 270, 11738-11740.

Bandyopadhyay, G., Sajan, M.P., Kanoh, Y., Standaert, M.L., Quon, M.J., Reed, B.C., Dikic, I., and Farese, R.V. (2001). Glucose activates protein kinase C-zeta /lambda through proline-rich tyrosine kinase-2, extracellular signal-regulated kinase, and phospholipase D: a novel mechanism for activating glucose transporter translocation. J Biol Chem 276, 35537-35545.

Banz, A., Peixoto, A., Pontoux, C., Cordier, C., Rocha, B., and Papiernik, M. (2003). A unique subpopulation of CD4+ regulatory T cells controls wasting disease, IL-10 secretion and T cell homeostasis. European journal of immunology 33, 2419-2428.

Baratelli, F., Lin, Y., Zhu, L., Yang, S.C., Heuze-Vourc'h, N., Zeng, G., Reckamp, K., Dohadwala, M., Sharma, S., and Dubinett, S.M. (2005). Prostaglandin E2 induces FOXP3 gene expression and T regulatory cell function in human CD4+ T cells. J Immunol 175, 1483-1490.

Basu, S., Golovina, T., Mikheeva, T., June, C.H., and Riley, J.L. (2008). Cutting edge: Foxp3-mediated induction of pim 2 allows human T regulatory cells to preferentially expand in rapamycin. J Immunol 180, 5794-5798.

Beissert, S., Schwarz, A., and Schwarz, T. (2006). Regulatory T cells. The Journal of investigative dermatology 126, 15-24.

Page 171: Phospholipase D Signaling in T Cells

154

Bennett, C.L., Christie, J., Ramsdell, F., Brunkow, M.E., Ferguson, P.J., Whitesell, L., Kelly, T.E., Saulsbury, F.T., Chance, P.F., and Ochs, H.D. (2001). The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nature genetics 27, 20-21.

Bettelli, E., Korn, T., and Kuchroo, V.K. (2007). Th17: the third member of the effector T cell trilogy. Current opinion in immunology 19, 652-657.

Beyer, M., and Schultze, J.L. (2006). Regulatory T cells in cancer. Blood 108, 804-811.

Billah, M.M., Eckel, S., Mullmann, T.J., Egan, R.W., and Siegel, M.I. (1989). Phosphatidylcholine hydrolysis by phospholipase D determines phosphatidate and diglyceride levels in chemotactic peptide-stimulated human neutrophils. Involvement of phosphatidate phosphohydrolase in signal transduction. J Biol Chem 264, 17069-17077.

Blin, N., and Stafford, D.W. (1976). A general method for isolation of high molecular weight DNA from eukaryotes. Nucleic acids research 3, 2303-2308.

Bopp, T., Becker, C., Klein, M., Klein-Hessling, S., Palmetshofer, A., Serfling, E., Heib, V., Becker, M., Kubach, J., Schmitt, S., et al. (2007). Cyclic adenosine monophosphate is a key component of regulatory T cell-mediated suppression. The Journal of experimental medicine 204, 1303-1310.

Borsellino, G., Kleinewietfeld, M., Di Mitri, D., Sternjak, A., Diamantini, A., Giometto, R., Hopner, S., Centonze, D., Bernardi, G., Dell'Acqua, M.L., et al. (2007). Expression of ectonucleotidase CD39 by Foxp3+ Treg cells: hydrolysis of extracellular ATP and immune suppression. Blood 110, 1225-1232.

Brown, H.A., Gutowski, S., Moomaw, C.R., Slaughter, C., and Sternweis, P.C. (1993). ADP-ribosylation factor, a small GTP-dependent regulatory protein, stimulates phospholipase D activity. Cell 75, 1137-1144.

Page 172: Phospholipase D Signaling in T Cells

155

Brunkow, M.E., Jeffery, E.W., Hjerrild, K.A., Paeper, B., Clark, L.B., Yasayko, S.A., Wilkinson, J.E., Galas, D., Ziegler, S.F., and Ramsdell, F. (2001). Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse. Nature genetics 27, 68-73.

Burelout, C., Thibault, N., Levasseur, S., Simard, S., Naccache, P.H., and Bourgoin, S.G. (2004). Prostaglandin E2 inhibits the phospholipase D pathway stimulated by formyl-methionyl-leucyl-phenylalanine in human neutrophils. Involvement of EP2 receptors and phosphatidylinositol 3-kinase gamma. Molecular pharmacology 66, 293-301.

Cantor, H., and Boyse, E.A. (1975). Functional subclasses of T-lymphocytes bearing different Ly antigens. I. The generation of functionally distinct T-cell subclasses is a differentiative process independent of antigen. The Journal of experimental medicine 141, 1376-1389.

Cao, Q., Batey, R., Pang, G., and Clancy, R. (1998). Altered T-lymphocyte responsiveness to polyclonal cell activators is responsible for liver cell necrosis in alcohol-fed rats. Alcohol Clin Exp Res 22, 723-729.

Cerottini, J.C., Nordin, A.A., and Brunner, K.T. (1970). Specific in vitro cytotoxicity of thymus-derived lymphocytes sensitized to alloantigens. Nature 228, 1308-1309.

Chadha, K.C., Stadler, I., Albini, B., Nakeeb, S.M., and Thacore, H.R. (1991). Effect of alcohol on spleen cells and their functions in C57BL/6 mice. Alcohol (Fayetteville, N.Y 8, 481-485.

Chatila, T.A., Blaeser, F., Ho, N., Lederman, H.M., Voulgaropoulos, C., Helms, C., and Bowcock, A.M. (2000). JM2, encoding a fork head-related protein, is mutated in X-linked autoimmunity-allergic disregulation syndrome. The Journal of clinical investigation 106, R75-81.

Chen, J. (2004). Novel regulatory mechanisms of mTOR signaling. Curr Top Microbiol Immunol 279, 245-257.

Page 173: Phospholipase D Signaling in T Cells

156

Chen, J., and Fang, Y. (2002). A novel pathway regulating the mammalian target of rapamycin (mTOR) signaling. Biochem Pharmacol 64, 1071-1077.

Chen, W., Jin, W., Hardegen, N., Lei, K.J., Li, L., Marinos, N., McGrady, G., and Wahl, S.M. (2003). Conversion of peripheral CD4+CD25- naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. The Journal of experimental medicine 198, 1875-1886.

Chess, L. (2006). The birth of functionally distinct T cell subsets. J Immunol 176, 3859-3860.

Claman, H.N., and Chaperon, E.A. (1969). Immunologic complementation between thymus and marrow cells--a model for the two-cell theory of immunocompetence. Transplantation reviews 1, 92-113.

Cockcroft, S., Way, G., O'Luanaigh, N., Pardo, R., Sarri, E., and Fensome, A. (2002). Signalling role for ARF and phospholipase D in mast cell exocytosis stimulated by crosslinking of the high affinity FcepsilonR1 receptor. Mol Immunol 38, 1277-1282.

Colley, W.C., Sung, T.C., Roll, R., Jenco, J., Hammond, S.M., Altshuller, Y., Bar-Sagi, D., Morris, A.J., and Frohman, M.A. (1997). Phospholipase D2, a distinct phospholipase D isoform with novel regulatory properties that provokes cytoskeletal reorganization. Curr Biol 7, 191-201.

Constant, S., Pfeiffer, C., Woodard, A., Pasqualini, T., and Bottomly, K. (1995). Extent of T cell receptor ligation can determine the functional differentiation of naive CD4+ T cells. The Journal of experimental medicine 182, 1591-1596.

Cook, R.T. (1998). Alcohol abuse, alcoholism, and damage to the immune system--a review. Alcoholism, clinical and experimental research 22, 1927-1942.

Page 174: Phospholipase D Signaling in T Cells

157

Cortese, M.M., Wolff, M., Almeido-Hill, J., Reid, R., Ketcham, J., and Santosham, M. (1992). High incidence rates of invasive pneumococcal disease in the White Mountain Apache population. Arch Intern Med 152, 2277-2282.

Coutinho-Silva, R., Stahl, L., Raymond, M.N., Jungas, T., Verbeke, P., Burnstock, G., Darville, T., and Ojcius, D.M. (2003). Inhibition of chlamydial infectious activity due to P2X7R-dependent phospholipase D activation. Immunity 19, 403-412.

Cronstein, B.N., Kramer, S.B., Weissmann, G., and Hirschhorn, R. (1983). Adenosine: a physiological modulator of superoxide anion generation by human neutrophils. The Journal of experimental medicine 158, 1160-1177.

Cua, D.J., Sherlock, J., Chen, Y., Murphy, C.A., Joyce, B., Seymour, B., Lucian, L., To, W., Kwan, S., Churakova, T., et al. (2003). Interleukin-23 rather than interleukin-12 is the critical cytokine for autoimmune inflammation of the brain. Nature 421, 744-748.

Cummings, R., Parinandi, N., Wang, L., Usatyuk, P., and Natarajan, V. (2002). Phospholipase D/phosphatidic acid signal transduction: role and physiological significance in lung. Mol Cell Biochem 234-235, 99-109.

Deaglio, S., Dwyer, K.M., Gao, W., Friedman, D., Usheva, A., Erat, A., Chen, J.F., Enjyoji, K., Linden, J., Oukka, M., et al. (2007). Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. The Journal of experimental medicine 204, 1257-1265.

Degos, F. (1999). Hepatitis C and alcohol. J Hepatol 31 Suppl 1, 113-118.

