the immunogenetics of natural killer cell alloreactivity · the immunogenetics of natural killer...

186
I The Immunogenetics of Natural Killer Cell Alloreactivity Bree Amanda Foley BSc (Hons) This thesis is presented for the degree of Doctor of Philosophy of the University of Western Australia 2008 School of Pathology and Laboratory Medicine

Upload: others

Post on 26-Mar-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

I

The Immunogenetics of Natural Killer Cell

Alloreactivity

Bree Amanda Foley BSc (Hons)

This thesis is presented for the degree of Doctor of Philosophy of the University of

Western Australia

2008 School of Pathology and Laboratory Medicine

II

Statement of Candidate Contribution

All work was performed by the author unless otherwise stated in the thesis ……………………………….. Bree Foley Student ……………………………….. Campbell Witt Supervisor ……………………………….. Frank Christiansen Supervisor

III

Declaration from UWA website

IV

The data presented in chapter 4 has been submitted for publication. The bibliographic

details and percentage contribution of each author to the work are set out below

The Reactivity of Bw4-positive HLA-B and HLA-A Alleles with KIR3DL1: Implications

for Patient and Donor Suitability for Haploidentical Stem Cell Transplants.

Foley BA, De Santis D, van Beelen E, Lathbury LJ, Christiansen FT and Witt CS

Blood 2008 (In Press)

Author Signature

Bree Foley (76%)

Dianne De Santis (4%)

Els Van Beelen (4%)

Louise Lathbury (2%)

Frank Christiansen (4%)

Campbell Witt (10%)

V

The data presented in chapter 5 has been accepted for publication. The bibliographic

details and contribution of each author to the work are set out below.

KIR2DS1-Mediated Activation Overrides NKG2A-Mediated Inhibition in HLA-C C2

Negative Individuals. Foley B, De Santis D, Lathbury L, Christiansen F and Witt, C.

International Immunology Accepted 23rd January 2008 Author Signature

Bree Foley (80%)

Dianne De Santis (5%)

Louise Lathbury (5%)

Frank Christiansen (5%)

Campbell Witt (5%)

VI

Acknowledgments

First and foremost I would like to thank my two supervisors, Campbell Witt and Frank

Christiansen. Thank you for allowing me the opportunity to work on this project. I thank

you both for all of your guidance, advice, patience, wisdom, helpfulness and

encouragement over the past four years. I don’t think that there any words that can

truly describe how thankful I am for all of your efforts in helping me complete this PhD, I

will always be eternally grateful.

I would like to thank the three members of my lab group: Jodie Goodridge, Louise

Lathbury and Dianne De Santis. Jodie I thank you for tolerating my many questions,

always encouraging me and always being there when I needed help. Louise I thank

you for teaching me the ‘ins and outs’ of NK cell cloning and for all your guidance over

the past four years. Dianne I thank you for all of your help in the lab, I really

appreciated having someone else in the lab who knew exactly what studying NK cell

alloreactivity really involved.

A special thank you to Els van Beelen from the Department of Immunohematology and

Blood Transfusion at Leiden University Medical Center for all of your help in generating

the results presented in chapter 4.

Thank you to all the staff at the Department of Clinical Immunology at RPH for all your

help over the years. I thank you for performing class I HLA typing on the donors used in

this thesis and sequencing of KIR genes. I would especially like to thank Catena

Causerano for teaching me the basics of tissue culture, for your infinite wisdom and for

allowing me to work in the cell lab.

VII

Thank you to the staff of the Flow Cytometry Unit (RPH) particularly to Rom Krueger.

Thank you for all of your patience in teaching me the basics of flow cytometry and

helping me to optimise protocols.

Thank you to the staff of the Department of Medical Physics (RPH). I owe you a huge

thank you for your help with the 51Cr-realse cytotoxicity assays and for counting my

many thousands of samples.

Thank you to the staff at the Research Center (RPH). Thank you for allowing me to

work in the radioactive area and work in your tissue culture lab.

Thank you to all of the current and past PhD students, post-doctoral researchers,

research assistants and honours students: Andrew, Ann, Coral-Ann, Craig, Darren,

Diana, Dino, Hayley, Julius, Karey, Leo, Niamh, Nicole, Silvia, Sonia and Steve for all

of your advice over the years, I really do appreciate it all. A special thank you to my

‘lunch buddies’, Coral-Ann, Hayley, Karey, Silvia and Sonia for all the entertaining

lunches over the years. An extra thank you to Coral-Ann for help with formatting this

thesis. A special thank you to Julius for helping me ‘escape’ when experiments didn’t

work out. A special thank you to Diana who always knew exactly what I was going

through and for always being there when I needed someone the most.

Finally I would like to thank my family. Thank you Mum and Dad for your constant

support and encouragement, not just during this PhD but throughout my whole life. I

know it sounds cliché but you always told me that I could do anything that I wanted and

you’ve always believed in me. To my brother Dylan and sister Alyssa, thank you for

always supporting me and for always being there for me.

VIII

Abstract

Natural killer (NK) cell alloreactivity can be exploited in haploidentical haematopoietic

stem cell transplantation (HSCT) to improve graft survival, reduce graft versus host

disease and decrease leukaemic relapse. NK cells lyse cells that have reduced

expression of class I HLA molecules. In an allogeneic setting, donor NK cells may be

activated by the absence of donor (self) class I HLA molecules on recipient cells; the

absence of self-epitopes being detected by inhibitory KIR receptors on donor NK cells.

The way in which genetic polymorphism of the receptors and ligands affects NK

allorecognition of missing self, has not been fully elucidated.

HLA-C molecules are divided into two groups, C1 and C2, with KIR2DL1 recognising

cells expressing C2 and KIR2DL2 and KIR2DL3 recognising cells expressing C1.

Donor NK cells expressing KIR2DL2 or KIR2DL3 can be alloreactive towards a

recipient if they lack the C1 epitope and donor NK cells expressing KIR2DL1 can be

alloreactive towards a recipient if they lack the C2 epitope. KIR3DL1 recognises the

Bw4 epitope present on one-third of HLA-B alleles and certain HLA-A alleles. NK cells

from donors expressing KIR3DL1 can be alloreactive towards recipients whose cells

lack Bw4.

Mismatches of KIR related HLA epitopes does not always results in NK alloreactivity.

Therefore it is not possible to reliably predict NK alloreactivity based solely on the

donor’s HLA type and KIR repertoire and the recipient’s HLA type. The current

selection of suitable donors for a recipient undergoing a haploidentical HSCT involves

long, labourious NK cell cloning assays to determine if the donor’s NK cells can lyse

the recipient’s cells. This thesis has attempted to gain a better understanding of the

interaction of HLA alleles and KIR receptors, which may eventually allow suitable

donors to be predicted from HLA type and KIR genotypes.

IX

To date there has been no systematic survey of all the common HLA-B and HLA-C

alleles to interact with KIR. This thesis examined the ability of all the common HLA-C

alleles to interact with two different alleles of KIR2DL3; KIR2DL3*001 and

KIR2DL3*002. Whilst NK cells expressing KIR2DL3*002 were equally inhibited by

target cells expressing any of the common C1 alleles, NK cells expressing

KIR2DL3*001 were inhibited to a variable degree. HLA-C*1402, a C1 allele, only

weakly inhibited NK cells expressing KIR2DL3*001 or KIR2DL2*001. Other alleles such

as HLA-C*0303 and HLA-C*0304 gave modest inhibition of cells expressing

KIR2DL3*001.

This thesis also undertook a systematic analysis of the ability of the most common

HLA-B alleles and HLA-A alleles with Bw4 serological reactivity to inhibit KIR3DL1-

dependent NK cells. All Bw4-negative HLA-B alleles failed to inhibit target cells from

lysis. All Bw4-positive HLA-B alleles, with the exception of HLA-B*1301 and –B*1302,

protected targets from lysis. HLA-A*2402 and HLA-A*3201 unequivocally protected

target cells from lysis whereas HLA-A*2501 and HLA-A*2301 provided only weak

protection from lysis. KIR3DL1-dependent alloreactive NK clones were identified in

donors whose only Bw4 positive allele was HLA-A*2402 but not in donors whose only

Bw4 positive HLA allele was HLA-B*1301 or B*1302.

Finally this thesis demonstrated that an activating KIR can control NK cell alloreactivity.

Donors who are C2 negative and KIR2DS1 positive had NK cells that expressed the

activating receptor KIR2DS1 and were capable of lysing cells expressing the C2

epitope. More so, KIR2DS1 dependent NK clones were shown to override inhibitory

signals generated by NKG2A interacting with its ligand, HLA-E. The identification of

these NK clones has important implications for haploidentical HSCT in that recipient

expressing all three NK epitopes, C1, C2 and Bw4 were previously thought to be

resistant to alloreactive NK cells controlled by inhibitory receptors. Such patients may

be amenable to haploidentical HSCT from C2 negative, KIR2DS1 positive donors.

X

These results will improve the ability to predict NK cell alloreactivity based on a donor’s

HLA type and KIR repertoire and the recipient’s HLA type.

XI

Table of Contents

THESIS DECLARATION............................................................................................................ II

ACKNOWLEDGMENTS............................................................................................................VI

ABSTRACT .........................................................................................................................VIII

TABLE OF CONTENTS…………………………………………………... ......................................XI

LIST OF TABLES..................................................................................................................XVI

LIST OF FIGURES..............................................................................................................XVIII

CHAPTER 1: LITERATURE REVIEW........................................................................................ 1

1.1 SCOPE OF THE LITERATURE REVIEW............................................................................. 2

1.2 NATURAL KILLER CELLS ................................................................................................... 2

1.2.1 FUNCTIONS OF NK CELLS ............................................................................................. 3

1.2.1.1 Natural Killer Cell Cytotoxicity.......................................................................... 4

1.3 NATURAL KILLER CELL ALLORECOGNITION................................................................... 5

1.3.1 THE “MISSING SELF” HYPOTHESIS.................................................................................. 5

1.3.2 NATURAL KILLER CELLS RECEPTORS ............................................................................. 6

1.3.2.1 Killer Cell Immunoglobulin-like Receptors (KIR) .............................................. 7

1.3.2.2 CD94/NKG2 Receptor Family .......................................................................... 9

1.3.2.3 Leukocyte Immunoglobulin-like Receptors (LILR).......................................... 10

1.3.2.4 Natural Cytotoxicity Receptors....................................................................... 10

1.4 KIR ..................................................................................................................................... 11

1.4.1 KIR HAPLOTYPES ....................................................................................................... 12

1.4.2 KIR GENE POLYMORPHISM ......................................................................................... 13

1.4.3 KIR RECOGNITION OF CLASS I HLA ............................................................................. 13

1.5 DEPARTURES FROM THE SIMPLE C1/C2, BW4/BW6 INHIBITORY MODEL ................. 15

1.5.1 HLA-C*1402 AS A LIGAND FOR KIR2DL2 AND KIR2DL3 .............................................. 15

1.5.2 KIR3DL1 AND HLA-B................................................................................................. 16

1.6 KIR3DL2 AND HLA-A ......................................................................................................... 18

1.7 RULES GOVERNING NK ALLOREACTIVITY .................................................................... 21

1.8 ACTIVATING KIR ............................................................................................................... 22

1.9 ALLOGENEIC BONE MARROW TRANSPLANTATION..................................................... 24

1.9.1 COMPLICATIONS OF BMT ............................................................................................ 24

1.9.1.1 Leukemic Relapse ......................................................................................... 24

XII

1.9.1.2 Rejection........................................................................................................ 25

1.9.1.3 GvHD............................................................................................................. 25

1.9.2 MATCHING DONOR AND RECIPIENT FOR BMT ................................................................ 26

1.9.3 CELLS MEDIATING GRAFT VERSUS HOST DISEASE ........................................................ 26

1.9.3.1 Role of T cells in Graft versus Host Disease.................................................. 26

1.9.3.2 Role of Natural Killer cells in GvHD ............................................................... 28

1.9.4 CELLS MEDIATING THE GRAFT VERSUS LEUKAEMIA EFFECT........................................... 29

1.9.4.1 Role of T cells in GvL effect ........................................................................... 31

1.9.4.2 Role of Natural Killer cells in GvL effect ......................................................... 31

1.10 RECENT EVIDENCE OF THE ROLE OF NK CELLS IN BMT .......................................... 32

1.10.1 EVIDENCE THAT ALLOREACTIVE NK CELLS CAN ELIMINATE LEUKAEMIC CELLS .............. 32

1.10.2 EVIDENCE THAT ALLOREACTIVE NK CELLS CAN PREVENT GVHD.................................. 33

1.10.3 PREDICTION OF NK ALLOREACTIVITY USING HLA AND KIR GENETICS ........................... 34

1.11 HYPOTHESES AND AIMS ............................................................................................... 35

CHAPTER 2: MATERIALS AND METHODS ........................................................................... 37

2.1 CELL CULTURE................................................................................................................. 38

2.1.1 CELL LINES ................................................................................................................ 38

2.1.2 SEPARATION OF PMBC............................................................................................... 39

2.1.2.1 Isolation of PBMC.......................................................................................... 39

2.1.2.2 Isolation of PBMC from Unusable Red Cell Bags as Allogeneic Feeder Cells 39

2.1.2.3 Storage of Pooled Allogeneic PBMC ............................................................. 39

2.1.2.4 Thawing of PBMC.......................................................................................... 40

2.1.2.5 Irradiation of Cells.......................................................................................... 40

2.2 NATURAL KILLER CELL CLONING................................................................................... 40

2.2.1 ENRICHMENT OF NATURAL KILLER CELLS ..................................................................... 40

2.2.2 NATURAL KILLER CELL CLONING.................................................................................. 41

2.2.3 NATURAL KILLER CELL POLYCLONAL CULTURES ........................................................... 41

2.2.4 RESTIMULATION OF NATURAL KILLER CELL CLONES AND POLYCLONAL CULTURES .......... 42

2.3 IMMUNOPHENOTYPING................................................................................................... 42

2.3.1 FLOW CYTOMETRY ON WHOLE BLOOD ......................................................................... 42

2.3.2 NK CELL PHENOTYPYING-LABELLED PRIMARY ANTIBODIES ........................................... 42

2.3.3 NK CELL PHENOTYPYING AND CLASS I HLA STAINING-LABELLED SECONDARY ANTIBODIES

.......................................................................................................................................... 43

2.3.4 ANALYSIS OF FLOW CYTOMETRY DATA......................................................................... 43

2.4 NATURAL KILLER CELL CYTOTOXICITY......................................................................... 43

2.4.1 TARGET CELL PREPARATION ....................................................................................... 44

2.4.2 EFFECTOR CELL PREPARATION ................................................................................... 44

XIII

2.4.3 4 HOUR 51CR RELEASE ASSAY .................................................................................... 44

2.4.4 CD107A ASSAY ......................................................................................................... 45

2.5 DNA AND RNA................................................................................................................... 46

2.5.1 DNA EXTRACTION ...................................................................................................... 46

2.5.2 RNA EXTRACTION ...................................................................................................... 46

2.5.3 REVERSE TRANSCRIPTASE PCR.................................................................................. 46

2.6 POLYMERASE CHAIN REACTION.................................................................................... 46

2.6.1 AMPLIFICATION OF KIR2DL2, KIR2DL3 AND KIR2DS1 FROM CDNA............................. 47

2.6.2 SSP-PCR FOR KIR2DS1 ON GENOMIC DNA AND SSP-PCR FOR KIR2DL1 FROM CDNA48

2.6.3 KIR3DL1 ALLELE TYPING ........................................................................................... 50

2.6.4 KIR GENOTYPING BY SSP- PCR................................................................................. 50

2.6.5 VISUALISING PCR PRODUCTS ..................................................................................... 51

2.6.6 DNA SEQUENCING ..................................................................................................... 51

CHAPTER 3: KIR2DL3*001 AND KIR2DL3*002 DIFFER IN THEIR ABILITY TO INTERACT WITH C1-GROUP HLA-C ALLELES ....................................................................................... 52

3.1 INTRODUCTION ................................................................................................................ 53

3.2 MATERIALS AND METHODS ............................................................................................ 54

3.3 RESULTS ........................................................................................................................... 55

3.3.1 SELECTION OF NK CLONES ......................................................................................... 55

3.3.1.1 Characterisation of Clones from Donor 1 ....................................................... 56

3.3.1.1 Characterisation of Clones from Donor 2 ....................................................... 57

3.3.1.1 Characterisation of Clones from Donor 3 ....................................................... 58

3.3.1.1 Characterisation of Clones from Donor 4 ....................................................... 58

3.3.2 SUMMARY OF NK CLONES........................................................................................... 59

3.3.3 KIR2DL3 MEDIATED INHIBITION BY THE COMMON C1-GROUP AND C2-GROUP HLA-C

ALLELES ............................................................................................................................. 61

3.3.4 HLA-C*1402 INHIBITS NK CELLS FROM SOME DONORS ............................................... 68

3.3.5 HLA-C*1402 ONLY INHIBITS NK CELLS EXPRESSING KIR2DL3*002............................... 70

3.3.6 DO HLA-C ALLELES THAT INTERACT WEAKLY WITH KIR2DL3*001 SHARE AMINO ACID

MOTIFS? ............................................................................................................................. 72

3.4 DISCUSSION ..................................................................................................................... 74

CHAPTER 4: THE REACTIVITY OF BW4-POSITIVE HLA-B AND HLA-A ALLELES WITH KIR3DL1: IMPLICATIONS FOR PATIENT AND DONOR SUITABILITY FOR HAPLOIDENTICAL STEM CELL TRANSPLANTS.................................................................. 79

4.1 INTRODUCTION ................................................................................................................ 80

XIV

4.2 MATERIALS AND METHODS ............................................................................................ 82

4.3 RESULTS ........................................................................................................................... 85

4.3.1 KIR3DL1 GENOTYPE OF NK CELLS............................................................................. 85

4.3.2 KIR3DL1 DEPENDENT NK CELLS LYSE CELLS LACKING THE BW4 EPITOPE.................... 87

4.3.3 HLA-B*13 IS NOT A KIR3DL1 LIGAND BUT HLA-B*5101 IS ........................................... 93

4.3.4 HLA-A*2402 AND HLA-A*3201 ARE LIGANDS FOR KIR3DL1 ........................................ 96

4.3.5 DONORS WHO HAVE HLA-B*1301 OR B*1302 CAN MAKE KIR3DL1 DEPENDENT NK CLONES

BUT THESE NK CLONES ARE INFREQUENT.............................................................................. 98

4.3.6 DONORS WHO LACK BW4 EXPRESSING HLA-B ALLELES BUT EXPRESS HLA-A*2402 CAN

MAKE KIR3DL1 DEPENDENT NK CLONES ............................................................................ 101

4.4 DISCUSSION ................................................................................................................... 102

CHAPTER 5: KIR2DS1-MEDIATED ACTIVATION OVERRIDES NKG2A-MEDIATED INHIBITION IN HLA-C C2 NEGATIVE INDIVIDUALS ........................................................... 108

5.1 INTRODUCTION .............................................................................................................. 109

5.2 MATERIALS AND METHODS .......................................................................................... 111

5.3 RESULTS ......................................................................................................................... 114

5.3.1 ALLOREACTIVE NK CLONES EXPRESSING CD158A LYSE C2 EXPRESSING TARGETS ..... 114

5.3.2 THE ALLOSPECIFICITY OF CLONE C9 CANNOT BY EXPLAINED BY KIR3DL2.................... 116

5.3.3 THE ALLOSPECIFICITY OF CLONE C9 IS DETERMINED BY THE PRESENCE OF THE C2 EPITOPE

IN A DOSE DEPENDENT MANNER ........................................................................................ 118

5.3.4 CLONE C9 EXPRESSES KIR2DS1*002 ...................................................................... 119

5.3.5 ACTIVATION BY C2 POSITIVE TARGETS IS MEDIATED BY KIR2DS1 AND OVER-RIDES

NKG2A MEDIATED INHIBITION............................................................................................ 120

5.3.6 KIR2DS1 ACTIVATED NK CLONES REPRESENT 1% OF ALL NK CELLS IN C1 HOMOZYGOUS

INDIVIDUALS...................................................................................................................... 122

5.3.7 NK CELLS ACTIVATED BY C2 KILL EBV NON-INFECTED TARGETS................................. 125

5.4 DISCUSSION ................................................................................................................... 127

CHAPTER 6: FINAL DISCUSSIONS AND CONCLUSIONS ................................................. 130

6.1 THE AIMS OF THIS THESIS............................................................................................ 132

6.2 SUMMARY OF NEW DATA ON HLA:KIR INTERACTION PROVIDED IN THIS THESIS. 132

6.2.1 KIR2DL2/3 AND HLA-C............................................................................................ 132

6.2.2 KIR3DL1 AND BW4 .................................................................................................. 133

6.2.3 KIR2DS1 AND C2 GROUP HLA-C ALLELES ................................................................ 133

6.3 LIMITATIONS TO THE CONCLUSIONS DRAWN IN THIS THESIS ................................ 134

XV

6.4 FURTHER STUDIES ........................................................................................................ 136

6.5 IMPLICATIONS FOR HAPLOIDENTICAL HSCT.............................................................. 137

CHAPTER 7: REFERENCES ................................................................................................. 138

APPENDICES ........................................................................................................................ 159

APPENDIX 1: SOLUTIONS AND BUFFERS .............................................................................. 160

APPENDIX 2: ANTIBODIES................................................................................................... 164

APPENDIX 3: PRIMER SEQUENCES...................................................................................... 165

XVI

List of Tables

Table 1.1 The five-year probability of leukaemic relapse for AML and ALL patients with

and without ‘NK mismatches’. ...................................................................... 32

Table 2.1 Master Mix for KIR2DL2, KIR2DL3 and KIR2DS1 (cDNA) Full Length PCR 47

Table 2.2 KIR2DL2 cDNA and KIR2DL3 cDNA Full Length PCR Thermocycling

Conditions .................................................................................................. 48

Table 2.3 KIR2DS1 Full Length PCR Thermocycling Conditions................................. 48

Table 2.4 Mater Mix for KIR2DS1 SSP (Genomic DNA) .............................................. 49

Table 2.5 Master Mix for KIR2DL1 SSP (cDNA) .......................................................... 49

Table 2.6 KIR2DS1 SSP (Genomic DNA) and KIR2DL1 SSP (cDNA) Thermocycling

Conditions .................................................................................................... 49

Table 2.7 Master Mix for KIR3DL1 Allele Typing.......................................................... 50

Table 2.8 KIR3DL1 Allele Typing Thermocycling Conditions ....................................... 50

Table 3.1 NK Donor Class I HLA Typing...................................................................... 55

Table 3.2 NK Donor KIR Genotype .............................................................................. 55

Table 3.3 Class I HLA Typing of Target Cell Panel Used To Screen NK Clones......... 56

Table 3.4 NK Clones from Donor 2 for which the Alloreactivity Pattern was Consistent

with Receptor Expression ............................................................................ 57

Table 3.5 NK Clones from Donor 3 for which the Alloreactivity Pattern was Consistent

with Receptor Expression .......................................................................... 58

Table 3.6 HLA-C Target Cell Panel .............................................................................. 61

Table 3.7 HLA-C Target Cell Panel 2 ........................................................................... 64

Table 3.8 HLA-C Target Cell Panel 3 ........................................................................... 66

Table 3.9 Amino Acid Differences between KIR2DL3*002, KIR2DL3*001 and

KIR2DL2*001 ............................................................................................... 71

Table 3.11 Residue 7 Binding Environment of Strong, Moderate and Low Inhibiting

HLA-C .......................................................................................................... 73

Table 3.12 Residue 8 Binding Environment of Strong, Moderate and Low Inhibiting

HLA-C .......................................................................................................... 73

XVII

Table 4.1 HLA Class I and KIR3DL1 Allele Typing of NK Cell Donors........................ 83

Table 4.2. HLA Class I Typing of BLCL Target Cell Panel ........................................... 84

Table 5.1. Class I HLA Typing and NK Epitopes of BLCL Target Cell Panel ............. 112

Table 5.2. HLA Typing NK Epitopes and KIR Typing of NK Cell Donors ................... 113

XVIII

List of Figures

Figure 1.1 KIR Genomic Structure ............................................................................... 12

Figure 1.2 KIR Haplotypes ........................................................................................... 13

Figure 1.4 Recognition of HLA-C by KIR2DL2 (left) and KIR2DL1 (right). ................... 14

Figure 3.1 NK clones selected for use in testing different HLA-C alleles and their

staining with CD158b and their cytotoxicity patterns ................................. 60

Figure 3.2 Clone A6 and YTS-2DL3 differ in their interaction with HLA-C alleles with the

C1 epitope.................................................................................................. 63

Figure 3.3 Pattern of Inhibition is Reproduced with a second panel of C1-group HLA-C

alleles......................................................................................................... 65

Figure 3.4 C1-group HLA-C alleles differ in their ability to inhibit YTS-2DL3 ............... 67

Figure 3.5 HLA-C*0702 inhibits all NK clones whereas HLA-C*1402 only inhibits some

NK clones................................................................................................... 69

Figure 3.6 Amplification of KIR2DL2 or KIR2DL3 cDNA from each clone.................... 70

Figure 4.1 Low level KIR3DL1 expression differs in two donors due to a

nonsynonymous mutation at nucleotide position 115 in exon 3 encoding the

D0 domain of the KIR3DL1*005 allele. ...................................................... 86

Figure 4.2. CD107a expression by KIR3DL1-positive polyclonal NK cells is inhibited by

most Bw4-positive targets.......................................................................... 89

Figure 4.3. Level of Bw4 expression does not correlate with percentage of CD107a

expression induced on KIR3DL1 positive NK cells. ................................... 92

Figure 4.4. HLA-B*1302 and HLA-B*1301 do not inhibit cytotoxicity of KIR3DL1-

dependent NK cells.................................................................................... 95

Figure 4.5 Inhibition through Bw4 expressing HLA-A alleles can be reversed by

addition of anti-KIR3DL1............................................................................ 97

Figure 4.6 Donors who express HLA-B*1302 or HLA-B*1301 make very few KIR3DL1-

dependent NK cells.................................................................................. 100

Figure 5.1. Receptor expression and cytotoxicity pattern of CD158b negative clones

showing specificity for C2 positive targets. .............................................. 115

XIX

Figure 5.2. Despite expression of KIR3DL2, clone C9 was inhibited equally well by

HLA-A*0301 and HLA-A*0201 homozygous BLCL target. ...................... 117

Figure 5.3. NK clone C9 only lyses C2 positive BLCL targets and shows a C2 dose

effect. ....................................................................................................... 118

Figure 5.4. NK clone C9 transcribes KIR2DS1*002. .................................................. 119

Figure 5.5 Target cell lysis by NK clone C9 is enhanced by anti-NKG2A antibody and

inhibited by anti-CD158a (anti-KIR2DL1/KIR2DS1)................................. 121

Figure 5.6. NK cell activation through KIR2DS1 (CD158a) only occurs in C2 negative,

KIR2DS1 positive individuals. .................................................................. 124

Figure 5.7. C2 positive EBV transformed and PHA blast cells both activate CD158a

positive NK cells....................................................................................... 126

XX

Chapter 1

1

Chapter 1

Literature Review

Chapter 1

2

Chapter 1: Literature Review

This literature review will cover literature available at the time of commencement of this

thesis (February 2004). Literature published after 2004 but which is relevant to the

topics in this thesis will be discussed in the relevant chapter.

1.1 Scope of the Literature Review

Allogeneic bone marrow transplantation (BMT) or haematopoietic stem cell

transplantation (HSCT) is commonly used in clinical practice as a curative treatment for

haematological malignancies as well as non-malignant disorders and diseases of the

haematopoietic system, such as severe aplastic anaemia. Despite advances in human

leucocyte antigen (HLA) matching, BMT remains a high-risk treatment for these

diseases and disorders. In addition, many recipients will not have an HLA identical

sibling donor but most will have a haploidentical related donor. The discovery (Ruggeri

et al, 2002) that certain HLA mismatches result in the generation of alloreactive natural

killer (NK) cells that can not only prevent leukemic relapse, but also enhance bone

marrow engraftment and eliminate graft versus host disease (GvHD), has greatly

increased the efficacy of haploidentical BMT. NK alloreactivity cannot always be

predicted from KIR and HLA genotypes. A clearer understanding of the interactions

between KIR and HLA alleles will improve prediction of NK alloreactivity. This literature

review will introduce NK cells, their receptors and their interaction with class I HLA,

followed by an overview of bone marrow transplantation and a review of the role of NK

cell alloreactivity in BMT.

1.2 Natural Killer Cells

NK cells are components of the innate immune response. They are important in the

initial phase of infections and play an important role in tumour surveillance. They

received their name from their ability to kill target cells without prior sensitisation

(Kiessling et al, 1975). Recognised morphologically as relatively large lymphocytes

Chapter 1

3

containing cytoplasmic azurophilic granules, NK cells were also once known as large

granular lymphocytes (LGL) (Herberman 1986). Comprising 10-15% of circulating

blood lymphocytes, NK cells are also found in the bone marrow, spleen, lymph nodes,

liver, lung, omentum, intestine and placenta (reviewed in Vivier 2006).

NK cells are thought to originate from the same lineage as T lymphocytes in the bone

marrow (Spits et al, 1995). However, they do not rearrange immunoglobulin (Ig) or T

cell receptor genes to make antigen receptors like T and B cells (Lanier and Phillips

1992). Resting peripheral blood NK cells can be identified from T and B lymphocytes

based on their expression of CD56 (neural cell adhesion molecule-1, NCAM-1), CD16

(FcγRIII) and CD160. Since all three makers are expressed by other immune cells it is

the combination of the three markers and lack of the T cell marker CD3 that effectively

identify NK cells.

NK cells can be further characterised based on the surface level of expression of CD56

and CD16. Approximately 90% of NK cells circulating in peripheral blood have low

density CD56 (CD56dim) and express high levels of CD16 whereas the other 10%

(CD56bright) express higher levels of CD56 (Lanier et al, 1986). CD56dim expressing NK

cells are more strongly cytotoxic towards target cells than are CD56bright cells. CD56bright

NK cells produce greater quantities of cytokines, but are weakly cytotoxic relative to

CD56dim cell.

1.2.1 Functions of NK Cells

NK cells are pivotal members of the innate immune response, playing a crucial role in

the elimination of virus-infected and tumour cells as well as the recruitment of cells of

the adaptive immune response. NK cells mediate their activities through the release of

cytokines or through cytotoxic pathways (section 1.2.1.1). Cytokines produced by NK

cells in response to virus-infected or tumour cells play a major role in shaping the

Chapter 1

4

adaptive immune response. For example IFNγ production by NK cells influences the

TH1/TH2 bias of the adaptive immune response by activating TH1 cells (Seaman 2000).

Recently the relationship between dendritic cells (DC) and NK cells has been examined

in the early stages of the immune response. Many of the cytokines (eg. Interleukin-12

(IL-12) and interleukin-18 (IL-18)) known to enhance NK cell function are naturally

produced by DC in response to microbial infections (reviewed Andoniou et al, 2006).

NK cells also reciprocally enhance DC function. NK cells have been shown to eliminate

immature DC, perhaps to ensure that only activated DC are involved in antigen

presentation (Ferlazzo and Munz, 2004). NK cells also activate DCs through the

production of IFNγ and tumour necrosis factor-α (TNFα) and other mechanisms that

require cell to cell contact (Gerosa et al, 2002). The interaction between NK cells and

DC demonstrate that effective immune responses rely on relationships between the

cells of both the innate and adaptive immune response.

NK cells have also been shown to play a role in the regulation of haematopoiesis

(Trinchieri, 1995). They influence the growth of haematopoietic cells through the

release of cytokines, including GM-CSF and TNFα which influence stem cell

proliferation and differentiation (Seaman 2000).

1.2.1.1 Natural Killer Cell Cytotoxicity

NK cells lyse susceptible target cells (eg. virus-infected cells or tumour cells), by one of

two mechanisms: “natural killing” (no prior sensitisation), or by antibody dependent

cellular cytoxicity (ADCC). Natural killing is initiated through a variety of activating

receptors and can be inhibited by a variety of inhibitory receptors. In ADCC, the

activating receptor, FcγRIII (CD16) binds to the Fc piece of antibodies bound to the

target cell (Roitt et al, 2000). In both pathways, NK cells can lyse the target via two

Chapter 1

5

mechanisms; either perforin-mediated cytotoxicity or induction of apoptosis through

Fas (Bosi and Griffiths, 1999).

1.3 Natural Killer Cell Allorecognition

1.3.1 The “Missing self” Hypothesis

Until relatively recently, it was thought that NK cells had receptors for foreign antigens

on target cells. However, the nature of the receptors and foreign antigen remained

elusive. Subsequently a correlation was observed between the susceptibility of target

cells to NK cell lysis and low, or absent, expression of class I MHC molecules on the

target cell (Snell, 1976). Low or absent expression of class I MHC molecules is

common in tumour cells and virus-infected cells, the usual targets for NK cell lysis.

Therefore, it was proposed that NK cells may interact with MHC class I molecules, but

perhaps somehow recognise the absence of MHC class I. This idea was consistent

with research conducted by Cudkowicz and Bennet (1971) who investigated the

phenomenon of ‘F1 hybrid resistance’ in which it was observed that homozygous

parental bone-marrow transplants were rejected by their heterozygous (absence of one

parental antigen) F1 offspring. Thus they were responding to the absence of MHC class

I. Rejection could be prevented by depletion of recipient NK cells demonstrating that

NK cells were responsible for the rejection.

Karre and colleagues (1986) studied NK cell mediated rejection of H-2 incompatible

lymphomas and bone marrow grafts in mice. Lymphoma cells selected for the loss of

H-2 were less malignant than the wild-type and were rejected by innate mechanisms

rather than adaptive mechanisms. NK cells were postulated to be the effector. It was

hypothesised that NK cells recognise and eliminate cells that fail to express self MHC

class I molecules (Ljunggren and Karre 1990) and this was formally known as the

“missing self” hypothesis.

Chapter 1

6

Pursuing the ‘missing self’ hypothesis, Storkus and colleagues (1987) demonstrated

that transfection of class I HLA genes into class I bare target cells rendered the target

cells totally or partially resistant to NK cell mediated lysis. This further supported the

idea that NK cells sense the presence of MHC class I molecules on target cells

resulting in inhibition of target lysis.

In 1988, Ciccone and colleagues demonstrated that human NK cells would proliferate

in the presence of allogeneic stimulator cells. Lymphocytes were treated in vitro with

monoclonal antibodies (mAbs) against CD3, CD4 and CD8 followed by treatment with

rabbit complement to eliminate T cells. The T cell depleted population was cultured

with irradiated allogeneic cells. The CD3-, CD2+, CD7+ lymphocytes, which proliferated,

lysed target cells bearing the same HLA type as the stimulator cells, but did not lyse

autologous cells. This was the first evidence that human NK cells could be alloreactive.

Moretta and colleagues (1990) described four distinct NK cell cytotoxic specificities

based on the lysis of different allogeneic target cells and showed that the four

specificities could be correlated with the expression on the NK cells of molecules

recognised by the mAb, EB6 and GL183. The four different effector populations were

EB6+GL183-, EB6-GL183+, EB6+GL183+ and EB6-GL183-. Using an informative

family, Ciccone and colleagues (1990) mapped susceptibility of target cells to lysis by

alloreactive NK cells, to a region in the MHC between BF and HLA-B.