Dejaco, C., Duftner, C., Grubeck-Loebenstein, B., and Schirmer, M. (2006). Imbalance of regulatory T cells in human autoimmune diseases. Immunology 117, 289-300.

Page 175: Phospholipase D Signaling in T Cells

158

Delgoffe, G.M., Kole, T.P., Zheng, Y., Zarek, P.E., Matthews, K.L., Xiao, B., Worley, P.F., Kozma, S.C., and Powell, J.D. (2009). The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity 30, 832-844.

Delon, C., Manifava, M., Wood, E., Thompson, D., Krugmann, S., Pyne, S., and Ktistakis, N.T. (2004). Sphingosine kinase 1 is an intracellular effector of phosphatidic acid. J Biol Chem 279, 44763-44774.

Dittmer, U., He, H., Messer, R.J., Schimmer, S., Olbrich, A.R., Ohlen, C., Greenberg, P.D., Stromnes, I.M., Iwashiro, M., Sakaguchi, S., et al. (2004). Functional impairment of CD8(+) T cells by regulatory T cells during persistent retroviral infection. Immunity 20, 293-303.

Dragone, L.L., Myers, M.D., White, C., Gadwal, S., Sosinowski, T., Gu, H., and Weiss, A. (2006). Src-like adaptor protein (SLAP) regulates B cell receptor levels in a c-Cbl-dependent manner. Proceedings of the National Academy of Sciences of the United States of America 103, 18202-18207.

Du, G., Altshuller, Y.M., Kim, Y., Han, J.M., Ryu, S.H., Morris, A.J., and Frohman, M.A. (2000). Dual requirement for rho and protein kinase C in direct activation of phospholipase D1 through G protein-coupled receptor signaling. Molecular biology of the cell 11, 4359-4368.

Eberl, G. (2010). A new vision of immunity: homeostasis of the superorganism. Mucosal immunology.

El Bawab, S., Macovschi, O., Sette, C., Conti, M., Lagarde, M., Nemoz, G., and Prigent, A.F. (1997). Selective stimulation of a cAMP-specific phosphodiesterase (PDE4A5) isoform by phosphatidic acid molecular species endogenously formed in rat thymocytes. Eur J Biochem 247, 1151-1157.

English, D. (1996). Phosphatidic acid: a lipid messenger involved in intracellular and extracellular signalling. Cell Signal 8, 341-347.

Page 176: Phospholipase D Signaling in T Cells

159

Ermann, J., Hoffmann, P., Edinger, M., Dutt, S., Blankenberg, F.G., Higgins, J.P., Negrin, R.S., Fathman, C.G., and Strober, S. (2005). Only the CD62L+ subpopulation of CD4+CD25+ regulatory T cells protects from lethal acute GVHD. Blood 105, 2220-2226.

Esposito, A.L. (1984). Community-acquired bacteremic pneumococcal pneumonia. Effect of age on manifestations and outcome. Arch Intern Med 144, 945-948.

Exton, J.H. (2002). Phospholipase D-structure, regulation and function. Reviews of physiology, biochemistry and pharmacology 144, 1-94.

Fang, Y., Park, I.H., Wu, A.L., Du, G., Huang, P., Frohman, M.A., Walker, S.J., Brown, H.A., and Chen, J. (2003). PLD1 regulates mTOR signaling and mediates Cdc42 activation of S6K1. Curr Biol 13, 2037-2044.

Fang, Y., Vilella-Bach, M., Bachmann, R., Flanigan, A., and Chen, J. (2001). Phosphatidic acid-mediated mitogenic activation of mTOR signaling. Science (New York, N.Y 294, 1942-1945.

Farese, R.V. (1988). Phospholipid signaling systems in insulin action. The American journal of medicine 85, 36-43.

Fathman, C.G., and Hengartner, H. (1978). Clones of alloreactive T cells. Nature 272, 617-618.

Fehervari, Z., and Sakaguchi, S. (2004). Development and function of CD25+CD4+ regulatory T cells. Current opinion in immunology 16, 203-208.

Fernandez-Botran, R., Sanders, V.M., Mosmann, T.R., and Vitetta, E.S. (1988). Lymphokine-mediated regulation of the proliferative response of clones of T helper 1 and T helper 2 cells. The Journal of experimental medicine 168, 543-558.

Page 177: Phospholipase D Signaling in T Cells

160

Fernandez-Sola, J., Junque, A., Estruch, R., Monforte, R., Torres, A., and Urbano-Marquez, A. (1995). High alcohol intake as a risk and prognostic factor for community-acquired pneumonia. Arch Intern Med 155, 1649-1654.

Fiorentini, C., Falzano, L., Travaglione, S., and Fabbri, A. (2003). Hijacking Rho GTPases by protein toxins and apoptosis: molecular strategies of pathogenic bacteria. Cell death and differentiation 10, 147-152.

Firestein, G.S., Roeder, W.D., Laxer, J.A., Townsend, K.S., Weaver, C.T., Hom, J.T., Linton, J., Torbett, B.E., and Glasebrook, A.L. (1989). A new murine CD4+ T cell subset with an unrestricted cytokine profile. J Immunol 143, 518-525.

Fontenot, J.D., Gavin, M.A., and Rudensky, A.Y. (2003). Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nature immunology 4, 330-336.

Fontenot, J.D., Rasmussen, J.P., Gavin, M.A., and Rudensky, A.Y. (2005). A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nature immunology 6, 1142-1151.

Forssell, J., Nilsson, A., and Sideras, P. (2000). Reduced formation of phosphatidic acid upon B-cell receptor triggering of mouse B-lymphocytes lacking Bruton's tyrosine kinase. Scand J Immunol 52, 30-38.

Frank, C., Keilhack, H., Opitz, F., Zschornig, O., and Bohmer, F.D. (1999). Binding of phosphatidic acid to the protein-tyrosine phosphatase SHP-1 as a basis for activity modulation. Biochemistry 38, 11993-12002.

Fredholm, B.B., AP, I.J., Jacobson, K.A., Klotz, K.N., and Linden, J. (2001). International Union of Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacological reviews 53, 527-552.

Page 178: Phospholipase D Signaling in T Cells

161

Freyberg, Z., Sweeney, D., Siddhanta, A., Bourgoin, S., Frohman, M., and Shields, D. (2001). Intracellular localization of phospholipase D1 in mammalian cells. Molecular biology of the cell 12, 943-955.

Friedman, L.N., Sullivan, G.M., Bevilaqua, R.P., and Loscos, R. (1987). Tuberculosis screening in alcoholics and drug addicts. Am Rev Respir Dis 136, 1188-1192.

Frohman, M.A., Sung, T.C., and Morris, A.J. (1999). Mammalian phospholipase D structure and regulation. Biochimica et biophysica acta 1439, 175-186.

Gajewski, T.F., and Fitch, F.W. (1988). Anti-proliferative effect of IFN-gamma in immune regulation. I. IFN-gamma inhibits the proliferation of Th2 but not Th1 murine helper T lymphocyte clones. J Immunol 140, 4245-4252.

Gambineri, E., Torgerson, T.R., and Ochs, H.D. (2003). Immune dysregulation, polyendocrinopathy, enteropathy, and X-linked inheritance (IPEX), a syndrome of systemic autoimmunity caused by mutations of FOXP3, a critical regulator of T-cell homeostasis. Current opinion in rheumatology 15, 430-435.

Gavin, M.A., Clarke, S.R., Negrou, E., Gallegos, A., and Rudensky, A. (2002). Homeostasis and anergy of CD4(+)CD25(+) suppressor T cells in vivo. Nature immunology 3, 33-41.

Genth, H., Dreger, S.C., Huelsenbeck, J., and Just, I. (2008). Clostridium difficile toxins: more than mere inhibitors of Rho proteins. The international journal of biochemistry & cell biology 40, 592-597.

Gershon, R.K., and Kondo, K. (1970). Cell interactions in the induction of tolerance: the role of thymic lymphocytes. Immunology 18, 723-737.

Gessi, S., Cattabriga, E., Avitabile, A., Gafa, R., Lanza, G., Cavazzini, L., Bianchi, N., Gambari, R., Feo, C., Liboni, A., et al. (2004a). Elevated expression of A3 adenosine receptors in human colorectal cancer is reflected in peripheral blood cells. Clin Cancer Res 10, 5895-5901.

Page 179: Phospholipase D Signaling in T Cells

162

Gessi, S., Merighi, S., Varani, K., Leung, E., Mac Lennan, S., and Borea, P.A. (2008). The A3 adenosine receptor: an enigmatic player in cell biology. Pharmacology & therapeutics 117, 123-140.

Gessi, S., Varani, K., Merighi, S., Cattabriga, E., Avitabile, A., Gavioli, R., Fortini, C., Leung, E., Mac Lennan, S., and Borea, P.A. (2004b). Expression of A3 adenosine receptors in human lymphocytes: up-regulation in T cell activation. Molecular pharmacology 65, 711-719.

Gewirtz, A.T., and Simons, E.R. (1997). Phospholipase D mediates Fc gamma receptor activation of neutrophils and provides specificity between high-valency immune complexes and fMLP signaling pathways. J Leukoc Biol 61, 522-528.

Ghosh, S., and Bell, R.M. (1997). Regulation of Raf-1 kinase by interaction with the lipid second messenger, phosphatidic acid. Biochem Soc Trans 25, 561-565.

Ghosh, S., Strum, J.C., Sciorra, V.A., Daniel, L., and Bell, R.M. (1996). Raf-1 kinase possesses distinct binding domains for phosphatidylserine and phosphatidic acid. Phosphatidic acid regulates the translocation of Raf-1 in 12-O-tetradecanoylphorbol-13-acetate-stimulated Madin-Darby canine kidney cells. J Biol Chem 271, 8472-8480.

Gidwani, A., Brown, H.A., Holowka, D., and Baird, B. (2003). Disruption of lipid order by short-chain ceramides correlates with inhibition of phospholipase D and downstream signaling by FcepsilonRI. Journal of cell science 116, 3177-3187.

Giel, J.L., Sorg, J.A., Sonenshein, A.L., and Zhu, J. (2010). Metabolism of bile salts in mice influences spore germination in Clostridium difficile. PloS one 5, e8740.

Golstein, P., Wigzell, H., Blomgren, H., and Svedmyr, E.A. (1972). Cells mediating specific in vitro cytotoxicity. II. Probable autonomy of thymus-processed lymphocytes (T cells) for the killing of allogeneic target cells. The Journal of experimental medicine 135, 890-906.

Goodnow, C.C. (2001). Pathways for self-tolerance and the treatment of autoimmune diseases. Lancet 357, 2115-2121.

Page 180: Phospholipase D Signaling in T Cells

163

Gordon, A.S., Collier, K., and Diamond, I. (1986). Ethanol regulation of adenosine receptor-stimulated cAMP levels in a clonal neural cell line: an in vitro model of cellular tolerance to ethanol. Proceedings of the National Academy of Sciences of the United States of America 83, 2105-2108.

Govaerts, A. (1960). Cellular antibodies in kidney homotransplantation. J Immunol 85, 516-522.

Gowans, J.L. (1996). The lymphocyte--a disgraceful gap in medical knowledge. Immunology today 17, 288-291.

Grange, M., Picq, M., Prigent, A.F., Lagarde, M., and Nemoz, G. (1998). Regulation of PDE-4 cAMP phosphodiesterases by phosphatidic acid. Cell Biochem Biophys 29, 1-17.

Grange, M., Sette, C., Cuomo, M., Conti, M., Lagarde, M., Prigent, A.F., and Nemoz, G. (2000). The cAMP-specific phosphodiesterase PDE4D3 is regulated by phosphatidic acid binding. Consequences for cAMP signaling pathway and characterization of a phosphatidic acid binding site. J Biol Chem 275, 33379-33387.

Gross, L. (1951). Pathogenic properties, and "vertical" transmission of the mouse leukemia agent. Proceedings of the Society for Experimental Biology and Medicine. Society for Experimental Biology and Medicine (New York, N.Y 78, 342-348.

Groux, H. (2003). Type 1 T-regulatory cells: their role in the control of immune responses. Transplantation 75, 8S-12S.

Groux, H., O'Garra, A., Bigler, M., Rouleau, M., Antonenko, S., de Vries, J.E., and Roncarolo, M.G. (1997). A CD4+ T-cell subset inhibits antigen-specific T-cell responses and prevents colitis. Nature 389, 737-742.

Page 181: Phospholipase D Signaling in T Cells

164

Gruchalla, R.S., Dinh, T.T., and Kennerly, D.A. (1990). An indirect pathway of receptor-mediated 1,2-diacylglycerol formation in mast cells. I. IgE receptor-mediated activation of phospholipase D. J Immunol 144, 2334-2342.

Gudermann, T., Hofmann, T., Mederos y Schnitzler, M., and Dietrich, A. (2004). Activation, subunit composition and physiological relevance of DAG-sensitive TRPC proteins. Novartis Found Symp 258, 103-118; discussion 118-122, 155-109, 263-106.

Guy, C.S., and Vignali, D.A. (2009). Organization of proximal signal initiation at the TCR:CD3 complex. Immunological reviews 232, 7-21.

Haeffner, E.W. (1993). Diacylglycerol: formation and function in phospholipid-mediated signal transduction. Comp Biochem Physiol C 105, 337-345.

Hammond, S.M., Jenco, J.M., Nakashima, S., Cadwallader, K., Gu, Q., Cook, S., Nozawa, Y., Prestwich, G.D., Frohman, M.A., and Morris, A.J. (1997). Characterization of two alternately spliced forms of phospholipase D1. Activation of the purified enzymes by phosphatidylinositol 4,5-bisphosphate, ADP-ribosylation factor, and Rho family monomeric GTP-binding proteins and protein kinase C-alpha. The Journal of biological chemistry 272, 3860-3868.

Harris, D.R., Gonin, R., Alter, H.J., Wright, E.C., Buskell, Z.J., Hollinger, F.B., and Seeff, L.B. (2001). The relationship of acute transfusion-associated hepatitis to the development of cirrhosis in the presence of alcohol abuse. Ann Intern Med 134, 120-124.

Hasko, G., and Cronstein, B.N. (2004). Adenosine: an endogenous regulator of innate immunity. Trends in immunology 25, 33-39.

Hasko, G., Linden, J., Cronstein, B., and Pacher, P. (2008). Adenosine receptors: therapeutic aspects for inflammatory and immune diseases. Nat Rev Drug Discov 7, 759-770.

Page 182: Phospholipase D Signaling in T Cells

165

Hayakawa, K., and Hardy, R.R. (1988). Murine CD4+ T cell subsets defined. The Journal of experimental medicine 168, 1825-1838.

Heinzel, F.P., Sadick, M.D., Holaday, B.J., Coffman, R.L., and Locksley, R.M. (1989). Reciprocal expression of interferon gamma or interleukin 4 during the resolution or progression of murine leishmaniasis. Evidence for expansion of distinct helper T cell subsets. The Journal of experimental medicine 169, 59-72.

Hekman, M., Hamm, H., Villar, A.V., Bader, B., Kuhlmann, J., Nickel, J., and Rapp, U.R. (2002). Associations of B- and C-Raf with cholesterol, phosphatidylserine, and lipid second messengers: preferential binding of Raf to artificial lipid rafts. The Journal of biological chemistry 277, 24090-24102.

Hirst, J.A., Beverley, P.C., Kisielow, P., Hoffmann, M.K., and Oettgen, H.F. (1975). Ly antigens: markers of T cell function on mouse spleen cells. J Immunol 115, 1555-1557.

Hitomi, T., Yanagi, S., Inatome, R., and Yamamura, H. (1999). Cross-linking of the B cell receptor induces activation of phospholipase D through Syk, Btk and phospholipase C-gamma2. FEBS Lett 445, 371-374.

Hodgkin, M.N., Clark, J.M., Rose, S., Saqib, K., and Wakelam, M.J. (1999). Characterization of the regulation of phospholipase D activity in the detergent-insoluble fraction of HL60 cells by protein kinase C and small G-proteins. The Biochemical journal 339 ( Pt 1), 87-93.

Hori, S., Nomura, T., and Sakaguchi, S. (2003). Control of regulatory T cell development by the transcription factor Foxp3. Science (New York, N.Y 299, 1057-1061.

Hosken, N.A., Shibuya, K., Heath, A.W., Murphy, K.M., and O'Garra, A. (1995). The effect of antigen dose on CD4+ T helper cell phenotype development in a T cell receptor-alpha beta-transgenic model. The Journal of experimental medicine 182, 1579-1584.

Page 183: Phospholipase D Signaling in T Cells

166

Houslay, M.D., and Adams, D.R. (2003). PDE4 cAMP phosphodiesterases: modular enzymes that orchestrate signalling cross-talk, desensitization and compartmentalization. Biochem J 370, 1-18.

Howard, J.G., Hale, C., and Chan-Liew, W.L. (1980). Immunological regulation of experimental cutaneous leishmaniasis. 1. Immunogenetic aspects of susceptibility to Leishmania tropica in mice. Parasite immunology 2, 303-314.

Howard, J.G., Hale, C., and Liew, F.Y. (1981). Immunological regulation of experimental cutaneous leishmaniasis. IV. Prophylactic effect of sublethal irradiation as a result of abrogation of suppressor T cell generation in mice genetically susceptible to Leishmania tropica. The Journal of experimental medicine 153, 557-568.

Hsieh, C.S., Heimberger, A.B., Gold, J.S., O'Garra, A., and Murphy, K.M. (1992). Differential regulation of T helper phenotype development by interleukins 4 and 10 in an alpha beta T-cell-receptor transgenic system. Proceedings of the National Academy of Sciences of the United States of America 89, 6065-6069.

Huang, W., Na, L., Fidel, P.L., and Schwarzenberger, P. (2004). Requirement of interleukin-17A for systemic anti-Candida albicans host defense in mice. The Journal of infectious diseases 190, 624-631.

Huber, B., Cantor, H., Shen, F.W., and Boyse, E.A. (1976). Independent differentiative pathways of Ly1 and Ly23 subclasses of T cells. Experimental production of mice deprived of selected T-cell subclasses. The Journal of experimental medicine 144, 1128-1133.

Hynie, S., Lanefelt, F., and Fredholm, B.B. (1980). Effects of ethanol on human lymphocyte levels of cyclic AMP. In vitro: Potentiation of the response to isoproterenol, prostaglandin E2 or adenosine stimulation. Acta pharmacologica et toxicologica 47, 58-65.

Page 184: Phospholipase D Signaling in T Cells

167

Ishii, N., Nagy, Z.A., and Klein, J. (1982). Absence of Ir gene control of T cells recognizing foreign antigen in the context of allogenic MHC molecules. Nature 295, 531-533.