1.3.2 Natural Killer Cells Receptors

NK cell receptors for target cell ligands fall into two broad groups based on their

structure (Yokoyama, 1998): C-type lectin-like receptors and Ig-like receptors. C-type

lectin-like receptors are disulfide-linked, dimeric, type II, integral membrane proteins.

They include the CD94/NKG2 family and the Ly49 family (found only in mice). Ig-like

receptors are usually monomeric type I integral membrane proteins. Ig-like receptors

Chapter 1

7

include the killer immunoglobulin-like receptors (KIR), natural cytotoxicity receptors and

the LILR family of receptors.

Subsets of NK cells differ in their receptor expression. (Yokoyama, 1998). Receptor

expression differs not only between NK clones within an individual but also between

individuals (Valiante et al, 1997). Within an individual, NK cells differ in the number and

type of receptors expressed. However, every NK cell in a given individual that is

capable of cytotoxicity has at least one inhibitory receptor for self-HLA molecules to

prevent lysis of autologous cells. The inhibitory receptor may be a KIR with specificity

for allelic epitopes of the classical class I HLA loci or another inhibitory receptor, for

example NKG2A, the ligand for which is the monomorphic, non-classical class I

molecule, HLA-E.

1.3.2.1 Killer Cell Immunoglobulin-like Receptors (KIR)

Following the identification of a region in the MHC that determined the susceptibility of

targets to killing by different NK cell clones, Colonna and colleagues (1992) identified a

correlation between the expression of particular HLA-C alleles on target cells and

resistance to NK cell mediated lysis. NK cell clones with different specificities termed

NK-1 and NK-2 were identified. NK-1 clones were inhibited by targets expressing HLA-

C alleles with an asparagine at amino acid 77 and lysine at amino acid 80 in the α1

domain of HLA-C molecules. NK cells, with the NK-2 specificity, were inhibited by

target cells expressing HLA-C alleles with a serine at amino acid 77 and asparagine at

amino acid 80. All HLA-C alleles have one or the other of these two sets of amino

acids. The NK receptor p58 (different forms of which were recognised by the EB6 and

GL183 antibodies) was identified as the receptor that recognised HLA-C (Moretta et al,

1993). NK cells, with the specificity NK-1, expressed a p58 receptor that reacted with

the mAb EB6, and NK cells, with the NK-2 specificity expressed a p58 receptor that

reacted with the mAb GL183. Other NK clones that were inhibited by target cells

Chapter 1

8

expressing certain HLA-B alleles were also discovered. D’Andrea and colleagues

(1995) showed that these NK cells expressed the NKB1 surface antigen, or p70. NKB1

was structurally similar to the p58 receptors but recognised HLA-B alleles with the Bw4

public epitope at amino acids 77-83 of the α1 domain of HLA-B molecules.

Colonna and Samaridis (1995) and Wagtmann et al (1995) described the structure of

the p58 and p70 receptors. Utilising molecular techniques, p58 and p70 were shown to

posses two and three immunoglobulin-like domains, respectively. Pende and

colleagues (1996) described an additional receptor, p140, a dimer of a p70-like

receptor, with three immunoglobulin-like domains that had a high degree of homology

to p58 and p70. The p140 receptor was shown to interact with the -A3 allele at the

HLA-A locus and it was postulated that p140 may recognise an allelic determinant of

HLA-A alleles.

The p58, p70 and p140 NK cell receptors are encoded on the leukocyte receptor

complex (LRC) on chromosome 19q13.4 in man and recognise allelic determinants of

HLA-C, HLA-B and HLA-A alleles, respectively. The discovery that p58, p70 and p140

interacted with cells expressing the appropriate HLA class I molecules causing

inhibition of NK mediated lysis, revealed that NK recognition of “non-self” was based on

a mechanism that was different to T and B cell recognition of non-self. These inhibitory

receptors were initially called “killer cell inhibitory receptors”. However the later

discovery of activating versions of these receptors led to them being renamed “killer

cell immunoglobulin-like receptors” or KIR. Inhibitory KIR interact with HLA class I

molecules. Activating KIR show a high degree of sequence homology to inhibitory KIR,

often differing by only a few amino acids in the Ig-domains that are responsible for

ligand binding (Wagtmann et al, 1995; Biassoni et al, 1996). They produce activation

signals when cross-linked with antibodies. Their ligands are believed to be the same as

for their inhibitory counterparts because of the structural similarity between the

Chapter 1

9

receptors. However this has been difficult to demonstrate (Vales-Gomez et al, 1998;

Winter et al, 1998; Saulquin et al, 2003).

Inhibitory KIR have immuno-receptor tyrosine–based inhibition motifs (ITIMs) in their

cytoplasmic tail (Moretta et al, 2000). Upon engagement of HLA class I, the tyrosine

residues are phosphorylated and bind to the protein tyrosine phosphatase, SHP-1,

leading to negative cell signalling (Burshtyn et al, 1996). Activating receptors lack

ITIMs, but have a positively charged amino acid residue in their transmembrane

domain. This associates with the negatively charged adaptor molecule DAP-12. DAP-

12 contains an immuno-receptor tyrosine-based activation motif (ITAM). Upon ligand

engagement or antibody cross-linking, phosphorylation occurs through Src tyrosine

kinases, which recruit and activate further tyrosine kinases such as ZAP-70 which

activates signalling pathways (Lanier et al, 1998).

1.3.2.2 CD94/NKG2 Receptor Family

The CD94/NKG2 receptor family is encoded on chromosome 12p13 in humans and

chromosome 6 in mice (Yabe et al, 1993). The NKG2 family consists of six genes:

NKG2A, NKG2C, NKG2E, NKG2D and NKG2F and CD94. Each member of the family

forms a disulfide-linked heterodimer with CD94, except for NKG2D which is self-

dimerizing (Lazetic et al, 1996). The CD94/NKG2A heterodimer forms an inhibitory

receptor which binds to the genetically invariant non-classical class I HLA molecule,

HLA-E (Yokoyama, 1998). As the expression of HLA-E is promoted by binding of

peptides clipped from leader-sequence of classical HLA class I molecules, it is thought

that HLA-E expression acts as a barometer of classical class I protein expression. The

purpose of CD94/NKG2A may, therefore, be to monitor overall class I expression

whereas KIR receptors monitor the expression of individual alleles. CD94/NKG2B is a

splice variant of NKG2A, which lacks the stem region but still associates with CD94

and recognises HLA-E (Braud et al, 1998). CD94/NKG2C and NKG2D are activating

Chapter 1

10

receptors. CD94/NKG2C recognises HLA-E, but with lower affinity than CD94/NKG2A

(Vales-Gomez et al, 1999). NKG2D is a homodimer and unlike the other members of

the NKG2 family does not associate with CD94 (Lazetic et al, 1996). NKG2D

recognises the non-classical MHC molecules, MICA and MICB, and other non-MHC

molecules. The NKG2D ligands, MICA and MICB, ULBP1, ULBP2 and ULBP3 and the

RAE family, are up regulated by cells in times of stress and in virus-infected cells and

tumour cells (Farrell et al, 2000).

1.3.2.3 Leukocyte Immunoglobulin-like Receptors (LILR)

Most LILRs are expressed on a wide variety of cell types including NK cells, TH cells,

cytotoxic T lymphocytes (CTL), B cells and monocytes (Colonna et al, 1997; Meyaard

et al, 1997) and the genes encoding these receptors are in the LRC. The LILR deliver

an inhibitory signal to NK cells. LILRB1 binds to an array of classical and non-classical

HLA class I molecules, including HLA-F and HLA-G. LILRB2 and LILRA1 also bind

class I molecules (reviewed Brown et al, 2004). As the epitopes recognised by the LILR

family are genetically monomorphic, it seems unlikely that the LILRB1 receptors play

any role in allorecognition.

1.3.2.4 Natural Cytotoxicity Receptors

Natural cytotoxicity receptors (NCR) are activating receptors on NK cells (Moretta et al,

2000). Three receptors, NKp46, NKp44 and NKp30 are classified as NCR. Although

belonging to the immunoglobulin-gene superfamily, natural cytotoxicity receptors have

little homology with other receptors encoded by the LRC or with each other and there is

very little known about the genetic polymorphism of these receptors. Sivori and

colleagues (1999) reported a positive correlation with NKp46 surface density and

natural cytotoxicity of fresh and cultured NK cells against various FcγR-negative and

positive targets such as the class I negative cell lines, 721.221 and K562. The ligands

for NKp30, NKp44 and NKp46 however remain elusive. Some investigators have

Chapter 1

11

suggested viral antigens as being the ligands for the NCR (Arnon et al, 2001;

Mandelboim et al, 2001; Arnon et al, 2005) based on their role in the lysis of virally-

infected cells. Similarly, NCR have also been shown to mediated lysis of tumour cells

of various histotypes suggesting their ligands may be up-regulated or induced following

tumour transformation (reviewed Bottino et al, 2005).

1.4 KIR

Genes encoding the KIR receptors are found on chromosome 19q13.4 in the LRC

(Suto et al, 1998). KIR are classified as 2-domain (2D) or 3-domain (3D) according to

the number of immunoglobulin-like extracellular domains in the translated protein. They

are further subdivided on the basis of the length of their cytoplasmic tail (long - L) or

(short-S) (Vilches and Parham, 2002). Different genes sharing the same number of Ig

domains and the same kind of cytoplasmic tail are distinguished by a number at the

end of their name eg. KIR2DL3. KIR receptors are encoded by nine exons with exon 3

existing as a pseudoexon that is not translated in any of the 2D genes except KIR2DL4

and KIR2DL5 (Vilches et al, 2000a) (Figure 1.1). The Ig-domain encoded by exons 3, 4

and 5 are referred to as the D0, D1 and D2 domains, respectively. Skipping of exon 3

during transcription of the 2D KIR results in these KIR only translating the D1 and D2

domains. KIR2DL4 and KIR2DL5 are encoded by only eight exons (Vilches et al,

2000b) and lack the exon encoding the D1 Ig domain (Figure 1.1).

Chapter 1

12

(taken from http://www.ebi.ac.uk/ipd/kir/)

Figure 1.1 KIR Genomic Structure

Each KIR gene has been grouped based on structural homology, with exons shown in blue and

the pseudoexon shown in red. The size of each exon in base pairs is indicated above each

exon.

1.4.1 KIR haplotypes

The KIR gene content of human chromosomes 19 varies, but there are two broad

groups of KIR haplotypes (Uhrberg et al, 1997). One haplotype is particularly common

(~40% haplotype frequency in Caucasian populations (Uhrberg et al, 1997; Witt et al,

1999)) and is composed of the genes for KIR2DL1, KIR2DL3, KIR2DL4, KIR3DL1,

KIR3DL2, KIR3DL3 and KIR2DS4. It also includes the pseudogenes 2DP1 and 3DP1.

This haplotype and minor variants of it have been defined as the A haplotype. All other

haplotypes are referred to as B haplotypes. B haplotypes generally have a greater

number of activating KIR and are less conserved in terms of gene content than group A

haplotypes (Uhrberg et al, 1997). KIR genes that are common to both A and B

Chapter 1

13

haplotypes are known as framework genes. These include: KIR3DL3, the pseudogene

KIR3DP1, KIR2DL4 and KIR3DL2 (Figure 1.2)

(Adapted from Martin et al, 2000)

Figure 1.2 KIR Haplotypes

KIR genes are organised into two haplotypes, A haplotype and B haplotype. The bold lines

indicate the more common haplotypes.

1.4.2 KIR Gene Polymorphism

Whilst KIR haplotypes contribute to the diversity of an individual’s KIR repertoire, allelic

variation within each KIR gene also exists (Vilches and Parham, 2002). Allelic

polymorphisms have been described for all inhibitory KIR, with the receptors KIR3DL1

and KIR3DL2 showing the most extensive polymorphism. The influence of polymorphic

positions on ligand binding and other functions will be discussed in sections 1.4.3 and

1.5.

1.4.3 KIR Recognition of Class I HLA

KIR recognise allelic determinants of classical class I HLA molecules. HLA-C alleles

can be divided into two groups, C1 and C2, based on their amino acids sequences in

the α1 helix. (Colonna et al, 1993). C1-group alleles have the amino acid serine (Ser)

at residue 77 and asparagine (Asn) at residue 80. C2-group alleles have the amino

Haplotype ‘A’

3DL3 2DP1

2DL3 2DL1

3DP1 2DL4

3DL1 2DS4 3DL2

3DS1 2DS12DL1V

2DL5.1 2DS3

2DS2 2DL2 2DS2 2DL2 2DL5.2 2DS5

2DL5.1 2DS3

3DL1

2DL1

Haplotype ‘B’

Centromeric variable Telomeric variable

2DP1

Chapter 1

14

acid Asn at residue 77 and Lysine (Lys) at residue 80 (Colonna et al, 1993). HLA-B

alleles can similarly be split into two groups based on the Bw4 and Bw6 epitopes at

amino acids 77-83 (Muller et al, 1989).

The residues involved in the KIR/HLA binding interface are highly conserved in all KIR

and HLA alleles (Boyington and Sun, 2002) except for those residues determining

locus or allele specificity. Only the amino acid at position 80 of HLA-C determines

whether it binds to KIR2DL1 or KIR2DL2/3 (KIR2DL2 or KIR2DL3) (Mandelboim et al,

1996). The specificity of KIR2DL2 and KIR2DL3 for C1-group alleles is determined by

Lys at residue 44 of the receptor interacting specifically with amino acid Asn at position

80 of HLA-C (Figure 1.3 (left)). KIR2DL1 encodes a methionine (Met) at the same

residue. In C2-group alleles, Lys80 forms a hydrogen bond with Ser at residue 184 and

a salt bridge with glutamine (Glu) at residue 187 in KIR2DL1 (Boyington and Sun,

2002) (Figure 1.3 (right)). Mutation of residue 44 from Met to Lys and vice-versa is

sufficient to switch the HLA-C specificity of the 2D KIR (Winter and Long, 1997).

Boyington and Sun, 2002

Figure 1.4 Recognition of HLA-C by KIR2DL2 (left) and KIR2DL1 (right).

KIR are shown in green and HLA-C in brown. The interaction between amino acid 80 on HLA-C

and amino acid 44 (KIR2DL2), 184 and 187 (KIR2DL1) is shown in red with hydrogen bonds

and salt bridges represented by dotted lines.

Chapter 1

15

It has been reported that KIR are unable to recognise HLA class I molecules that have

an empty peptide binding groove (Boyington and Sun, 2002) suggesting that KIR

recognition is dependent on the peptide folded in the class I molecule. Peruzzi and

colleagues in 1996 demonstrated that residues 7 and 8 of the peptide bound by the

Bw4 positive HLA-B allele, HLA-B*27, influenced KIR3DL1 binding. Mutation of these

residues conferred or abolished the interaction between KIR3DL1 and HLA-B*27.

Similarly, residues 7 and 8 are likely to influence binding of KIR2DL1, KIR2DL2 and

KIR2DL3 to HLA-C. Crystal structures of KIR2DL2 in complex with HLA-Cw3, show the

KIR molecule is situated directly over residues 7-9 of the peptide resulting in direct

contact between KIR and residues 7 and 8 (Boyington et al, 2000). In contrast mouse

studies suggest that MHC binding to the Ly49 family of receptors is generally

independent of peptide although Ly49C and Ly49I exhibit a degree of peptide

specificity in their recognition of H-2Kb and H-2Kd (reviewed Deng and Mariuzza 2006).

1.5 Departures from the Simple C1/C2, Bw4/Bw6 Inhibitory Model

1.5.1 HLA-C*1402 as a Ligand for KIR2DL2 and KIR2DL3

HLA-C*1402, has a serine at position 77 and an asparagine at position 80 (C1-group)

and would therefore be expected to bind to the NK receptors KIR2DL2 and KIR2DL3.

In 1993, Colonna and colleagues reported that HLA-C*1402 behaved in an unexpected

manner. They established the specificity of NK clones as NK-1 and NK-2 (equivalent of

C1-dependent and C2-dependent clones in current terminology) using the chromium-

51 cytotoxicity release assay and PHA blast targets from individuals with different HLA-

C alleles. However, they found that five group 1/group 2 HLA-C heterozygous target

cells were not protected from lysis by NK clones that were inhibited by target cells

expressing HLA-C alleles with Ser77 and Asn80 (C1-dependent clones). It was

determined by sequence analysis that each of these targets had HLA-C*1402, which

has Ser77 and Asn80. Due to the absence of an anti-HLA-C*1402 specific antibody for

Chapter 1

16

HLA-C*1402 it could not be determined whether the allele was expressed on the cell

surface. Therefore it was suggested that lack of inhibition via HLA-C*1402 may be due

to poor or no expression of the allele at the cell surface or alternatively, due to the

allele being unable, or only weakly able, to bind to its receptor.

1.5.2 KIR3DL1 and HLA-B

Amino acids 77-83 contribute to the Bw4 and Bw6 public epitopes. Only amino acids

82 (leucine) and 83 (arginine) are common to all Bw4 alleles but other amino acids

between 77-83 are typically present in Bw4 positive alleles and not in Bw6 positive

alleles. In 1994, Cella and colleagues defined a new NK cell clone allo-specificity, NK-

3. NK cell clones with the NK-3 specificity were inhibited by PHA blasts expressing the

Bw4 epitope. The NK receptor interacting with Bw4 was identified as NKB1 (current

nomenclature KIR3DL1) (Litwin et al, 1994). Using the chromium-51 cytotoxicity

release assay, Cella et al (1994) reported greater levels of inhibition of NK3 specific NK

clones by target cells whose Bw4 allele included an isoleucine at residue 80 compared

to target cells expressing an HLA-Bw4 allele with a threonine at this position. They also

showed that NK-3 specific NK cell clones were also inhibited by cells expressing HLA-

A alleles that share the Bw4 motif including isoleucine at residue 80. This suggested

that some HLA-A alleles could bind to KIR3DL1. Subsequently, D’Andrea et al (1995)

showed that the predominant influence which determines binding to KIR3DL1 is the

amino acid at position 80. HLA-Bw4 alleles express either an isoleucine (Ile) or

threonine (Thr) at this position.

Gumperz and colleagues (1995) also demonstrated the inhibition of NK cells

expressing the NKB1 receptor by targets expressing Bw4-positive alleles. They also

investigated influence of the amino acid at position 80. NKB1 positive NK clones

generated from two donors were tested by chromium release against a panel of

721.221 cells transfected with Bw4 alleles (B*1513 (80Ile), B*2705(80Thr),

B*5101(80Ile) and B*5801 (80Ile)) or Bw6 alleles (B*0702, B*1501, B*1502, B*1508

Chapter 1

17

and B*3501) as well as HLA-A and HLA-C transfectants. NKB1-positive NK clones

were all inhibited by Bw4 expressing transfectants and not by the Bw6 expressing

transfectants. NKB1-positive NK clones were less effectively inhibited by the HLA-

B*2705 (80Thr) transfectant than the HLA-B*5801 (80Ile) transfectant. This was in

accordance with the data presented by Cella and colleagues (1994). However, HLA-

B*1513, which has an isoleucine at position 80 and would therefore be expected to

bind strongly to NKB1 gave inhibition comparable to that of HLA-B*2705. Whilst this

data was not entirely consistent with the results of Cella and colleagues, it did support

the idea that different HLA-B alleles have different affinities for KIR3DL1.

Gumperz and colleagues also investigated the potential of HLA-A alleles to inhibit

NKB1-positive (KIR3DL1) NK clones. The Bw4-positive HLA-A alleles, HLA-A*2403

and HLA-A*2501, were transfected into the 721.221 cell line. Expression of HLA-

A*2403 and HLA-A*2501 protected the transfectants from lysis by NKB1-positive NK

clones, to a similar degree as transfectants expressing Bw4-positive HLA-B alleles.

However, addition of DX9, an antibody specific for NKB1, did not result in reversal of

inhibition by the HLA-A transfectants. This was in stark contrast to HLA-B alleles in

which the DX9 antibody always reversed the inhibition seen. Thus it was concluded

that either NK cell binding to HLA-A alleles utilised a different part of the receptor or

that the binding to HLA-A alleles was via a different, perhaps unknown receptor

altogether.

Currently 17 different alleles of KIR3DL1 have been described, some of which differ in

density of surface expression. KIR3DS1 an activating receptor with high homology to

KIR3DL1 in the Ig-domains, also segregates as an allele of KIR3DL1. KIR3DL1 surface

density may be high, low or absent depending on the allele (Gardiner et al, 2001).

KIR3DL1*004 is retained in the cytoplasm due to two substitutions in the first two

immunoglobulin domains and is therefore not expressed on the surface of the cell. It

has been speculated that these substitutions effect the folding of the protein and

Chapter 1

18

consequently its ability to be expressed on the cell surface (Pando et al, 2003). The

reason for differences in expression between the “low” and “high” expression alleles is

less clear. It is possible that there is no real difference in membrane density between

“low” and “high” alleles. Rather differences in amino acid substitutions in the

immunoglobulin domains between the two types of alleles may influence binding of the

antibody DX9 to the receptor due to failure to form the DX9 epitope (Rojo et al, 1997;

Gardiner et al, 2001). Gardiner and colleagues also suggested that it is possible that

NK cells with the low expression phenotype simply have a lower number of KIR3DL1

molecules on the surface or that a large majority of those expressed did not bind to the

DX9 antibody. The low expression of other alleles, such as KIR3DL1*007, can not

easily be explained by immunoglobulin domain substitutions as the only amino acids

unique to KIR3DL1*007 are in the transmembrane region and cytoplasmic domain.

Gardiner and colleagues suggested that cytoplasmic domain substitutions may alter

folding of the receptor or translocation to the cell surface resulting in the low expressing

phenotype.

1.6 KIR3DL2 and HLA-A

KIR3DL2 is a framework gene present on all examples of chromosome 19 (Valiante et

al, 1997). It has been suggested that KIR3DL2 may recognise allelic determinants of

HLA-A alleles, but this remains controversial. Prior to discovery of KIR3DL2, Storkus

and colleagues (1991) found that the transfectants expressing HLA-A2 allele did not

protect target cells from NK mediated lysis whereas transfectants expressing HLA-A3, -

Aw68, and -Aw69 were all resistant to NK cell lysis. Storkus et al, attributed the

inhibition to the fact that HLA-A3, -Aw68, and -Aw69 all have an Asp-74 residue in the

α1 domain whereas HLA-A2 has a His-74 residue. Site-directed mutation of the His-74

to Asp-74 conferred resistance to NK cell lysis. This suggested that the Asp-74 residue

was important in binding to the inhibitory ligand on the NK cell. These studies were

undertaken prior to the discovery of the KIR genes and it was not known which KIR or

other receptor was mediating inhibition.

Chapter 1

19

Other studies have provided evidence that HLA-A3 (Dohring et al, 1996; Pende et al,

1996) and HLA–A11 (Gavioli et al, 1996; Pende et al, 1996) are ligands for KIR3DL2.

Both HLA-A3 and -A11 have the Asp-74 residue that was suggested to be important by

Storkus et al (1991). Dohring et al (1996) investigated the specificity of binding of a 70-

kDA receptor encoded by NKAT-4 (later to be known as KIR3DL2). They transfected

class I HLA negative target cells with different HLA alleles. NK clones were derived

from peripheral blood lymphocytes (PBL) and KIR expression determined by flow

cytometry using 5.133 mAb against the 70kDA receptor (KIR3DL2). NK clones that

were KIR3DL2 positive were specifically inhibited by HLA-A3 transfectants, but not by

transfectants expressing any of the other HLA-A, -B or -C alleles tested. Consistent

with the data of Storkus et al (1991), target cells expressing HLA-A2 were lysed. Target

cells expressing HLA-A11 were not tested. Dohring and colleagues did not test NK

clones that were negative for the NKAT-4 receptor against transfectants expressing

HLA-A3. It would have been important to determine whether such NK clones that did

not express NKAT-4 were able to lyse target cells expressing HLA-A3 in order to

determine whether another receptor could also bind to HLA-A3. However, blocking

NKAT-4 with an anti-NKAT-4 antibody increased NK cytotoxicity of target cells

expressing HLA-A3 suggesting that the inhibition observed for HLA-A3 was mediated

by NKAT-4. Dohring and colleagues did not test for the expression of CD94/NKG2A,

which binds to HLA-E. As HLA-A3 is one of the alleles whose signal peptide promotes

HLA-E expression, HLA-A3 may have induced HLA-E expression and any inhibition of

cytotoxicity may have occurred via CD94/NKG2A. However, transfectants expressing

HLA-A2, and -68 (which also promote HLA-E expression), were susceptible to killing by

NK cell clones expressing NKAT-4. Therefore, it appears more likely that HLA-A3

binding to NKAT-4 was responsible for the inhibition of cytotoxicity rather than

CD94/NKG2A.

Chapter 1

20

Pende and colleagues also examined the interaction between HLA-A3 and KIR3DL2.

After analysis of receptor expression, NK cell clones were selected based on the

expression of a then unidentified receptor (later shown to be KIR3DL2). Expression

was determined by two mAbs termed Q66 and Q241. These mAbs were derived from a

BALB/c mouse immunised with an NK cell clone that was inhibited by HLA-A3. Further

NK clones were identified using these mAbs and these clones were tested against a

panel of target cells expressing various HLA alleles: HLA-A1, -A2, -A3, -A11, -A24, -B7,

-B27, -B35, -B44, -B51, -Cw1 -Cw4 and –Cw5. Lysis of all target cells occurred except

where the target expressed HLA-A3 or -A11. Addition of the mAb Q66 resulted in

reversal of inhibition of lysis of targets expressing HLA-A3 or –A11. These NK clones

did not express the p58 or p70 receptor, but they did express CD94/NKG2A. However,

blocking of CD94/NKG2A with a mAb did not restore lysis of target cells expressing

HLA-A3. This evidence supports the findings of Dohring et al (1996) and Storkus et al

(1991) and suggests that KIR3DL2 binds an epitope expressed on HLA-A3.

Whilst Storkus demonstrated that site-directed mutation of His-74 in HLA-A2 to Asp-74

conferred resistance to NK cell lysis and concluded from this the importance of Asp-74,

it seems highly unlikely that Asp-74 is the only critical amino acid. All common HLA-A

alleles express Asp-74, with the HLA-A2 family being the only alleles to express His-

74. If this position was the only critical position, all HLA-A alleles, (apart from HLA-A2)

would bind to KIR3DL2, but as Pende and colleagues have demonstrated that HLA-A1

and HLA-A24 alleles do not protect targets from lysis by NK cells expressing KIR3DL2.

This suggests that other residues are important in binding of HLA-A alleles to KIR3DL2.

As the amino acid encoded at position 80 is critical in KIR recognition of HLA-B and

HLA-C alleles it is worth considering amino acid in relation to KIR3DL2. All HLA-A

alleles have either isoleucine or threonine at position 80 with HLA-A2, HLA-A3, HLA-

A11 and HLA-A68 all encoding threonine. As HLA-A3 and HLA-A2 differ in their

inhibition of KIR3DL2 expressing NK clones, this suggests that amino acid 80 may not

be critical for KIR3DL2 recognition of HLA-A.

Chapter 1

21

Valiante and colleagues (1997) were unable to demonstrate that HLA-A3 interacts with

KIR3DL2 as shown by Dohring et al (1996) and Pende et al (1996). Valiante and

colleagues tested NK cells from two donors against a panel of target cells expressing

various HLA alleles (section 1.7.1). Both donors had NK cell clones that expressed

KIR3DL2 and that were inhibited by cells expressing HLA-A*0201 and HLA-A*0301.

However this inhibition was shown not to be due to the expression of KIR3DL2 as

addition of anti-KIR3DL2 did not reverse inhibition. NK clones that were inhibited by

HLA-A*0301 also expressed CD94/NKG2A. They concluded that any inhibition of

target cells expressing HLA-A3 occurred via CD94/NKG2A. The identification of the

epitope that constitutes a ligand for KIR3DL2 remains controversial.

1.7 Rules Governing NK Alloreactivity

Valiante and colleagues (1997) elucidated some of the roles governing cytotoxic NK

cells and NK alloreactivity. They studied the NK cell receptor expression of two donors:

PP and NV. These donors differed in their KIR gene repertoire and HLA type. NK cell

clones from each donor selected for cytotoxic activity against the class I negative target

721.221 were tested for cytotoxicity against target cells transfected with HLA class I

genes. Some NK clones could be inhibited by HLA-C alleles. Cytotoxic NK clones from

donor PP that expressed KIR2DL3 were inhibited by cells expressing the C1 epitope.

NK clones from donor NV that expressed KIR2DL2 were also inhibited by cells

expressing the C1 epitope. Other NK clones from NV expressed KIR2DL1 and these

clones were inhibited by cells expressing the C2 epitope. NV also had clones that could

be inhibited by HLA-B alleles with the Bw4 epitope. These clones expressed KIR3DL1.

Still other clones could be inhibited by HLA-A alleles. Both PP and NV had NK cell

clones that were inhibited by cells expressing HLA-A*0201 and HLA-A*0301. However

this inhibition was not due to the expression of KIR3DL2 (Section 1.5.3), but rather

occurred via the CD94/NKG2A receptor present in both donor’s NK cell receptor

repertoire.

Chapter 1

22

In an allogeneic BMT setting NK cells from one individual may be able to kill cells from

another individual if the targets lack the appropriate class I HLA ligands. Valiante et al

(1997) demonstrated that NK clones from NV could kill cells from PP. PP lacks the

HLA-C ligand for KIR2DL1, an inhibitory receptor present on some NK clones from NV.

Any alloreactive clones from NV that could kill PP targets expressed KIR2DL1. NK

clones from PP displayed no alloreactivity when mixed with cells from NV. This was

because NV possessed all the HLA class I ligands for PP’s inhibitory receptors.

The general rules governing NK cell cytotoxicity that emerged from these studies were:

1. Every NK cell capable of cytotoxicity expresses at least one inhibitory receptor for

self-class I HLA.

2. The inhibitory receptor can be a KIR, which recognises allelic determinants of class

I HLA molecules, but if this KIR does not recognise self class I HLA, then it cannot

be the only inhibitory receptor on that NK cell.

3. The inhibitory receptor on those NK cells may be CD94/NKG2A, which recognises

HLA-E, or ILT-2, which recognises HLA-G and certain other class I HLA.

1.8 Activating KIR

Structurally similar to their inhibitory counterparts, it is thought that the activating KIR

(KIR2DS1, KIR2DS2, KIR2DS3, KIR2DS4, KIR2DS5 and KIR3DS1) may also

recognise class I HLA but this has been difficult to demonstrate. In 1995, Morretta and

colleagues demonstrated that some NK clones that lyse C2 expressing targets react

with the anti-CD158a antibody which recognises KIR2DL1 and its activating

counterpart, KIR2DS1. If these clones expressed KIR2DL1 inhibition of lysis would be

Chapter 1

23

expected. Activation of these NK clones by C2 homozygous targets was abolished by

addition of anti-CD158a therefore suggesting that these NK clones are activated by C2

binding to the activating receptor KIR2DS1. Despite this early data there has been little

additional evidence of activation of NK cells through KIR2DS1 recognition of the C2

ligand. Both Biassoni et al (1997) and Vales-Gomez et al (1998) using binding assays

demonstrated weak binding of KIR2DS1 with HLA-Cw4 (C2) comapred with strong

binding of KIR2DL1 with HLA-Cw4 questioning the physiological relevance of this

interaction.

Likewise, an interaction between KIR2DS2 and C1 group HLA-C alleles has been

difficult to demonstrate. KIR2DS2-Ig fusion proteins were used to study the interaction

between KIR2DS2 and C1 (Winter et al, 1998). The KIR2DS1-Ig fusion protein failed to

bind to 721.221 cells transfected with HLA-C*0304 (C1). However, mutation of the

amino acid at position 45, which is unique to KIR2DS2, from a tyrosine to a

phenylalanine (present in KIR2DL2) resulted in binding to HLA-C*0304. Saulquin and

colleagues (2003) solved the crystal strucutre of KIR2DS2 thereby providing supporting

evidence of a lack of interaction between KIR2DS2 and C1. The tyrosine at position 45

and the glutamine at position 71 of KIR2DS2 slightly alter the structure of KIR2DS2 in

such a way as to make interaction with C1 unlikely.

An interaction between KIR3DS1 and Bw4 positive HLA-B alleles also remains to be

formally demonstrated however a correlation between carriage of both KIR3DS1 and

Bw4 and AIDS progression suggests a biological interaction (Martin et al, 2002). It is

possible that the ligands for the activating KIR may not even be class I HLA. Rather

they may recognise stress related or pathogen enduced molecules or viral antigens. If

HLA moleucles are shown to be ligands for the activating KIR, it may be possible for

activating KIR to be exploited in NK cell alloreactivity.

Chapter 1

24

1.9 Allogeneic Bone Marrow Transplantation

This section of the lit review deals with mechanisms of rejection, GvHD and the graft

versus leukemia (GvL) effect with a particular focus on the role of NK cells. A review of

the recent evidence for a beneficial role of NK cell alloreactivity in HSCT will follow.

BMT is commonly used in clinical practice as a curative treatment for haematological

malignancies as well as non-malignant disorders and diseases of the haematopoietic

system, such as severe aplastic anaemia. Over the past 50 years the use of BMT has

increased greatly, paralleled by our understanding of the MHC and HLA genes.

When leukaemic patients fail conventional chemotherapy and radiation therapy, high-

dose chemotherapy and/or myeloablative irradiation is used to destroy the patient’s

leukaemic cells but it also destroys the patient’s haematopoietic system. The purpose

of a BMT is to replace the patient’s own haematopoietic system with a new one from

the donor.

1.9.1 Complications of BMT

1.9.1.1 Leukemic Relapse

Leukaemic relapse is a major problem after BMT. The risk of relapse depends on the

type of leukaemia, the stage of the leukaemia (eg, first or second remission), the age of

the recipient and whether or not it is the first BMT or subsequent BMT (Kernan et al,

1993). Interestingly, the risk of relapse is influenced by compatibility between donor

and recipient. An increase in mismatching between donor and recipient lowers the

probability of leukaemic relapse because donor immune cells are better able to

recognise the patient’s incompatible leukaemic cells.

Chapter 1

25

1.9.1.2 Rejection

Despite its current wide usage, BMT still remains a high-risk treatment. Graft rejection

occurs in about 5% of transplants and usually results in the death of the patient. Graft

rejection is mediated by the recipient’s residual immune system reacting against

incompatibilities present on the donor’s stem cells. Residual host T cells play an

important role in the process of acute rejection (reviewed in Buckley, 2003).

Coordinated response between residual activated T cells and APC results in acute

rejection. Evidence from murine models dating back to the early 1970s (Cudkowicz and

Bennett 1971; Murphy et al, 1987) suggest that NK cells may also mediate graft

rejection. Depletion of donor T cells from the donor’s marrow before transplantation has

been shown to increase the likelihood of rejection (Poynton 1988), suggesting that the

likelihood of rejection is determined by a battle between the host’s residual T and NK

cells and the donor T cells.