Ivanov, II, Frutos Rde, L., Manel, N., Yoshinaga, K., Rifkin, D.B., Sartor, R.B., Finlay, B.B., and Littman, D.R. (2008). Specific microbiota direct the differentiation of IL-17-producing T-helper cells in the mucosa of the small intestine. Cell host & microbe 4, 337-349.

Iwashima, M. (2003). Kinetic perspectives of T cell antigen receptor signaling. A two-tier model for T cell full activation. Immunological reviews 191, 196-210.

Iwashiro, M., Messer, R.J., Peterson, K.E., Stromnes, I.M., Sugie, T., and Hasenkrug, K.J. (2001). Immunosuppression by CD4+ regulatory T cells induced by chronic retroviral infection. Proceedings of the National Academy of Sciences of the United States of America 98, 9226-9230.

Jacobson, K.A., and Gao, Z.G. (2006). Adenosine receptors as therapeutic targets. Nat Rev Drug Discov 5, 247-264.

Jacobson, N.G., Szabo, S.J., Weber-Nordt, R.M., Zhong, Z., Schreiber, R.D., Darnell, J.E., Jr., and Murphy, K.M. (1995). Interleukin 12 signaling in T helper type 1 (Th1) cells involves tyrosine phosphorylation of signal transducer and activator of transcription (Stat)3 and Stat4. The Journal of experimental medicine 181, 1755-1762.

Jetten, A.M. (2009). Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism. Nuclear receptor signaling 7, e003.

Jin, X., Shepherd, R.K., Duling, B.R., and Linden, J. (1997). Inosine binds to A3 adenosine receptors and stimulates mast cell degranulation. The Journal of clinical investigation 100, 2849-2857.

Page 185: Phospholipase D Signaling in T Cells

168

Jonuleit, H., and Schmitt, E. (2003). The regulatory T cell family: distinct subsets and their interrelations. J Immunol 171, 6323-6327.

Jose Lopez-Andreo, M., Gomez-Fernandez, J.C., and Corbalan-Garcia, S. (2003). The simultaneous production of phosphatidic acid and diacylglycerol is essential for the translocation of protein kinase Cepsilon to the plasma membrane in RBL-2H3 cells. Mol Biol Cell 14, 4885-4895.

Kang, Y., Xu, L., Wang, B., Chen, A., and Zheng, G. (2008). Cutting edge: Immunosuppressant as adjuvant for tolerogenic immunization. J Immunol 180, 5172-5176.

Khan, K.N., and Yatsuhashi, H. (2000). Effect of alcohol consumption on the progression of hepatitis C virus infection and risk of hepatocellular carcinoma in Japanese patients. Alcohol Alcohol 35, 286-295.

Khattri, R., Cox, T., Yasayko, S.A., and Ramsdell, F. (2003). An essential role for Scurfin in CD4+CD25+ T regulatory cells. Nature immunology 4, 337-342.

Kikkawa, U., Kishimoto, A., and Nishizuka, Y. (1989). The protein kinase C family: heterogeneity and its implications. Annu Rev Biochem 58, 31-44.

Kim, J.M., Rasmussen, J.P., and Rudensky, A.Y. (2007). Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nature immunology 8, 191-197.

Kisielow, P., Bluthmann, H., Staerz, U.D., Steinmetz, M., and von Boehmer, H. (1988). Tolerance in T-cell-receptor transgenic mice involves deletion of nonmature CD4+8+ thymocytes. Nature 333, 742-746.

Knoechel, B., Lohr, J., Kahn, E., Bluestone, J.A., and Abbas, A.K. (2005). Sequential development of interleukin 2-dependent effector and regulatory T cells in response to endogenous systemic antigen. The Journal of experimental medicine 202, 1375-1386.

Page 186: Phospholipase D Signaling in T Cells

169

Kobie, J.J., Shah, P.R., Yang, L., Rebhahn, J.A., Fowell, D.J., and Mosmann, T.R. (2006). T regulatory and primed uncommitted CD4 T cells express CD73, which suppresses effector CD4 T cells by converting 5'-adenosine monophosphate to adenosine. J Immunol 177, 6780-6786.

Koch, W.J., Hawes, B.E., Inglese, J., Luttrell, L.M., and Lefkowitz, R.J. (1994). Cellular expression of the carboxyl terminus of a G protein-coupled receptor kinase attenuates G beta gamma-mediated signaling. The Journal of biological chemistry 269, 6193-6197.

Korn, T., Bettelli, E., Oukka, M., and Kuchroo, V.K. (2009). IL-17 and Th17 Cells. Annual review of immunology 27, 485-517.

Korn, T., Reddy, J., Gao, W., Bettelli, E., Awasthi, A., Petersen, T.R., Backstrom, B.T., Sobel, R.A., Wucherpfennig, K.W., Strom, T.B., et al. (2007). Myelin-specific regulatory T cells accumulate in the CNS but fail to control autoimmune inflammation. Nature medicine 13, 423-431.

Kronenberg, M., Steinmetz, M., Kobori, J., Kraig, E., Kapp, J.A., Pierce, C.W., Sorensen, C.M., Suzuki, G., Tada, T., and Hood, L. (1983). RNA transcripts for I-J polypeptides are apparently not encoded between the I-A and I-E subregions of the murine major histocompatibility complex. Proceedings of the National Academy of Sciences of the United States of America 80, 5704-5708.

Ktistakis, N.T., Delon, C., Manifava, M., Wood, E., Ganley, I., and Sugars, J.M. (2003). Phospholipase D1 and potential targets of its hydrolysis product, phosphatidic acid. Biochemical Society transactions 31, 94-97.

Kusner, D.J., Hall, C.F., and Jackson, S. (1999). Fc gamma receptor-mediated activation of phospholipase D regulates macrophage phagocytosis of IgG-opsonized particles. J Immunol 162, 2266-2274.

LaLonde, M., Janssens, H., Yun, S., Crosby, J., Redina, O., Olive, V., Altshuller, Y.M., Choi, S.Y., Du, G., Gergen, J.P., and Frohman, M.A. (2006). A role for Phospholipase D in Drosophila embryonic cellularization. BMC developmental biology 6, 60.

Page 187: Phospholipase D Signaling in T Cells

170

Lan, R.Y., Ansari, A.A., Lian, Z.X., and Gershwin, M.E. (2005). Regulatory T cells: development, function and role in autoimmunity. Autoimmunity reviews 4, 351-363.

Langrish, C.L., Chen, Y., Blumenschein, W.M., Mattson, J., Basham, B., Sedgwick, J.D., McClanahan, T., Kastelein, R.A., and Cua, D.J. (2005). IL-23 drives a pathogenic T cell population that induces autoimmune inflammation. The Journal of experimental medicine 201, 233-240.

Lappas, C.M., Rieger, J.M., and Linden, J. (2005). A2A adenosine receptor induction inhibits IFN-gamma production in murine CD4+ T cells. J Immunol 174, 1073-1080.

Laso, F.J., Madruga, J.I., Lopez, A., Ciudad, J., Alvarez-Mon, M., San Miguel, J., and Orfao, A. (1996a). Distribution of peripheral blood lymphoid subsets in alcoholic liver cirrhosis: influence of ethanol intake. Alcohol Clin Exp Res 20, 1564-1568.

Laso, F.J., Madruga, J.I., San Miguel, J.F., Ciudad, J., Lopez, A., Alvarez Mon, M., and Orfao, A. (1996b). Long lasting immunological effects of ethanol after withdrawal. Cytometry 26, 275-280.

Lassegue, B., Alexander, R.W., Clark, M., Akers, M., and Griendling, K.K. (1993). Phosphatidylcholine is a major source of phosphatidic acid and diacylglycerol in angiotensin II-stimulated vascular smooth-muscle cells. Biochem J 292 ( Pt 2), 509-517.

Le Stunff, H., Dokhac, L., and Harbon, S. (2000). The roles of protein kinase C and tyrosine kinases in mediating endothelin-1-stimulated phospholipase D activity in rat myometrium: differential inhibition by ceramides and cyclic AMP. The Journal of pharmacology and experimental therapeutics 292, 629-637.

Lee, H.Y., Park, J.B., Jang, I.H., Chae, Y.C., Kim, J.H., Kim, I.S., Suh, P.G., and Ryu, S.H. (2004). Munc-18-1 inhibits phospholipase D activity by direct interaction in an epidermal growth factor-reversible manner. The Journal of biological chemistry 279, 16339-16348.

Page 188: Phospholipase D Signaling in T Cells

171

Lee, Y.K., Turner, H., Maynard, C.L., Oliver, J.R., Chen, D., Elson, C.O., and Weaver, C.T. (2009). Late developmental plasticity in the T helper 17 lineage. Immunity 30, 92-107.

Levings, M.K., Bacchetta, R., Schulz, U., and Roncarolo, M.G. (2002). The role of IL-10 and TGF-beta in the differentiation and effector function of T regulatory cells. International archives of allergy and immunology 129, 263-276.

Levy, B.D., Fokin, V.V., Clark, J.M., Wakelam, M.J., Petasis, N.A., and Serhan, C.N. (1999). Polyisoprenyl phosphate (PIPP) signaling regulates phospholipase D activity: a 'stop' signaling switch for aspirin-triggered lipoxin A4. Faseb J 13, 903-911.

Lieber, C.S. (1997). Ethanol metabolism, cirrhosis and alcoholism. Clinica chimica acta; international journal of clinical chemistry 257, 59-84.

Liew, F.Y. (2002). T(H)1 and T(H)2 cells: a historical perspective. Nature reviews 2, 55-60.