1.9.1.3 GvHD

GvHD is a common complication of allogeneic BMT occurring in 20-50% of allogeneic

HLA-identical sibling transplants (Tabbara et al, 2002). It is caused by donor T cells

present in the infused marrow reacting against incompatibilities present on recipient

tissues. GvHD can be acute or chronic with different pathologies for each. GvHD is

more common when donor and recipient are mismatched for HLA alleles, but can still

occur even if the recipient is HLA identical to the donor due to mismatching of minor

histocompatibility loci (Petersdorf et al, 1999). The onset and severity of GvHD is

dependent on the degree of HLA mismatching between the recipient and the donor

(Hansen et al, 1999).

Chapter 1

26

1.9.2 Matching donor and recipient for BMT

The recognition of histocompatibility antigens greatly improved the efficacy of

transplantation of both solid organs and bone marrow. The HLA antigens are the most

important determinants of compatibility in allogeneic transplantation (Hansen et al,

1999). Therefore it is preferable to chose donor and recipient pairs with identical HLA

types. However only 25-30% of patients have an HLA-identical sibling and therefore

require an HLA matched unrelated donor. For the 25% of patients who can not find an

unrelated HLA identical match, a haploidentical BMT can be performed. A

“haploidentical” match refers to donors who share only one haplotype with the

recipient. As both parents share one haplotype with an individual, most patients will

have a haploidentical relative available. Therefore research has been undertaken to

investigate methods that will enable use of haploidentical donors. The potential for

complications arising from donor-recipient incompatibility are, of course, even greater

after haploidentical BMT than after HLA matched related or HLA matched unrelated

BMT due to the large number of HLA incompatibilities on the non-shared haplotype. It

is in this setting that NK cells may play a role responding to the absence of self-ligands

on the recipient’s cells.

1.9.3 Cells Mediating Graft versus Host Disease

1.9.3.1 Role of T cells in Graft versus Host Disease

GvHD is thought to be initiated when recipient APCs, in particular DC, present host

antigens to donor T cells (Reddy and Ferrara, 2003). Acute GvHD can be divided into

three different phases: phase 1- the pre-graft patient conditioning regime; phase 2-

donor T cell activation; phase 3: inflammatory effectors.

Before infusing the donor stem cells, the patient’s own bone marrow is destroyed by a

combination of intensive chemotherapy and radiotherapy. This results in a severe

Chapter 1

27

immune deficiency in the patient. Phase 1 of acute GvHD begins during this process.

Matzinger developed the “danger signal theory” that postulates that the immune system

is up-regulated in times of tissue stress or damage (the immune system senses

‘danger’) and this leads to the immune system being on a heightened alert for foreign

antigens (Matzinger, 1997). Damage to the host’s tissues caused by the conditioning

regime prior to transplantation causes the production of inflammatory cytokines and up-

regulates adhesion and MHC molecule expression predisposing the recipient to acute

GvHD following transplantation.

In Phase 2 of acute GvHD, donor T cells are activated subsequent to their interaction

with host APC (Reddy and Ferrara, 2003; Ferrara et al, 1999). TH cells recognise

foreign peptide in the context of class II HLA molecules on the surface of the APC and

CTL recognise foreign peptide in the context of MHC class I on the surface of donor

cells. Murine studies (Shlomchik et al, 1999) have demonstrated that the removal of

host APC reduces the severity of GvHD. Once activated, donor T cells produce an

array of cytokines and effector molecules. TH1 cytokines are produced preferentially

(Reddy and Ferrara, 2003) with IL-2 and IFN-γ being two of the most important

cytokines implicated in the pathogenesis of acute GvHD. IL-2 is important in the

activation of T cells and other effector cells such as NK cells. IFN-γ up-regulates

expression of adhesion molecules and MHC molecules and the production of

chemokines, thus increasing antigen presentation of foreign peptides (Ferrara et al,

1999).

Phase 3 of acute GvHD is initiated after the production of cytokines and effector

molecules by T cells. Activated T cells recruit an array of immune effector cells

including mononuclear phagocytes and NK cells. These effector cells, attack the host’s

organs and, in particular, the skin, the gastrointestinal tract and the liver (Sviland,

2000). Both CTL and NK cells are thought to be responsible for tissue damage (Ghayur

et al, 1987; Ferrara et al, 1989; Hill and Ferrara 2000). Tissue damage is mediated via

Chapter 1

28

the Fas/Fas ligand and the perforin/granzyme pathways (Kagi et al, 1994). T cells also

recruit mononuclear phagocytes.

As T cells are the primary initiators of GvHD, various studies, both human and mouse,

have attempted to reduce GvHD by T cell depletion of donor marrow. Early murine

studies (Uphoff, 1958) demonstrated that a graft that did not contain immunologically

competent cells resulted in less GvHD. Dicke et al (1968) confirmed in murine models

that the selective removal of donor T cells resulted in decreased frequency and severity

of GvHD.

The success of T cell depletion in murine studies led to its use in human BMT where it

has been shown to be associated with a major reduction in the risk of GvHD (reviewed

in Poynton, 1988). T cell depleted BMT was first described in relation to mismatched

transplants as these transplants would otherwise have an increased risk of GvHD

(Hansen et al, 1999). However, since GvHD also occurs with HLA identical donors, T

cell depletion has also been used for these transplants with equal efficacy in terms of

reducing GvHD (Atkinson et al, 1987). The beneficial effect of T cell depletion on GvHD

has been reviewed by Poynton (1998) and Butturini and Gale (1999). Unfortunately T

cell depletion also leads to an increase in graft rejection and leukaemic relapse.

Therefore T cell depletion is now generally reserved for use with HLA mismatched

donors.

1.9.3.2 Role of Natural Killer cells in GvHD

The role of NK cells in the pathogenesis of GvHD is less clear. Little is known about the

role of NK cells in GvHD in humans and most evidence for any role comes from murine

models. NK cells are implicated in the third phase of acute GvHD (Reddy and Ferrara,

2003). There is evidence from both humans and animal models (reviewed Reddy and

Ferrara, 2003), that NK cells, along with CTL, may be important effectors in the

Chapter 1

29

destruction of tissue. Ghayur and colleagues (1987) investigated the role of NK cells in

GvHD in strains of beige mutant mice (mice homozygous for the beige gene lack NK

cells). Mice were injected with lymphoid cells from either homozygous beige (bg/bg) or

heterozygous mice (bg/+). Early splenomegaly and pronounced immunosuppression

were observed in both bg/bg and bg/+ mice suggesting these effects were mediated by

donor T cells. However, only the mice injected with lymphoid cells from the bg/+ mice

had persistent GvH reactions and associated tissue damage which correlated with an

increase in NK cell cytotoxicity (measured by the 51Cr release assay) of donor origin.

NK cells were further implicated in GvH-associated tissue damage by Ferrara et al

(1989). Murine donor LGL were found infiltrating epithelial cells, mainly of the skin, liver

and intestinal tract, tissues typically associated with GvHD. Rhoades and colleagues

(1993) provided evidence for a role of NK cells in GvH-associated tissue damage in

humans. They found a strong association between high numbers of circulating NK cells

during acute GvHD. NK cells were the predominant cell infiltrating skin biopsies taken

from patients with GvHD with no increase in CD3+ T cells nor B cells. Thus suggests

that NK cells are the mediators of tissue destruction in GvHD.

In summary, the evidence outlined above suggests that donor T cells are the initiators

of GvHD after BMT and there is little evidence that NK cells play a role in initiating

GvHD. However, NK cells appear to play an important role in GvH-associated tissue

damage.

1.9.4 Cells Mediating the Graft versus Leukaemia Effect

The main purpose of a BMT is to replace the patient’s haematopoietic system which is

destroyed by lethal doses of irradiation used to kill the leukaemic cells. Leukaemic

relapse is quite common following BMT and it is much harder to treat recurrent

leukaemia then the primary leukemia. One of the advantages of allogeneic BMT over

Chapter 1

30

autologous BMT is the lower incidence of leukaemic relapse following allogeneic BMT.

This has been termed the GvL effect. It is thought that donor lymphocytes infused with

the transplanted bone marrow destroy any remaining leukaemic cells (Farag et al,

2002).

Early murine studies (Barnes et al, 1956) established the concept that bone marrow

allografts mediate an anti-leukaemia effect. Leukaemic mice infused with syngeneic

marrow could not be cured, whereas mice infused with allogeneic marrow were able to

clear the leukaemia but subsequently died from GvHD. Weiden and colleagues (1979)

were among the first to propose a relationship between leukaemia relapse and GvHD

in humans. In allogeneic (HLA-identical siblings) BM, they found that the leukemic

relapse rate was 2.5 times lower in transplants with GvHD compared to those without

GvHD (P<0.01). Other authors have reported that the prevalence of GvHD is greater

and leukemic relapse lower in HLA mismatched BMT than in HLA matched BMT,

supporting the idea that the same immune responses that causes GvHD also reduces

leukaemic relapse (Weiden et al, 1979; Weiden et al, 1981; Horowitz et al, 1990).

The GvL effect exhibited in HLA-identical siblings is thought to occur due to differences

in minor histocompatibility antigens because donor and recipient are matched for the

major histocompatibility antigens. Donor T cells recognise these antigens as foreign.

Therefore, the GvL effect can be mediated by T cells against minor histocompatibility

antigens (Riddell et al, 2003). The GvL effect in HLA mismatched BMT is likely to be

directed against the major histocompatibility antigens. Given class I HLA mismatch,

both T cells and NK cells are potentially involved. The GvL effect is also more profound

in certain leukemias. Aversa and colleagues (1998) studied patients with leukaemia

who underwent T cell depleted BMT from haploidentical relatives. They concluded that

AML and chronic myelogenous leukaemia (CML) in the chronic phase exhibit a greater

GvL effect than acute lymphocytic leukaemia (ALL).

Chapter 1

31

1.9.4.1 Role of T cells in GvL effect

Section 1.6.3.1 discussed T cell depletion of donor bone marrow as a method of

preventing GvHD. Although successful, it is associated with an unacceptable increase

in the rate of leukaemia relapse (Butturini and Gale, 1988). In one study of HLA

identical sibling BMT, the risk of relapse was 20% without T cell depletion and 40%

with T cell depletion (Butturini and Gale, 1988). This suggests an important role for T

cells in the GvL effect. Donor T cells are capable of recognising both major and minor

histocompatibility molecules on the surface of the leukaemic cells (reviewed Riddell et

al, 2003). It seems likely that the recognition of minor histocompatibility antigens is

responsible for the GvL effect exhibited in HLA-identically matched related individuals,

as there is no MHC incompatibility. In vitro assays have demonstrated that T cells

specific for minor histocompatibility antigens can lyse recipient leukaemic cells and

prevent leukaemic colony formation (Warren et al, 1998).

1.9.4.2 Role of Natural Killer cells in GvL effect

Until very recently the role of NK cells in the GvL effect has been less clear and

evidence in humans has been mainly circumstantial. For example, any GvL effect seen

after T cell depletion has been postulated to be due to NK cells (Poynton, 1988). NK

cells are one of the first cells to recover following both allogeneic and autologous BMT

(Poynton, 1988). Therefore they could potentially be involved in the elimination of

residual leukemic cells. Evidence for NK cells mediating the GvL effect in a

haploidentical setting in humans has recently been described and will be reviewed in

detail in section 1.7

Chapter 1

32

1.10 Recent Evidence of the Role of NK Cells in BMT

1.10.1 Evidence that Alloreactive NK Cells can Eliminate Leukaemic Cells

Alloreactive T cells have been implicated in mediating the GvL effect observed in

allogeneic BMT (Poynton, 1988). Ruggeri and colleagues (2002) performed BMT on 92

patients with advanced AML or ALL. All donors shared only one haplotype with the

patient and some donors were mismatched for the known NK epitopes (C1, C2 and

Bw4) allowing allorecognition of recipient cells by the donor NK cells. To determine

whether or not a GvL effect was exhibited, the five-year probability of leukaemic

relapse in both AML and ALL patients was examined (Table 1.1).

Table 1.1 The five-year probability of leukaemic relapse for AML and ALL

patients with and without ‘NK mismatches’.

KIR Ligand Incompatibility in the GvH Direction

NO YES

Relapse in AML 75% 0%

Relapse in ALL 90% 85%

(Adapted from Ruggeri et al, 2002)

‘NK mismatches’ decrease the chance of leukaemic relapse for AML patients but not ALL

patients

As shown in Table 1.1, in transplants for AML patients, in which there was KIR ligand

incompatibility in the GvH direction, a strong GvL effect was exhibited. This GvL effect

was postulated to be mediated by NK cell killing of leukaemic cells due to the absence

of the appropriate inhibitory ligand on the target leukaemic cell. However, in transplants

for ALL patients, the five-year probability of leukaemic relapse only slightly decreased

with KIR ligand incompatibility in the GvH direction. Thus the GvL effect was evident in

AML patients, but not in ALL patients. This finding correlated with in vitro evidence that

ALL cells were relatively resistant to NK cell lysis (Ruggeri et al, 1999). Alloreactive NK

Chapter 1

33

cells that had previously been shown to be able to kill allogeneic PHA lymphoblasts or

EBV-transformed B lymphoblastoid cell lines (BLCL) of leukemia patients were then

tested against AML, CML or ALL cells from the same patients. AML and CML cells

lacking the relevant HLA epitopes were lysed, but ALL cells from only 2 out of 5

patients were lysed. To determine why ALL cells were less susceptible to NK cells

mediated lysis, the expression of several adhesion molecules was determined by flow

cytometry. ALL cells resistant to lysis by NK cells exhibited low expression of the

lymphocyte function antigen-1 (LFA-1) compared with susceptible CML and AML cells.

This data suggests that NK cells require the expression of LFA-1 on the surface of the

target cell, in addition to the absence of a ligand for the inhibitory KIR, to be able to

bind and lyse the target cell.

Having shown that NK mismatches may induce the GvL effect, murine studies were

conducted to investigate the mechanisms behind these results. Ruggeri and colleagues

(2002) injected human CML cells into severe combined immunodeficient (SCID) mice.

Mice developed advanced leukemia within 6 weeks and died if left untreated or infused

with human NK cells that were not alloreactive towards the human leukemic cells.

However, mice infused with human NK cells that were alloreactive towards leukemic

cells cleared the leukaemia and survived. Thus alloreactive NK cells were not only able

to kill leukaemic cells in vitro, but they were also capable of clearing leukaemia in an

animal model.

1.10.2 Evidence that Alloreactive NK cells can Prevent GvHD

Haploidentical transplants performed by Ruggeri et al (2002) also suggested that

alloreactive NK cells protect against GvHD. The prevalence of GvHD was 0% in

transplants with potential for NK alloreactivity and 13.5% in transplants without the

potential for NK alloreactivity. Ruggeri and colleagues investigated the mechanisms by

which GvHD was reduced in a murine model. H-2b mice transplanted with non-T cell

Chapter 1

34

depleted marrow from H-2d mice died from GvHD. However, if recipient mice were

infused with alloreactive NK cells from H-2d mice as part of the conditioning regime

prior to infusion of stem cells, 100% of the mice survived without developing GvHD.

Even with escalating doses of donor T cells, all mice still survived. Mice infused with

non-alloreactive NK cells prior to transplant developed GvHD. They provided evidence

that alloreactive NK cells prevent GvHD by eliminating recipient APC, thereby

preventing interaction with donor T cells which would otherwise initiate GvHD. These

experimental findings were consistent with lower prevalence of GvHD observed in

human transplants, in which there was potential for NK alloreactivity.

In summary, Ruggeri and colleagues provided evidence that alloreactive NK cells could

eliminate leukaemic cells in vitro and in animal models. They also postulated that the

GvL effect exhibited in human haploidentical BMT was mediated by alloreactive NK

cells. The low occurrence of GvHD evident in the human haploidentical BMT was also

postulated to be due to alloreactive NK cells lysing recipient APC.

1.10.3 Prediction of NK alloreactivity using HLA and KIR Genetics

Not all mismatches of KIR related HLA epitopes result in NK alloreactivity. Therefore it

is not possible to reliably predict NK alloreactivity based on the donor’s HLA type and

KIR repertoire and the recipient’s HLA type. It has been reported that HLA-C*1402 (C1)

may not be a ligand for KIR2DL2/3 despite having the appropriate amino acid at

position 80 for a C1-group allele (Colonna et al, 1993). It might be predicted therefore

that donors who have HLA-C*1402 as their only C1 allele may not be able to generate

alloreactive NK clones dependent on KIR2DL2/3. Other examples of HLA-C alleles that

do not behave as their amino acid sequence at position 80 would predict, may exist but

a survey of all common alleles has not been undertaken. Genetic polymorphisms of the

inhibitory KIR for HLA-C alleles (KIR2DL1, KIR2DL2 and KIR2DL3) also exists and the

Chapter 1

35

influence of this allelic variation on the ability to generate alloreactive NK cells has not

been examined.

Only two-thirds of HLA-Bw4 positive and KIR3DL1 positive individuals can generate NK

clones that are alloreactive towards targets that are Bw4-negative (reviewed in Ruggeri

et al, 2006). The reason for this is not clear, but it could be due, at least in part, to the

different levels of expression of the KIR3DL1 alleles (Gardiner et al, 2001).

KIR3DL1*004 is the second most common KIR3DL1 allele in Caucasions (Halfpenny et

al, 2004) but it is not expressed on the cell surface and therefore could account for a

large proportion of patients with Bw4 and KIR3DL1 who do not make KIR3DL1

dependent NK clones. It is also not known whether the high and low expression

KIR3DL1 alleles respond in the same way to targets lacking Bw4 nor whether high or

low KIR3DL1 expression affects a donor’s ability to generate KIR3DL1 dependent NK

clones. The different Bw4-positive HLA alleles may also interact differently with

KIR3DL1. Bw4-positive HLA alleles with an 80I have been reported to be better ligands

then those with an 80T (Cella et al, 1994). Therefore the ability of donors to make

KIR3DL1 dependent NK clones may be effected by their Bw4-positive HLA alleles.

The role of activating KIR in mediating alloreactivity is less well defined. The ligands for

the activating KIR remain controversial but are believed to be the same as for their

inhibitory counterparts because of the structural similarity between the receptors.

However this has been difficult to demonstrate (Vales-Gomez et al, 1998; Winter et al,

1998; Saulquin et al, 2003). The role of activating KIR in mediating NK alloreactivity

needs investigating.

1.11 Hypotheses and Aims

Long, labour intensive functional NK cloning assays are currently used to select donors

who are able to generate alloreactive NK clones towards a patient requiring a

haploidentical transplant. A better understanding of the interaction of HLA alleles and

Chapter 1

36

KIR receptors may eventually allow suitable donors to be predicted from HLA type and

KIR genotypes.

Hypothesis

The current understanding of HLA ligands and KIR receptors will not reliably predict

HLA:KIR interaction and therefore, the ability to generate alloreactive NK clones

Aims

1. To develop a more efficient assay for the detection of alloreactive NK cells

2. To determine the ability of common HLA-C alleles to interact with KIR2DL1 and

KIR2DL2/3

3. To determine the ability of common HLA-B alleles and Bw4-positive HLA-A alleles

to interact with KIR3DL1

4. To identify a role for activating KIR in mediating NK alloreactivity

Chapter 2

37

Chapter 2

Materials and Methods

Chapter 2

38

Chapter 2: Materials and Methods

2.1 Cell Culture

2.1.1 Cell Lines

EBV-transformed B-Lymphoblastoid Cell Lines

EBV-transformed BLCL were selected from an extensive panel at the Department of

Clinical Immunology and Immunogenetics at Royal Perth Hospital (RPH). Cell lines

were maintained in RPMI 1640 and 10% sterile heat inactivated FCS (RPMI/10%) and

split every 3-4 days to 1x105 cells/mL.

721.221 Cell Line

The 721.221 cell line is a class I negative human BLCL (kindly provided by Jim

McCluskey, University of Melbourne). 721.221 cells were maintained in RPMI/10%FCS

and split every 2-3 days at 1x105 cells/mL.

RPMI-8866 Cell Line

RPMI-8866 cells (American Type Cell Collection, Manassas, USA) were maintained in

RPMI/10%FCS and split every 2-3 days at 1x105 cells/mL.

NK Cell Lines

The YTS NK cell line transfected with KIR2DL3 (kindly provided by Eric Long, NIAID)

was maintained in IMDM with 10%FSC and 1ug/mL puromycin (Sigma-Aldrich, St

Louis USA) and split every 3 days at 1x105 cells/mL.

Chapter 2

39

2.1.2 Separation of PMBC

2.1.2.1 Isolation of PBMC

Blood was obtained from laboratory volunteers and blood donors from The Australian

Red Cross Blood Service with informed consent. Blood was diluted 1/3 with 1xPBS

containing 2%FCS (PBS2%) and mixed well. 1/3 Ficoll Paque (Pharmacia, Uppsala

Sweden) was under-layed carefully. Blood was then centrifuged at 1200g for 20

minutes with the brake-off. PBMC were harvested from the interface of the ficoll

gradient and washed three times in PBS2% at 300g for 7 minutes. Cells were then

resuspended in 1mL RPMI/10%. Cells were diluted either 1/2 or 1/5 in trypan blue and

counted using a Neubauer Haemocytometer (GmbH, Munich Germany).

2.1.2.2 Isolation of PBMC from Unusable Red Cell Bags as Allogeneic Feeder Cells

Ten unusable red cell bags were obtained from normal donors from the Australian Red

Cross Blood Bank. Buffy coats were made from the blood by centrifugation at 1200g for

15 minutes. The white cell layer (buffy coat) was removed with a pasteur pipette and

diluted 1/3 with PBS2%. PBMC were isolated on a gradient as in section 2.1.2.1. After

isolation and before cell counting, PMBC from each donor were pooled.

2.1.2.3 Storage of Pooled Allogeneic PBMC

Pooled allogeneic PBMC were stored at concentrations varying from 20-50 x 106

cells/mL in 1mL freezing medium in CryoTubeTM Vials (Nalgene Nunc International,

Albertslund Denmark). The vials were initially frozen at -80°C to allow the gradual

cooling of the cells before transfer to liquid nitrogen.

Chapter 2

40

2.1.2.4 Thawing of PBMC

Cells were removed from liquid nitrogen and placed on dry ice. Each vial was thawed in

a waterbath at 37°C and immediately transferred to RPMI/10%. The cells were then

centrifuged at 300g for 7 minutes. The cells were resuspended in 1mL RPMI/10%.

Cells were diluted either 1/2 or 1/5 in trypan blue and counted using a Neubauer

Haemocytometer.

2.1.2.5 Irradiation of Cells

Pooled allogeneic PBMC were irradiated at 30Gy using the GammaCell 3000 ELAN

(MDS Nordion, Ottawa Canada) at the Institute for Child Health Research. RPMI-8866

cells were likewise irradiated at 60Gy. After radiation, both PBMC and RMPI-8866 cells

were washed twice in RMPI/10% at 300g for 7 minutes.

2.2 Natural Killer Cell Cloning

2.2.1 Enrichment of Natural Killer Cells

Prior to dilution with PBS, 50μL of RosetteSep Antibody Cocktail (Stemcell

Technologies, Vancouver Canada) per mL of blood was mixed with the blood and the

mixture was incubated for 20 minutes. The RosetteSep Antibody Cocktail is a

combination of bispecific mouse and rat monoclonal antibodies for which one arm of

the antibody is specific for cell surface antigens on human haematopoietic cells (CD3,

CD4, CD19, CD36, CD66b) and the other is specific for glycophorin A on red blood

cells. The antibodies cause non-NK cells to be complexed to red blood cells and then

eliminated by ficoll centrifugation. It is used to deplete blood of T and B lymphocytes,

monocytes, granulocytes and dendritic cells. After incubation, the NK cells were

harvested from the interface of a ficoll gradient as described in section 2.1.2.1.

Chapter 2

41

2.2.2 Natural Killer Cell Cloning

After separation, NK cells were plated out at 20 cells/100μL, 10 cells/100μL, 5 cells/

100μL and 2 cells/100μL by limiting dilutions into 96-well round bottom plates (BD,

Franklin Lakes USA). Irradiated allogeneic PBMC were added to each well at a

concentration of 1x105 cells/100μL. On day 1 (24 hours after plating), 100μL of medium

was removed and replaced with fresh medium containing 400IU/mL IL-2 (Chiron,

Emeryville USA) resulting in a final concentration of 200IU/mL. On day 4, 100μL of

medium was removed and replaced with irradiated PBMC added to each well at a

concentration of 8 x 104 cells/100μL. On day 11, 100μL of medium was removed and

replaced with 100ul fresh medium containing 200IU/mL IL-2, such that the final

concentration was 100IU/mL. On days 14-20 cell growth was monitored by light

microscopy and cells were split if necessary. When splitting cells, 100μL of fresh

medium containing 2 x 104 irradiated RPMI-8866 cells were added to each well. Cells

were screened for NK receptor expression and alloreactivity by flow cytometry and

cytotoxicity, respectively. Once NK clones were fully characterised, selected NK clones

were frozen at a concentration between 1-3 x 106 cells/mL and stored in liquid nitrogen

at -80°C.

2.2.3 Natural Killer Cell Polyclonal Cultures

After isolation, polyclonal NK cells were diluted to a concentration of 3 x 104 cells/mL.

100μL of NK cells were added to each well of a 96-well round bottom tray. 100μL of

irradiated allogeneic PBMC were added to each well at a concentration of 3 x 105

cells/mL. Every 2-3 days, 100μL of medium was removed and replaced with 100μL

fresh medium containing 200IU/mL IL-2, such that the final concentration was

100IU/mL. After 12 days of culture, the polyclonal NK cells were counted and frozen at

a concentration between 2-3 x 106 cells/mL.

Chapter 2

42

2.2.4 Restimulation of Natural Killer Cell Clones and Polyclonal Cultures

NK cells were thawed rapidly in a waterbath at 37°C and immediately transferred to

8mL warm NK medium. The cells were washed at 300g for 7 minutes and resuspended

in 1mL for counting. Cells were diluted 1/2 in trypan blue and counted using a

Neubauer Haemocytometer. The NK cells were diluted to 5 x 104 cells/mL in NK

medium containing 200IU/mL IL-2 and 100μL was added to each well of a 96-well

round bottom tray. 100μL of irradiated RPMI-8866 (section 2.1.2.5) were added to each

well at a concentration of 5x105 cells/mL in NK medium. Every 2-3 days 100μL medium

was removed and 100μL fresh NK medium containing 200IU/mL IL-2 was added. Every

12 days after restimulation the NK cells were subcultured at 5 x 104 cells/mL and fresh

irradiated RMPI-8866 was added to the NK cells at a concentration of 5 x 105 cells/mL.

2.3 Immunophenotyping

2.3.1 Flow Cytometry on Whole Blood

5 – 10μL of the appropriate conjugated antibody (appendix 2) was added to a FACS

tube. 100μL whole blood was added to the tube and incubated at room temperature for

15 minutes. Red cells were lysed after incubation using the Q-Prep Workstation

(Beckman Coulter, Fullerton USA). Flow cytometric analysis was then carried out on a

Coulter XL-EPICSTM Flow Cytometer (Beckman Coulter, Fullerton USA) and the BD

Biosciences FACSCantoTM Flow Cytometer (BD, Franklin Lakes USA).

2.3.2 NK Cell Phenotypying-Labelled Primary Antibodies

104 - 105 NK cells were added to one well of a 96-well round bottom tray and the plate

was washed twice in flow buffer (appendix 1) by centrifugation at 200g for 5 minutes.

50μL of the appropriate conjugated antibody was added to each well and incubated on

ice in the dark for 20 minutes. Cells were than washed twice and resuspended in

Chapter 2

43

100μL of flow buffer with 100μL of 1% paraformaldehyde. Cells were then stored at

4°C in the dark until analysis.

2.3.3 NK Cell Phenotypying and class I HLA Staining-Labelled Secondary

Antibodies

Staining for class I HLA antigens and certain NK cell markers required the use of

unconjugated antibodies which involved a two-step staining process. 105 – 106 cells

were added to FACS tubes. 2mL flow buffer was added and the cells washed twice by

centrifugation at 200g for 5 minutes. 50μL of the appropriate unconjugated antibody

(appendix 2) was added to each FACS tube and incubated on ice in the dark for 20

minutes. After incubation the cells were washed twice. 50ul of the appropriate

secondary antibody (appendix 2) was then added to each FACS tubes and the cells

incubated on ice in the dark for 20 minutes. After incubation the cells were washed

twice and resuspended in a final volume of 400μL containing 1% paraformaldehyde.

2.3.4 Analysis of Flow Cytometry Data

Analysis of flow cytometry was performed using BD FACSDivaTM Software (BD

Biosciences, Franklin Lakes, USA) and FlowJo Software (Tree Star Inc, Ashland USA).

2.4 Natural Killer Cell Cytotoxicity

NK cell cytotoxicity was measured by one of two methods: the 4 hour 51Cr release

assay or the CD107a assay. Briefly, NK cells were incubated with target cells in both

assays. The 4 hour 51Cr release assay involved labelling target cells with 51Cr. Effector

cells (NK cells) killing of the target cell results in the release of 51Cr into the

supernatant. The amount of 51Cr released can be sampled as a measure of

cytotoxicity. The CD107a assay involves the use of an antibody specific for the

Chapter 2

44

lysosomal-associated membrane protein-1 (LAMP-1 or CD107a) which is expressed on

degranulating (cytotoxic) NK cells.

2.4.1 Target Cell Preparation

EBV-transformed B cell lines were used as target cells in the 4 hour 51Cr release

assay. Cell lines were subcultured by making a 1 in 3 dilution two days before the

assay to ensure that they were rapidly growing when labelled with 51Cr. Approximately

16 hours before the start of the assay, the cell lines were counted and resuspended at

5 x 105 cells/mL in a 24-well plate (BD, Franklin Lakes USA). 25uCi 51Cr was added per

0.5 x 106 cells (calculated as 30μL 51Cr). The amount of 51Cr added increased as the

half-life of the radioisotope decreased and the amount added was calculated

proportionately. Target cells were incubated overnight at 37°C in 5% CO2. After

incubation cells were washed three times in RPMI/10% by centrifugation at 300g for 7

minutes. Cells were than counted and resuspended at 5 x 104 cells/mL.

2.4.2 Effector Cell Preparation

NK cell clones or NK cell lines were used as effector cells in the 4 hour 51Cr release

assay. Cells were counted and adjusted to varying concentrations in order to achieve

the appropriate effector: target ratios. A 1:1 ratio was used when screening NK clones

for alloreactivity. All other assays were performed at a 2:1 ratio

2.4.3 4 hour 51Cr Release Assay

Target cells were plated out into a 96-well round-bottom plate. Effector cells were

added at varying ratios (section 2.4.2). Spontaneous release was determined in wells

containing target cells and medium alone without effector cells. Maximum release was

determined by incubating 100μL of target cells with 100μL of 0.5% Triton X-100. Plates

were incubated at 37°C in 5% CO2 for 4 hours. After incubation, plates were

Chapter 2

45

centrifuged at 200g for 3 minutes to pellet cells. 100μL of supernatant was carefully

removed from each well without disturbing the pellet. 51Cr release was then measured

in a 1282 Compugamma Universal Gamma Counter (LKB Wallace, Turku Finland) and

expressed in counts per minute. The relative specific lysis of targets was calculated as:

Relative specific lysis = (specific release (cpm) - background release (cpm)

(maximum release (cpm) - background release)

If blocking of certain NK cell receptors was required, 2μL of receptor specific antibody

or isotype control diluted appropriately (appendix 2) was added to each well. 100μL of

effector cells were added to each well and incubated at 37°C for 20 minutes. After

incubation 100μL of target cells were added to each well and the assay proceeded as

above.

2.4.4 CD107a Assay

105 NK cells were mixed with 105 target cells in a 96 well-round bottom tray at a ratio of

1:1. 5μL of CD107a-FITC antibody was added to each well and the plate was

incubated for 1 hour at 37°C in 5%CO2. 6ug/mL monensin (Golgi-Stop, BD, Franklin

Lakes USA) was added to each well and the cells were incubated for a further 5 hours

at 37°C in 5%CO2. The cells were then either transferred to flow tubes or staining was

continued in the 96 well-round bottom tray. Cells were washed twice in flow buffer by

centrifugation at 200g for 5 minutes. The appropriate KIR antibody (appendix 2) and

CD56-PeCy7 (BD, Franklin Lakes USA) were added to the cells and the tubes or plate

incubated on ice in the dark for 15 minutes. Cells were then washed twice in flow buffer

by centrifugation at 200g for 5 minutes. Cells were resuspended in 300μL flow buffer

and analysed.

X 100

Chapter 2

46

2.5 DNA and RNA

2.5.1 DNA Extraction

DNA was either isolated from 200µL of whole blood or, up to 5 x 106 cells, using the

QIAamp DNA Blood Mini Kit (Qiagen, Valencia USA). After extraction the concentration

of DNA was read by placing 1µl of the sample on the Nanodrop (ND 1000

Spectrophotometer, Biolab, Victoria AUS). DNA was stored in 1.5mL Eppendorf tubes

at 4°C.

2.5.2 RNA Extraction

RNA was extracted from up to 5 x 106 NK cells or EBV-transformed B cell lines using

the RNeasy Protect Mini Kit (Qiagen, Valencia USA) as per the manufacturer’s

instructions. After extraction the concentration of DNA was read by placing 1µl of the

sample on the Nanodrop. RNA was stored in 1.5mL eppendorf tubes at -80°C.

2.5.3 Reverse Transcriptase PCR

cDNA was synthesised from 1μg of mRNA using oligo(dT) (Promega, Madison USA)

and omniscript (Qiagen, Valencia USA) and amplified for 90 minutes at 37°C. cDNA

was stored at -20°C.

2.6 Polymerase Chain Reaction

The polymerase chain reaction (PCR) was used to amplify specific regions of DNA

using sets of oligonucleotides complementary for each DNA strand, which at specific

temperatures, anneal to the DNA. PCR was used throughout the work described in this

thesis to amplify KIR2DL1, KIR2DS1, KIR2DL2 and KIR2DL3 cDNA and KIR3DL1

genomic DNA for allele typing by DNA sequencing. SSP-PCR was also used to

determine complete KIR genotypes. Primer sequences are outlined in appendix 3.

Chapter 2

47

2.6.1 Amplification of KIR2DL2, KIR2DL3 and KIR2DS1 from cDNA

KIR2DL2 and KIR2DL3 cDNA was produced from RNA isolated from NK cell clones

generated in section 2.2.2. Primers were designed to amplify full length cDNA of each

KIR gene. cDNA was diluted 1/10 for NK cell clones and 1/100 for polyclonal NK cell

cDNA. After PCR amplification, each PCR product was sequenced to determine which

allele each NK clone transcribed (section 2.6.6). Master mixes used in the PCR

amplification of KIR2DL2, KIR2DL3 and KIR2DS1 full length cDNA are listed in Table

2.1. Thermocycling conditions for amplification of full length KIR2DL2 and KIR2DL3

cDNA are listed in Table 2.2 and thermocycling conditions for amplification of full length

KIR2DS1 cDNA are listed in Table 2.3.

Table 2.1 Master Mix for KIR2DL2, KIR2DL3 and KIR2DS1 (cDNA) Full Length

PCR

Master Mix (1 Sample) Reagent Volume 5' primer (20pmol/ul) 0.5ul 3' primer (20pmol/ul) 0.5ul 40mM dNTP 1ul 10 x PCR buffer 25 mM MgCl2 2.5ul Sterile water 7.7ul AmpliTaq polymerase* 0.3ul cDNA 12.5ul Total Volume 25ul *(Applied Biosystems, Foster City USA)

Chapter 2

48

Table 2.2 KIR2DL2 cDNA and KIR2DL3 cDNA Full Length PCR Thermocycling

Conditions

KIR2DL2 KIR2DL3 Temperature/Time Cycle Number Temperature/Time Cycle Number 95°C x 5 min 1 95°C x 5 min 1 97°C x 20 s 97°C x 20 s 66°C x 45 s 5 66.6°C x 45 s 5 72°C x 90 s 72°C x 90 s 95°C x 20 s 95°C x 20 s 62°C x 45 s 25 62.6°C x 45 s 25 72°C x 90 s 72°C x 90 s 72°C x 10 min 1 72°C x 10 min 1 4°C HOLD 4°C HOLD Table 2.3 KIR2DS1 Full Length PCR Thermocycling Conditions

Temperature/Time Cycle Number 95°C x 5 min 1 97°C x 20 s 64°C x 45 s 5 72°C x 90 s 95°C x 20 s 60°C x 45 s 25 72°C x 90 s 72°C x 10 min 1 4°C HOLD

2.6.2 SSP-PCR for KIR2DS1 on genomic DNA and SSP-PCR for KIR2DL1 from

cDNA

Detection of KIR2DS1 from genomic DNA and KIR2DL1 from cDNA (chapter 5) was

performed using SSP primers designed by (Uhrberg et al, 1997). The master mix used

in the PCR amplification of KIR2DS1 genomic DNA is listed in Table 2.4 and in Table

2.5 for the amplification of KIR2DL1. Thermocycling conditions for amplification of both

KIR2DS1 genomic DNA and KIR2DL1 cDNA are listed in Table 2.6.

Chapter 2

49

Table 2.4 Mater Mix for KIR2DS1 SSP (Genomic DNA)

Master Mix (1 Sample) Reagent Volume 5' primer (20pmol/ul) 0.5ul 3' primer (20pmol/ul) 0.5ul HGH I (3pmol/ul) 0.5uL HGH II (3pmol/ul) 0.5uL 40mM dNTP 1ul 10 x PCR buffer 25 mM MgCl2 2.5ul Sterile water 6.7ul AmpliTaq polymerase 0.3ul Genomic DNA 12.5ul Total Volume 25ul Table 2.5 Master Mix for KIR2DL1 SSP (cDNA)

Master Mix (1 Sample) Reagent Volume 5' primer (20pmol/ul) 0.5ul 3' primer (20pmol/ul) 0.5ul 40mM dNTP 1ul 10 x PCR buffer 25 mM MgCl2 2.5ul Sterile water 7.7ul AmpliTaq polymerase 0.3ul cDNA 12.5ul Total Volume 25ul Table 2.6 KIR2DS1 SSP (Genomic DNA) and KIR2DL1 SSP (cDNA) Thermocycling

Conditions

Temperature/Time Cycle Number 95°C x 5 min 1 97°C x 20 s 64°C x 45 s 5 72°C x 90 s 95°C x 20 s 60°C x 45 s 25 72°C x 90 s 72°C x 10 min 1 4°C HOLD

Chapter 2

50

2.6.3 KIR3DL1 Allele Typing

There are currently 17 identified alleles of the KIR3DL1 gene. KIR3DL1 specific

primers situated in intronic sequence (Norman et al, 2007) and DNA sequencing using

M13 forward and reverse primers was used to assign the different KIR3DL1 alleles.

The Master mix used for the PCR amplification of KIR3DL1 genomic DNA is listed in

Table 2.7. Thermocycling conditions for amplification of KIR3DL1 genomic DNA is

listed in Table 2.8.

Table 2.7 Master Mix for KIR3DL1 Allele Typing

Master Mix (1 Sample) Reagent Volume 5' primer (20pmol/ul) 1ul 3' primer (20pmol/ul) 1ul 10mM dNTP 2ul 10 x PCR buffer 25mM MgCl2 5ul Sterile water 37.4ul AmpliTaq polymerase 0.6ul DNA (30ng/ul) 3ul Total Volume 50ul

Table 2.8 KIR3DL1 Allele Typing Thermocycling Conditions

Temperature/Time Cycle Number 94°C x 5 min 1 94°C x 10 s 65°C x 60 s 10 94°C x 10 s 61°C x 50 s 20 72°C x 30 s 72°C x 10 min 1 4°C HOLD

2.6.4 KIR Genotyping by SSP- PCR

The KIR genotype of different individuals was determined using a multiplex PCR-SSP

system developed by Sun et al (2004). Four multiplex reactions amplify all KIR genes

including the KIR2DS4 variant, KIRDS4v.

Chapter 2

51

2.6.5 Visualising PCR Products

After amplification, PCR products were visualised by electrophoresis on a 1% agarose

gel. 3ul of loading buffer was added to 10ul of PCR product and loaded into the wells

within 1% agarose gel containing ethidium bromide. 10ul of 1kb Plus Lambda DNA

ladder (Invitrogen, Carlsbad USA) was also added to one well. The gel was run at 150

volts for 20-30 minutes in an electrophoresis tank containing 0.5 x TBE. PCR products

were visualised on an UV-light transilluminator using a Bio-Rad Gel Doc (Bio-Rad,

Hercules USA). PCR products were analysed using the Quantity One software from

Bio-Rad.

2.6.6 DNA Sequencing

DNA sequencing was carried out using Big Dye terminator technology (Applied

Biosystems, Foster City USA). HLA-A, B and C typing and KIR allele typing was

performed by the Department of Clinical Immunology and Immunogenetics, RPH.

Sequence was analysed using SeqScape Software (Applied Biosystems, Foster City

USA).

Chapter 3

52

Chapter 3

KIR2DL3*001 and KIR2DL3*002 Differ in Their Ability to Interact with C1-group HLA-C Alleles

Chapter 3

53

Chapter 3: KIR2DL3*001 and KIR2DL3*002 Differ in Their Ability

to Interact with C1-group HLA-C Alleles

3.1 Introduction

The NK cell KIR receptor family, recognise allelic epitopes of class I HLA molecules. All

HLA-C alleles are divided into two groups, C1 and C2, based on their amino acids at

position 80 of the α1 domain (Mandelboim et al, 1996). C1-group alleles bind to

KIR2DL2 and KIR2DL3 while C2-group alleles bind to KIR2DL1. Individuals with an

HLA-C allele that includes the C1 epitope and whose KIR repertoire includes KIR2DL2

and/or KIR2DL3 are able to generate NK clones dependent on that receptor alone for

inhibition. In an allogeneic setting these NK clones will lyse cells from donors whose

HLA type lacks the C1 epitope. Likewise, individuals whose HLA-C alleles include the

C2 epitope and whose KIR repertoire includes KIR2DL1 are able to generate NK

clones dependent on that receptor and will lyse allogeneic targets lacking the C2

epitope. If an individual has both the C1 and C2 epitope as well as KIR2DL1 and

KIR2DL2/3, they are able to generate both KIR2DL1 dependent and KIR2DL2/3

dependent NK clones.

Although the amino acid at residue 80 is known for all HLA-C alleles, not all HLA-C

alleles have been shown empirically to bind to the predicted KIR receptor. An example

of an allele that does not behave as expected has been reported. Colonna and

colleagues (1993) demonstrated in chromium release assays that the HLA-C*1402

allele did not inhibit NK clones inhibited by other C1 alleles, despite the fact that HLA-

C*1402 has asparganine at amino acid 80 which would confer the C1 epitope. In

addition to the lack of empirical evidence for many HLA-C alleles, there are currently

five known alleles of KIR2DL2 and seven known alleles of KIR2DL3 (Robinson et al,

2003) and it is not known whether allelic differences between KIR alleles influence their

interactions with HLA-C.

Chapter 3

54

In bone marrow transplant patients, an HLA-C allele that did not interact with its

predicted receptor might confer susceptibility to alloreactive NK cells where it might not

otherwise have existed. Bone marrow transplant donors with such alleles might not be

able to generate NK alloreactivity as expected; eg if HLA-C*1402 is not a ligand for

KIR2DL2/3, it might be predicted that individuals who express HLA-C*1402 as their

only C1 positive allele would not be able to generate alloreactive NK cells dependent

on KIR2DL2/3.

Individual NK cells may express multiple inhibitory and activating receptors. To

examine the interaction of KIR2DL2 and KIR2DL3 with HLA-C alleles having the C1

epitope, NK clones were generated from several individuals to find NK clones

expressing KIR2DL2 or KIR2DL3 as their only inhibitory receptor. KIR2DL2 and

KIR2DL3 dependent NK clones were used to investigate the interaction of the common

HLA-C alleles with KIR2DL2 and KIR2DL3 alleles. The YTS NK cell line transfected

with KIR2DL3 (YTS-2DL3), represents a second approach to examining an individual

KIR receptor. The YTS NK line itself, does not express any endogenous KIR or NKG2-

family receptors. In order to investigate the interaction of different KIR2DL2 and

KIR2DL3 alleles with HLA-C, KIR2DL2 or KIR2DL3 dependent clones and the YTS NK

cell line transfected with KIR2DL3*001, were tested for inhibition of cytotoxicity against

targets expressing the common C1-group and C2-group alleles including HLA-C*1402.

3.2 Materials and Methods

Detailed materials and methods relating to this chapter are described in chapter 2.

Class I HLA typing was performed on each NK donor by the Department of Clinical

Immunology and Immunogenetics, RPH and their full KIR genotype was determined by

myself.

Chapter 3

55

Table 3.1 NK Donor Class I HLA Typing

HLA-A HLA-B HLA-C NK Epitopes

Donor 1 A*03, 68 B*07, 14 C*07, 08 C1 Donor 2 nt nt nt Donor 3 A*02, 33 B*4601, 5801 C*0102, 0302 C1 Bw4 Donor 4 A*02, 1101 B*1501, 35 C*0303, 0401 C1 C2

Donor 2 was a HLA unknown donor from the Red Cross Blood Bank. Table 3.2 NK Donor KIR Genotype

KIR2DL1 KIR2DL2 KIR2DL3 KIR3DL1 KIR2DS1 KIR2DS2 KIR2DS3 KIR3DS1

Donor 1 + - + - + - NT NT Donor 2 + - + + - - - - Donor 3 + + + + - - - + Donor 4 + + - + + + + +

KIR2DS3 and KIR3DS1 typing of Donor 1 was not performed 3.3 Results

3.3.1 Selection of NK Clones

NK clones were generated from several individuals in order to find NK clones

expressing KIR2DL2 or KIR2DL3 as their only inhibitory receptor. Such NK clones

should be alloreactive towards targets not expressing the C1 epitope and be non-lytic

towards targets expressing the C1 epitope. This section outlines the selection and

characterisation of NK clones from four donors (donors 1-4) three of which have at

least one C1 positive HLA-C allele and have KIR2DL2 and/or KIR2DL3 in their KIR

genotype.

Alloreactive NK clones from each donor were screened by testing for cytotoxicity

against a panel of BLCL targets with different C1/C2 epitope expression in the four-

hour 51Cr-release assay. On some occasions, NKG2A-positive clones were excluded

by flow cytometry prior to the 51Cr-release assay. This was due to the fact that all BLCL

used in the screening panel would be expected to express HLA-E, the ligand for the

Chapter 3

56

inhibitory receptor NKG2A. It might therefore be expected that any NK cell clone

expressing NKG2A would be inhibited by all BLCL targets and therefore could not be

used to study the interaction of KIR with HLA-C. Therefore NK clones were screened

for NKG2A expression and positive NK clones were discarded.

All NKG2A negative NK clones were screened against a panel of four BLCL in the 4-

hour 51Cr-release assay at an E:T ratio of 1:1 (Table 3.3). An NK clone was classified

as alloreactive if it was able to lyse the 721.221 cell line (relative specific lysis > 60%),

lyse at least one of the other targets (relative specific lysis > 20%) and was inhibited by

one of the other targets (relative specific lysis < 10%). Following detection of NK

alloreactivity, flow cytometry was used to determine the expression of CD158a

(KIR2DL1/KIR2DS1), CD158b (KIR2DL2/KIR2DL3/KIR2DS2) and KIR3DL1. Receptor

expression was then correlated with the cytotoxicity pattern.

Table 3.3 Class I HLA Typing of Target Cell Panel Used To Screen NK Clones

Target Cell HLA-A HLA-B HLA-C NK Epitopes 721.221 - - - - (IHW 9034) A*0301 B*0702 C*0702 C1 (IHW 9019) A*3002 B*1801 C*0501 C2 (IHW 9004) A*0201 B*2705 C*0102 C1, Bw4

3.3.1.1 Characterisation of Clones from Donor 1

Over 100 NK clones from donor 1 were screened for NKG2A expression by flow

cytometry. Those clones that were negative for NKG2A were tested against the panel

of BLCL target cells in the four-hour 51Cr release assay at an E:T ratio of 1:1. Donor 1’s

HLA type only included one NK epitope, C1, and therefore was expected to generate

only KIR2DL3 dependent NK clones. Nine NK clones were negative for NKG2A and

four of these clones had a cytotoxicity pattern consistent with KIR2DL3 dependence.

That is, they were inhibited by BLCL targets expressing the C1 epitope and lysed BLCL

Chapter 3

57

targets not expressing the C1 epitope. All four NK clones were positive for the CD158b

receptor, consistent with KIR2DL2/3 expression, and two clones also expressed

CD158a consistent with KIR2DL1 or KIR2DS1 expression. As these NK clones were

not inhibited by C2 positive targets, CD158a positivity was possibly due to expression

of KIR2DS1. The other five NK clones could not be characterised as being dependent

on KIR2DL1, KIR2DL3 or KIR3DL1 because the cytotoxicity pattern did not correlate

with receptor expression. Clone A6 from donor 1 (Figure 3.1, page 60) was selected for

further analysis.

3.3.1.1 Characterisation of Clones from Donor 2

35 NKG2A negative NK clones from donor 2 were tested in the four-hour 51Cr release

assay. Six of these clones had cytotoxicity patterns consistent with KIR2DL3

dependence and receptor expression correlated with cytotoxicity. An additional 11 NK

clones were identified to be dependent on KIR2DL1, KIR3DL1 or more than one KIR

receptor. These clones had cytotoxicity patterns consistent with KIR receptor

expression determined by flow cytometry and could be classified as being dependent

on one or more particular KIR receptor (Table 3.4).

Table 3.4 NK Clones from Donor 2 for which the Alloreactivity Pattern was

Consistent with Receptor Expression

Alloreactivity Pattern Number of Clones KIR2DL1 2KIR2DL3 6KIR3DL1 3KIR2DL3/KIR3DL1 2KIR2DL1/KIR3DL1 4KIR2DL1/KIR2DL3 0

Although each NK clone was selected because it was negative for NKG2A expression

prior to testing in the four-hour 51Cr release assay, it was noticed that some NK clones

Chapter 3

58

up-regulated expression of NKG2A in culture in between initial cytotoxicity screening

and secondary testing but this did not influence their ability to lyse or be inhibited by

particular target cells. This suggested that screening out of NKG2A expression could

eliminate potentially useful alloreactive NK clones. Therefore in subsequent cloning

experiments flow cytometry for NKG2A was omitted and all clones were screened for

cytotoxicity. The KIR2DL2/3 dependent NK clone, L10 (Figure 3.1, page 60) from donor

2 was selected for further analysis.

3.3.1.1 Characterisation of Clones from Donor 3

132 NK clones from donor 3 were screened in the four-hour 51Cr release assay. NK

clones that had a KIR dependent cytotoxicity pattern were screened for KIR receptor

expression by flow cytometry and correlated with cytotoxicity pattern. Seven NK clones

were identified that were dependent on KIR2DL2/3. Four of these clones co-expressed

NKG2A. An additional 7 NK clones were also identified and their reactivity patterns are

summarised in Table 3.5. Clone J6 (Figure 3.1), from donor 3 was selected for further

analysis.

Table 3.5 NK Clones from Donor 3 for which the Alloreactivity Pattern was

Consistent with Receptor Expression

Pattern of Alloreactivity Number of Clones NKG2A PositiveKIR2DL1 0 0KIR2DL2/3 7 4KIR3DL1 2 2KIR2DL2/3/KIR3DL1 3 1KIR2DL1/KIR3DL1 2 1

3.3.1.1 Characterisation of Clones from Donor 4

The screening of NK clones from donor 4 was performed by another scientist in the

laboratory. Six NK clones were identified with an alloreactive pattern that appeared to

Chapter 3

59

be KIR2DL2 (the donor’s KIR genotype included KIR2DL2 but not KIR2DL3). These

clones were selected for further characterisation. Each NK clone was confirmed by

myself to be KIR2DL2 dependent by testing against the four target cells used for

donors 1-3 and were shown to be CD158b positive by flow cytometry. Clone C9 was

selected for further analysis (Figure 3.1).

3.3.2 Summary of NK Clones

NK clones from four individuals were characterised and KIR2DL2 or KIR2DL3

dependent NK clones were identified from each donor (Donors 1-4). Figure 3.1A shows

NKG2A and CD158 expression of each NK clone selected for further analysis. The

cytotoxicity pattern of each NK clone was consistent with an NK clone dependent on

KIR2DL2 or KIR2DL3 and this pattern was consistent with receptor expression

determined by flow cytometry (Figure 3.1B). Clone A6 also expressed CD158a

(KIR2DL1/KIR2DS1) but was not inhibited by the target expressing the C2 epitope.

Therefore it is more likely that it expressed KIR2DS1 rather than KIR2DL1.

Chapter 3

60

Figure 3.1 NK clones selected for use in testing different HLA-C alleles and their

staining with CD158b and their cytotoxicity patterns

(A) CD158 and NKG2A expression on clones A6, L10, J6 and C9. (B), Cytotoxicity patterns for

each clone. All NK clones lysed the class I HLA negative target (721.221 cell line) and the C2

homozygous cell line (Table 3.3) but not the C1 homozygous cell line or the C1Bw4

homozygous cell line (Table 3.3).

Spe

cific

Lys

is (%

)

0

20

40

60

80

Target Cell

0

20

40

60

Target Cell 0

20

40

60

80

100

Target Cell

0

20

40

60

80

Target Cell

C1 - + - +C2 - - + -Bw4 - - - +

Clone A6

Clone C9Clone J6

Clone L10

A

B

NK Clone CD158a(KIR2DL1/KIR2DS1)

CD158b(KIR2DL2/KIR2DL3/

KIR2DS2)

CD158e(KIR3DL1/KIR3DS1)

NKG2A

Donor 1 A6 + + - -Donor 2 L10 - + - +Donor 3 J6 - + - +Donor 4 C9 - + - +

C1 - + - +C2 - - + -Bw4 - - - +

C1 - + - +C2 - - + -Bw4 - - - +

C1 - + - +C2 - - + -Bw4 - - - +

Chapter 3

61

3.3.3 KIR2DL3 mediated inhibition by the common C1-group and C2-group HLA-

C Alleles

To assess the interaction of different HLA-C alleles with KIR2DL3, a panel of BLCL

target cells was assembled representing all the common C1-group and C2-group HLA-

C alleles (Table 3.6).

Table 3.6 HLA-C Target Cell Panel

Lab Number HLA-A HLA-B HLA-C C1/C2 IHW 9004 0201 2705 0102 C1 IHW 9157 33 5801 0302 C1 IHW 9064 0217 1501 0303 C1 IHW 9069 0201 4001 0304 C1 IHW 9014 2601 0801 0701 C1 IHW 9034 0301 0702 0702 C1 IHW 9055 0301 1402 0802 C1 IHW 9011 0101 5201 1202 C1 IHW 9026 2601 3801 1203 C1 R05 307566X 11, 3101 5101 1402 C1 IHW 9051 2902 4403 1601 C1 IHW 9084 0201 4002 0202 C2 IHW 9068 0201 3501 0401 C2 IHW 9019 3002 1801 0501 C2 IHW 9046 0201 1302 0602 C2 IHW 9016 0204 5101 1502 C2 IHW 9043 0101 4101 1701 C2

Clone A6 (donor 1) and the NK cell line YTS, transfected with KIR2DL3, were selected

for screening against the target cell panel in the four-hour 51Cr-release assay. Clone A6

was selected due to large cell numbers of this clone. The YTS-2DL3 cell line was

selected due to it having expanded sufficiently that cell numbers were not limited and

experiments could be easily repeated. Since the chromium release assay may be

influenced by the fragility of the target cell, specific lysis of a target cell may vary from

cell to cell as a function of fragility rather than KIR:HLA-C interaction. Targets that are

inherently difficult to lyse may be misinterpreted as showing strong HLA-C mediated

inhibition of lysis. In order to derive a better measure of HLA-C mediated inhibition, the

Chapter 3

62

extent to which anti-CD158b blocking antibody increased lysis was used as a measure

of HLA-C mediated inhibition via KIR2DL3. Percentage inhibition was calculated as:

percentage inhibition = ((specific lysis with anti-CD158b - specific lysis with isotype

control)/specific lysis with anti-CD158b))*100

Figure 3.2 shows that clone A6 and YTS-2DL3 were not inhibited by interaction with

any of the C2-group alleles in panel 1. ie. had low percentage inhibition. For clone A6,

all C1-group alleles from panel 1 inhibited NK cell lysis with greater than 80%

percentage inhibition. For the YTS-2DL3 cell line however, inhibition differed amongst

the different C1 alleles. HLA-C*0302, -C*0701, -C*0702 and -C*1601 were all strong

inhibitors of NK cell cytotoxicity. HLA-C*0102, -C*0303, -C*0304 and -C*0802 were all

moderate inhibitors of NK cell cytotoxicity and, of the C1-group alleles, HLA-C*1402

was the poorest inhibitor. It is interesting to note that for clone A6, whilst HLA-C*1402

did strongly inhibit lysis (82%), it was nevertheless the weakest of all C1 alleles.

Chapter 3

63

Figure 3.2 Clone A6 and YTS-2DL3 differ in their interaction with HLA-C alleles

with the C1 epitope

Clone A6 and YTS-2DL3 were tested against the common C1- and C2 -group HLA-C alleles

(Table 3.6) in the four-hour 51Cr-release assay. Clone A6 was tested in triplicate wells. YTS-

2DL3 was also tested in triplicate wells on two different occasions and results for both occasions

are shown. Both clone A6 and YTS-2DL3 lysed all target cells expressing a C2-group allele.

Clone A6 was inhibited by all HLA-C alleles expressing the C1 epitope with percentage of

inhibition being greater then 80% with HLA-C*1402 being the weakest inhibitor at 82%.

Percentage inhibition greater than 100% occurred where specific lysis of the isotype control was

lower than the background. YTS-2DL3 was inhibited equally well by HLA-C*0302, HLA-C*0701,

HLA-C*0702 and HLA-C*1601 and was not inhibited at all by HLA-C*1402. YTS-2DL3 was

moderately inhibited by HLA-C*0102, HLA-C*0303, HLA-C*0304 and HLA-C*0802.

0102 0302 0303 0304 0701 0702 0802 1202 1203 1402 1601 0202 0401 0501 0602 1502 17010

20

40

60

80

100

120

140

A6 (1st Panel)YTS-2DL3 (1st Panel)

HLA-C

Perc

enta

ge In

hibi

tion

C1 C2

Chapter 3

64

The different levels of inhibition of YTS-2DL3 may have been due to differences

between the different HLA-C alleles or other unidentified ligands that differed between

targets. Therefore a second set of C1-group HLA-C alleles was assembled to test the

reproducibility of the different levels of inhibition (Table 3.7).

Table 3.7 HLA-C Target Cell Panel 2

Lab Number HLA-A HLA-B HLA-C C1/C2 IHW 9141 24 54 01 C1 IHW 9156 33 5801 0302 C1 IHW 9099 021701 1501 0303 C1 IHW 9098 3101 4001 0304 C1 IHW 9088 0101 0801 0701 C1 IHW 9065 0301 0702 0702 C1 IHW 9049 3301 1405 0802 C1 IHW 9142 2402 5201 1202 C1 IHW 9008 2501 1801 1203 C1 IHW 9053 3303 4403 1403 C1 IHW 9027 2902 4403 1601 C1

Unfortunately, there were not enough cells from clone A6 to test against this second

target cell panel. Clone J6, from donor 3, was therefore tested alongside YTS-2DL3

against the second HLA-C panel and against the C2-group HLA-C alleles. Clone J6

was also tested against the HLA-C*1402 target from panel 1 (Table 3.6). Figure 3.3

shows that both clone J6 and YTS-2DL3 were not inhibited through interaction with any

of the C2-group alleles in panel 1. Like clone A6, clone J6 was inhibited by all C1-group

alleles from panel 2 with greater than 75% inhibition. There was again variation

amongst the percentage inhibition by different C1 alleles for the YTS-2DL3 cell line.

HLA-C*0302, -C*0701, -C*0702 and –C*1601 were again shown to be strong inhibitors.

HLA-C*0303, -C*0304 and –C*0802 were shown again to be moderate inhibitors. The

fact that targets that had the same HLA-C alleles in panel 1 and panel 2 gave similar

levels of inhibition suggested that it was the HLA-C allele that was responsible for the

differing levels of inhibition. YTS-2DL3 was not tested against HLA-C*1402 again, but

its closest related allele, HLA-C*1403 was tested against both clone J6 and YTS-2DL3.

HLA-C*1403 inhibited clone J6, but was the poorest inhibitor for YTS-2DL3.

Chapter 3

65

Figure 3.3 Pattern of Inhibition is Reproduced with a second panel of C1-group

HLA-C alleles

Clone J6 (donor 3) and YTS-2DL3 were tested in triplicate against the second panel of C1-

group HLA-C alleles (Table 3.7) and the C2-group HLA-C alleles from panel 1 (Table 3.6) in the

four-hour 51Cr-release assay. YTS-2DL3 was tested on two occasions and results for both

occasions shown. Both Clone J6 and YTS-2DL3 lysed targets expressing the C2 epitope. Clone

J6 was inhibited by all C1-group HLA-C alleles with percentage inhibition greater than 75%

similar to clone A6. YTS-2DL3 was strongly inhibited by target cells expressing HLA-C*0302, -

C*0701 and -C*0702 from the second panel and moderately inhibited by HLA-C*0303, -C*0304,

-C*0802 and –C*1203. HLA-C*1403, the closest related allele to HLA-C*1402, weakly inhibited

YTS-2DL3.

0102 0302 0303 0304 0701 0702 0802 1202 1203 1402 1403 1601 0202 0401 0501 0602 1502 17010

20

40

60

80

100

120

140

J6 (2nd Panel)YTS-2DL3 (2nd Panel)

HLA-C

Perc

enta

ge In

hibi

tion

C1 C2

Chapter 3

66

To further confirm the different levels of inhibition of YTS-2DL3 exhibited by targets

expressing particular HLA-C alleles a third panel of selected C1 alleles was assembled

(Table 3.8).

Table 3.8 HLA-C Target Cell Panel 3

Lab Number HLA-A HLA-B HLA-C C1/C2 IHW 9067 0201 2705 0102 C1 IHW 9060 0101 1501 0303 C1 IHW 9032 0201 1501 0304 C1 IHW 9086 0101 0801 0701 C1 IHW 9029 2301 1402 0802 C1

Figure 3.4 shows the combined results for YTS-2DL3 against all three target cell

panels (Tables 3.6, 3.7 and 3.8). Targets having HLA-C*0302, -C*0701, -C*0702 and –

C*1601 were always strong inhibitors. Targets having HLA-C*0303, -C*0304 and -

C*0802 were moderate inhibitors. HLA-C*0102 appeared to be a moderate inhibitor,

but the results were less consistent between the three different HLA-C*0102

expressing BLCL. Results for HLA-C*1202 and –C*1203 were also less consistent

between different targets and further testing of these two alleles is required to more

precisely determine the strength of interaction of these two alleles with YTS-2DL3.

Chapter 3

67

Figure 3.4 C1-group HLA-C alleles differ in their ability to inhibit YTS-2DL3

The YTS-2DL3 cell line was tested against three panels of target cells representing the common

C1-group HLA-C alleles. Duplicate cell lines expressing HLA-C*0302, -C*0701, -C*0702, -

C*1601 demonstrate that these alleles all strongly inhibited YTS-2DL3. Three sets of cell lines

expressing either HLA-C*0102, -C*0303, -C*0304 or –C*0802 were tested and all moderately

inhibited lysis however the results for the three HLA-C*0102 cell lines were less consistent.

Both HLA-C*1402 and -C*1403 weakly inhibited lysis.

0102 0302 0303 0304 0701 0702 0802 120202 1203 1402 1403 16010

20

40

60

80

100

120

140

1st Panel2nd Panel

3rd Panel

HLA-C Allele

Perc

enta

ge In

hibi

tion

Chapter 3

68

3.3.4 HLA-C*1402 Inhibits NK Cells From Some Donors

Both NK clones, A6 and J6, were equally well inhibited by all C1 alleles, whereas the

YTS-2DL3 cell line was inhibited to different degrees by different C1 targets. To

determine whether the behaviour of YTS-2DL3 was an artefact of a transfected cell line

or whether other examples of NK clones might also behave this way, two additional NK

clones (clones L10 from donor 2 and C9 from donor 4) were tested in the four-hour 51Cr

release assay against a target expressing a C1-group allele that strongly inhibited all

NK cells (HLA-C*0702) and a target cell expressing HLA-C*1402. All NK clones and

YTS-2DL3 were inhibited by the HLA-C*0702 target and inhibition was reversed by

addition of anti-CD158b (Figure 3.5). HLA-C*1402 poorly inhibited the YTS-2DL3 cell

line as previously shown and was also found to poorly inhibit clone C9 from donor 4.

Addition of anti-CD158b resulted in only weak reversal of inhibition for clone C9 and

YTS-2DL3 (Figure 3.5B). However, clone A6, L10 and J6 were all inhibited through

interaction with HLA-C*1402 and inhibition was reversed by addition of anti-CD158b.

Chapter 3

69

Figure 3.5 HLA-C*0702 inhibits all NK clones whereas HLA-C*1402 only inhibits

some NK clones.

NK clones from 4 different individuals (donors 1-4) and YTS-2DL3 were incubated with BLCL

target cells expressing either HLA-C*0702 or HLA-C*1402 in the presence of

KIR2DL2/KIR2DL3 blocking antibody (anti-CD158b) or isotype control. (A) All NK clones and

YTS-2DL3 were inhibited by HLA-C*0702 and inhibition was reversed by addition of anti-

CD158b. (B), NK clones, A6, L10 and J6 (donors 1-3) were inhibited by HLA-C*1402 and this

was reversed by addition of anti-CD158b. NK clones C9 (donor 4) and the YTS-2DL3 cell line

were weakly bound to HLA-C*1402 with a slight increase in specific lysis with addition of anti-

CD158b.

YTS-2DL3 Clone A6 Clone L10 Clone J6 Clone C9-10

0

10

20

30

40

50

60 isotype control anti-CD158b

NK Cell Effector

Spe

cific

Lys

is (%

)

YTS-2DL3 Clone A6 Clone L10 Clone J6 Clone C9-10

0

10

20

30

40

50

60 isotype control anti-CD158b

NK Cell Effector

Spe

cific

Lys

is (%

)

A

B

HLA-C*0702 Target

HLA-C*1402 Target

Chapter 3

70

3.3.5 HLA-C*1402 only inhibits NK cells expressing KIR2DL3*002

The survey of targets with different HLA-C alleles and the further investigation of HLA-

C*1402 demonstrated that different NK cells expressing CD158b may not interact with

HLA-C in the same way. To determine if polymorphisms within the KIR gene can

distinguish between NK clones inhibited or not inhibited by HLA-C*1402, mRNA was

extracted from each clone and YTS-2DL3. KIR2DL2 or KIR2DL3 cDNA was amplified

(Figure 3.6) from the mRNA using a generic KIR2D forward primer and a KIR2DL2 or

KIR2DL3 specific reverse primer. Each cDNA PCR product was sequenced to

determine which allele of KIR2DL2 or KIR2DL3 each NK clone transcribed. Clones A6,

L10 and J6, which were all inhibited by HLA-C*1402, all transcribed KIR2DL3*002. The

KIR2DL3 expressed by the YTS cell line was KIR2DL3*001 and clone C9 transcribed

KIR2DL2*001. These results were consistent with the KIR gene repertoires of the

donors as donors 1-3 all had KIR2DL3 while donor 4 had KIR2DL2 and not KIR2DL3.

Figure 3.6 Amplification of KIR2DL2 or KIR2DL3 cDNA from each clone

cDNA was amplified from YTS-2DL3 (lane 1), clones A6 (lane 2), L10 (lane 3) and J6 (lane 4)

using full-length KIR2DL3 primers resulting in the expected 1044bp product. cDNA was

amplified from clone C9 (donor 4) using full length KIR2DL2 primers resulting in the expected

1047bp product.

In order to determine whether the alleles that were inhibited by HLA-C*1402 shared a

polymorphism that was not present in alleles that were not inhibited, the amino acid

sequences were aligned (Table 3.9).

1044b

1 2 31047b

1

Chapter 3

71

Table 3.9 Amino Acid Differences between KIR2DL3*002, KIR2DL3*001 and KIR2DL2*001

Signal D1 D2 stem transmembrane cytoplasmic domain peptide

Inhibited by HLA-C*1402

Examples Tested

-13 16 35 148 199 208 216 225 240 248 254 260 266 268 282 297 312 319

KIR2DL3*002 yes 3 V P Q R T L E V L C V P V R A H T S KIR2DL3*001 no 1 V P Q R T P E V L C V P V R A R T S KIR2DL2*001 no 1 A R E C I P K I F S A S A S T R A F

The amino acid differences between KIR2DL3*002, KIR2DL3*001 and KIR2DL2*001 are shown. The position of each amino acid in the gene is also

shown. KIR2DL3*002 is separated from KIR2DL3*001 at amino acid 208 (bold) in the stem and amino acid 297 (bold) in the cytoplasmic domain.

KIR2DL2*001 is identical to KIR2DL3*001 at these positions.

Chapter 3

72

KIR2DL3*001 and KIR2DL3*002 differ at only two nucleotide positions one in the stem

and one in the cytoplasmic domain. KIR2DL2*001 differs from both KIR2DL3 alleles at

several amino acids, but shares the stem and cytoplasmic amino acids with

KIR2DL3*001 at positions 208 and 297, respectively. Thus, either amino acid 208

(stem) or 297 (cytoplasmic domain) which were shared by both alleles that were not

inhibited by HLA-C*1402 and differed in the allele that was inhibited, could explain the

difference in reactivity with HLA-C*1402.

3.3.6 Do HLA-C alleles that interact weakly with KIR2DL3*001 share amino acid

motifs?

Amino acid 80 in the α1 domain of HLA-C is the critical amino acid in determining

specificity for KIR2DL1 or KIR2DL2 and KIR2DL3. On the KIR molecule, specificity for

C1-group or C2-group HLA-C alleles is determined by the amino acid at position 44.

Neither of these positions would explain why, for example, HLA-C*1402 binds poorly to

KIR2DL3*001 because these positions are conserved in both KIR and HLA-C.