Liew, F.Y., and Parish, C.R. (1974). Lack of a correlation between cell-mediated immunity to the carrier and the carrier-hapten helper effect. The Journal of experimental medicine 139, 779-784.

Lim, H.K., Choi, Y.A., Park, W., Lee, T., Ryu, S.H., Kim, S.Y., Kim, J.R., Kim, J.H., and Baek, S.H. (2003). Phosphatidic acid regulates systemic inflammatory responses by modulating the Akt-mammalian target of rapamycin-p70 S6 kinase 1 pathway. The Journal of biological chemistry 278, 45117-45127.

Lima, M.P., Pedro, R.J., and Rocha, M.D. (2000). Prevalence and risk factors for hepatitis C virus (HCV) infection among pregnant Brazilian women. Int J Gynaecol Obstet 70, 319-326.

Page 189: Phospholipase D Signaling in T Cells

172

Limatola, C., Schaap, D., Moolenaar, W.H., and van Blitterswijk, W.J. (1994). Phosphatidic acid activation of protein kinase C-zeta overexpressed in COS cells: comparison with other protein kinase C isotypes and other acidic lipids. Biochem J 304 ( Pt 3), 1001-1008.

Linden, J. (2001). Molecular approach to adenosine receptors: receptor-mediated mechanisms of tissue protection. Annual review of pharmacology and toxicology 41, 775-787.

Littman, D.R., and Rudensky, A.Y. (2010). Th17 and regulatory T cells in mediating and restraining inflammation. Cell 140, 845-858.

Liu, G.L., Shaw, L., Heagerty, A.M., Ohanian, V., and Ohanian, J. (1999). Endothelin-1 stimulates hydrolysis of phosphatidylcholine by phospholipases C and D in intact rat mesenteric arteries. J Vasc Res 36, 35-46.

Liu, M.Y., Gutowski, S., and Sternweis, P.C. (2001). The C terminus of mammalian phospholipase D is required for catalytic activity. The Journal of biological chemistry 276, 5556-5562.

Livy, D.J., Parnell, S.E., and West, J.R. (2003). Blood ethanol concentration profiles: a comparison between rats and mice. Alcohol (Fayetteville, N.Y 29, 165-171.

Loegering, D.J., and Lennartz, M.R. (2004). Signaling pathways for Fc gamma receptor-stimulated tumor necrosis factor-alpha secretion and respiratory burst in RAW 264.7 macrophages. Inflammation 28, 23-31.

Malcolm, K.C., Elliott, C.M., and Exton, J.H. (1996). Evidence for Rho-mediated agonist stimulation of phospholipase D in rat1 fibroblasts. Effects of Clostridium botulinum C3 exoenzyme. The Journal of biological chemistry 271, 13135-13139.

Page 190: Phospholipase D Signaling in T Cells

173

Mamoon, A.M., Smith, J., Baker, R.C., and Farley, J.M. (1999). Activation of protein kinase A increases phospholipase D activity and inhibits phospholipase D activation by acetylcholine in tracheal smooth muscle. The Journal of pharmacology and experimental therapeutics 291, 1188-1195.

Marrack, P., and Kappler, J. (1987). The T cell receptor. Science (New York, N.Y 238, 1073-1079.

Marrack, P.C., and Kappler, J.W. (1975). Antigen-specific and nonspecific mediators of T cell/B cell cooperation. I. Evidence for their production by different T cells. J Immunol 114, 1116-1125.

Martini, A., and Burgio, G.R. (1999). Tolerance and auto-immunity: 50 years after Burnet. European journal of pediatrics 158, 769-775.

Martins, T.C., and Aguas, A.P. (1999). A role for CD45RBlow CD38+ T cells and costimulatory pathways of T-cell activation in protection of non-obese diabetic (NOD) mice from diabetes. Immunology 96, 600-605.

Masopust, D., Vezys, V., Wherry, E.J., and Ahmed, R. (2007). A brief history of CD8 T cells. European journal of immunology 37 Suppl 1, S103-110.

McKeever, U., Mordes, J.P., Greiner, D.L., Appel, M.C., Rozing, J., Handler, E.S., and Rossini, A.A. (1990). Adoptive transfer of autoimmune diabetes and thyroiditis to athymic rats. Proceedings of the National Academy of Sciences of the United States of America 87, 7618-7622.

Meadows, G.G., Blank, S.E., and Duncan, D.D. (1989). Influence of ethanol consumption on natural killer cell activity in mice. Alcoholism, clinical and experimental research 13, 476-479.

Melendez, A.J., and Allen, J.M. (2002). Phospholipase D and immune receptor signalling. Seminars in immunology 14, 49-55.

Page 191: Phospholipase D Signaling in T Cells

174

Miller, J.F. (1961). Analysis of the thymus influence in leukaemogenesis. Nature 191, 248-249.

Miller, J.F. (1999). Discovering the origins of immunological competence. Annual review of immunology 17, 1-17.

Miller, J.F. (2002). The discovery of thymus function and of thymus-derived lymphocytes. Immunological reviews 185, 7-14.

Miller, J.F., and Mitchell, G.F. (1967). The thymus and the precursors of antigen reactive cells. Nature 216, 659-663.

Mills, K.H. (2004). Regulatory T cells: friend or foe in immunity to infection? Nature reviews 4, 841-855.

Mills, K.H., and McGuirk, P. (2004). Antigen-specific regulatory T cells--their induction and role in infection. Seminars in immunology 16, 107-117.

Milojevic, D., Nguyen, K.D., Wara, D., and Mellins, E.D. (2008). Regulatory T cells and their role in rheumatic diseases: a potential target for novel therapeutic development. Pediatric rheumatology online journal 6, 20.

Mitchell, G.F., and Miller, J.F. (1968). Immunological activity of thymus and thoracic-duct lymphocytes. Proceedings of the National Academy of Sciences of the United States of America 59, 296-303.

Mollinedo, F., Gajate, C., and Flores, I. (1994). Involvement of phospholipase D in the activation of transcription factor AP-1 in human T lymphoid Jurkat cells. J Immunol 153, 2457-2469.

Montesinos, M.C., Desai, A., Delano, D., Chen, J.F., Fink, J.S., Jacobson, M.A., and Cronstein, B.N. (2003). Adenosine A2A or A3 receptors are required for inhibition of inflammation by methotrexate and its analog MX-68. Arthritis and rheumatism 48, 240-247.

Page 192: Phospholipase D Signaling in T Cells

175

Morris, A.J., Frohman, M.A., and Engebrecht, J. (1997). Measurement of phospholipase D activity. Analytical biochemistry 252, 1-9.

Morrissey, P.J., Charrier, K., Braddy, S., Liggitt, D., and Watson, J.D. (1993). CD4+ T cells that express high levels of CD45RB induce wasting disease when transferred into congenic severe combined immunodeficient mice. Disease development is prevented by cotransfer of purified CD4+ T cells. The Journal of experimental medicine 178, 237-244.

Mosmann, T.R., Cherwinski, H., Bond, M.W., Giedlin, M.A., and Coffman, R.L. (1986). Two types of murine helper T cell clone. I. Definition according to profiles of lymphokine activities and secreted proteins. J Immunol 136, 2348-2357.

Moss, M., Bucher, B., Moore, F.A., Moore, E.E., and Parsons, P.E. (1996). The role of chronic alcohol abuse in the development of acute respiratory distress syndrome in adults. Jama 275, 50-54.

Mozzicato, S., Joshi, B.V., Jacobson, K.A., and Liang, B.T. (2004). Role of direct RhoA-phospholipase D1 interaction in mediating adenosine-induced protection from cardiac ischemia. Faseb J 18, 406-408.

Mudter, J., Wirtz, S., Galle, P.R., and Neurath, M.F. (2002). A new model of chronic colitis in SCID mice induced by adoptive transfer of CD62L+ CD4+ T cells: insights into the regulatory role of interleukin-6 on apoptosis. Pathobiology 70, 170-176.

Mullmann, T.J., Siegel, M.I., Egan, R.W., and Billah, M.M. (1990). Complement C5a activation of phospholipase D in human neutrophils. A major route to the production of phosphatidates and diglycerides. J Immunol 144, 1901-1908.

Murphy, C.A., Langrish, C.L., Chen, Y., Blumenschein, W., McClanahan, T., Kastelein, R.A., Sedgwick, J.D., and Cua, D.J. (2003). Divergent pro- and antiinflammatory roles for IL-23 and IL-12 in joint autoimmune inflammation. The Journal of experimental medicine 198, 1951-1957.

Page 193: Phospholipase D Signaling in T Cells

176

Naganuma, M., Wiznerowicz, E.B., Lappas, C.M., Linden, J., Worthington, M.T., and Ernst, P.B. (2006). Cutting edge: Critical role for A2A adenosine receptors in the T cell-mediated regulation of colitis. J Immunol 177, 2765-2769.

Nakamura, Y., Nakashima, S., Ojio, K., Banno, Y., Miyata, H., and Nozawa, Y. (1996). Ceramide inhibits IgE-mediated activation of phospholipase D, but not of phospholipase C, in rat basophilic leukemia (RBL-2H3) cells. J Immunol 156, 256-262.

Nakayama, T., and Yamashita, M. (2010). The TCR-mediated signaling pathways that control the direction of helper T cell differentiation. Seminars in immunology.

Nelson, S., and Kolls, J.K. (2002). Alcohol, host defence and society. Nature reviews 2, 205-209.

Nishizuka, Y. (1992). Intracellular signaling by hydrolysis of phospholipids and activation of protein kinase C. Science 258, 607-614.