However there are other invariant amino acids in both HLA-C and KIR2DL2/3 that

contribute to KIR:HLA-C interaction (HLA-C amino acids - 69, 75, 145, 146 and 151)

and may determine differences between the different C1 alleles and KIR alleles.

However these amino acids are conserved in both HLA-C and KIR2DL2/3 and are

therefore unlikely to play a role in the differences seen in this study. Section 1.3.2 of

the Literature Review described the role of peptide in influencing KIR binding to HLA-C.

Amino acid residues 7 and 8 of the peptide are thought to be the most critical. In order

to determine whether polymorphisms within the peptide binding pockets could be partly

responsible for differences in HLA-C:KIR interaction, a comprehensive analysis

(Chelvanayagam, 1996) of the amino acids of HLA-C that make up the peptide binding

environment for residue 7 (Table 3.11) and 8 (Table 3.12) of the peptide was therefore

undertaken.

Chapter 3

73

Table 3.11 Residue 7 Binding Environment of Strong, Moderate and Low

Inhibiting HLA-C Alleles

Inhibition conferred by KIR2DL3*001

C* 73 77 97 114 116 133 147 152 155 156

Strong 0701 A S R D S W L A Q L Strong 0702 A S R D S W L A Q L Strong 0302 T S R D S W W E Q L Strong 1601 T S W D S C W A Q Q Moderate 0802 T S R N F W W E Q R Moderate 0303 T S R D Y W W E Q L Moderate 0304 T S R D Y W W E Q L Weak 1402 T S W D S W W E Q R Weak 1403 T S W D S W W E Q R

Table 3.12 Residue 8 Binding Environment of Strong, Moderate and Low

Inhibiting HLA-C Alleles

Inhibition conferred by KIR2DL3*001

C* 73 77 80 97 143 147

Strong 0701 A S N R T L Strong 0702 A S N R T L Strong 0302 T S N R T W Strong 1601 T S N W T W Moderate 0802 T S N R T W Moderate 0303 T S N R T W Moderate 0304 T S N R T W Weak 1402 T S N W T W Weak 1403 T S N W T W

Examination of both groups of amino acids reveals no amino acids unique to the

different inhibitory levels of C1-group HLA-C alleles although the combined peptide 7

and peptide 8 binding environments of HLA-C*1402 and HLA-C*1403 are identical to

each other and not shared with any other allele. Therefore it is more likely that a

combination of polymorphisms confer high, moderate and low binding to KIR2DL3.

Chapter 3

74

3.4 Discussion

Despite extensive polymorphism within the HLA-C locus, only one polymorphic

position, amino acid 80, determines HLA-C specificity for KIR2DL1 or KIR2DL2 and

KIR2DL3. However this study suggests that other positions may influence the

interaction between HLA-C and KIR. KIR2DL2 and KIR2DL3 dependent NK clones

were generated from donors who express the C1 epitope and these clones were used

to test the ability of all the common HLA-C alleles to inhibit NK cell cytotoxicity via

these receptors. The data confirmed a previous report by Colonna and colleagues that

HLA-C*1402 was a poor C1 ligand. However, the data in this thesis show that this was

the case for KIR2DL3*001 and KIR2DL2*001, but not for KIR2DL3*002. As was

expected, HLA-C alleles with the C2 epitope did not inhibit any KIR2DL3 or KIR2DL2

dependent NK cells. In addition to the different reactivities of KIR2DL3*001 and

KIR2DL3*002 with HLA-C*1402, this study also provided evidence that these two

alleles interact differently with other HLA-C alleles. All the common C1-group alleles

inhibited cytotoxicity of NK cells expressing KIR2DL3*002 to a relative uniform degree

whereas different HLA-C alleles differed in their level of inhibition of the YTS-2DL3*001

cell line. Consistent results with multiple examples of each BLCL target expressing

particular HLA-C alleles suggest that the differences observed are more likely to be

due to the C1-group allele expressed rather than other differences between the BLCL.

It is possible the HLA-C expression is lower for alleles that showed weak inhibition. As

there are no monoclonal antibodies specific only for HLA-C this could not be tested

directly. However, since NK clones expressing KIR2DL3*002 are inhibited uniformly by

all C1-group HLA-C alleles this explanation seems unlikely.

The structures of C1-group HLA-C alleles in complex with KIR2DL2, of C2-group HLA-

C alleles in complex with KIR2DL1, and the structures for the receptors KIR2DL3 and

KIR2DL1 alone have been solved by crystallography (Fan et al, 1997; Maenaka et al,

1999; Boyington et al, 2000; Fan et al, 2001). The two-immunoglobulin domains of the

Chapter 3

75

KIR molecules are important in binding to HLA-C and polymorphisms within these

regions could result in differential binding. The two most common KIR2DL3 alleles,

KIR2DL3*001 and KIR2DL3*002, differ at two amino acids: one in the stem region and

one in the cytoplasmic domain. The amino acid at these positions are shared with

KIR2DL3*001 and KIR2DL2*001 both of which interact poorly with HLA-C*1402.

Neither of these would be expected to have a major influence on KIR binding to HLA-C.

However the data in this study suggests that these alleles differ in their ability to bind to

HLA-C*1402. When analysing the crystal structure of KIR2DL1, Fan and colleagues

generated two structures: one comprised of amino acids forming the D1 and D2

domains alone (amino acids 1-200) and one comprised of amino acids forming the D1,

D2 and the stem (amino acids 1-224). They reported an influence of the stem on ligand

binding with higher affinity binding for structures that included the stem. It was

suggested that the stem region improved binding to HLA-C by stabilising dimerisation

of the KIR receptor. KIR2DL3*002 has a leucine at amino acid 208 in the stem region

and KIR2DL3*001 and KIR2DL2*001 have a proline at the same position. Proline, a

relatively small amino acid, contains an imino group rather than an amino group and

thus can not easily substitute for other amino acids. Proline is often found in very tight

turns in protein structure and can introduce kinks into alpha helices (Betts and Russell,

2003). Leucine prefers to be buried in hydrophobic cores and is rarely involved in

protein function. It is uncertain whether a proline at amino acid 208 of the stem region

would alter the ability of KIR2DL3*001 and KIR2DL2*001 to interact with HLA-C.

However, it is possible that it alters the structure of the stem region such that when the

receptor binds to HLA-C the overall structure is weaker than for KIR2DL3*002.

Inhibitory KIR contain two ITIMs in their cytoplasmic domain and are phosphorylated

upon engagement with class I HLA molecules thereby delivering an inhibitory signal

(Burshtyn et al, 1996). Amino acid 297 sits between the ITIMs and is not directly

involved in signalling. Although the possibility that a histidine-arginine substitution at

Chapter 3

76

this position could affect ligand binding cannot be excluded, it seems less likely than

the leucine-proline substitution in the stem region.

Final proof of the importance of this position will require site-directed mutagenesis. This

was attempted during this thesis but successful transfection of the mutant construct

into YTS could not be achieved. Site-directed mutagenesis of the two positions will

need to be performed and the resulting construct transfected into the YTS cell line.

After generating stable cell lines expressing wild-type and mutant KIR2DL3, all

transfected cell lines would be tested against the panel of C1-group and C2-group

alleles in the chromium release assay to determine the effect of each mutation.

HLA-C*1402 was also a poor ligand for KIR2DL2*001. KIR2DL2*001 shares both the

stem and cytoplasmic domain polymorphisms with KIR2DL3*001. Therefore it is

possible that KIR2DL2*001, like KIR3DL1*001, will also show interactions of different

strengths with other C1-group HLA-C alleles, but this requires further investigation.

As not all C1-group alleles are poor ligands for KIR2DL3*001 (and KIR2DL2*001) it is

unlikely that the polymorphisms within the KIR allele are the only positions involved in

poor KIR:HLA-C interaction. Rather, the poor interaction is likely due to a combination

of polymorphisms within both the KIR2DL3*001 (and KIR2DL2*001) and HLA-C alleles.

As described in 3.3.7, the amino acids involved at the KIR:HLA-C interface are

conserved in both genes, but the peptide bound may influence the interaction.

Residues 7 and 8 are the most critical peptide amino acids as KIR receptors directly

contact both (Boyington et al, 2000). Particular amino acids of the HLA-C molecule

make up the binding environment for P7 and P8 and this environment is highly

polymorphic (Chelvanayagam,1996). The C1 alleles were divided into strong, moderate

and weak inhibitors and the amino acids comprising the peptide binding environments

were examined. There were no polymorphisms that could separate the three groups.

This suggests that for these peptide binding environments to influence and result in

Chapter 3

77

differing KIR:HLA-C interaction it is through a group of polymorphisms within HLA-C

rather than a single one. This preliminary data would suggest that polymorphic

differences within both KIR and HLA-C are responsible for the differing levels of

KIR:HLA-C interaction.

As HLA-C*1402 and HLA-C*1403 do not appear to act as ligands for KIR2DL3*001 or

KIR2DL2*001, it could be hypothesised that donors who express one of these KIR

alleles and HLA-C*1402 or –C*1403 as their only C1-group HLA-C allele may not be

able to generate very many NK clones dependent on KIR2DL2 or KIR2DL3. Donors

who expressed either HLA-C*1402 or HLA-C*1403, which is more common in

Japanese populations, as their only C1-group HLA-C allele were sought to determine if

they can generate alloreactive NK clones dependent on KIR2DL2/3. Two donors were

selected however blood could only be obtained from one and after NK cell isolation, the

NK cells failed to expand and therefore could not be tested for their ability to generate

KIR2DL2 or KIR2DL3 dependent NK clones.

The data in this chapter suggests that different HLA-C alleles differ in their ability to

inhibit different alleles of KIR2DL2 and KIR2DL3. It is possible that the apparent

correlation of HLA-C interaction with the different KIR alleles is purely coincidental.

Other receptors expressed by the clones cannot be ignored as possible reasons for the

differences. Although NKG2A appeared to not influence NK alloreactivity in the

screening assay, it is possible that NKG2A may influence reactivity with different target

cells. However, this seems unlikely as HLA-E is expressed by all BLCL used in this

study and therefore uniform inhibition would be expected if NKG2A was functional on

the cell and this was not seen as all targets expressing C2-group HLA-C alleles were

lysed. There may be other receptors expressed by these NK clones or the YTS cell line

that are currently not known that may influence lysis of target cells with particular HLA.

Chapter 3

78

The common HLA-C alleles were analysed for their ability to inhibit two NK clones

expressing KIR2DL3*002 and the YTS cell line transfected with KIR2DL3*001.

Therefore the comparison between the KIR2DL3 alleles was not strictly controlled. It

will be important to replicate this data using NK clones expressing KIR2DL3*001. A

similar screening process also needs to be conducted with NK clones expressing

KIR2DL2 to determine if KIR2DL2 interacts with HLA-C alleles similarly to KIR2DL3. It

will also be important to screen NK clones expressing KIR2DL1 against the C1 and C2

panels to determine if all C2 alleles act as appropriate ligands for KIR2DL1 and also

determine if any C1 alleles interact with KIR2DL1. These additional experiments will

provide important information needed to enable better identification of potential donor

and recipient pairs from HLA and KIR repertoires.

Chapter 4

79

Chapter 4

The Reactivity of Bw4-positive HLA-B and HLA-A Alleles with

KIR3DL1: Implications for Patient and Donor Suitability for

Haploidentical Stem Cell Transplants

Foley BA, De Santis D, van Beelen E, Lathbury LJ, Christiansen FT and Witt CS

Blood 2008 (In Press)

The work in this chapter has been submitted for publication in Blood. It has been

reproduced in whole with one alteration. The materials and methods section has been

simplified to include only the NK cell donors and cell lines used in this chapter. For

other methods refer to chapter 2.

Chapter 4

80

Chapter 4: The Reactivity of Bw4-positive HLA-B and HLA-A

Alleles with KIR3DL1: Implications for Patient and Donor

Suitability for Haploidentical Stem Cell Transplants

4.1 Introduction

Natural killer (NK) cell alloreactivity can be exploited in haploidentical haematopoietic

stem cell transplantation (HSCT) to improve graft survival, reduce graft versus host

disease and decrease leukaemic relapse (Ruggeri et al, 2002). Infusion of alloreactive

NK cells without stem cells has also been shown to result in haematological remission

in AML patients (Miller et al, 2005). NK cells lyse cells that have reduced expression of

class I HLA molecules. In an allogeneic setting, donor NK cells are activated by the

absence of donor (self) class I HLA molecules on recipient cells (Karre et al, 1986;

Ljunggren and Karre 1990), the absence of self-epitopes being detected by inhibitory

killer immunoglobulin-like receptors (KIR) on donor NK cells. HLA-C molecules with an

asparagine at amino acid 80 provide the C1 epitope for the KIR2DL2 and KIR2DL3

inhibitory receptors and those with a lysine at amino acid 80 provide the self-epitope,

C2, for the KIR2DL1 inhibitory receptor. The self-epitope of relevance to the KIR3DL1

receptor is the Bw4 epitope at amino acids 77 – 83 on some HLA-B alleles. All HLA-B

alleles have either the Bw4 or Bw6 epitope. NK cells from donors, whose HLA-B

alleles include the Bw4 epitope, lyse cells lacking the Bw4 epitope because such target

cells cannot supply the ligand for the NK inhibitory receptor KIR3DL1. However, it has

been reported (reviewed in Ruggeri et al, 2006) that only two thirds of donors with Bw4-

positive HLA-B alleles make NK clones that lyse Bw4-negative targets. Therefore,

selecting an NK or stem cell donor for a particular patient currently requires a lengthy,

labour intensive in vitro NK cloning procedure to confirm the donor has NK cells

alloreactive towards the patient (Ruggeri et al, 2002).

Chapter 4

81

It is not clear why some Bw4 positive donors cannot make NK clones that lyse Bw4

negative targets. A likely explanation in some cases is that a common allele of

KIR3DL1 (KIR3DL1*004) is not expressed at the cell surface (Pando et al, 2003).

However, a systematic examination of the ability of HLA-B alleles to bind to KIR3DL1

has not been undertaken. Amino acid 80 in the HLA-B protein critically determines

binding to KIR3DL1 (D'Andrea et al, 1995). Alleles with the Bw6 epitope have

asparagine (N) at this position and do not bind to KIR3DL1 while alleles with Bw4 have

either an isoleucine (I) or threonine (T) and are ligands for KIR3DL1. It has been

reported that alleles with an isoleucine (80I) are stronger ligands than alleles with a

threonine (80T) (Cella et al, 1994; Draghi et al, 2005) whilst other investigators have

noted differences within the Bw4 80I family of alleles (Gumperz et al, 1995) and

peptide-dependent differences in the binding of Bw4 tetramers to different KIR3DL1

variants (Thananchai et al, 2007). Several HLA-A alleles are also known to react with

anti-Bw4 antibodies but their ability to behave as a ligand for KIR3DL1 is controversial

(Cella et al, 1994; Gumperz et al, 1995; Thananchai et al, 2007). These studies utilised

HLA-bare EBV cell lines transfected with individual HLA alleles or tetramers loaded

with individual peptides to compare the effect of different alleles. A related question that

has not been addressed is whether donors with the different forms of Bw4 epitope are

all capable of producing NK clones that lyse cells lacking Bw4.

In order to improve the ability to select donors for haploidentical HSCT or NK cell

infusions based on HLA rather than NK cloning assays, we undertook a systematic

analysis of the ability of common HLA-B alleles and Bw4-positive HLA-A alleles to

inhibit KIR3DL1-dependent NK clones. In addition, we tested the ability of individuals

with Bw4-positive HLA-A alleles to produce KIR3DL1-dependent clones. As it is difficult

to know whether results from HLA-bare cells transfected with single HLA alleles, or

tetramers loaded with single peptides, accurately reflect the complex array of HLA

ligands presented to NK cells (in vivo), we chose to utilise EBV cell lines that express a

normal array of HLA antigens but which were homozygous for the HLA alleles of

Chapter 4

82

interest. Our results confirm that most, but not all, of the common Bw4-positive HLA-B

alleles are ligands for KIR3DL1. In addition, the status of the Bw4-positive HLA-A

alleles is clarified and we demonstrate that individuals with a Bw4-positive HLA-A allele

are able to generate KIR3DL1-dependent clones. These results have implications for

donor selection for haploidentical HSCT.

4.2 Materials and Methods

The materials and methods section has been simplified to include only the NK cell

donors and cell lines used in this chapter. For other methods refer to chapter 2.

Chapter 4

83

Table 4.1 HLA Class I and KIR3DL1 Allele Typing of NK Cell Donors

Donor HLA-A HLA-B Bw4/6 HLA-C C1/2 KIR3DL1 Typing KIR3DL1 Expression

1 0201, 2902 4403, 5703 4 0701, 1601 1 01501/017, 008 high 2 0201, 2601 0702, 3801 4,6 0702, 1203 1 002, 00101 high 3 0101, 2402 0801, 5109 4,6 0102, 0701 1 00501-like, 01502 high, low 4 0201, 3303 4601, 5801 4,6 0102, 0302 1 00501, 01502 high, low 5 1101 1301, 1501 4,6 0304, 0401 1,2 007, 01502 high, low 6 1101 1301, 1501 4,6 0304, 0401 1,2 00501, 007 high, low 7 0301, 3101 1302, 3501 4,6 0401, 0602 2 001/016, 002 high 8 0101, 0301 0801 6 0701 1 nt high 9 0201, 1101 1501, 3501 6 0303, 0401 1,2 nt low

10 0101, 2402 0801, 5501 6 0303, 0701 1 nt high

The KIR3DL1*005-like allele in donor 3 differs from KIR3DL1*005 by a valine to leucine substitution at amino acid 18 in the D0 domain. In all cases,

HLA alleles were assigned on the basis of sequencing of exons 2 and 3. In some cases, the assignment of class I HLA alleles is based on the most

common allele consistent with the sequence in exons 2 and 3 and alleles differing in other exons cannot be excluded. As the signal peptide of KIR3DL1

was not sequenced, KIR3DL1*01501 cannot be distinguished from KIR3DL1*017 in donor 1 and KIR3DL1*001 cannot be distinguished from

KIR3DL1*016 in donor 7. nt = not tested.

Chapter 4

84

Table 4.2. HLA Class I Typing of BLCL Target Cell Panel

ID SHORT ID

HLA-B AA 80

Bw4/6 HLA-A HLA-C C1/2

IHW 9084 B13 1302 T 4 3001 0602 2 IHW 9067 B27 2705 T 4 0201 0102 1 IHW 9009 B37 3701 T 4 0101 0602 2 IHW 9090 B4402 4402 T 4 0201 0501 2 IHW 9027 B4403 4403 T 4 2902 1601 1 IHW 9047 B47 4701 T 4 0301 0602 2 IHW 9062 B38 3801 I 4 0201 1203 1 IHW 9040 B49 4901 I 4 0101 0701 1 IHW 9016 B51 5101 I 4 0204 1502 2 IHW 9011 B52 5201 I 4 0101 1202 1 IHW 9010 B53 5301 I 4 6802 0401 2 IHW 9052 B57 5701 I 4 0201 0602 2 IHW 9157 B58 5801 I 4 33 0302 1 IHW 9029 A23 1402 I 6 2301 0802 1 IHW 9001 A24 0702 I 6 2402 0702 1 IHW 9008 A25 1801 I 6 2501 1203 1 Q94 0055016Y A32 1501, 3901 I 6,6 3201, 0201 0303, 1203 1,1 IHW 9065 B7 0702 N 6 0301 0702 1 IHW 9088 B8 0801 N 6 0101 0701 1 IHW 9029 B14 1402 N 6 2301 0802 1 IHW 9099 B15 1501 N 6 0217 0303 1 IHW 9019 B18 1801 N 6 3002 0501 2 IHW 9068 B35 3501 N 6 0201 0401 2 Q94 0052722Q B39 0702, 3901 N 6,6 0201, 0301 0702, 1203 1,1 IHW 9084 B40 4002 N 6 0201 0202 2 IHW 9043 B41 4101 N 6 0101 1701 2 IHW 9021 B42 4201 N 6 3001, 6802 1701 2 IHW 9058 B45 4501 N 6 0201 1601 1 IHW 9076 B46 4601 N 6 0206, 0207 0102, 0801 1,1 R98 0903165W B50 5001, 0702 N 6,6 0301, 0201 R04 0901000J B55 5502, 0801 N 6,6 0101, 0206 0102, 0701 1,1 R97 0330581F B56 5601, 0801 N 6,6 01, 34

HLA types of target cells used in flow cytometric and chromium release assays. In all cases,

HLA alleles were assigned on the basis of sequencing of exons 2 and 3. In some cases, the

assignment of class I HLA alleles is based on the most common allele consistent with the

sequence in exons 2 and 3 and alleles differing in other exons cannot be excluded. Targets

that were homozygous for HLA-B50, B55 and B56 were not available. Therefore, cells that were

heterozygous for these antigens and had a common Bw6 antigen as the second antigen were

selected. AA80 indicates the amino acid present at residue 80 of the HLA-B alleles. Where

HLA-C typing has not been performed the table entry is left blank.

Chapter 4

85

4.3 Results

4.3.1 KIR3DL1 Genotype of NK Cells

As different alleles of KIR3DL1 have high or low expression at the cell surface, we

determined the KIR3DL1 alleles and receptor expression pattern on 10 NK donors

(Table 4.1). To compare the behaviour of high and low expressing alleles two donors

with only high expression alleles and two donors heterozygous for high and low

expression alleles (donors 1, 2 and 3, 4 respectively in Table 4.1) were selected. Also,

as the high expressing allele in both donors 3 and 4 was the same (KIR3DL1*01502),

between-donor reproducibility could be assessed. All four donors had at least one

Bw4-positive HLA-B allele (Table 4.1). Interestingly, the low expression receptors in

donors 3 and 4 differed in their level of expression (Figure 4.1). This difference may be

due to the fact that the low expression receptor in donor 3 differed from the low

expression receptor encoded by KIR3DL1*005 in donor 4 by a valine to leucine

substitution at amino acid 18 in the D0 domain.

Chapter 4

86

Figure 4.1 Low level KIR3DL1 expression differs in two donors due to a

nonsynonymous mutation at nucleotide position 115 in exon 3 encoding the D0

domain of the KIR3DL1*005 allele.

NK cells from each donor were stained with anti-KIR3DL1 antibody (DX9) (solid lines) or isotype

control (IgG1) (dotted lines). KIR3DL1 surface expression correlated with KIR3DL1 allele typing,

with donors 1 and 2 having unimodal high level expression and donors 3 and 4 having bimodal

(high and low level) expression. The low level receptor expression differed between donors 3

and 4. Comparison of both KIR3DL1 sequences from donors 3 and 4 revealed a single

nonsynonymous nucleotide mutation at position 115 of the KIR3DL1*005 allele in donor 3.

Chapter 4

87

4.3.2 KIR3DL1 Dependent NK Cells Lyse Cells Lacking the Bw4 Epitope

KIR expression on polyclonally expanded NK cells has been demonstrated not to alter

the frequency of NK cells expressing particular KIR receptors (Draghi et al, 2005). In

order to detect NK cell cytotoxicity towards Bw4-negative targets mediated by

KIR3DL1-dependent clones, polyclonal NK cells from donors 1 to 4 were expanded by

in vitro culture and then incubated with BLCL targets expressing common HLA-B

alleles or the Bw4-positive HLA-A alleles A*2301, A*2402, A*2501 and A*3201 (see

Table 4.2). Cytotoxicity was measured by CD107a expression on KIR3DL1 (DX9)

positive, CD158b (KIR2DL2, KIR2DL3, KIR2DS2) negative NK cells. As all four NK

donors were homozygous for C1-group HLA-C alleles and a significant proportion of

KIR3DL1-positive NK cells co-expressed CD158b, the inhibitory receptor for the C1-

epitope, only KIR3DL1-positive, CD158b-negative cells were examined. KIR3DL1-

positive, CD158b-negative NK cells up-regulated CD107a when incubated with either

the class I HLA negative target (721.221-positive control) or targets homozygous for

the Bw6 epitope (Figure 4.2). Bw4-positive targets tended to inhibit CD107a

expression. Overall, there was little difference between the four donors in terms of the

target cells that showed greatest inhibition of CD107a expression. CD107a expression

was inhibited by all Bw4 expressing targets with four exceptions: those expressing

HLA-B*1302, HLA-B*5101, HLA-A*2301 and HLA-A*2501. There were some small

differences in the ability of the other Bw4 expressing targets to inhibit CD107a

expression but these differences did not relate to the presence of 80I or 80T alleles.

Chapter 4

88

NA

721

B13

B27

B37

B44

02B

4403

B47

B38

B49

B51

B52

B53

B57

B58

A23

A24

A25

A32 B7

B8

B14

B15

B18

B35

B39

B40

B41

B42

B45

B46

B50

B55

B56

0

20

40

60

80

KIR3DL1 high/high

NA

721

B13

B27

B37

B44

02B

4403 B47

B38

B49

B51

B52

B53

B57

B58

A23

A24

A25

A32 B7 B8 B14

B15

B18

B35

B39

B40

B41

B42

B45

B46

B50

B55

B56

0

20

40

60

80

KIR3DL1 high/high

NA

721

B13

B27

B37

B44

02B

4403

B47

B38

B49

B51

B52

B53

B57

B58

A23

A24

A25

A32 B

7B

8B

14B

15B

18B

35B

39B

40B

41B

42B

45B

46B

50B

55B

56

0

20

40

60

80

KIR3DL1 high KIR3DL1 low

NA

721

B13

B27

B37

B44

02B

4403

B47

B38

B49

B51

B52

B53

B57

B58

A23

A24

A25

A32 B

7B

8B

14B

15B

18B

35B

39B

40B

41B

42B

45B

46B

50B

55B

56

0

20

40

60

80

KIR3DL1 high KIR3DL1 lowP

erce

ntag

e of

KIR

3DL1

pos

itive

, 158

b ne

gativ

e ce

lls e

xpre

ssin

g C

D10

7a

Target Cells

80T

80T

80T

80T

80I

80I

80I

80I

Bw4Donor 1

Donor 4

Donor 3

Bw6

Donor 2

Chapter 4

89

Figure 4.2. CD107a expression by KIR3DL1-positive polyclonal NK cells is

inhibited by most Bw4-positive targets.

Polyclonally expanded NK cells from four donors (donors 1-4) were incubated alone (NA), with

a positive control, the class I negative BLCL 721.221 (721) and a range of Bw4 and Bw6

positive targets (Table 4.2) and CD107a expression was measured on KIR3DL1 positive,

CD158b negative NK cells. Donors 3 and 4 had NK cells which showed high or low expression

of DX9 and for each the percentage of CD107a positive cells amongst either the NK cells

bearing the high level (solid bars) or low level (open bars) of DX9 expression are shown. All

donors lysed the 721.221 cell line and all Bw6-homozygous (Bw4-negative) targets. All donors

were inhibited by most of the Bw4-positive targets except for targets B13, B51, A23 and A25.

For donor 3, the low level expression allele was less cytotoxic against Bw6 expressing targets

and not as well inhibited by most Bw4 expressing targets compared to the high expression

allele. By contrast for donor 4, the high and low level expression alleles behaved similarly.

Chapter 4

90

Of the two donors with high KIR3DL1 expression, donor 1 had a higher percentage of

NK cells expressing CD107a in response to Bw4-negative targets and therefore

appeared to better discriminate between Bw4-positive and negative targets. However,

this may simply reflect the fact that donor 1 had a higher proportion of NK cells

dependent on KIR3DL1 for inhibition than donor 2. The high expression KIR3DL1

receptor in donors 3 and 4 and the low expression allele (KIR3DL1*005) in donor 4

behaved in a similar way to NK cells in donor 1. However, in donor 3, NK cells with the

low expression KIR3DL1*005-like variant up-regulated CD107a relatively weakly in

response to Bw6 targets and were less completely inhibited by Bw4 targets such that

there was relatively poor discrimination between the Bw4 and Bw6 targets.

Interestingly, in this donor, those NK cells expressing the KIR3DL1*005-like variant

also exhibited less up-regulation of CD107a in response to the class I bare 721.221

cells than the NK cells expressing the KIR3DL1*01502 allele. These results suggest

that the poor discrimination between Bw4 and Bw6 targets exhibited by the NK cells

expressing the receptor encoded by the KIR3DL1*005-like variant, may be due to

these cells being relatively weakly armed rather than a smaller proportion being

KIR3DL1-dependent for inhibition. Donor 3 was particularly informative in terms of the

Bw4 alleles that are KIR3DL1 ligands. For almost all Bw4 targets, NK cells with the

high expression allele KIR3DL1*01502 were more strongly inhibited than NK cells with

the weakly expressed KIR3DL1*005-like variant. For each Bw6 target, NK cells with

the high expression allele KIR3DL1*01502 were less inhibited than NK cells expressing

the weakly expressed KIR3DL1*005-like variant. In this respect, HLA-B*1302, HLA-

A*2301 and HLA-A*2501, all behaved like Bw6 alleles, reinforcing the conclusion that

these alleles are not KIR3DL1 ligands. Interestingly, in donor 4, the only target that

appeared to discriminate between NK cells expressing KIR3DL1*01502 and

KIR3DL1*005 was the HLA-A*23 target. This suggests that some KIR3DL1 alleles

may provide exceptions to the more general rules.

Chapter 4

91

The ability of the different targets to inhibit CD107a expression on KIR3DL1 expressing

effectors could be related not so much to the affinity of the ligand but rather, differences

in the level of expression of the ligand on the various targets. Therefore the level of

Bw4 expression was checked by staining each target with an anti-Bw4 mAb. As shown

in Figure 4.3, the average CD107a expression induced on KIR3DL1-positive NK cells

was not related to the level of Bw4 expression on the target. The A23, A24 and A32

targets also stained strongly whereas the A25 target did not stain with the Bw4 mAb

and did not inhibit NK cell cytotoxicity despite being known to have the Bw4 motif.

However, this mAb also did not react with two additional cell lines with HLA-A25

indicating that this mAb does not react with the HLA-A25-associated Bw4 epitope. The

B51 target did have reduced HLA-B expression, which could explain the poor inhibition

mediated by this target. However, the B13 target, which was also poorly inhibitory, had

levels of Bw4 expression comparable with the other Bw4 expressing targets. Therefore,

reduced HLA-B expression was not responsible for its lack of inhibition. There was no

correlation between the level of Bw4 expression and ability to suppress CD107a

expression when the HLA-A*2501 and B*5101 targets were excluded (r = -0.13,

p=0.68).

Chapter 4

92

Figure 4.3. Level of Bw4 expression does not correlate with percentage of

CD107a expression induced on KIR3DL1 positive NK cells.

Each Bw4-positive target cell was stained with an anti-Bw4 mAb to detect Bw4 expression. The

mean channel fluorescence (MCF) for each target was plotted against the percentage of

CD107a positive NK cells induced by the target on NK cells with a high expression KIR3DL1

receptor, averaged across the four NK cell donors. Target A25 did not stain with the Bw4 mAb

and does not inhibit NK cell cytotoxicity. Target B51, which weakly inhibited NK cell cytotoxicity,

has lower expression of Bw4 than the other Bw4 expressing alleles. However, the B13 target,

which also weakly inhibited NK cell cytotoxicity, expressed levels of Bw4 comparable to most

Bw4-positive targets. After excluding the A25 and B51 targets, there was no significant

correlation between the percentage of KIR3DL1-positive cells expressing CD107a and Bw4

expression on the target cell (r = -0.13, p=0.68).

0 200 400 600 800 1000 1200 1400 1600 1800 2000 22000

5

10

15

20

A25

B51B13

Mean FITC (anti-Bw4)

Per

cent

age

of D

X9

(KIR

3DL1

) pos

itive

cells

exp

ress

ing

CD

107a

Chapter 4

93

4.3.3 HLA-B*13 is not a KIR3DL1 ligand but HLA-B*5101 is

To confirm the lack of inhibition seen with the targets bearing HLA-B*1302 and HLA-

B*5101, additional examples of BLCL expressing B*1302 and B*5101, and one

expressing B*1301 (Figure 4.4A) were tested in the CD107a assay against NK cells

from donor 1. KIR3DL1-positive NK cells up-regulated CD107a when incubated with

either the positive control (721.221) or the Bw6 control (B8) as expected (Figure 4.4B).

The Bw4 control (B27) inhibited CD107a expression as expected. Both examples of

targets expressing HLA-B*1302 and the single example of a target expressing HLA-

B*1301 failed to inhibit CD107a expression. In contrast, the original HLA-B*5101 target

with reduced HLA-B expression (5101a) inhibited CD107a expression less strongly

than the two additional examples of B*5101 targets. The additional B*13 and B*5101

targets had HLA-B expression comparable to the other Bw4-positive targets as

determined by staining with the Bw4 mAb (data not shown). These data suggest that

the observation in relation to the original B51 target was due to reduced expression of

HLA-B*5101 on that target whereas HLA-B*13 appears to be a poor ligand for

KIR3DL1. Lack of inhibition of cytotoxicity by HLA-B*1302 was confirmed using NK

clones from donors 3 and 4 in a four hour chromium-release assay (Figure 4.4C). Two

KIR3DL1-positive clones (clone C3 with low KIR3DL1 expression from donor 4 and

clone D2 with high KIR3DL1 expression from donor 3) were tested. As shown in

Figure 4.4C, cytotoxicity of clones C3 and D2 against the class I negative cell line

(721.221) and a Bw6 control (B35) (Table 4.2) was not affected by blocking KIR3DL1

with DX9 antibody whereas inhibition of cytotoxicity by a Bw4 control (B57) (Table 4.2)

was reversed by blocking with DX9 (Fig 4.4C). Cytotoxicity against the two HLA-

B*1302 homozygous targets was relatively strong in the absence of blocking antibody

and only weakly enhanced in the presence of DX9, confirming that HLA-B*1302 is a

poor ligand for KIR3DL1.

Chapter 4

94

Donor 3

721.221 B35 B57 1302a 1302b0

20

40

60

80

100 Isotype controlAnti-KIR3DL1

Target Cell

Spec

ific

Lysi

s (%

)

Donor 4

721.221 B35 B57 1302a 1302b0

20

40

60

80

100 Isotype controlAnti-KIR3DL1

Target Cell

Spe

cific

Lys

is (%

)

NA72

1 B8B27

5101

a51

01b

5101

c13

02a

1302

b13

010

10

20

30

40

50

60 Donor 1

Target Cell

Perc

enta

ge o

f KIR

3DL1

posi

tive

cells

expr

essi

ng C

D10

7a

Lab Number Short ID HLA-B AA 80 HLA-A HLA-CIHW 9016 5101a 5101 I 0204 1502R05 0307566X 5101b 5101 I 1101, 3101 1402IHW 9045 5101c 5101 I 0216, 0301 0704, 1502IHW 9084 1302a 1302 T 3001 0602IHW 9093 1302b 1302 T 0201 0602R03 0242430X 1301 1301, 5501 T 0207, 1101 0102, 0304

B

C

A

Chapter 4

95

Figure 4.4. HLA-B*1302 and HLA-B*1301 do not inhibit cytotoxicity of KIR3DL1-

dependent NK cells.