Northrop, J.P., Ullman, K.S., and Crabtree, G.R. (1993). Characterization of the nuclear and cytoplasmic components of the lymphoid-specific nuclear factor of activated T cells (NF-AT) complex. The Journal of biological chemistry 268, 2917-2923.

O'Shea, J.J., and Paul, W.E. (2010). Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science (New York, N.Y 327, 1098-1102.

Obradovic, T., and Meadows, G.G. (2002). Chronic ethanol consumption increases plasma leptin levels and alters leptin receptors in the hypothalamus and the perigonadal fat of C57BL/6 mice. Alcoholism, clinical and experimental research 26, 255-262.

Page 194: Phospholipase D Signaling in T Cells

177

Onizuka, S., Tawara, I., Shimizu, J., Sakaguchi, S., Fujita, T., and Nakayama, E. (1999). Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor alpha) monoclonal antibody. Cancer research 59, 3128-3133.

Ottlinger, M.E., and Lin, S. (1988). Clostridium difficile toxin B induces reorganization of actin, vinculin, and talin in cultured cells. Experimental cell research 174, 215-229.

Oude Weernink, P.A., Lopez de Jesus, M., and Schmidt, M. (2007). Phospholipase D signaling: orchestration by PIP2 and small GTPases. Naunyn-Schmiedeberg's archives of pharmacology 374, 399-411.

Owen, R.D. (1945). Immunogenetic Consequences of Vascular Anastomoses between Bovine Twins. Science (New York, N.Y 102, 400-401.

Pandiyan, P., Zheng, L., Ishihara, S., Reed, J., and Lenardo, M.J. (2007). CD4+CD25+Foxp3+ regulatory T cells induce cytokine deprivation-mediated apoptosis of effector CD4+ T cells. Nature immunology 8, 1353-1362.

Panther, E., Corinti, S., Idzko, M., Herouy, Y., Napp, M., la Sala, A., Girolomoni, G., and Norgauer, J. (2003). Adenosine affects expression of membrane molecules, cytokine and chemokine release, and the T-cell stimulatory capacity of human dendritic cells. Blood 101, 3985-3990.

Parrott, D.V., De Sousa, M.A., and East, J. (1966). Thymus-dependent areas in the lymphoid organs of neonatally thymectomized mice. The Journal of experimental medicine 123, 191-204.

Penhale, W.J., Farmer, A., McKenna, R.P., and Irvine, W.J. (1973). Spontaneous thyroiditis in thymectomized and irradiated Wistar rats. Clinical and experimental immunology 15, 225-236.

Page 195: Phospholipase D Signaling in T Cells

178

Plo, I., Lautier, D., Levade, T., Sekouri, H., Jaffrezou, J.P., Laurent, G., and Bettaieb, A. (2000). Phosphatidylcholine-specific phospholipase C and phospholipase D are respectively implicated in mitogen-activated protein kinase and nuclear factor kappaB activation in tumour-necrosis-factor-alpha-treated immature acute-myeloid-leukaemia cells. Biochem J 351 Pt 2, 459-467.

Powrie, F., Carlino, J., Leach, M.W., Mauze, S., and Coffman, R.L. (1996). A critical role for transforming growth factor-beta but not interleukin 4 in the suppression of T helper type 1-mediated colitis by CD45RB(low) CD4+ T cells. The Journal of experimental medicine 183, 2669-2674.

Powrie, F., Correa-Oliveira, R., Mauze, S., and Coffman, R.L. (1994). Regulatory interactions between CD45RBhigh and CD45RBlow CD4+ T cells are important for the balance between protective and pathogenic cell-mediated immunity. The Journal of experimental medicine 179, 589-600.

Powrie, F., Leach, M.W., Mauze, S., Caddle, L.B., and Coffman, R.L. (1993). Phenotypically distinct subsets of CD4+ T cells induce or protect from chronic intestinal inflammation in C. B-17 scid mice. International immunology 5, 1461-1471.

Preininger, A.M., Henage, L.G., Oldham, W.M., Yoon, E.J., Hamm, H.E., and Brown, H.A. (2006). Direct modulation of phospholipase D activity by Gbetagamma. Molecular pharmacology 70, 311-318.

Quintana, F.J., Basso, A.S., Iglesias, A.H., Korn, T., Farez, M.F., Bettelli, E., Caccamo, M., Oukka, M., and Weiner, H.L. (2008). Control of T(reg) and T(H)17 cell differentiation by the aryl hydrocarbon receptor. Nature 453, 65-71.

Raff, M.C. (2008). Theta isoantigen as a marker of thymus-derived lymphocytes in mice. 1969. J Immunol 180, 4353-4354.

Page 196: Phospholipase D Signaling in T Cells

179

Ramstad, C., Sundvold, V., Johansen, H.K., and Lea, T. (2000). cAMP-dependent protein kinase (PKA) inhibits T cell activation by phosphorylating ser-43 of raf-1 in the MAPK/ERK pathway. Cellular signalling 12, 557-563.

Rao, A., Allard, W.J., Hogan, P.G., Rosenson, R.S., and Cantor, H. (1983). Alloreactive T-cell clones. Ly phenotypes predict both function and specificity for major histocompatibility complex products. Immunogenetics 17, 147-165.

Read, S., Malmstrom, V., and Powrie, F. (2000). Cytotoxic T lymphocyte-associated antigen 4 plays an essential role in the function of CD25(+)CD4(+) regulatory cells that control intestinal inflammation. The Journal of experimental medicine 192, 295-302.

Read, S., Mauze, S., Asseman, C., Bean, A., Coffman, R., and Powrie, F. (1998). CD38+ CD45RB(low) CD4+ T cells: a population of T cells with immune regulatory activities in vitro. European journal of immunology 28, 3435-3447.

Redpath, S., Ghazal, P., and Gascoigne, N.R. (2001). Hijacking and exploitation of IL-10 by intracellular pathogens. Trends in microbiology 9, 86-92.

Reid, P.A., Gardner, S.D., Williams, D.M., and Harnett, M.M. (1997). The antigen receptors on mature and immature T lymphocytes are coupled to phosphatidylcholine-specific phospholipase D activation. Immunology 90, 250-256.

Rizzo, M., and Romero, G. (2002). Pharmacological importance of phospholipase D and phosphatidic acid in the regulation of the mitogen-activated protein kinase cascade. Pharmacol Ther 94, 35-50.

Rizzo, M.A., Shome, K., Vasudevan, C., Stolz, D.B., Sung, T.C., Frohman, M.A., Watkins, S.C., and Romero, G. (1999). Phospholipase D and its product, phosphatidic acid, mediate agonist-dependent raf-1 translocation to the plasma membrane and the activation of the mitogen-activated protein kinase pathway. J Biol Chem 274, 1131-1139.

Page 197: Phospholipase D Signaling in T Cells

180

Rizzo, M.A., Shome, K., Watkins, S.C., and Romero, G. (2000). The recruitment of Raf-1 to membranes is mediated by direct interaction with phosphatidic acid and is independent of association with Ras. J Biol Chem 275, 23911-23918.

Roncarolo, M.G., Levings, M.K., and Traversari, C. (2001). Differentiation of T regulatory cells by immature dendritic cells. The Journal of experimental medicine 193, F5-9.

Rouse, B.T., and Suvas, S. (2004). Regulatory cells and infectious agents: detentes cordiale and contraire. J Immunol 173, 2211-2215.

Ruiz, M., Ewig, S., Marcos, M.A., Martinez, J.A., Arancibia, F., Mensa, J., and Torres, A. (1999). Etiology of community-acquired pneumonia: impact of age, comorbidity, and severity. Am J Respir Crit Care Med 160, 397-405.

Sacanella, E., Estruch, R., Gaya, A., Fernandez-Sola, J., Antunez, E., and Urbano-Marquez, A. (1998). Activated lymphocytes (CD25+ CD69+ cells) and decreased CD19+ cells in well-nourished chronic alcoholics without ethanol-related diseases. Alcohol Clin Exp Res 22, 897-901.

Saito, S., Sasaki, Y., and Sakai, M. (2005). CD4(+)CD25high regulatory T cells in human pregnancy. Journal of reproductive immunology 65, 111-120.

Sakaguchi, S. (2004). Naturally arising CD4+ regulatory t cells for immunologic self-tolerance and negative control of immune responses. Annual review of immunology 22, 531-562.

Sakaguchi, S., Fukuma, K., Kuribayashi, K., and Masuda, T. (1985). Organ-specific autoimmune diseases induced in mice by elimination of T cell subset. I. Evidence for the active participation of T cells in natural self-tolerance; deficit of a T cell subset as a possible cause of autoimmune disease. The Journal of experimental medicine 161, 72-87.

Page 198: Phospholipase D Signaling in T Cells

181

Sakaguchi, S., Sakaguchi, N., Asano, M., Itoh, M., and Toda, M. (1995). Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 155, 1151-1164.

Sakaguchi, S., Wing, K., and Miyara, M. (2007). Regulatory T cells - a brief history and perspective. European journal of immunology 37 Suppl 1, S116-123.

Salvatore, C.A., Tilley, S.L., Latour, A.M., Fletcher, D.S., Koller, B.H., and Jacobson, M.A. (2000). Disruption of the A(3) adenosine receptor gene in mice and its effect on stimulated inflammatory cells. The Journal of biological chemistry 275, 4429-4434.

Santos-Perez, J.L., Diez-Ruiz, A., Luna-Casado, L., Soto-Mas, J.A., Wachter, H., Fuchs, D., and Gutierrez-Gea, F. (1996). T-cell activation, expression of adhesion molecules and response to ethanol in alcoholic cirrhosis. Immunol Lett 50, 179-183.

Savany, A., Abriat, C., Nemoz, G., Lagarde, M., and Prigent, A.F. (1996). Activation of a cyclic nucleotide phosphodiesterase 4 (PDE4) from rat thymocytes by phosphatidic acid. Cell Signal 8, 511-516.

Schiff, E.R. (1999). The alcoholic patient with hepatitis C virus infection. Am J Med 107, 95S-99S.

Schmidt, M., Rumenapp, U., Bienek, C., Keller, J., von Eichel-Streiber, C., and Jakobs, K.H. (1996). Inhibition of receptor signaling to phospholipase D by Clostridium difficile toxin B. Role of Rho proteins. The Journal of biological chemistry 271, 2422-2426.

Schnurr, M., Toy, T., Shin, A., Hartmann, G., Rothenfusser, S., Soellner, J., Davis, I.D., Cebon, J., and Maraskovsky, E. (2004). Role of adenosine receptors in regulating chemotaxis and cytokine production of plasmacytoid dendritic cells. Blood 103, 1391-1397.

Page 199: Phospholipase D Signaling in T Cells

182

Schubert, L.A., Jeffery, E., Zhang, Y., Ramsdell, F., and Ziegler, S.F. (2001). Scurfin (FOXP3) acts as a repressor of transcription and regulates T cell activation. The Journal of biological chemistry 276, 37672-37679.

Sciorra, V.A., Rudge, S.A., Prestwich, G.D., Frohman, M.A., Engebrecht, J., and Morris, A.J. (1999). Identification of a phosphoinositide binding motif that mediates activation of mammalian and yeast phospholipase D isoenzymes. The EMBO journal 18, 5911-5921.

Scott, P., Natovitz, P., Coffman, R.L., Pearce, E., and Sher, A. (1988). Immunoregulation of cutaneous leishmaniasis. T cell lines that transfer protective immunity or exacerbation belong to different T helper subsets and respond to distinct parasite antigens. The Journal of experimental medicine 168, 1675-1684.

Selvatici, R., Falzarano, S., Mollica, A., and Spisani, S. (2006). Signal transduction pathways triggered by selective formylpeptide analogues in human neutrophils. European journal of pharmacology 534, 1-11.

Sempere, J.M., Soriano, V., and Benito, J.M. (2007). T regulatory cells and HIV infection. AIDS reviews 9, 54-60.

Sette, A., Sidney, J., Gaeta, F.C., Appella, E., Colon, S.M., del Guercio, M.F., Guery, J.C., and Adorini, L. (1993). MHC class II molecules bind indiscriminately self and non-self peptide homologs: effect on the immunogenicity of non-self peptides. International immunology 5, 631-638.

Shao, H., Zhou, J., and Ewald, S.J. (1995). Regulation of signal transduction and DNA fragmentation in thymocytes by ethanol. Cellular immunology 164, 11-19.

Shimizu, J., Yamazaki, S., Takahashi, T., Ishida, Y., and Sakaguchi, S. (2002). Stimulation of CD25(+)CD4(+) regulatory T cells through GITR breaks immunological self-tolerance. Nature immunology 3, 135-142.

Page 200: Phospholipase D Signaling in T Cells

183

Sibley, D., and Jerrells, T.R. (2000). Alcohol consumption by C57BL/6 mice is associated with depletion of lymphoid cells from the gut-associated lymphoid tissues and altered resistance to oral infections with Salmonella typhimurium. The Journal of infectious diseases 182, 482-489.

Silverstein, A.M. (2001). Autoimmunity versus horror autotoxicus: the struggle for recognition. Nature immunology 2, 279-281.

Singh, N., Seki, Y., Takami, M., Baban, B., Chandler, P.R., Khosravi, D., Zheng, X., Takezaki, M., Lee, J.R., Mellor, A.L., et al. (2006). Enrichment of regulatory CD4(+)CD25(+) T cells by inhibition of phospholipase D signaling. Nature methods 3, 629-636.

Song, K., Coleman, R.A., Zhu, X., Alber, C., Ballas, Z.K., Waldschmidt, T.J., and Cook, R.T. (2002). Chronic ethanol consumption by mice results in activated splenic T cells. Journal of leukocyte biology 72, 1109-1116.

Steinman, L. (2007). A brief history of T(H)17, the first major revision in the T(H)1/T(H)2 hypothesis of T cell-mediated tissue damage. Nature medicine 13, 139-145.

Strauss, L., Bergmann, C., Gooding, W., Johnson, J.T., and Whiteside, T.L. (2007a). The frequency and suppressor function of CD4+CD25highFoxp3+ T cells in the circulation of patients with squamous cell carcinoma of the head and neck. Clin Cancer Res 13, 6301-6311.

Strauss, L., Bergmann, C., Szczepanski, M., Gooding, W., Johnson, J.T., and Whiteside, T.L. (2007b). A unique subset of CD4+CD25highFoxp3+ T cells secreting interleukin-10 and transforming growth factor-beta1 mediates suppression in the tumor microenvironment. Clin Cancer Res 13, 4345-4354.

Strauss, L., Whiteside, T.L., Knights, A., Bergmann, C., Knuth, A., and Zippelius, A. (2007c). Selective survival of naturally occurring human CD4+CD25+Foxp3+ regulatory T cells cultured with rapamycin. J Immunol 178, 320-329.

Page 201: Phospholipase D Signaling in T Cells

184

Sugars, J.M., Cellek, S., Manifava, M., Coadwell, J., and Ktistakis, N.T. (1999). Fatty acylation of phospholipase D1 on cysteine residues 240 and 241 determines localization on intracellular membranes. The Journal of biological chemistry 274, 30023-30027.

Sugars, J.M., Cellek, S., Manifava, M., Coadwell, J., and Ktistakis, N.T. (2002). Hierarchy of membrane-targeting signals of phospholipase D1 involving lipid modification of a pleckstrin homology domain. The Journal of biological chemistry 277, 29152-29161.

Sung, T.C., Roper, R.L., Zhang, Y., Rudge, S.A., Temel, R., Hammond, S.M., Morris, A.J., Moss, B., Engebrecht, J., and Frohman, M.A. (1997). Mutagenesis of phospholipase D defines a superfamily including a trans-Golgi viral protein required for poxvirus pathogenicity. The EMBO journal 16, 4519-4530.

Sung, T.C., Zhang, Y., Morris, A.J., and Frohman, M.A. (1999). Structural analysis of human phospholipase D1. The Journal of biological chemistry 274, 3659-3666.

Suvas, S., Azkur, A.K., Kim, B.S., Kumaraguru, U., and Rouse, B.T. (2004). CD4+CD25+ regulatory T cells control the severity of viral immunoinflammatory lesions. J Immunol 172, 4123-4132.

Suvas, S., Kumaraguru, U., Pack, C.D., Lee, S., and Rouse, B.T. (2003). CD4+CD25+ T cells regulate virus-specific primary and memory CD8+ T cell responses. The Journal of experimental medicine 198, 889-901.

Szabo, G., Verma, B., and Catalano, D. (1993). Selective inhibition of antigen-specific T lymphocyte proliferation by acute ethanol exposure: the role of impaired monocyte antigen presentation capacity and mediator production. Journal of leukocyte biology 54, 534-544.

Szabo, S.J., Kim, S.T., Costa, G.L., Zhang, X., Fathman, C.G., and Glimcher, L.H. (2000). A novel transcription factor, T-bet, directs Th1 lineage commitment. Cell 100, 655-669.

Page 202: Phospholipase D Signaling in T Cells

185

Taams, L.S., Smith, J., Rustin, M.H., Salmon, M., Poulter, L.W., and Akbar, A.N. (2001). Human anergic/suppressive CD4(+)CD25(+) T cells: a highly differentiated and apoptosis-prone population. European journal of immunology 31, 1122-1131.

Tada, T., Takemori, T., Okumura, K., Nonaka, M., and Tokuhisa, T. (1978). Two distinct types of helper T cells involved in the secondary antibody response: independent and synergistic effects of Ia- and Ia+ helper T cells. The Journal of experimental medicine 147, 446-458.

Takeda, K., Tanaka, T., Shi, W., Matsumoto, M., Minami, M., Kashiwamura, S., Nakanishi, K., Yoshida, N., Kishimoto, T., and Akira, S. (1996). Essential role of Stat6 in IL-4 signalling. Nature 380, 627-630.

Tamura, T., Ariga, H., Kinashi, T., Uehara, S., Kikuchi, T., Nakada, M., Tokunaga, T., Xu, W., Kariyone, A., Saito, T., et al. (2004). The role of antigenic peptide in CD4+ T helper phenotype development in a T cell receptor transgenic model. International immunology 16, 1691-1699.

Tan, S.L., Zhao, J., Bi, C., Chen, X.C., Hepburn, D.L., Wang, J., Sedgwick, J.D., Chintalacharuvu, S.R., and Na, S. (2006). Resistance to experimental autoimmune encephalomyelitis and impaired IL-17 production in protein kinase C theta-deficient mice. J Immunol 176, 2872-2879.

Tang, Q., Adams, J.Y., Penaranda, C., Melli, K., Piaggio, E., Sgouroudis, E., Piccirillo, C.A., Salomon, B.L., and Bluestone, J.A. (2008). Central role of defective interleukin-2 production in the triggering of islet autoimmune destruction. Immunity 28, 687-697.