(A) Polyclonally expanded NK cells from donor 1 were incubated with the class I HLA negative

cell line, 721.221, (positive control), a Bw6 control (B8, Table 4.2), a Bw4 control (B27, Table

4.2), two HLA-B*1302 targets, one HLA-B*1301 target and three HLA-B*5101 targets. (B)

CD107a expression was measured on KIR3DL1-positive, CD158b-negative NK cells incubated

with various target cells or alone (NA). CD107a expression was induced by the 721.221 cell line

and the Bw6 control (B8) and not by the Bw4 control (B27) as expected. All targets expressing

either HLA-B*1302 or HLA-B*1301 failed to inhibit CD107a expression. HLA-B*5101 expressing

targets inhibited CD107a expression. (C) Two KIR3DL1-positive NK clones (clone C3 from

donor 4 expressing the low expression KIR3DL1*005 allele and clone D2 from donor 3

expressing the high expression KIR3DL1*01502 allele were used in the four hour 51Cr release

assay against the class I negative cell line (721.221), a Bw6 control (B35, Table 4.2), a Bw4

control (B57, Table 4.2) and two HLA-B*1302 homozygous targets (Table 4A) to confirm lack of

inhibition through KIR3DL1. KIR3DL1 was blocked with anti-DX9 (KIR3DL1) (open bars) and

isotype control (IgG1) (solid bars). Both NK clones lysed 721.221 and the Bw6 control (B35).

Both NK clones were inhibited by the Bw4 control (B57) and inhibition was reversed in the

presence of anti-KIR3DL1 mAb. Neither clone was inhibited by either HLA-B*1302 target with

very little or no reversal of inhibition in the presence of the anti-KIR3DL1 mAb.

Chapter 4

96

4.3.4 HLA-A*2402 and HLA-A*3201 are ligands for KIR3DL1

Four HLA-A alleles react with anti-Bw4 antibodies: HLA-A*23, HLA-A*24, HLA-A*25

and HLA-A*32. When assessed in terms of the ability of target cells expressing these

alleles to inhibit CD107a expression of KIR3DL1 positive NK cells (Figure 4.2), HLA-

A*2402 and HLA-A*3201 were clearly KIR3DL1 ligands whilst HLA-A*2501 appeared

to have some weak inhibitory activity. HLA-A*2301 appeared devoid of inhibitory

activity particularly for NK cells expressing the high level allele (solid bars – Figure 4.2).

Gumperz et al (1995) were unable to show reversibility of HLA-A*24 mediated inhibition

of cytotoxicity using an anti-KIR3DL1 antibody. We therefore investigated the

reversibility of inhibition by HLA-A alleles in a chromium release assay with an NK

clone from donor 4 expressing the KIR3DL1*005 allele. As shown in Figure 4.5, the

HLA bare (721.221) and Bw6-negative control target (B8) were lysed and lysis was not

enhanced by blocking KIR3DL1. The Bw4 control target (B27) completely inhibited

lysis and inhibition was reversed substantially by blocking with mAb. The A24 and A32

targets provided just as effective inhibition as the B27 control and again, inhibition was

blocked by the mAb. In contrast, the A25 target inhibited lysis less effectively and

showed only slightly enhanced lysis after blocking KIR3DL1 suggesting that HLA-

A*2501 may have some weak binding to KIR3DL1. The A23 target provided

intermediate inhibition, which was substantially reversed by blocking the receptor.

Interestingly, in donor 4, A23 was the only target which showed a difference in CD107a

expression between the high and low allele expressing cells suggesting a difference in

the interaction of HLA-A*2301 with these two alleles.

Chapter 4

97

Figure 4.5 Inhibition through Bw4 expressing HLA-A alleles can be reversed by

addition of anti-KIR3DL1.

Clone C3 from donor 4 expressing the low allele KIR3DL1*005 was tested in a four hour 51Cr

release cytotoxicity assay against the class I negative cell line (721.221), a Bw6 control (HLA-

B*0801) (Table 4.2), a Bw4 control (HLA-B*2705) (Table 4.2) and targets expressing one of the

four Bw4 expressing HLA-A alleles, A23, A24, A25 and A32 (Table 4.2). KIR3DL1 was blocked

with either anti-KIR3DL1 antibody (DX9) (open bars) or isotype control (IgG1) (solid bars). Clone

C3 lysed both 721.221 and the Bw6 control, and inhibition was not enhanced by anti-KIR3DL1

antibody. Clone C3 was inhibited by the Bw4 control, the A24, A23 and A32 targets and this

inhibition was reversed by addition of anti-KIR3DL1 antibody. Clone C3 lysed the A25 target

and anti-KIR3DL1 antibody only weakly enhanced specific lysis.

721.221 B8 B27 A24 A23 A25 A32-200

20406080

100 Isotype control Anti-KIR3DL1

Target Cell

Spec

ific

Lysi

s(%

)

Donor 4 (Clone C3 - KIR3DL1*005)

Chapter 4

98

4.3.5 Donors who have HLA-B*1301 or B*1302 can make KIR3DL1 dependent NK

clones but these NK clones are infrequent

As HLA-B*1302 and HLA-B*1301 are poor ligands for KIR3DL1, we asked whether

donors who have HLA-B*13 as their only Bw4-positive allele, can generate KIR3DL1-

dependent NK clones capable of lysing Bw4 negative targets. Using the CD107a

assay, we tested the ability of four donors with the Bw4-positive alleles (donors 1-4,

Table 4.1), three donors with B*1301 or B*1302 as their only Bw4-positive allele

(Donors 5-7, Table 4.1) and two donors with only Bw6 positive alleles (donors 8 and 9,

Table 4.1) to generate KIR3DL1-dependent alloreactive NK clones. NK cells from each

donor were incubated alone, with 721.221, a target lacking only the Bw4 epitope (C1+,

C2+, Bw4-), a target lacking only the C1 epitope (C1-, C2+, Bw4+) and a target lacking

only the C2 epitope (C1+, C2-, Bw4+). KIR3DL1-positive, CD158a,b-negative cells

were selected and CD107a expression measured against the range of target cells.

Representative cytofluorograms of CD107a expression in the presence of 721.221 and

the Bw4-negative target are shown in Figure 4.6A.

Chapter 4

99

1 2 3 4 5 6 7 8 90

25

50

75

100

Donor

Perc

enta

geof

KIR

3DL1

+,C

D15

8a/b

-N

K C

ells

1 2 3 4 5 6 7 8 90123456

Donor

Perc

enta

ge o

fKI

R3D

L1D

epen

dent

NK

Clo

nes

1 2 3 4 5 6 7 8 90

20

40

60Bw4- Target

Donor

Perc

enta

ge o

fKI

R3D

L1 C

ells

Expr

essi

ngC

D10

7a

Bw4 B13 Bw6

30% 70% 46% 54%5% 2%

3%90%

37% 63% 91% 9%7% 9%

59%

25%

Donor 1

Donor 2

721.221 Bw4-

B

C

D

A

Chapter 4

100

Figure 4.6 Donors who express HLA-B*1302 or HLA-B*1301 make very few

KIR3DL1-dependent NK cells.

NK cells from nine donors (donor 1-9) were tested in the CD107a assay to determine their

ability to make KIR3DL1 dependent NK clones. Each donor was incubated with the class I bare

target 721.221, targets lacking either the C1 or C2 epitopes (data not shown) or targets lacking

only the Bw4 epitope (HLA-A*0301, 1101, B*0702, 3501, C*0401, 0702). (A) Representative

examples of flow cytometry showing identification of KIR3DL1-positive, CD158a,b negative NK

cells and their subsequent CD107a expression when incubated with 721.221 or the Bw4-

negative target. (B) Following incubation with the Bw4-negative target, the proportion of

KIR3DL1-positive, 158a,b-negative NK cells that were CD107a-positive did not distinguish

donors with HLA-B*13 from donors with other Bw4-positive alleles and donors homozygous for

Bw6. (C) The proportion of KIR3DL1-positive, CD158a,b-negative NK cells does not distinguish

HLA-B*13 donors from donors with other Bw4-positive alleles and donors homozygous for Bw6.

(D) The proportion of KIR3DL1 dependent NK clones out of the total NK cell population

distinguishes HLA-B*13 donors from donors with other Bw4-positive alleles. 2-3% of NK cells

from donors with other Bw4-positive alleles are KIR3DL1-dependent whereas this is true of less

then 1% of NK cells from HLA-B*13 donors. Less than 0.5% of NK clones from donors

homozygous for Bw6 are KIR3DL1 dependent, slightly less than donors with HLA-B13.

Chapter 4

101

As shown in Figure 4.6B, among the donors with Bw4 alleles other than B*13, donors 1

and 4 up regulated CD107a expression on a large proportion of KIR3DL1-positive NK

cells in response to the Bw4-negative target (KIR3DL1-dependent) whereas for donors

2 and 3, a much lower percentage of KIR3DL1-positive NK cells were KIR3DL1-

dependent. In fact, donors 2 and 3 had similar proportions of KIR3DL1-dependent NK

cells to the three HLA-B*13 and the two Bw6 homozygous donors. Thus, by this

measure, HLA-B*13 donors were not distinguishable from some donors expressing

other Bw4 alleles. However, as shown in Figure 4.6C, the proportion of KIR3DL1-

positive, CD158a,b-negative NK cells also differed considerably between donors.

When the proportion of KIR3DL1-positive, CD158a,b-negative NK cells that were

KIR3DL1-dependent (Figure 6B) was multiplied by the proportion of all NK cells that

were KIR3DL1-positive, CD158a,b-negative (Figure 4.6C) to determine the percentage

of all NK cells from each donor that were KIR3DL1-dependent (Figure 4.6D), HLA-B*13

positive donors could be distinguished from other Bw4 positive donors. Donors with

Bw4 alleles other than HLA-B*13 had 2-3% KIR3DL1-dependent NK clones whereas

donors with HLA-B*13 or homozygous for Bw6 had less than 0.5%. Donors with HLA-

B*13 were, however, able to generate C1-dependent and C2-dependent clones (data

not shown).

4.3.6 Donors who lack Bw4 expressing HLA-B alleles but express HLA-A*2402

can make KIR3DL1 dependent NK clones

As HLA-A*2402 appears to be a ligand for KIR3DL1, we asked whether donors who

lack Bw4 expressing alleles other than HLA-A*2402 are able to make KIR3DL1-

dependent NK clones which could be exploited in haploidentical transplantation. A total

of 129 NK clones were generated from a C1 homozygous, Bw6 homozygous, HLA-

A*2402 positive donor (donor 10, Table 4.1). Each NK clone was tested against four

targets: 721.221 (positive control), a C1+, C2-, Bw4- target (IHW 9065), a C1-, C2+,

Bw4- target (IHW 9019) and a C1+, C2-, Bw4+ target (IHW 9157) in the four hour 51Cr

Chapter 4

102

release assay. 14% of all NK clones killed the targets in a pattern consistent with

KIR2DL2/KIR2DL3-dependent NK clones and expressed CD158b by flow cytometry

indicating that this donor could make KIR2DL2/KIR2DL3-dependent clones. None of

the clones killed targets in a KIR2DL1-dependent manner, as would be expected for an

individual who does not have a C2-positive HLA-C allele. However, a further 12.5% of

all NK clones killed the targets in a pattern consistent with KIR3DL1-dependent NK

clones (killed both Bw4–negative targets but not the Bw4-positive target) and

expressed KIR3DL1 by flow cytometry. Thus, donors who express HLA-A*2402 and

lack Bw4-positive HLA-B alleles can generate KIR3DL1 dependent NK clones.

4.4 Discussion

Our data confirm that KIR3DL1-dependent NK cells lyse targets that are homozygous

for common Bw6 alleles but do not lyse targets with most of the common Bw4 alleles.

These findings are consistent with the rule that the amino acid at position 80 of HLA-B

molecules has the predominant influence on specificity for KIR3DL1 (Cella et al, 1994;

D'Andrea et al, 1995). It has been reported that Bw4 alleles with an isoleucine at

position 80 (80I) are better inhibitors of KIR3DL1 mediated lysis than those with a

threonine at position 80 (80T) (Cella et al, 1994). In particular, class I HLA-bare cells

transfected with HLA-B*5801 (80I) have been reported to be stronger inhibitors of

KIR3DL1-dependent clones than cells transfected with HLA-B*2705 and HLA-B*3701

(80T) (O'Connor et al, 2007). We were unable to confirm these findings. The

interpretation of the data from Cella and colleagues may have been complicated by the

fact that most of the targets used in their study were not homozygous at the HLA-B

locus. In addition, the difference between 80I and 80T alleles was only apparent when

the Bw4 expressing HLA-A alleles were included. It is also possible that the effector

cells used in their study had a KIR3DL1 allele that does interact differently with 80I or

80T and was not represented in our study. Nevertheless, our study included five high

and two low expression alleles of KIR3DL1, including the common KIR3DL1*001, *015

and *005 alleles, and no evidence for an 80I/80T effect was observed. Whilst the

Chapter 4

103

hierarchy of different Bw4-positive HLA-B alleles reported in earlier publications may

well exist if analysed by the particular read outs used in those studies, our data suggest

that these subtleties may not be significant for target cells expressing a normal array of

ligands and polyclonal NK cells.

Among Bw4-positive HLA-B alleles, we found only HLA-B*1302, and B*1301 which is

more common in oriental populations, to be poor KIR3DL1 ligands. Three individuals,

in whom HLA-B*1302 or B*1301 was the only Bw4-positive allele, had a very low

frequency (<0.5%) of alloreactive NK cells dependent on KIR3DL1 compared to donors

with other Bw4-positive alleles (2-3%). These flow cytometric estimates of clonal

frequency are comparable with data from Ruggeri and colleagues obtained by direct

NK cloning (Ruggeri et al, 2006). HLA-B*1301 and HLA-B*1302 are unique among

HLA-B and HLA-C alleles in having a leucine rather than arginine at position 145, a

position that is important in salt bridge formation between HLA-C and KIR (Boyington et

al, 2000). Mutation of asparagine 135 of KIR2DL2, which forms the salt bridge with

arginine 145 of HLA-C, lowered HLA binding affinity by twenty-fold relative to the wild-

type receptor, suggesting a vital role for proper salt bridge formation in stabilising

KIR/HLA interaction. Whilst the structure of the KIR3DL1/HLA-B complex has not been

solved, it is thought that KIR3DL1 interaction with HLA-B may be similar to KIR2DL2

interaction with HLA-C, as many of the positions important in KIR recognition of HLA

are conserved amongst HLA-B and HLA-C alleles (Boyington et al, 2000). It is

possible that the leucine at position 145, which prefers to be buried within the protein

hydrophobic core and which also has a non-reactive side-chain, may not form an

adequate salt bridge with KIR3DL1.

In the initial screen of common Bw4-positive and Bw6-positive HLA alleles, HLA-

B*5101 poorly inhibited NK cell cytotoxicity. This weak inhibition could be low

expression of HLA-B*5101 and hence, low expression of Bw4 on the 5101a cell. When

additional examples of HLA-B*5101 were tested along with the original cell line

Chapter 4

104

B5101a, all three inhibited NK cell cytotoxicity albeit with the original example of B51

giving the weakest inhibition of the three. It is unclear why B5101a exhibited somewhat

better inhibition in the second experiment. However it may be relevant that in the initial

screen, the standard error was large for this target.

Conclusions from previous reports addressing the role of Bw4 positive HLA-A alleles as

ligands for KIR3DL1 have been inconsistent (Cella et al, 1994; Gumperz et al, 1995;

Thananchai et al, 2007). In particular, Gumperz et al (1995) observed inhibition of a

KIR3DL1 positive NK clone by HLA-A*2501 and HLA-A*2403 but inhibition was not

reversed by blocking with anti-KIR3DL1. Our data agree with the findings of

Thananchai et al (2007) showing that HLA-A*2402 (the common subtype of HLA-A24)

is an effective ligand. We show that inhibition by HLA-A24 is reversed by blocking with

anti-KIR3DL1 and furthermore show that an individual whose only Bw4-positive allele

was HLA-A*2402 was able to generate KIR3DL1-dependent NK clones indicating that,

at least in this individual, A*2402 was an effective ligand and capable of “arming” NK

cells for effector function. HLA-A*3201 is also an effective ligand. The status of HLA-

A*2301 and -A*2501 is less clear. HLA-A*2501 expressing targets exhibited, at best, a

weak inhibitory effect in both the CD107a and chromium release assays. The

interaction of HLA-A*2301 with KIR3DL1 is particularly interesting. The A23 target was

virtually ineffective in the CD107a assay with NK cells expressing the KIR3DL1 high

allele but was the only target which clearly showed greater inhibition of NK cells

expressing KIR3DL1*005 than those expressing KIR3DL1*01502 in donor 4. The

inhibitory capacity of HLA-A*2301 for the KIR3DL1*005 allele was confirmed in the

chromium release assay using an NK clone expressing KIR3DL1*005 from this same

donor. These data suggest that the effectiveness of HLA-A*2301 as a KIR3DL1 ligand

may be more dependent on the KIR3DL1 allele than other Bw4 alleles. However

studies using additional A23 targets and further examples of NK cells expressing

KIR3DL1*005 and other alleles are required before definitive conclusions can be

drawn.

Chapter 4

105

Other groups have reported that different KIR3DL1 alleles interact with Bw4 to differing

degrees (Carr et al, 2005; Draghi et al, 2005; Yawata et al, 2006; O'Connor et al,

2007). For alleles other than HLA-A*2301 we found little difference between the

behaviour of the high expression allele KIR3DL1*01502 and the low expression

KIR3DL1*005 allele in donor 4. This result is consistent with the findings of Draghi et al

(2005), except they compared KIR3DL1*005 with KIR3DL1*002 rather than

KIR3DL1*01502. (KIR3DL1*002 differs from KIR3DL1*01502 by a single amino acid in

the D2 domain). However for donor 3, there was a clear difference between the NK

cells expressing the high expression and low expression alleles. The KIR3DL1*005-

like variant differs from KIR3DL1*005 by a single non-synonymous mutation at

nucleotide position 115, is weakly expressed, and the mutation appears to alter the

ability of this receptor to interact with Bw4. Not only is it poorly inhibited by Bw4 alleles,

but NK cells expressing this allele only weakly lyse cells lacking Bw4. Thus KIR3DL1-

dependent clones in donors whose only KIR3DL1 allele is the KIR3DL1*005-like

variant, may be weakly armed and may not be particularly effective alloreactive NK

cells.

One third of Bw4-positive potential stem cell donors cannot make KIR3DL1- dependent

clones. Many of these may have the KIR3DL1*004 allele which is not expressed at the

cell surface. However, our data suggest additional explanations. For example, our data

suggest that individuals with HLA-B*1301 or -B*1302 as their only Bw4-positive allele

will make relatively few KIR3DL1-dependent clones and may therefore be a poor

choice of donor for a haploidentical transplant. On the other hand, individuals who have

HLA-A*2402 or -A*3201 may well be suitable. The influence of KIR3DL1 polymorphism

is also likely to be relevant. Our data suggesting KIR3DL1 polymorphism influences

the arming of NK cells on the one hand and the effectiveness of HLA-A*2301 as a

ligand on the other hand is consistent with reports of others demonstrating KIR3DL1

Chapter 4

106

allele-dependent variation in strength of interaction with different Bw4 alleles (Carr et

al, 2005; Draghi et al, 2005; Yawata et al, 2006; O'Connor et al, 2007).

The exploitation of alloreactive NK cells in haploidentical transplants requires both a

patient amenable to such therapy and a suitable donor. In addition to Bw4, the HLA-

C1 and –C2 epitopes can also mediate NK allorecognition through their corresponding

KIR receptor (Ruggeri et al, 2006). Only cells from patients who lack either Bw4 and or

the C1 or C2 ligand can be lysed by alloreactive NK cells. Only donors who have the

corresponding epitope are capable of NK mediated alloreactivity. The changed status

of HLA-B*13, -A*24 and -A*32 shown in our study means that a number of individuals

(patients and donors) will need reclassification. Individuals currently classified as Bw4

negative (Bw6 homozygous) would now be classified as Bw4 positive if they have HLA-

A24 or –A32 whilst those whose only Bw4 positive allele is HLA-B13 would now be

classified as Bw4 negative. Although we do not have a database of HLA-C typed,

haploidentical pairs in which to directly analyse these effects, we can make some

predictions from the general population. In a database of 200 Caucasian Western

Australians typed at HLA-A, -B and –C, by sequencing, based on the current definition

of Bw4 positivity and the HLA-C alleles present, 28% of individuals would have all three

NK cell epitopes (Bw4, C1, C2) and thus not be amenable to NK allotherapy. However,

HLA-A24, -A32 and –B13 have frequencies of 20%, 6% and 6%, respectively in this

population resulting in 15% of individuals (thus patients and donors) requiring

reclassification. The net effect when examined empirically in this population is such

that the proportion of individuals (and thus patients) who would now be considered to

have all three NK cell epitopes will increase from 28% to 32%. However, for Bw4

negative patients an additional 11% of individuals (and thus donors) would now be

expected to be Bw4 positive and therefore suitable donors. Such effects will also be

evident to varying degrees in other ethnic groups depending upon the frequencies of

the relevant alleles. HLA-B13 for instance, has a frequency of 25% in some Chinese

populations (www.allelefrequencies.net).

Chapter 4

107

Until the rules governing the interaction of HLA alleles and KIR3DL1 alleles are better

understood, methods to accurately estimate the frequency of alloreactive clones are

required. Our data indicate that the simple flow cytometric assay we have used can do

this allowing faster identification of donors suitable for haploidentical transplants and

other NK allo-therapeutic applications. A formal evaluation of the CD107a assay for

detecting C1, C2 and Bw4-dependent alloreactive clones is currently underway.

Chapter 5

108

Chapter 5

KIR2DS1-Mediated Activation Overrides NKG2A-Mediated Inhibition in

HLA-C C2 Negative Individuals

Foley B, De Santis D, Lathbury L, Christiansen F and Witt, C.

International Immunology Accepted 23rd January 2008 This chapter is presented in the form accepted for publication with one alteration. The

materials and methods section has been simplified to include only the NK cell donors

and cell lines used in this chapter. For more detailed methods refer to chapter 2.

Chapter 5

109

Chapter 5: KIR2DS1-Mediated Activation Overrides NKG2A-

Mediated Inhibition in HLA-C C2 Negative Individuals

5.1 Introduction

Natural killer (NK) cell cytotoxicity is controlled by a balance of both activating and

inhibitory signals. To prevent autoreactivity, NK cells do not become competent for

cytotoxicity (armed or licensed) unless they express at least one inhibitory receptor

recognising self-ligands (Raulet et al, 2001; Kim et al, 2005). As the ligands for some

NK inhibitory receptors are allelic epitopes on class I HLA molecules, NK cell clones

that depend on such receptors for inhibition exhibit alloreactivity. It is thought that the

inhibitory signal mediated by these receptors under normal conditions are dominant

over signals generated by activating receptors (Biassoni et al, 1997; Vales-Gomez et

al, 1998)

The killer immunoglobulin-like receptors (KIR) recognise allelic epitopes present on

HLA-A, HLA-B or HLA-C molecules. HLA-C alleles can be divided into two groups, C1

and C2, based on their amino acid sequences in the α1 helix. C1-group alleles have

the amino acid serine (Ser) at residue 77 and asparagine (Asn) at residue 80 whereas

C2-group alleles have the amino acid Asn at residue 77 and Lysine (Lys) at residue 80

(Colonna et al, 1993). The inhibitory receptor KIR2DL1 recognises C2-group alleles

while KIR2DL2 and KIR2DL3 (CD158b) recognise C1-group alleles. Although the

amino acid sequences of activating KIR (KIR2DS1, KIR2DS2, KIR3DS1) suggest that

they might bind to similar ligands as their inhibitory counterparts (KIR2DL1, KIR2DL2/3,

KIR3DL1), it has been difficult to show that activating receptors bind to these ligands.

Alloreactive NK clones from C1 homozygous individuals lyse C2 homozygous EBV-

transformed B lymphoblastoid cell lines (BLCL). These clones have been shown to be

alloreactive due to expression of the inhibitory receptor KIR2DL2 or KIR2DL3

Chapter 5

110

(CD158b) for which the C2 homozygous targets cannot supply the C1 self-ligand, that

is they recognise “missing-self” (Moretta et al, 1995). However, it has been shown that

some NK clones that lyse C2 expressing targets lack CD158b and react with the anti-

CD158a antibody which recognises KIR2DL1 or KIR2DS1. Moreover, activation of

these NK clones by C2 homozygous targets was abolished by addition of anti-CD158a

suggesting that these NK clones mediate alloreactivity by recognition of C2 ligands by

the activating receptor KIR2DS1 (Moretta et al, 1995). Despite this early data there has

been little additional evidence of activation of NK cells through KIR2DS1 recognition of

the C2 ligand. Stewart and colleagues in 2005 (Stewart et al, 2005), used KIR2DL1

and KIR2DS1 tetramers to determine whether these receptors bound to C2. KIR2DL1

tetramers bound to C2 positive uninfected B cells and EBV-infected B cells. In contrast,

KIR2DS1 tetramers were found to bind only to C2 positive B cells after EBV infection. It

was postulated that this difference in KIR2DS1 binding was due to increased HLA-C

density on the surface of EBV-infected cells.

In 2007, Chewning and colleagues provided evidence to support the earlier work of

Moretta and colleagues that KIR2DS1 binding could be responsible for NK cell lysis of

C2 positive BLCL target cells showing that such lysis could be blocked by anti-CD158a

antibody.

Here we provide further evidence supporting the findings of Chewning et al, that

KIR2DS1 positive NK clones are activated by recognition of C2 on BLCL targets. In

addition we show that the activation signal generated by engagement of KIR2DS1

overrides the inhibitory signal generated by engagement of CD94/NKG2A in these

clones. Furthermore we report that C2 homozygous targets undergo greater cell lysis

than C1/C2 heterozygous targets consistent with the idea that activation through

KIR2DS1 is dependent on class I density at the cell surface. We also demonstrate that

KIR2DS1 activated cells can lyse PHA blasts in addition to EBV-transformed cells from

the same individual, indicating that non-transformed cells express enough HLA-C to

Chapter 5

111

activate NK cells through KIR2DS1 highlighting a potential role for these NK clones in

mediating NK alloreactivity.

5.2 Materials and Methods

The materials and methods section has been simplified to include only the NK cell

donors and cell lines used in this chapter. For more detailed methods refer to chapter

2.

Chapter 5

112

Table 5.1. Class I HLA Typing and NK Epitopes of BLCL Target Cell Panel

Cell Identifier HLA-A HLA-B HLA-C NK Epitopes 721.221 - - - - IHW9034 A*0301 B*0702 C*0702 C1 Q94056412J A*0201, 1101 B*0702 C*0702 C1 IHW9065 A*0301 B*0702 C*0702 C1 IHW9157 A*3301 B*5801 C*0302 C1, Bw4 R050307566X A*1101, 3101 B*5101 C*1402 C1, Bw4 R020349726A A*0301, 1101 B*07, 35 C*0401, 0702 C1, C2 R020358799H A*0201 B*4402, 5101 C*0501, 1402 C1, C2, Bw4 Q9454288T A*01, 0301 B*2705, 4403 C*04, 0102 C1, C2, Bw4 R97349692S A*0301, 1101 B*4001, 5701 C*0304, 0602 C1, C2, Bw4 IHW9019 A*3002 B*180101 C*0501 C2 IHW9084 A*0201 B*4002 C*0202 C2 IHW9090 A*0201 B*4402 C*0501 C2, Bw4 IHW9016 A*0201 B*5101 C*1502 C2, Bw4 Q95050308M A*0201 B*4402 C*0501 C2, Bw4 HLA types of target cells used in flow cytometric and chromium release assays unless otherwise stated in the figure legend. In all cases, HLA alleles

were assigned on the basis of sequencing of exons 2 and 3. Cell lines from the 10th International Histocompatibility Workshop are identified with their

workshop identification, all other EBV cell lines are described in Chapter 2.1.1.

Chapter 5

113

Table 5.2. HLA Typing NK Epitopes and KIR Typing of NK Cell Donors

Donor HLA-A HLA-B HLA-C NK Epitopes

KIR2DL1 KIR2DL2 KIR2DL3 KIR3DL1 KIR2DS1

1 A*24, 32 B*18, 51 C*07, 1402 C1, Bw4 + + + + + 2 A*03, 68 B*07, 14 C*07, 08 C1 + - + - + 3 A*1102, 2410 B*1802, 4601 C*01, 07 C1 + + + + + 4 A*01, 03 B*08 C*07 C1 + + + + - 5 A*02, 1101 B*1501, 35 C*0303, 0401 C1, C2 + + - + +

In all cases, HLA alleles were assigned on the basis of sequencing of exons 2 and 3. Full KIR gene repertoire was performed as described in Chapter

2.6.4 for all donors however only relevant KIR genes are shown.

Chapter 5

114

5.3 Results

5.3.1 Alloreactive NK Clones Expressing CD158a Lyse C2 Expressing Targets

As expected, we found that individuals who express the C1 epitope have alloreactive

NK clones expressing KIR2DL2 or KIR2DL3 (CD158b) which are inhibited by BLCL

targets expressing C1 and lyse targets which lack C1. Of the122 NK clones tested from

a C1 homozygous donor (Donor 1), 28 NK clones lysed C1 negative (C2 homozygous)

targets. 26 of these 28 clones expressed CD158b. However, 2 clones (clone C9 and

E11) were CD158b negative (Figure 5.1) suggesting that another receptor must explain

their alloreactivity. Similarly, 4 out of 100 NK clones from another C1 homozygous

donor (Donor 2) lysed C1 negative (C2 homozygous) targets. Of these, 3 were CD158b

positive but 1 (clone P10) was CD158b negative, suggesting that another receptor

must explain its alloreactivity. All three CD158b negative NK clones expressed CD158a

(KIR2DL1 or KIR2DS1). Clones C9 and E11 also expressed NKG2A and clones C9

and P10 also expressed CD158k (KIR3DL2).

Chapter 5

115

Figure 5.1. Receptor expression and cytotoxicity pattern of CD158b negative

clones showing specificity for C2 positive targets.

(A) Receptor expression on NK clones C9, E11 and P10. (B) Flow cytometry histograms

showing NK receptor expression on representative NK clone C9 showing expression of NKG2A

and KIR3DL2. (C) Initial screen cytotoxicity patterns of the three CD158a positive (CD158b

negative) clones. Each NK clone lysed the class I HLA negative target (721.221 cell line) and

the C2 homozygous BLCL target (IHW9019) but not the C1 homozygous BLCL target

(IHW9034) (Table 5.1).

0

20

40

60

80

Target Cell

Spec

ific

Lysi

s(%

)

0

20

40

60

80

Target Cell

0

20

40

60

Target CellC1 - + -C2 - - +Bw4 - - -A3 - + -

C1 - + -C2 - - +Bw4 - - -A3 - + -

C1 - + -C2 - - +Bw4 - - -A3 - + -

Clone C9 Clone P10

Donor Clone NKG2A/B CD158b(KIR2DL2/KIR2DL3/

KIR2DS2)

CD158a(KIR2DL1/KIR2DS1)

CD158e(KIR3DL1)

CD158k(KIR3DL2)

1 C9 + - + - +1 E11 + - + - ND2 P10 - - + - +

Count

NKG2A/B CD158aKIR2DL1/KIR2DS1

CD158bKIR2DL2/KIR2DL3/

KIR2DS2

CD158eKIR3DL1

CD158kKIR3DL2

Clone E11

A

B

C

Chapter 5

116

5.3.2 The Allospecificity of Clone C9 cannot by Explained by KIR3DL2

It has been reported that KIR3DL2 recognises HLA-A*0301 and HLA-A*1101 but not

HLA-A*0201 (Gavioli et al, 1996; Pende et al, 1996). As the C1 homozygous target (C2

negative) used for screening NK clones also expressed HLA-A3 and the C2

homozygous target (C1 negative) did not, we considered the possibility that the lack of

cytotoxicity against the C1 homozygous target was due to HLA-A3 mediated inhibition

through KIR3DL2. NK clone C9 was therefore tested against 6 HLA-A3 homozygous

and 6 HLA-A2 homozygous (ie. HLA-A3 negative) BLCL targets (Figure 5.2). All of

these targets lacked the C2 epitope. Whilst clone C9 lysed the HLA class I negative

cell line, 721.221, it was inhibited by all A*0301 and A*0201 homozygous cell lines

indicating that KIR3DL2 mediated inhibition was unlikely to explain this clone’s

allospecificity.

Chapter 5

117

Figure 5.2. Despite expression of KIR3DL2, clone C9 was inhibited equally well

by HLA-A*0301 and HLA-A*0201 homozygous BLCL target.

(A) NK clone C9 was tested against a target cell panel of 12 BLCL including six targets

homozygous for HLA-A*0301 and six targets homozygous for HLA-A*0201. All targets were

homozygous for the C1 epitope (ie. lacked the C2 epitope). (B) HLA alleles of the BLCL panel.

-101030507090

Target CellSp

ecifi

c Ly

sis

(%)

A*0301 A*0201 721.221

Identifier HLA-A HLA-B HLA-CR8612350C A*0301 B*0702 C*0702R8612340F A*0301 B*1402 C*0802R8612319W A*0301 B*0702 C*0702R8612298B A*0301 B*0702 C*0702R8612318Y A*0301 B*0702 C*0702R8612302Q A*0301 B*0702 C*0702R8612343Z A*0201 B*4501 C*1601R8612289C A*0201 B*2705 C*0102R8612317A A*0201 B*1501 C*0304R8612347R A*0201 B*3801 C*1203R8612346T A*0201 B*1801 C*0702R8612323F A*0201 B*1801 C*0701

Class I HLA Alleles of BLCL Target Panel

A

B

Chapter 5

118

5.3.3 The Allospecificity of Clone C9 is Determined by the Presence of the C2

epitope in a Dose Dependent Manner

To further characterise the allospecificity of clone C9 we tested a more comprehensive

panel of BLCL including C1 homozygous, C2 homozygous and C1/C2 heterozygous

targets (Table 5.1). Clone C9 lysed all five C2 positive targets and none of the C2

negative targets (Figure 5.3). Interestingly, cytotoxicity was stronger against C2

homozygous targets than the C1/C2 heterozygous targets.

Figure 5.3. NK clone C9 only lyses C2 positive BLCL targets and shows a C2

dose effect.

NK clone C9 was tested against a panel of BLCL target cells (IHW9090, IHW9019,

Q95050308M, R020358799H, R020349726A, IHW9034, R050307566X, IHW9157 and

Q94056412J) in the four-hour 51Cr release assay at an E:T ratio of 2:1. NK clone C9 strongly

lysed the HLA class I negative 721.221 cell line (positive control), and the BLCL targets

homozygous for HLA-C group 2. C1/C2 heterozygous targets were moderately lysed

whereas C1 homozygous targets were essentially negative.

0

25

50

75

100

Target Cell

Spec

ific

Lysi

s (%

)

C1 - - + +C2 - + + -

C1-C2-

C1+C2-

C1+C2+

C1-C2+

Chapter 5

119

5.3.4 Clone C9 Expresses KIR2DS1*002

We postulated that the alloreactivity of clone C9 was conferred by the activating

receptor KIR2DS1 (CD158a) interacting with the C2 epitope. In order to confirm that

clone C9 transcribes a functional KIR2DS1 gene, cDNA was prepared from clone C9

and KIR2DS1 was amplified using KIR2DS1 sequence-specific primers (Uhrberg et al,

1997) (Figure 5.4) and primers that would amplify the full length KIR2DS1 cDNA (data

not shown).

Figure 5.4. NK clone C9 transcribes KIR2DS1*002.

Genomic DNA from donor 1 (lane 1), cDNA from NK clone c9 (lane 2) and cDNA from a

CD158a negative NK clone (Donor 2) (lane 3) were amplified with KIR2DS1 specific primers

(Uhrberg et al, 1997). The genomic DNA resulted in an expected 1800bp product (growth

hormone amplification control shown as a 433bp product) confirming the presence of KIR2DS1.

The cDNA resulted in a 353bp product as expected.

The sequence-specific primers detected KIR2DS1 transcript. The full length KIR2DS1

PCR product was also amplified and sequenced. The sequence was identical to

KIR2DS1*002. Since CD158a stains both KIR2DS1 and it’s inhibitory counterpart

KIR2DL1, we also tested the mRNA using KIR2DL1-specific primers. Clone C9 did not

transcribe KIR2DL1 although a KIR2DL1 transcript could be amplified from a positive

control (data not shown).

Chapter 5

120

5.3.5 Activation by C2 Positive Targets is Mediated by KIR2DS1 and Over-rides

NKG2A Mediated Inhibition

In order to prove that lysis of C2 positive targets by the CD158b negative, CD158a

positive clone C9 was mediated by KIR2DS1, we determined the effect on cytotoxicity

of blocking KIR2DS1 using anti-CD158a mAb. The addition of anti-CD158a completely

abolished lysis of C2 positive targets and had no effect on the lysis of C2 negative

targets confirming the role of KIR2DS1 (Figure 5.5). Clone C9 also expressed the NK

inhibitory receptor NKG2A. As all the BLCL target cells used in these experiments

express HLA class I alleles that provide the necessary leader peptides for expression

of HLA-E, NKG2A on clone C9 might be expected to bind to its ligand HLA-E and

inhibit lysis of the target cell. To determine whether NKG2A was functional in clone C9,

we used mAb z199 to block any NKG2A mediated inhibition. As shown in Figure 5.5,

the addition of anti-NKG2A had no effect on lysis of the class I deficient control

(721.221 cell line) but moderately enhanced lysis of 6 out of 7 target cells, indicating

that NKG2A was functional. This data indicates that in the absence of blocking mAbs,

KIR2DS1 mediated activation over-rides NKG2A mediated inhibition. Interestingly,

blocking of NKG2A did not result in enhanced lysis of one of the C1 homozygous target

cells (IHWS 9034). Blocking with the pan-HLA class I mAb, W6/32 resulted in lysis of

this target (data not shown) suggesting that the ligand inhibiting NK cell lysis of this

target cell is an HLA class I molecule but not HLA-E.

Chapter 5

121

Figure 5.5 Target cell lysis by NK clone C9 is enhanced by anti-NKG2A antibody

and inhibited by anti-CD158a (anti-KIR2DL1/KIR2DS1).

NK clone C9 was tested against the class I negative control (721.221 cell line) and a panel of

BLCL target cells with various C1 and C2 combinations (from left-right IHW9019, IHW9084,

IHW9016, R020358799H, R020349726A, IHW9034 and R050307566X). NKG2A and KIR2DS1

receptors were blocked with mAb z199 (NKG2A) (grey bars) and HP-3E4 (CD158a) (open

bars), respectively and isotype control (solid bars). Addition of anti-CD158a blocked lysis of C2

positive targets but had no effect on C2 negative targets. Addition of ant-NKG2A resulted in

moderate enhanced lysis of 6 of the 7 targets.

-10

10

30

50 anti-NKG2Aanti-CD158a

isotype

Target CellSpec

ific

Lysi

s (%

)

C1 - - - - + + + +C2 - + + + + + - -

Chapter 5

122

5.3.6 KIR2DS1 Activated NK Clones Represent 1% of all NK Cells in C1

Homozygous Individuals

To determine the frequency of NK clones that are activated by KIR2DS1 and whether

this varies amongst individuals of different HLA-C and KIR genotype we determined

CD107a expression on CD158a positive NK cells when incubated with C2 positive

BLCL targets. Polyclonal NK cells from a C1 homozygous, KIR2DS1 positive donor

(Donor 3), a C1 homozygous, KIR2DS1 negative donor (Donor 4) and a C1/C2

heterozygous, KIR2DS1 positive donor (Donor 5) were incubated with C1 homozygous

and C2 homozygous BLCL targets and the percentage of CD107a positive cells

determined. C2 positive targets were predicted to induce CD107a expression only on

CD158a positive NK cells from subjects who have the KIR2DS1 gene and in whom C2

is not a self-epitope (ie. C1 homozygous donors). As shown in Figure 5.6A, C2 positive

(either homozygous or heterozygous) targets induced CD107a expression on CD158a

positive NK cells only from the C1 homozygous, KIR2DS1 positive donor.

Representative cytofluorograms are shown in Figure 5.6, B1-6. Figure 5.6, B1-3 show

that a C1 homozygous target cell induced very little CD107a expression on CD158a

positive cells from a C1 homozygous, KIR2DS1 positive donor (1.9% of CD158a

positive cells Figure 5.6, B2) whereas CD107a expression was induced on 33.8% of

CD158a positive cells by a C2 homozygous target (Figure 5.6, B3). These represent

some 0.8% of all NK cells. Neither C1 homozygous nor C2 homozygous targets

induced CD107a expression on CD158a positive cells from a C1 homozygous

KIR2DS1 negative donor (Figure 5.6, B4-6). We performed similar experiments using

C2 homozygous, KIR2DS2 positive individuals and HLA-Bw6 homozygous, KIR3DS1

positive individuals to determine if NK cells could be activated by the C1 or Bw4 (both

80I and 80T) epitopes through KIR2DS2 or KIR3DS1, respectively. We were unable to

detect NK cell activation in such experiments (data not shown).

Chapter 5

123

0

10

20

30

40C1+ 2DS1+C1+ 2DS1-C1+C2+ 2DS1+

Target Cell

Perc

enta

ge o

f CD

158a

cells

exp

ress

ing

CD

107a

C1 - - + +C2 - + + -

NKAlone

721.221 C1+C2-

C1+C2+

C1-C2+

Genotype ofNK Donor

16.7%

70.5%

4.6%

8.2%36.6%

60.9%1.8%

0.7%

C1 HomozygousTarget

C2 HomozygousTarget

2.3%

11.3%

76.5%

10.0%

NK Donor C1+KIR2DS1+

8.4%

85.0%

6.4%

0.2%9.7%

87.7%

2.5%

0.1%1.6%

15.1%

81.6%

1.7%

NK Donor C1+KIR2DS1-

B1 B2 B3

B4 B6B5

A

B

Chapter 5

124

Figure 5.6. NK cell activation through KIR2DS1 (CD158a) only occurs in C2

negative, KIR2DS1 positive individuals.

(A) NK cells from three donors, Donor 3 (C1+C2- KIR2DS1+:solid bars), Donor 4 (C1+C2-

KIR2DS1-:grey bars) and Donor 5 (C1+C2+ KIR2DS1+:open bars) were incubated with 721.221

cell line (positive control), two C2 homozygous cell lines (IHW9019, IHW9084), two C1/C2

heterozygous cell lines (Q9454288T, R97349692S) and two C1 homozygous cell lines

(IHW9034, IHW9065) and CD107a expression was quantified on the CD158a positive

population. All three NK donors lyse 721.221 cells, but only the C1 homozygous, KIR2DS1

positive donor lyses C2 homozygous BLCLs and to a lesser extent C1/C2 heterozygous BLCLs.

NK cells from all donors are inhibited by the C1 homozygous (C2 negative) BLCL targets. (B)

Representative flow cytometric plots for the C1 homozygous, KIR2DS1 positive (Fig 5.6, B1-3)

donor and the C1 homozygous, KIR2DS1 negative (Fig 5.6, B4-6) donor with the percentage of

cells in each quadrant shown. After incubation with target cells, CD158b negative cells were

identified (B1, B4). CD107a and CD158a staining of the CD158b negative NK cells (selected

from B1 or B4) are shown after incubation with a C1 homozygous target (B2, B5) or C2

homozygous target (B3, B6) are shown. CD158a positive cells from the C1 positive, KIR2DS1

positive donor show CD107a staining (8.2% of the CD158a positive cells) only after incubation

with the C2 homozygous target (B3) but not the C1 homozygous target (0.7% of the CD158a

positive cells). The CD158a positive cells from the C1 homozygous, KIR2DS1 negative donor

do not show CD107a staining when incubated with C1 or C2 homozygous targets (0.1% and

0.2% of CD158a cells respectively).

Chapter 5

125

5.3.7 NK Cells Activated by C2 Kill EBV non-infected Targets

As it has been shown that KIR2DS1 binds to EBV-infected cells, but not to uninfected

cells (Stewart et al, 2005), we tested the ability of KIR2DS1 activated NK cells to kill

PHA blasts and EBV BLCL from the same individual. Polyclonally expanded NK cells

from a C1 homozygous, KIR2DS1 positive individual (Donor 3) were incubated with C1

homozygous, C2 homozygous and C1/C2 heterozygous PHA blasts and BLCL targets,

and CD107a expression measured on CD158a positive cells. CD107a expression was

induced on CD158a positive NK cells from Donor 3 by both C2 positive BLCL and PHA

blasts (Figure 5.7), although in all cases, C2 positive BLCL induced CD107a in a higher

percentage of NK cells than PHA blasts from the same individual. As expected CD107a

expression was not induced on CD158a positive NK cells from a KIR2DS1 positive

C1/C2 heterozygous donor (Donor 5) by either C2 positive BLCL or PHA target cells

(Figure 5.7). To determine whether the difference in the ability of BLCL and PHA blasts

to induce CD107a expression on NK cells might reflect differences in the level of class I

expression, BLCL and PHA blasts were stained using the class I HLA specific

monoclonal antibody W6/32. The mean fluorescence intensity of class I HLA

expression was 20-50% higher on BLCL then PHA blasts (data not shown).

Chapter 5

126

Figure 5.7. C2 positive EBV transformed and PHA blast cells both activate

CD158a positive NK cells.

Polyclonally expanded NK cells from a C1+ KIR2DS1+ donor (Donor 3) and a C1+C2+

KIR2DS1+ donor (Donor 5) were used in the CD107a assay against a panel of EBV

transformed BLCL and PHA blasts from the same individuals. (A) HLA typing and NK epitopes

of the BLCL and PHA blast donors. (B) CD158a positive (CD158b negative) NK cells from both

donors expressed CD107a when incubated with the 721.221 cell line (positive control). Only NK

cells from the C1+ KIR2DS1+ donor (Donor 3 solid bars) expressed CD107a in the presence of

both the C2 homozygous and C1/C2 heterozygous BLCL and PHA blasts but, as expected, not

in the presence of the C1 homozygous BLCL or PHA blasts. By contrast the CD158a positive

cells from the C1+C2+ KIR2DS1+ donor (Donor 5 open bars) did not express CD107a in the

presence of any of the target pairs.

0

10

20

30

40

C1+ 2DS1+C1+ C2+ 2DS1+

Target Cell

Perc

enta

ge o

f CD

158a

cel

lsex

pres

sing

CD

107a

C1 - - - + +C2 - + + + -

NKAlone

721 C1C1C2C2(a) C2(b) Genotype ofNK Donor

NK Epitope Cell Identifier HLA-A HLA-B HLA-CC2 (a) R020106262S A*1101, 68 B*1301, 1401 C*04C2 (b) R030309974F A*02, 3001 B*1302, 5701 C*0602C1C2 R030152571S A*0205, 24 B*1503, 3502 C*04, 1203C1 R030316728W A*02, 1101 B*0702, 27 C*0102, 0303

A

B

EBV PHA EBV PHAEBV PHAEBV PHA

Chapter 5

127

5.4 Discussion

We have demonstrated in this study that the specificity of alloreactive NK cells can be

determined by the activating receptor KIR2DS1. These results confirm the results of

other studies providing evidence that NK cells can be activated by KIR2DS1

recognition of the C2 epitope (Moretta et al, 1995; Stewart et al, 2005 and Chewning et

al, 2007). The current data also confirms the finding of Chewning et al, that only C1

homozygous (ie. C2 negative), KIR2DS1 positive individuals are capable of generating

such alloreactive NK clones dependent on KIR2DS1. This is intuitively logical as

KIR2DS1 activated clones would be autoreactive in individuals whose HLA-C alleles

include the C2 epitope. Thus, C1 homozygous individuals produce alloreactive NK

clones dependent on KIR2DL2/3 which are activated by the absence of the C1 epitope,

and also KIR2DS1 positive clones that are activated by the presence of the C2 epitope.

The latter population of cells represents approximately 1% of all NK cells in C1

homozygous individuals. Thus, in addition to detecting “missing-self”, NK cells are

capable of detecting “non-self”.

It is generally believed that under normal conditions inhibitory signals are dominant

over activating signals in controlling NK cell activation. NK cells are only “licensed” for

cytotoxicity if they express an inhibitory receptor for ‘self’. The majority of KIR2DS1

expressing NK clones characterised in this study, and in the studies by Moretta et al

and Chewning et al co-expressed NKG2A suggesting that KIR2DS1 dependent

alloreactive NK cells are “licensed” by NKG2A interaction with self. Our data suggests

that the interaction of activating receptors for non-self ligands, such as NK cells from a

KIR2DS1 positive, C2 negative individual interacting with C2, can over-ride NKG2A-

mediated inhibition. It has previously been reported that under cellular stress the

activating receptor NKG2D can over-ride inhibitory signals to allow NK cells to

eliminate tumour or infected cells (Bauer et al, 1999) but our data is the first evidence

of an activating KIR behaving in a similar manner. Why the inhibitory signal generated

through NKG2A does not suppress the activation signal generated through KIR2DS1 is

Chapter 5

128

unclear. It has been speculated that the ability of inhibitory signals to dominate over

activating signals is due to the low affinity with which activating receptors bind to their

ligand compared with their inhibitory counterparts (Vales-Gomez et al, 1999; Lopez-

Botet et al, 2000). The kinetics of NKG2A binding to HLA-E are similar to those for

inhibitory KIR binding to HLA-C (Vales-Gomez et al, 1999) so it seems unlikely that our

current data showing the inability of NKG2A to over-ride KIR2DS1 can be explained by

low affinity binding of NKG2A to HLA-E.

This study only examined one allele of KIR2DS1 in detail, KIR2DS1*002, which is

expressed by clone C9 and we do not know the KIR2DS1 alleles expressed by the

other donors. Four alleles of KIR2DS1 have been reported (Robinson et al, 2003).

KIR2DS1*001 and KIR2DS1*004 differ from KIR2DS1*002 at positions 70 and 90 of

the first immunoglobulin domain, respectively. KIR2DS1*003 differs from KIR2DS1*002

only in the leader sequence. Differences in the immunoglobulin domains may result in

different binding affinities. Therefore we cannot be sure that KIR2DS1*001 and

KIR2DS1*004 would behave like KIR2DS1*002 whereas it seems likely that

KIR2DS1*003 would have similar specificity to KIR2DS1*002 in its interaction with its

C2 ligand.

Stewart and colleagues (Stewart et al, 2005) demonstrated that KIR2DS1 tetramers

only bound to C2 on EBV-infected cells compared with normal B cells and B cells

infected with members of the human Herpesvirus family and postulated that this was

due to an increase in class I HLA expression induced by EBV infection. In the current

study lysis of C1/C2 heterozygous BLCL was weaker than for C2 homozygous BLCL,

suggesting that increased target cell density of HLA-C enhances KIR2DS1 mediated

activation. We also found that BLCL were more susceptible to lysis than PHA blasts

from the same person and that this correlated with lower class I HLA expression.

Nevertheless, KIR2DS1 activated NK cells were able to lyse PHA activated T cells

Chapter 5

129

(non-EBV infected) from both C2 homozygous and heterozygous individuals despite

lower class I expression than on BLCL from the same individuals.

It is unclear as to whether other activating KIR (KIR2DS2, KIR2DS3, KIR2DS4,

KIR2DS5 and KIR3DS1) can mediate NK alloreactivity. While we and others (Moretta

et al, 1995; Winter et al, 1998; Saulquin et al, 2003; Stewart et al, 2005; Chewning et

al, 2007) have been unable to demonstrate that NK cell activation mediated through

KIR2DS2 interacting with C1, or through KIR3DS1 interacting with Bw4 (O'Connor et

al, 2007), it is possible that this may be due to the fact that these epitopes are simply

not the cognate ligands for these receptors. Therfore furtherwork is required to resolve

this issue.

It is not surprising that KIR2DS1 dependent NK clones, which only arise in C1

homozygous individuals, have been largely overlooked. When considering potential

donors for haploidentical stem cell transplants (HSCT), C1 homozygous donors are

rarely considered because all haploidentical recipients would also express C1 thereby

preventing any inhibitory receptor-mediated NK alloreactivity by such donors.

Leukaemia patients with all three ligands (C1, C2 and Bw4) for inhibitory KIR are

thought to be unable to benefit from NK-mismatched SCT as all inhibitory KIR on

haploidentical donor NK cells would be inhibited by HLA ligands on the recipient’s cells.

The data described in this report suggest that it may be possible for patients with all

three epitopes to benefit from the use of alloreactive NK cells. NK cells from a C1

homozygous sibling donor sharing one haplotype with a C1/C2 heterozygous recipient

may provide an anti-leukaemic effect (and reduced graft versus host disease (GvHD))

through KIR2DS1 activated NK clones. Such NK cells may also be evident in HSCT

using an unrelated donor where the donor has KIR2DS1 and the recipient has C2.

Indeed there is evidence to suggest that C1/C2 heterozygous BMT recipients who

receive a transplant from a KIR2DS1 positive donor have a lower risk of developing

acute GvHD compared to the same transplant in a C1 homozygous recipient

Chapter 5

130

(McQueen et al, 2007). These findings have potentially important implications in the

selection of HSCT donors.

Chapter 6

131

Chapter 6

Final Discussion and Conclusions

Chapter 6

132

Chapter 6 Final Discussion and Conclusions

6.1 The Aims of this Thesis

Whilst it is clear that KIR recognise allelic epitopes present on class I HLA molecules,

there has not been a systematic survey to determine whether the common C1, C2 or

Bw4 positive HLA alleles bind to the expected KIR receptor. Therefore, in

haploidentical BMT, one cannot rely on HLA typing and KIR genotyping of the donor

and HLA typing of the recipient to predict the donor’s NK cells will be alloreactive

towards the recipient. The current protocol for selection of suitable donors for a given

recipient involves lengthy and labour intensive in vitro cloning of the donor’s NK cells to

determine whether they will lyse the recipient’s cells. A better understanding of HLA

and KIR interactions will enable selection of donors and recipient’s from HLA type and

KIR genotype.

6.2 Summary of New Data on HLA:KIR Interaction Provided in this Thesis

6.2.1 KIR2DL2/3 and HLA-C

The survey of common HLA-C alleles undertaken in chapter 3 suggests that the current

understanding of the interaction of HLA-C alleles with KIR2DL2 and KIR2DL3 requires

some revision. Consistent with current dogma, all C2 group alleles failed to inhibit, and

all C1 group alleles inhibited NK cells expressing KIR2DL3*002 to the same degree.

However, the NK cell line, YTS, transfected with KIR2DL3*001 was inhibited to

different degrees by different C1 group alleles. Furthermore, HLA-C*1402 and –C*1403

showed no inhibitory activity for NK cells expressing KIR2DL3*001 or KIR2DL2*001.

Whilst only amino acid 80 in the HLA-C molecule is thought to determine KIR2DL1,

KIR2DL2 and KIR2DL3 specificity, these studies suggest that other positions may

influence HLA-C affinity for KIR2DL2 and KIR2DL3.

Chapter 6

133

6.2.2 KIR3DL1 and Bw4

The survey of common HLA-B alleles undertaken in chapter 4 suggests that the current

understanding of the interaction of HLA-B alleles with KIR3DL1 requires some revision.

It was found that KIR3DL1 positive NK cells lysed target cells homozygous for all

common Bw6 positive HLA-B alleles as expected. Most Bw4 positive HLA-B alleles

inhibited NK cells expressing KIR3DL1 but HLA-B*1301 and –B*1302 were important

exceptions to this rule. The inability of HLA-B*13 to act as a ligand for KIR3DL1 was

confirmed by the inability of HLA-B*13 positive donors to make KIR3DL1 dependent

clones. There are 23 subtypes of HLA-B*13 and all but two subtypes (HLA-B*1303 and

–B*1304) share the lysine at position 145. This position, which is important in salt

bridge formation between KIR and HLA, encodes an arginine in all other HLA-B and

HLA-C alleles. HLA-B*1303 and –B*1304 also express an arginine at this position and

therefore may be expected to bind to KIR3DL1 whereas the other HLA-B*13 alleles

may not.

The controversy surrounding Bw4 positive HLA-A alleles was clarified. HLA-A*2402

and A*3201 clearly inhibited NK cells expressing KIR3DL1 and HLA-A*2402 positive

donors were shown to be capable of making KIR3DL1 dependent NK cells. HLA-

A*2301 and HLA-A*2501 did not appear to be ligands for KIR3DL1 although there was

a suggestion that this may be influenced by the particular KIR3DL1 allele in the case of

HLA-A*2301. 80I and 80T alleles were shown to inhibit equally well.

6.2.3 KIR2DS1 and C2 group HLA-C alleles

Until recently, NK alloreactivity has been attributed to the function of inhibitory KIR.

The ligands and biological function of activating KIR have been unclear. Chapter 5

described the activation of alloreactivity in KIR2DS1 positive NK clones by HLA-C

alleles with the C2 epitope. Such NK cells were only found in individuals whose HLA-C

alleles did not include the C2 epitope as might be predicted if autoaggression is to be

Chapter 6

134

avoided. KIR2DS1 dependent NK clones were shown to occur at a frequency of around

1% in such individuals. Furthermore, KIR2DS1 engagement with its ligand C2 could

override the inhibitory signal generated from NKG2A engagement with its ligand, HLA-

E.

6.3 Limitations to the Conclusions Drawn in this Thesis

The NK cell line, YTS, transfected with KIR2DL3*001 was used to screen the common

C1 group and C2 group HLA-C alleles and conclusions regarding the interaction of

KIR2DL3*001 with HLA-C alleles were based solely on this effector cell. The YTS

transfected cell line has undergone extensive laboratory culture and genetic

manipulation. Therefore it is possible that YTS-2DL3 may not function in exactly the

same way as an NK clone expressing the same allele even though the gene was re-

sequenced and shown not to be mutated. For that reason conclusions concerning the

function of KIR2DL3*001 should be replicated with NK clones expressing the same

receptor. During the course of these studies attempts to generate KIR2DL3*001

dependent NK clones were made but these were unsuccessful.

The conclusion that different alleles of KIR2DL2 and KIR2DL3 interact differently with

HLA-C*1402 were based on results from four NK clones and YTS-2DL3. It is possible

that although lysis of target cells expressing HLA-C*1402, appeared to correlate with

alleles of KIR2DL2 and KIR2DL3, this correlation may be purely coincidental and large

numbers of clones representing these receptors should be analysed. It is possible that

the differences observed between clones was due to other receptors that are

expressed on some NK clones and not others (eg NKG2A). These receptors could

modify responses on those particular clones. Target cells might inhibit NK cells

expressing NKG2A to varying degrees due to differences in HLA-E expression and the

interaction of NKG2A with HLA-E is known to be peptide dependent. Additional

examples of each KIR allele are required to confirm that different reactivities are due to

the allelic differences in the KIR receptors.

Chapter 6

135

While all but one Bw4 positive HLA-B allele tested was a ligand for KIR3DL1, there are

many other subtypes of each broad HLA-B allele that were not tested. It cannot be

assumed that each of these alleles will also be ligands for KIR3DL1. Likewise, HLA-

A*2402 was demonstrated to be a ligand for KIR3DL1, but it is not known whether

other HLA-A*24 subtypes interact with KIR3DL1. Similarly, not all alleles of KIR3DL1

were tested for their ability to interact with Bw4 and different alleles may interact to

differing degrees. In chapter 4 the following alleles were represented: KIR3DL1*001,

*002, *00501, *00501-like, *008, *01501 and *01502, although only KIR3DL1*01502,

*005 and the *005-like allele could be observed in isolation from the influence of other

alleles. Therefore only the ability of these alleles to interact with Bw4 is clear. Other

alleles of KIR3DL1 with appreciable population frequencies, such as KIR3DL1*007 and

*009, are yet to be assessed for their ability to interact with Bw4.

The results in chapter 5 demonstrated that NK clones expressing KIR2DS1*002 can

mediate NK alloreactivity. This study only investigated KIR2DS1*002 in detail and it is

not known whether the other alleles of KIR2DS1 will also interact with C2 to the same

degree (chapter 5.4). Similarly, a formal evaluation of the common C2 alleles was not

performed to assess their ability to interact with KIR2DS1. However in different

experiments, target cells expressing HLA-C*0202, -C*0401, -C*0501, -C*0602 and –

C*1502 all activated NK cells expressing KIR2DS1. These alleles account for the vast

majority of C2 alleles. The only other C2 allele with an appreciable population

frequency is HLA-C*1701.

The ability of a donor to generate alloreactive NK clones is routinely determined by NK

cloning and the chromium release assay. The preliminary data in chapters 4 and 5

suggest that the CD107a assay may be used to detect NK alloreactivity. However,

CD107a expression, a marker of NK cell degranulation, is associated with both

cytotoxicity and IFNγ secretion (Alter et al, 2004). Since IFNγ secretion may not be an

Chapter 6

136

effective alloresponse in a transplant setting, there is a need to distinguish cytokine

secretion from cytotoxicity. Alter and colleagues (2004) demonstrated that following

incubation of NK cells with class I HLA negative targets, CD107a expression correlated

quantatively with NK cell mediated lysis of the target cell using the chromium release.

They also showed that CD107a expression is associated with IFNγ production as less

than 1% of NK cells that secreted IFNγ did not co-express CD107a. However, although

CD107a was expressed both by cells secreting IFNγ and cytotoxic cells, the majority of

NK cells expressed CD107a without secreting IFNγ. Therefore whilst there was a

proportion of cells expressing CD107a that also secrete IFNy, the majority of CD107a

positive cells in that system were cytotoxic. It seems likely that this will also be the

situation in systems measuring KIR dependent alloreactivity, although this remains to

be proven.

The CD107a assay was used in chapter 4 to assess the ability of nine donors to

generate KIR3DL1 dependent NK clones. 2-3% of all NK cells were demonstrated to

be KIR3DL1 dependent, a frequency which correlates with the frequency of KIR3DL1

dependent cytotoxic clones determined by NK cloning and the chromium release assay

(Ruggeri at al, 2006). This supports the idea that the CD107a assay detects cytotoxic

allroeactive NK cells.

6.4 Further Studies

To enable accurate prediction of NK cell alloreactivity from KIR and HLA genetics it will

be necessary to catalogue each exception to the general rules. These studies have

investigated in detail the relationship between KIR3DL1 and Bw4 positive HLA alleles,

and between KIR2DL3 and C1-group HLA-C alleles. Further work is required to

document the interaction between different C1-group HLA-C alleles and KIR2DL2.

Whilst KIR2DL2*001, like KIR2DL3*001, was not inhibited by HLA-C*1402, it is not

clear how KIR2DL2*001 and other alleles of KIR2DL2 interact with the other C1 alleles.

Chapter 6

137

A systematic analysis of the interaction between KIR2DL1 and HLA-C alleles is also

required. It will be of interest to determine why HLA-C*1402 does not interact with

KIR2DL3*001 and KIR2DL2*001. Two polymorphisms in these receptors, one in the

stem and one in the cytoplasmic domain, may influence the interaction. Site-directed

mutagenesis of KIR2DL3*001 will enable the precise residue that determines the

degree of interaction between KIR and HLA-C to be determined. While HLA-C*1402

has no single amino acid that is unique among HLA-C alleles, site-directed

mutagenesis should reveal which amino acid or combination of amino acids influence

interaction with these KIR receptors.

NK clones dependent on KIR2DS1 were shown to mediate NK alloreactivity. The role

of the other activating KIR (KIR2DS2, KIR2DS3, KIR2DS4, KIR2DS5 and KIR3DS1) in

mediating alloreactivity is unknown. The study of the other activating KIR is

complicated by the lack of defined ligands for each of the receptors. It is believed that

C1 may act as a ligand for KIR2DS2, but no one (chapter 5.4) has been able to

demonstrate this interaction. Likewise, until very recently (Alter et al, 2007) no one has

been able to demonstrate a functional interaction between KIR3DS1 and Bw4.

Although the studies in this thesis could not detect KIR2DS2 or KIR3DS1 mediated NK

alloreactivity (considering C1 and Bw4 as their respective ligands), the possibility that

these activating KIR can mediate alloreactivity cannot be excluded. However it will first

be necessary to identify their ligands. When screening donors for NK alloreactivity

using the CD107a assay, any cytotoxicity that cannot be explained by known

receptor:ligand systems warrants further investigation. Investigation of these

alloreactive NK cells could reveal potential ligands for the other activating KIR.

6.5 Implications for Haploidentical HSCT

The findings in this thesis have several implications for the selection of donors for

haploidentical HSCT. KIR2DL3*001, KIR2DL3*002 and KIR2DL2*001 have been

Chapter 6

138

shown to interact differently with HLA-C*1402. This should be taken into consideration

when selecting donors. For example, donors who are homozygous for KIR2DL3*001

and express HLA-C*1402 as their only C1 ligand, may not generate NK clones

dependent on KIR2DL3 and therefore may not be capable of mounting an alloresponse

in vivo. Likewise, as demonstrated in chapter 4, donors who express HLA-B*1301 or

HLA-B*1302 as their only Bw4 positive HLA allele may not a significant number of

KIR3DL1-dependent NK clones and therefore would not be suitable donors to mediate

NK alloreactivity on the basis of Bw4 in vivo. On the other hand, recipients who

express HLA-B*1301 or HLA-B*1302 as their only Bw4 positive HLA allele could be

amenable to treatment with KIR3DL1 dependent alloreactive NK cells. Both HLA-

A*2402 and HLA-A*3201 appear to be strong ligands for KIR3DL1 and it has been

demonstrated that donors with HLA-A*2402 can generate KIR3DL1 dependent NK

clones (chapter 4.3.6). Therefore, expression of these Bw4 positive HLA-A alleles must

also be taken into consideration. HLA-A*2501 and HLA-A*2301 did not inhibit all alleles

of KIR3DL1 tested, and further investigation is still required to determine the interaction

of these alleles with different alleles of KIR3DL1 and therefore there influence on NK

alloreactivity.

Recipients whose HLA type includes all ligands (C1, C2 and Bw4) for the inhibitory KIR

are currently considered to be not amenable to treatment with alloreactive NK cells. as

any donor alloreactive NK cells. The finding that individuals who are homozygous for

the C1 epitope can generate KIR2DS1 dependent NK clones capable of lysing cells

expressing C2 suggests that these patients may actually be amenable to treatments

depending on NK alloreactivity. However, the ability to generate alloreactive NK cells

may not be an “all or nothing” phenomenon and it is not known how frequent

alloreactive clones must be in order to elicit a therapeutic effect in a stem cell

transplant. KIR2DS1 dependent NK clones represent only 1% of NK cells and it

remains to be seen if such small numbers of alloreactive NK cells can elicit a beneficial

alloresponse in a stem cell transplant.

Chapter 6

139

This thesis clarifies several aspects of the interaction of HLA alleles with KIR receptors

and will improve the ability of clinicians and scientists to more correctly identify suitable

donor and recipient pairs for haploidentical HSCT.

References

140

Chapter 7

References

References

141

Chapter 7 References

All author names are provided where there are fewer than 7: for 7 or more, the first 3

authors are listed followed by et al.

Alter, G., Malenfant, J. M. and Altfeld, M. (2004) CD107a as a functional marker for the

identification of natural killer cell activity J Immunol Methods 294(1-2): 15-22

Alter, G., Martin, M. P., Teigen, N., et al (2007) Differential natural killer cell-mediated

inhibition of HIV-1 replication based on distinct KIR/HLA subtypes J Exp Med 204(12):

3027-36

Andoniou, C. E., Andrews, D. M. and Degli-Esposti, M. A. (2006). Natural killer cells in

viral infection: more than just killers. Immunol Rev 214: 239-50.

Arnon, T. I., Achdout, H., Levi, O., et al. (2005). Inhibition of the NKp30 activating

receptor by pp65 of human cytomegalovirus. Nat Immunol 6(5): 515-23.

Arnon, T. I., Lev, M., Katz, G., Chernobrov, Y., Porgador, A. and Mandelboim, O.

(2001). Recognition of viral hemagglutinins by NKp44 but not by NKp30. Eur J

Immunol 31(9): 2680-9.

Atkinson, K., Cooley, M., Farrelly, H., O'Flaherty, E., Ashby, M. and Biggs, J. (1987).

CD4+ T cells appear capable of initiating graft-versus-host disease across non-major

histocompatibility complex (MHC) barriers in man. Bone Marrow Transplant 2(1): 79-

84.

References

142

Aversa, F., Tabilio, A., Velardi, A., et al. (1998) Treatment of high-risk acute leukemia

with T-cell-depleted stem cells from related donors with one fully mismatched HLA

haplotype N Engl J Med 339(17): 1186-93

Barnes, D. W., Corp, M. J., Loutit, J. F. and Neal, F. E. (1956). Treatment of murine

leukaemia with X rays and homologous bone marrow; preliminary communication. Br

Med J 2(4993): 626-7.

Bauer, S., Groh, V., Wu, J., et al. (1999). Activation of NK cells and T cells by NKG2D,

a receptor for stress-inducible MICA. Science 285(5428): 727-9.

Betts, M. and Russell, R. (2003). Amino acid properties and consequences of

subsitutions. Hoboken New Jersey, Wiley.

Biassoni, R.,Cantoni, C., Falco, M., et al. (1996) The human leukocyte antigen (HLA)-

C-specific "activatory" or "inhibitory" natural killer cell receptors display highly

homologous extracellular domains but differ in their transmembrane and

intracytoplasmic portions J Exp Med,183(2): 645-50

Biassoni, R., Pessino, A., Malaspina, A., et al. (1997). Role of amino acid position 70

in the binding affinity of p50.1 and p58.1 receptors for HLA-Cw4 molecules. Eur J

Immunol 27(12): 3095-9.

Bosi, G. and Griffiths, G. (1999). Degranulation plays an essential part in regulating

cell surface expression of Fas ligand in T cells and natural killer cells Nature Medicine

5: 90-96.

Bottino, C., Castriconi, R., Moretta, L. and Moretta, A. (2005). Cellular ligands of

activating NK receptors. Trends Immunol 26(4): 221-6.

References

143

Boyington, J. C., Motyka, S. A., Schuck, P., Brooks, A. G. and Sun, P. D. (2000).

Crystal structure of an NK cell immunoglobulin-like receptor in complex with its class I

MHC ligand. Nature 405(6786): 537-43.