Thibault, N., Burelout, C., Harbour, D., Borgeat, P., Naccache, P.H., and Bourgoin, S.G. (2002). Occupancy of adenosine A2a receptors promotes fMLP-induced cyclic AMP accumulation in human neutrophils: impact on phospholipase D activity and recruitment of small GTPases to membranes. Journal of leukocyte biology 71, 367-377.

Page 203: Phospholipase D Signaling in T Cells

186

Thibault, N., Harbour, D., Borgeat, P., Naccache, P.H., and Bourgoin, S.G. (2000). Adenosine receptor occupancy suppresses chemoattractant-induced phospholipase D activity by diminishing membrane recruitment of small GTPases. Blood 95, 519-527.

Thornton, A.M., and Shevach, E.M. (1998). CD4+CD25+ immunoregulatory T cells suppress polyclonal T cell activation in vitro by inhibiting interleukin 2 production. The Journal of experimental medicine 188, 287-296.

Toka, F.N., Suvas, S., and Rouse, B.T. (2004). CD4+ CD25+ T cells regulate vaccine-generated primary and memory CD8+ T-cell responses against herpes simplex virus type 1. Journal of virology 78, 13082-13089.

Tomic, S., Greiser, U., Lammers, R., Kharitonenkov, A., Imyanitov, E., Ullrich, A., and Bohmer, F.D. (1995). Association of SH2 domain protein tyrosine phosphatases with the epidermal growth factor receptor in human tumor cells. Phosphatidic acid activates receptor dephosphorylation by PTP1C. J Biol Chem 270, 21277-21284.

Veldhoen, M., Hirota, K., Westendorf, A.M., Buer, J., Dumoutier, L., Renauld, J.C., and Stockinger, B. (2008). The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 453, 106-109.

Vollaard, E.J., and Clasener, H.A. (1994). Colonization resistance. Antimicrobial agents and chemotherapy 38, 409-414.

Waldmann, T.A. (2006). Effective cancer therapy through immunomodulation. Annual review of medicine 57, 65-81.

Wan, Y.Y., and Flavell, R.A. (2009). How diverse--CD4 effector T cells and their functions. Journal of molecular cell biology 1, 20-36.

Page 204: Phospholipase D Signaling in T Cells

187

Wang, L., Cummings, R., Usatyuk, P., Morris, A., Irani, K., and Natarajan, V. (2002). Involvement of phospholipases D1 and D2 in sphingosine 1-phosphate-induced ERK (extracellular-signal-regulated kinase) activation and interleukin-8 secretion in human bronchial epithelial cells. Biochem J 367, 751-760.

Warner, N.L., Szenberg, A., and Burnet, F.M. (1962). The immunological role of different lymphoid organs in the chicken. I. Dissociation of immunological responsiveness. The Australian journal of experimental biology and medical science 40, 373-387.

Webster, K.E., Walters, S., Kohler, R.E., Mrkvan, T., Boyman, O., Surh, C.D., Grey, S.T., and Sprent, J. (2009). In vivo expansion of T reg cells with IL-2-mAb complexes: induction of resistance to EAE and long-term acceptance of islet allografts without immunosuppression. The Journal of experimental medicine 206, 751-760.

Weiner, H.L. (2001a). Induction and mechanism of action of transforming growth factor-beta-secreting Th3 regulatory cells. Immunological reviews 182, 207-214.

Weiner, H.L. (2001b). The mucosal milieu creates tolerogenic dendritic cells and T(R)1 and T(H)3 regulatory cells. Nature immunology 2, 671-672.

Weiss, L., Donkova-Petrini, V., Caccavelli, L., Balbo, M., Carbonneil, C., and Levy, Y. (2004). Human immunodeficiency virus-driven expansion of CD4+CD25+ regulatory T cells, which suppress HIV-specific CD4 T-cell responses in HIV-infected patients. Blood 104, 3249-3256.

Wildin, R.S., Ramsdell, F., Peake, J., Faravelli, F., Casanova, J.L., Buist, N., Levy-Lahad, E., Mazzella, M., Goulet, O., Perroni, L., et al. (2001). X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy. Nature genetics 27, 18-20.

Page 205: Phospholipase D Signaling in T Cells

188

Wing, K., and Sakaguchi, S. (2010). Regulatory T cells exert checks and balances on self tolerance and autoimmunity. Nature immunology 11, 7-13.

Winterbauer, R.H., Bedon, G.A., and Ball, W.C., Jr. (1969). Recurrent pneumonia. Predisposing illness and clinical patterns in 158 patients. Ann Intern Med 70, 689-700.

Wood, K.J., Bushell, A.R., and Jones, N.D. (2010). The discovery of immunological tolerance: now more than just a laboratory solution. J Immunol 184, 3-4.

Xie, Z., Ho, W.T., and Exton, J.H. (2000). Conserved amino acids at the C-terminus of rat phospholipase D1 are essential for enzymatic activity. European journal of biochemistry / FEBS 267, 7138-7146.

Xie, Z., Ho, W.T., and Exton, J.H. (2001). Requirements and effects of palmitoylation of rat PLD1. The Journal of biological chemistry 276, 9383-9391.

Yamashita, M., Hashimoto, K., Kimura, M., Kubo, M., Tada, T., and Nakayama, T. (1998). Requirement for p56(lck) tyrosine kinase activation in Th subset differentiation. International immunology 10, 577-591.

Yamashita, M., Kimura, M., Kubo, M., Shimizu, C., Tada, T., Perlmutter, R.M., and Nakayama, T. (1999). T cell antigen receptor-mediated activation of the Ras/mitogen-activated protein kinase pathway controls interleukin 4 receptor function and type-2 helper T cell differentiation. Proceedings of the National Academy of Sciences of the United States of America 96, 1024-1029.

Yang, C., and Kazanietz, M.G. (2003). Divergence and complexities in DAG signaling: looking beyond PKC. Trends Pharmacol Sci 24, 602-608.

Page 206: Phospholipase D Signaling in T Cells

189

Yao, L., Arolfo, M.P., Dohrman, D.P., Jiang, Z., Fan, P., Fuchs, S., Janak, P.H., Gordon, A.S., and Diamond, I. (2002). betagamma Dimers mediate synergy of dopamine D2 and adenosine A2 receptor-stimulated PKA signaling and regulate ethanol consumption. Cell 109, 733-743.

You, S., Slehoffer, G., Barriot, S., Bach, J.F., and Chatenoud, L. (2004). Unique role of CD4+CD62L+ regulatory T cells in the control of autoimmune diabetes in T cell receptor transgenic mice. Proceedings of the National Academy of Sciences of the United States of America 101 Suppl 2, 14580-14585.

Zarek, P.E., Huang, C.T., Lutz, E.R., Kowalski, J., Horton, M.R., Linden, J., Drake, C.G., and Powell, J.D. (2008). A2A receptor signaling promotes peripheral tolerance by inducing T-cell anergy and the generation of adaptive regulatory T cells. Blood 111, 251-259.

Zenclussen, A.C. (2005). CD4(+)CD25+ T regulatory cells in murine pregnancy. Journal of reproductive immunology 65, 101-110.

Zhang, H., and Meadows, G.G. (2005). Chronic alcohol consumption in mice increases the proportion of peripheral memory T cells by homeostatic proliferation. Journal of leukocyte biology 78, 1070-1080.

Zhao, Z., Shen, S.H., and Fischer, E.H. (1993). Stimulation by phospholipids of a protein-tyrosine-phosphatase containing two src homology 2 domains. Proceedings of the National Academy of Sciences of the United States of America 90, 4251-4255.

Zheng, W., and Flavell, R.A. (1997). The transcription factor GATA-3 is necessary and sufficient for Th2 cytokine gene expression in CD4 T cells. Cell 89, 587-596.

Zheng, X., Ray, S., and Bollag, W.B. (2003). Modulation of phospholipase D-mediated phosphatidylglycerol formation by differentiating agents in primary mouse epidermal keratinocytes. Biochimica et biophysica acta 1643, 25-36.

Page 207: Phospholipase D Signaling in T Cells

190

Zhou, X., Bailey-Bucktrout, S.L., Jeker, L.T., Penaranda, C., Martinez-Llordella, M., Ashby, M., Nakayama, M., Rosenthal, W., and Bluestone, J.A. (2009). Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nature immunology 10, 1000-1007.

Page 208: Phospholipase D Signaling in T Cells

VITA

The author, Uma Chandrasekaran was born in Chennai, India on March

13, 1983 to Mr. and Mrs.Chandrasekaran. She received a Bachelor of

Engineering in Electronics & Instrumentation and Masters in Biological Sciences

from Birla Institute of Technology & Sciences (BITS-Pilani), India in May 2005.

In August of 2005, Uma joined the graduate program in Biomedical

Sciences at Medical College of Georgia (Augusta, GA). Shortly, thereafter she

joined the laboratory of Dr.Makio Iwashima. Uma moved with Dr. Iwashima to

Loyola University, Chicago in Aug 2007. In Dr.Iwashima’s laboratory, Uma

studied mechanisms of regulation of Phospholipase D (PLD) signaling in T cells.

After completing her Ph.D., Uma will begin a post-doctoral position in Dr.

Alessandra Pernis’s laboratory at Weill Medical College of Cornell University

(New York, NY) where she will study the role of IL-17 producing T cells in the

pathogenesis of autoimmune disorders like Rheumatoid Arthritis and Lupus.

191

Page 209: Phospholipase D Signaling in T Cells