Boyington, J. C. and Sun, P. D. (2002). A structural perspective on MHC class I

recognition by killer cell immunoglobulin-like receptors. Mol Immunol 38(14): 1007-21.

Braud, V. M., Allan, D. S., O'Callaghan, C. A., et al. (1998). HLA-E binds to natural

killer cell receptors CD94/NKG2A, B and C. Nature 391(6669): 795-9.

Brown, D., Trowsdale, J. and Allen, R. (2004). The LILR family: modulators of innate

and adaptive immune pathways in health and disease. Tissue Antigens 64(3): 215-25.

Buckley, R. (2003). Transplantation immunology: Organ and bone marrow Journal of

Allergy and Clinical Immunology 111: S733-44.

Burshtyn, D. N., Scharenberg, A. M., Wagtmann, N., et al. (1996). Recruitment of

tyrosine phosphatase HCP by the killer cell inhibitor receptor. Immunity 4(1): 77-85.

Butturini, A. and Gale, R. P. (1988). T cell depletion in bone marrow transplantation for

leukemia: current results and future directions. Bone Marrow Transplant 3(3): 185-92.

Carr, W. H., Pando, M. J. and Parham, P. (2005). KIR3DL1 polymorphisms that affect

NK cell inhibition by HLA-Bw4 ligand. J Immunol 175(8): 5222-9.

Cella, M., Longo, A., Ferrara, G. B., Strominger, J. L. and Colonna, M. (1994). NK3-

specific natural killer cells are selectively inhibited by Bw4-positive HLA alleles with

isoleucine 80. J Exp Med 180(4): 1235-42.

References

144

Chelvanayagam, G. (1996). A roadmap for HLA-A, HLA-B, and HLA-C peptide binding

specificities. Immunogenetics 45(1): 15-26.

Chewning, J. H., Gudme, C. N., Hsu, K. C., Selvakumar, A. and Dupont, B. (2007).

KIR2DS1-positive NK cells mediate alloresponse against the C2 HLA-KIR ligand group

in vitro. J Immunol 179(2): 854-68.

Ciccone, E., Viale, O., Pende, D., et al. (1988). Specific lysis of allogeneic cells after

activation of CD3- lymphocytes in mixed lymphocyte culture. J Exp Med 168(6): 2403-

8.

Ciccone, E., Colonna, M., Viale, O., et al. (1990) Susceptibility or resistance to lysis by

alloreactive natural killer cells is governed by a gene in the human major

histocompatibility complex between BF and HLA-B Proc Natl Acad Sci U S A 87 (24):

9794-7

Colonna, M., Spies, T., Strominger, J. L., et al. (1992) Alloantigen recognition by two

human natural killer cell clones is associated with HLA-C or a closely linked gene Proc

Natl Acad Sci U S A 89 (17): 7983-5

Colonna, M., Brooks, E. G., Falco, M., Ferrara, G. B. and Strominger, J. L. (1993).

Generation of allospecific natural killer cells by stimulation across a polymorphism of

HLA-C. Science 260(5111): 1121-4.

Colonna, M. and Samaridis, J. (1995) Cloning of immunoglobulin-superfamily members

associated with HLA-C and HLA-B recognition by human natural killer cells Science

268 (5209): 405-8

References

145

Colonna, M., Navarro, F., Bellon, T., et al. (1997). A common inhibitory receptor for

major histocompatibility complex class I molecules on human lymphoid and

myelomonocytic cells. J Exp Med 186(11): 1809-18.

Cudkowicz, G. and Bennett, M. (1971). Peculiar immunobiology of bone marrow

allografts. I. Graft rejection by irradiated responder mice. J Exp Med 134(1): 83-102.

D'Andrea, A., Chang, C., Franz-Bacon, K., McClanahan, T., Phillips, J. H. and Lanier,

L. L. (1995). Molecular cloning of NKB1. A natural killer cell receptor for HLA-B

allotypes. J Immunol 155(5): 2306-10.

Deng, L. and Mariuzza, R. A. (2006). Structural basis for recognition of MHC and

MHC-like ligands by natural killer cell receptors. Semin Immunol 18(3): 159-66.

Dicke K, van Hooft J, and van Bekkum D (1968) The selective elimination of

immunologically competent cells from bone marrow and lymphatic cell mixtures

Transplantation 6: 562-70

Dohring, C. and Colonna, M. (1996). Human natural killer cell inhibitory receptors bind

to HLA class I molecules. Eur J Immunol 26(2): 365-9.

Dohring, C., Scheidegger, D., Samaridis, J., Cella, M. and Colonna, M. (1996). A

human killer inhibitory receptor specific for HLA-A1,2. J Immunol 156(9): 3098-101.

Draghi, M., Yawata, N., Gleimer, M., Yawata, M., Valiante, N. M. and Parham, P.

(2005). Single-cell analysis of the human NK cell response to missing self and its

inhibition by HLA class I. Blood 105(5): 2028-35.

References

146

Farag, S. S., Fehniger, T. A., Ruggeri, L., Velardi, A. and Caligiuri, M. A. (2002) Natural

killer cell receptors: new biology and insights into the graft-versus-leukemia effect

Blood 100(6): 1935-47

Farrell, H., Degli-Esposti, M., Densley, E., Cretney, E., Smyth, M. and Davis-Poynter,

N. (2000) Cytomegalovirus MHC class I homologues and natural killer cells: an

overview Microbes Infect 2(5):521-32

Fan, Q. R., Long, E. O. and Wiley, D. C. (2001). Crystal structure of the human natural

killer cell inhibitory receptor KIR2DL1-HLA-Cw4 complex. Nat Immunol 2(5): 452-60.

Fan, Q. R., Mosyak, L., Winter, C. C., Wagtmann, N., Long, E. O. and Wiley, D. C.

(1997). Structure of the inhibitory receptor for human natural killer cells resembles

haematopoietic receptors. Nature 389(6646): 96-100.

Ferlazzo, G. and Munz, C. (2004). NK cell compartments and their activation by

dendritic cells. J Immunol 172(3): 1333-9.

Ferrara, J. L., Guillen, F. J., van Dijken, P. J., Marion, A., Murphy, G. F. and Burakoff,

S. J. (1989). Evidence that large granular lymphocytes of donor origin mediate acute

graft-versus-host disease. Transplantation 47(1): 50-4.

Ferrara, J. L., Levy, R. and Chao, N. J. (1999) Pathophysiologic mechanisms of acute

graft-vs.-host disease Biol Blood Marrow Transplant 5(6): 347-56

Gardiner, C. M., Guethlein, L. A., Shilling, H. G., et al. (2001). Different NK cell surface

phenotypes defined by the DX9 antibody are due to KIR3DL1 gene polymorphism. J

Immunol 166(5): 2992-3001.

References

147

Gavioli, R., Zhang, Q. J. and Masucci, M. G. (1996). HLA-A11-mediated protection

from NK cell-mediated lysis: role of HLA-A11-presented peptides. Hum Immunol 49(1):

1-12.

Gerosa, F., Baldani-Guerra, B., Nisii, C., Marchesini, V., Carra, G. and Trinchieri, G.

(2002). Reciprocal activating interaction between natural killer cells and dendritic cells.

J Exp Med 195(3): 327-33.

Ghayur, T., Seemayer, T. A., Kongshavn, P. A., Gartner, J. G. and Lapp, W. S. (1987).

Graft-versus-host reactions in the beige mouse. An investigation of the role of host and

donor natural killer cells in the pathogenesis of graft-versus-host disease.

Transplantation 44(2): 261-7.

Gumperz, J. E., Litwin, V., Phillips, J. H., Lanier, L. L. and Parham, P. (1995). The Bw4

public epitope of HLA-B molecules confers reactivity with natural killer cell clones that

express NKB1, a putative HLA receptor. J Exp Med 181(3): 1133-44.

Halfpenny, I. A., Middleton, D., Barnett, Y. A. and Williams, F. (2004). Investigation of

killer cell immunoglobulin-like receptor gene diversity: IV. KIR3DL1/S1. Hum Immunol

65(6): 602-12.

Hansen, J. A., Yamamoto, K., Petersdorf, E. and Sasazuki, T. (1999). The role of HLA

matching in hematopoietic cell transplantation. Rev Immunogenet 1(3): 359-73.

Herberman, R. B. (1986). Natural killer cells. Annu Rev Med 37: 347-52.

Hill, G. R. and Ferrara, J. L. (2000). The primacy of the gastrointestinal tract as a

target organ of acute graft-versus-host disease: rationale for the use of cytokine shields

in allogeneic bone marrow transplantation. Blood 95(9): 2754-9.

References

148

Horowitz, M. M., Gale, R. P., Sondel, P. M., et al. (1990). Graft-versus-leukemia

reactions after bone marrow transplantation. Blood 75(3): 555-62.

Karre, K., Ljunggren, H. G., Piontek, G. and Kiessling, R. (1986). Selective rejection of

H-2-deficient lymphoma variants suggests alternative immune defence strategy.

Nature 319(6055): 675-8.

Kagi, D., Vignaux, F., Ledermann, B, et al. (1994) Fas and perforin pathways as major

mechanisms of T cell-mediated cytotoxicity Science 265(5171): 528-30

Kernan, N. A., Bartsch, G., Ash, R. C., et al (1993) Analysis of 462 transplantations

from unrelated donors facilitated by the National Marrow Donor Program N Engl J Med

328(9): 593-602

Kiessling, R., Klein, E. and Wigzell, H. (1975). "Natural" killer cells in the mouse. I.

Cytotoxic cells with specificity for mouse Moloney leukemia cells. Specificity and

distribution according to genotype. Eur J Immunol 5(2): 112-7.

Kim, S., Poursine-Laurent, J., Truscott, S. M., et al. (2005). Licensing of natural killer

cells by host major histocompatibility complex class I molecules. Nature 436(7051):

709-13.

Lanier, L. L., Le, A. M., Civin, C. I., Loken, M. R. and Phillips, J. H. (1986) The

relationship of CD16 (Leu-11) and Leu-19 (NKH-1) antigen expression on human

peripheral blood NK cells and cytotoxic T lymphocytes. J Immunol 136(12): 4480-6

References

149

Lanier, L. L., Corliss, B. C., Wu, J., Leong, C. and Phillips, J. H. (1998).

Immunoreceptor DAP12 bearing a tyrosine-based activation motif is involved in

activating NK cells. Nature 391(6668): 703-7.

Lanier, L. L. and Phillips, J. H. (1992). Natural killer cells. Curr Opin Immunol 4(1): 38-

42.

Lazetic, S., Chang, C., Houchins, J. P., Lanier, L. L., and Phillips, J. H. (1996) Human

natural killer cell receptors involved in MHC class I recognition are disulfide-linked

heterodimers of CD94 and NKG2 subunits J Immunol 157(11): 4741-5

Litwin, V., Gumperz, J., Parham, P., Phillips, J. H. and Lanier, L. L. (1994). NKB1: a

natural killer cell receptor involved in the recognition of polymorphic HLA-B molecules.

J Exp Med 180(2): 537-43.

Ljunggren, H. G. and Karre, K. (1990). In search of the 'missing self': MHC molecules

and NK cell recognition. Immunol Today 11(7): 237-44.

Lopez-Botet, M., Bellon, T., Llano, M., Navarro, F., Garcia, P. and de Miguel, M.

(2000). Paired inhibitory and triggering NK cell receptors for HLA class I molecules.

Hum Immunol 61(1): 7-17.

Martin A, Kulski J, Witt C, Pontarotti P and Christiansen F (2002). Leukocyte Ig-like

receptor complex (LRC) in mice and men. Trends in Immunology 23(2): 81-88.

Martin, M. P., Gao, X., Lee, J. H., et al (2002) Epistatic interaction between KIR3DS1

and HLA-B delays the progression to AIDS Nat. Genet. 31(4): 429-34

References

150

Maenaka, K., Juji, T., Stuart, D. I. and Jones, E. Y. (1999). Crystal structure of the

human p58 killer cell inhibitory receptor (KIR2DL3) specific for HLA-Cw3-related MHC

class I. Structure 7(4): 391-8.

Mandelboim, O., Lieberman, N., Lev, M., et al. (2001). Recognition of haemagglutinins

on virus-infected cells by NKp46 activates lysis by human NK cells. Nature 409(6823):

1055-60.

Mandelboim, O., Reyburn, H. T., Vales-Gomez, M., et al. (1996). Protection from lysis

by natural killer cells of group 1 and 2 specificity is mediated by residue 80 in human

histocompatibility leukocyte antigen C alleles and also occurs with empty major

histocompatibility complex molecules. J Exp Med 184(3): 913-22.

Matzinger, P. (1997). The immune system's role in graft loss: theoretic considerations.

Transplant Proc 29(8A): 11S-12S.

McQueen, K. L., Dorighi, K. M., Guethlein, L. A., Wong, R., Sanjanwala, B. and

Parham, P. (2007). Donor-recipient combinations of group A and B KIR haplotypes

and HLA class I ligand affect the outcome of HLA-matched, sibling donor

hematopoietic cell transplantation. Hum Immunol 68(5): 309-23.

Meyaard, L., Adema, G. J., Chang, C., et al. (1997). LAIR-1, a novel inhibitory receptor

expressed on human mononuclear leukocytes. Immunity 7(2): 283-90.

Middleton, D., Curran, M. and Maxwell, L. (2002). Natural killer cells and their

receptors. Transpl Immunol 10(2-3): 147-64.

References

151

Miller, J. S., Soignier, Y., Panoskaltsis-Mortari, A., et al. (2005). Successful adoptive

transfer and in vivo expansion of human haploidentical NK cells in patients with cancer.

Blood 105(8): 3051-7.

Moretta, A., Bottino, C., Pende, D., et al. (1990) Identification of four subsets of human

CD3-CD16+ natural killer (NK) cells by the expression of clonally distributed functional

surface molecules: correlation between subset assignment of NK clones and ability to

mediate specific alloantigen recognition J Exp Med 172 (6): 1589-98

Moretta, A., Vitale, M., Bottino, C., et al. (1993) P58 molecules as putative receptors for

major histocompatibility complex (MHC) class I molecules in human natural killer (NK)

cells. Anti-p58 antibodies reconstitute lysis of MHC class I-protected cells in NK clones

displaying different specificities J Exp Med 178(2): 597-604

Moretta, A., Sivori, S., Vitale, M., et al. (1995). Existence of both inhibitory (p58) and

activatory (p50) receptors for HLA-C molecules in human natural killer cells. J Exp

Med 182(3): 875-84.

Moretta, L., Biassoni, R., Bottino, C., Mingari, M. C. and Moretta, A. (2000) Human NK-

cell receptors Immunol Today 21(9): 420-2

Muller, C. A., Engler-Blum, G., Gekeler, V., Steiert, I., Weiss, E. and Schmidt, H.

(1989). Genetic and serological heterogeneity of the supertypic HLA-B locus

specificities Bw4 and Bw6. Immunogenetics 30(3): 200-7.

Murphy, W. J., Kumar, V. and Bennett, M. (1987). Rejection of bone marrow allografts

by mice with severe combined immune deficiency (SCID). Evidence that natural killer

cells can mediate the specificity of marrow graft rejection. J Exp Med 165(4): 1212-7.

References

152

Norman, P. J., Abi-Rached, L., Gendzekhadze, K., et al. (2007). Unusual selection on

the KIR3DL1/S1 natural killer cell receptor in Africans. Nat Genet 39(9): 1092-9.

O'Connor, G. M., Guinan, K. J., Cunningham, R. T., Middleton, D., Parham, P. and

Gardiner, C. M. (2007). Functional polymorphism of the KIR3DL1/S1 receptor on

human NK cells. J Immunol 178(1): 235-41.

Pando, M. J., Gardiner, C. M., Gleimer, M., McQueen, K. L. and Parham, P. (2003).

The protein made from a common allele of KIR3DL1 (3DL1*004) is poorly expressed at

cell surfaces due to substitution at positions 86 in Ig domain 0 and 182 in Ig domain 1.

J Immunol 171(12): 6640-9.

Pende, D., Biassoni, R., Cantoni, C., et al. (1996). The natural killer cell receptor

specific for HLA-A allotypes: a novel member of the p58/p70 family of inhibitory

receptors that is characterized by three immunoglobulin-like domains and is expressed

as a 140-kD disulphide-linked dimer. J Exp Med 184(2): 505-18.

Peruzzi, M., Wagtmann, N. and Long, E. O. (1996) A p70 killer cell inhibitory receptor

specific for several HLA-B allotypes discriminates among peptides bound to HLA-

B*2705 J Exp Med 184 (4): 1585-90

Petersdorf, E. W., Mickelson, E. M., Anasetti, C., Martin, P. J., Woolfrey, A. E. and

Hansen, J. A. (1999) Effect of HLA mismatches on the outcome of hematopoietic

transplants Curr Opin Immunol 11(5): 521-6

Poynton, C. H. (1988). T cell depletion in bone marrow transplantation. Bone Marrow

Transplant 3(4): 265-79.

References

153

Raulet, D. H., Vance, R. E. and McMahon, C. W. (2001). Regulation of the natural

killer cell receptor repertoire. Annu Rev Immunol 19: 291-330.

Reddy, P. and Ferrara, J. L. (2003). Immunobiology of acute graft-versus-host

disease. Blood Rev 17(4): 187-94.

Rhoades, J. L., Cibull, M. L., Thompson, J. S., et al. (1993) Role of natural killer cells in

the pathogenesis of human acute graft-versus-host disease Transplantation 56 (1):

113-20

Riddell, S. R., Berger, C., Murata, M., Randolph, S. and Warren, E. H. 2003 The graft

versus leukemia response after allogeneic hematopoietic stem cell transplantation

Blood Rev 17(3): 153-62

Robinson, J., Waller, M. J., Parham, P., et al. (2003). IMGT/HLA and IMGT/MHC:

sequence databases for the study of the major histocompatibility complex. Nucleic

Acids Res 31(1): 311-4.

Roitt I, Brostoff J and Male, D (2001) Immunology. London, Harcourt Publishers

Rojo, S., Wagtmann, N. and Long, E. O. (1997). Binding of a soluble p70 killer cell

inhibitory receptor to HLA-B*5101: requirement for all three p70 immunoglobulin

domains. Eur J Immunol 27(2): 568-71.

Ruggeri, L., Capanni, M., Casucci, M., et al. (1999). Role of natural killer cell

alloreactivity in HLA-mismatched hematopoietic stem cell transplantation. Blood 94(1):

333-9.

References

154

Ruggeri, L., Capanni, M., Urbani, E., et al. (2002). Effectiveness of donor natural killer

cell alloreactivity in mismatched hematopoietic transplants. Science 295(5562): 2097-

100.

Ruggeri, L., Mancusi, A., Burchielli, E., et al. (2006). Natural killer cell recognition of

missing self and haploidentical hematopoietic transplantation. Semin Cancer Biol

16(5): 404-11.

Saulquin, X., Gastinel, L. N. and Vivier, E. (2003). Crystal structure of the human

natural killer cell activating receptor KIR2DS2 (CD158j). J Exp Med 197(7): 933-8.

Seaman, W. E. (2000). Natural killer cells and natural killer T cells. Arthritis Rheum

43(6): 1204-17.

Shlomchik, W. D., Couzens, M. S., Tang, C. B., et al. (1999). Prevention of graft

versus host disease by inactivation of host antigen-presenting cells. Science

285(5426): 412-5.

Sivori, S., Pende, D., Bottino, C., et al. (1999) NKp46 is the major triggering receptor

involved in the natural cytotoxicity of fresh or cultured human NK cells. Correlation

between surface density of NKp46 and natural cytotoxicity against autologous,

allogeneic or xenogeneic target cells Eur J Immunol 29 (5): 1656-66

Snell, G. D. (1976). Recognition structures determined by the H-2 complex.

Transplant Proc 8(2): 147-56.

Spits, H., Lanier, L. L. and Phillips, J. H. (1995). Development of human T and natural

killer cells. Blood 85(10): 2654-70.

References

155

Stewart, C. A., Laugier-Anfossi, F., Vely, F., et al. (2005). Recognition of peptide-MHC

class I complexes by activating killer immunoglobulin-like receptors. Proc Natl Acad

Sci U S A 102(37): 13224-9.

Storkus, W. J., Howell, D. N., Salter, R. D., Dawson, J. R. and Cresswell, P. (1987) NK

susceptibility varies inversely with target cell class I HLA antigen expression J Immunol

138 (6): 1657-9

Storkus, W. J., Salter, R. D., Alexander, J., Ward, et al. (1991) Class I-induced

resistance to natural killing: identification of nonpermissive residues in HLA-A2 Proc

Natl Acad Sci U S A 88 (14): 5989-92

Sun, J.Y., Galdulis, L., Miller, M.M., et al (2004) Development of a multiplex PCR-SSP

method for Killer-cell immunoglobulin-like receptor genotyping Tissue Antigens 64:

462-68

Suto, Y., Ishikawa, Y., Kasahara, M., et al. (1998). Gene arrangement of the killer cell

inhibitory receptor family on human chromosome 19q13.4 detected by fiber-FISH.

Immunogenetics 48(4): 235-41.

Sviland, L. (2000). The Pathology of Bone Marrow Transplantation. Current

Diagnostic Pathology 6: 242-250.

Tabbara, I. A., Zimmerman, K., Morgan, C. and Nahleh, Z. (2002) Allogeneic

hematopoietic stem cell transplantation: complications and results Arch Intern Med

162(14): 1558-66

References

156

Thananchai, H., Gillespie, G., Martin, M. P., et al. (2007). Cutting Edge: Allele-specific

and peptide-dependent interactions between KIR3DL1 and HLA-A and HLA-B. J

Immunol 178(1): 33-7.

Trinchieri, G. (1995). Natural killer cells wear different hats: effector cells of innate

resistance and regulatory cells of adaptive immunity and of hematopoiesis. Semin

Immunol 7(2): 83-8.

Uhrberg, M., Valiante, N. M., Shum, B. P., et al. (1997). Human diversity in killer cell

inhibitory receptor genes. Immunity 7(6): 753-63.

Uphoff, D. E. (1958). Perclusion of secondary phase of irradiation syndrome by

inoculation of fetal hematopoietic tissue following lethal total-body x-irradiation. J Natl

Cancer Inst 20(3): 625-32.

Vales-Gomez, M., Reyburn, H. T., Erskine, R. A., Lopez-Botet, M. and Strominger, J. L.

(1999). Kinetics and peptide dependency of the binding of the inhibitory NK receptor

CD94/NKG2-A and the activating receptor CD94/NKG2-C to HLA-E. Embo J 18(15):

4250-60.

Vales-Gomez, M., Reyburn, H. T., Erskine, R. A. and Strominger, J. (1998).

Differential binding to HLA-C of p50-activating and p58-inhibitory natural killer cell

receptors. Proc Natl Acad Sci U S A 95(24): 14326-31.

Valiante, N. M., Uhrberg, M., Shilling, H. G., et al. (1997). Functionally and structurally

distinct NK cell receptor repertoires in the peripheral blood of two human donors.

Immunity 7(6): 739-51.

References

157

Vilches, C., Pando, M. J. and Parham, P. (2000a). Genes encoding human killer-cell

Ig-like receptors with D1 and D2 extracellular domains all contain untranslated

pseudoexons encoding a third Ig-like domain. Immunogenetics 51(8-9): 639-46.

Vilches, C. and Parham, P. (2002). KIR: diverse, rapidly evolving receptors of innate

and adaptive immunity. Annu Rev Immunol 20: 217-51.

Vilches, C., Rajalingam, R., Uhrberg, M., Gardiner, C. M., Young, N. T. and Parham, P.

(2000b). KIR2DL5, a novel killer-cell receptor with a D0-D2 configuration of Ig-like

domains. J Immunol 164(11): 5797-804.

Vivier, E. (2006). What is natural in natural killer cells? Immunol Lett 107(1): 1-7.

Wagtmann, N., Biassoni, R., Cantoni, C., et al (1995) Molecular clones of the p58 NK

cell receptor reveal immunoglobulin-related molecules with diversity in both the extra-

and intracellular domains Immunity 2 (5): 439-49

Warren, E. H., Greenberg, P. D. and Riddell, S. R. (1998). Cytotoxic T-lymphocyte-

defined human minor histocompatibility antigens with a restricted tissue distribution.

Blood 91(6): 2197-207.

Watzl, C. (2003). The NKG2D receptor and its ligands-recognition beyond the "missing

self"? Microbes Infect 5(1): 31-7.

Weiden, P. L., Flournoy, N., Thomas, E. D., et al. (1979). Antileukemic effect of graft-

versus-host disease in human recipients of allogeneic-marrow grafts. N Engl J Med

300(19): 1068-73.

References

158

Weiden, P. L., Sullivan, K. M., Flournoy, N., Storb, R. and Thomas, E. D. (1981).

Antileukemic effect of chronic graft-versus-host disease: contribution to improved

survival after allogeneic marrow transplantation. N Engl J Med 304(25): 1529-33.

Winter, C. C., Gumperz, J. E., Parham, P., Long, E. O. and Wagtmann, N. (1998).

Direct binding and functional transfer of NK cell inhibitory receptors reveal novel

patterns of HLA-C allotype recognition. J Immunol 161(2): 571-7.

Winter, C. C. and Long, E. O. (1997). A single amino acid in the p58 killer cell

inhibitory receptor controls the ability of natural killer cells to discriminate between the

two groups of HLA-C allotypes. J Immunol 158(9): 4026-8.

Witt, C. S., Dewing, C., Sayer, D. C., Uhrberg, M., Parham, P. and Christiansen, F. T.

(1999). Population frequencies and putative haplotypes of the killer cell

immunoglobulin-like receptor sequences and evidence for recombination.

Transplantation 68(11): 1784-9.

Yawata, M., Yawata, N., Draghi, M., Little, A. M., Partheniou, F. and Parham, P.

(2006). Roles for HLA and KIR polymorphisms in natural killer cell repertoire selection

and modulation of effector function. J Exp Med 203(3): 633-45.

Yabe, T., McSherry, C., Bach, F. H., et al. (1993) A multigene family on human

chromosome 12 encodes natural killer-cell lectins Immunogenetics 37(6): 455-60

Yokoyama, W. M. (1998). Natural killer cell receptors. Curr Opin Immunol 10(3): 298-

305.

Appendices

159

Appendices

Appendix 1: Solutions and Buffers

Appendix 2: Antibodies

Appendix 3: Primer Sequences

Appendices

160

Appendix 1 Solutions and Buffers

Cell Lines and Tissue Culture

Heat Inactivated Foetal Calf Serum (FCS).

FCS (ThermoTrace, Melbourne AUS) was incubated at 56°C with shaking for 30

minutes, aliquoted and stored at –20°C.

2-Mercaptoethanol (2-ME)

2-ME (ICN Biochemicals, OH USA) was diluted 1:250 in RPMI and stored at –20°C. A

1:1000 dilution of this stock was then added to Iscoves's Modified Dulbecco's Medium

(IMDM) and NK cell culture medium.

RPMI 1640

RPMI 1640 (Invitrogen, Carlsbad USA) supplemented with 500U/mL penicillin,

500μg/mL streptomycin, 2mM L-glutamine and 10% sterile FCS.

Iscoves's Modified Dulbecco's Medium (IMDM)

IMDM (1X) (Invitrogen, Carlsbad USA), liquid contains L-glutamine, 25mM HEPES

buffer, 3,024 mg/L sodium bicarbonate, supplemented with 2-ME and 1μg/mL

puromycin for transfectants.

NK Cell Culture Medium

RPMI 1640 supplemented with 500U/mL penicillin, 500μg/mL streptomycin, 2mM L-

glutamine, 0.1mM non-essential amino acids, 1mM pyruvate, 2-ME and 10% sterile

FCS.

Appendices

161

1 x PBS

17g NaCl, 2.68g Na2HPO42H2O (disodium hydrogen orthophosphate) and 0.78g

NaH2PO42H2O (sodium dihydrogen orthophosphate) were dissolved in 2L MilliQ water

and the pH adjusted to 7.2. Stored at room temperature.

Freezing Medium

Freezing medium was always made freshly with 10% DMSO (BDH, Poole UK) added

to 90% heat-inactivated FCS. Added to the cells drop-wise and always on ice.

Trypan Blue

0.5g Trypan Blue (ICN Biochemicals, Ohio USA) was dissolved in 100mL 1 x PBS. The

solution was sterilised using a 0.22μM filter and stored at room temperature.

Immunophenotyping

Flow Buffer 1%FCS and 0.2% sodium azide was mixed with 1 x PBS to make flow buffer and

stored at 4°C

4% Paraformaldehyde

4g of paraformaldehyde were dissolved in 100mL 1xPBS at 65°C with gentle shaking

to dissolve. Stored at 4°C.

Cytotoxicity Reagents

51Cr (Amersham, Buckinghamshire UK)

74MBq (2mCi) 51-Chromium sodium chromate in a sodium chloride solution was

stored at 4°C and used for up to 12 weeks.

Appendices

162

Triton X-100 (SigmaUltra, USA)

Triton X-100 was diluted 1 in 1000 in distilled water to obtain 0.1% Triton X-100.

PCR Reagents

40mM deoxynucleotide triphosphates (dNTPs) (Invitrogen, CA USA) 40mM dNTPs dATP 200μl dCTP 200μl dGTP 200μl dTTP 200μl Sterile Water 1200μl Total Volume 2000μl PCR Buffer Stock Solutions 1 M MgCl2 20.3g MgCl2.6H2O in 100ml sterile deionised water 1M Tris-HCl 60.6g Trizma Base in 500ml + concentrated HCl to

pH8.3 1 M KCl 37.3g KCl in 500ml sterile deionised water 20mg/ml gelatin 2g gelatin in 100ml sterile deionised water 10 x PCR Buffer (25mM MgCl2)

Stock Solution Volume of Stock Solution (mL)

Final Concentration of Stock

1 M MgCl2 0.625 25mM 1 M Tris-HCl (pH8.3) 2.5 100mM 1 M KCl 12.5 500mM 20mg/ml gelatin 0.125 0.1mg/ml Sterile deionised water 9.25 Total 25

10 x PCR Buffer (20mM MgCl2)

Stock Solution Volume of Stock

Solution (mL) Final Concentration of

Stock 1 M MgCl2 0.5 20mM 1 M Tris-HCl (pH8.3) 2.5 100mM 1 M KCl 12.5 500mM 20mg/mL gelatin 0.125 0.1mg/mL Sterile deionised water 9.38 Total 25

Appendices

163

0.5 x TBE 10 x TBE was initially prepared as follows: 215.6g of Trizma Base was added to 110g

of boric acid and 16.4g of EDTA. This was then diluted 1 in 20 to make 0.5 x TBE.

1% Agarose Gel

4g of agarose (Invitrogen, CA USA) was added to 400mL 0.5 x TBE. The agarose was

dissolved by heating in a microwave for 7 minutes. After heating 40ul of ethidium

bromide was added.

Type IV DNA Loading Buffer

5g of Ficoll 400 (Amersham Pharmacia Biotech, Sweden) and 400μl of 0.25%

bromophenol blue were dissolved in 10mL of sterile distilled water.

1kb Plus Lambda Ladder

1mL of 1kb Plus DNA ladder (Invitrogen, CA USA) was added to 2mL of Type IV DNA

loading buffer and 7mL of sterile, deionised water.

Appendices

164

Appendix 2 Antibodies

Antibody Fluorochrome Isotype Clone Dilution Concentration Supplier Use CD56 PC5 IgG1 N901(NKH1) 1/20 - Beckman Coulter Phenotyping CD56 PeCy7 IgG1 B159 1/40 - BD Biosciences CD107a assay CD158a PE IgG1 EB6B 1/10 - Beckman Coulter Phenotyping CD158b PE IgG1 GL183 1/10 - Beckman Coulter Phenotyping CD158a APC IgG1 EB6B 1/25 - Beckman Coulter CD107a assay CD158b APC IgG1 GL183 1/25 - Beckman Coulter Phenotyping/CD107a Assay CD158a PE IgM HP-3E4 1/10 - BD Biosciences Phenotyping/CD107a Assay CD158b PE IgG2B CH-L 1/10 - BD Biosciences Phenotyping/CD107a Assay CD158e PE IgG1 z27.3.7 1/10 - Beckman Coulter Phenotyping NKB1 PE IgG1 DX9 1/10 - BD Biosciences Phenotyping/CD107a Assay NKG2A PE IgG2b z199 1/20 - Beckman Coulter Phenotyping ILT-2 PE IgG2b GH1/75 1/10 - BD Biosciences Phenotyping W6/32 unconjugated IgG2a - neat - gift from McCluskey HLA expression Bw4 unconjugated IgG2a - neat - gift from McCluskey HLA expression Bw6 unconjugated IgG2a - neat - gift from McCluskey HLA expression CD158a unconjugated IgM HP-3E4 1/100 10μg/ml BD Biosciences Receptor blocking CD158b unconjugated IgG2B CH-L 1/500 2μg/ml BD Biosciences Receptor blocking Q66 unconjugated IgM Q66 neat - gift from Pende Phenotyping/receptor blocking NKB1 unconjugated IgG1 DX9 1/400 2.5μg/ml BD Biosciences Receptor blocking NKG2A unconjugated IgG2b z199 1/100 10μg/ml gift from M. Lopez-Botet Receptor blocking IgG isotype IgG1 679.1Mc7 1/20 - Beckman Coulter Isotype control IgG FITC IgG 1/100 - Chemicon Secondary antibody IgG isotype IgG1 1/100 10μg/ml eBiosciences Receptor blocking IgG2a isotype IgG2a 7T4-1F5 1/100 - Beckman Coulter Isotype control IgG2b unconjugated IgG2b 1/100 10μg/ml eBiosciences Receptor blocking IgM isotype IgM GC323 1/100 - Beckman Coulter Isotype control IgM isotype IgM 1/100 10μg/ml eBiosciences Receptor blocking IgM PE IgM GC323 1/100 - Beckman Coulter Secondary antibody CD107a FITC IgG1 H4A3 1/20 - BD Biosciences CD107a assay

Appendices

165

Appendix 3 – Primer Sequences

Primer Sequence 5' - 3' Target Sequence Reference KIR2DL3F CACCATGTCGCTCATGGTCGTCAGC full length KIR2DL3 cDNA In house KIR2DL3R CAGGAAACAGCTATGACCCAGGACAACTTTGGATCA SSP reverse primer Uhberg et al, 1997 KIR2DL2F CACCATGTCGCTCATGGTCGTCAGC full length KIR2DL2 cDNA In house KIR2DL2R CAGGAGACAACTTTGGATCTGGA full length KIR2DL2 cDNA In house KIR2DS1F TGTAAAACGACGGCCAGTATGTCGCTCAYGGTCGTC full length KIR2DS1 cDNA In house KIR2DS1R CAGGAAACAGCTATGACCGTGAAAACACAGTGATCCAA full length KIR2DS1 cDNA In house KIR2DS1(D)F TGTAAAACGACGGCCAGTTCTCCATCAGTCGCATGAR SSP primer used for genomic DNA Uhberg et al, 1997 KIR2DS1(D)R CAGGAAACAGCTATGACCAGGGCCCAGAGGAAAGTT SSP primer used for genomic DNA Uhberg et al, 1997 KIR2DL1(R)F TGTAAAACGACGGCCAGTGCAGCACCATGTCGCTCT SSP primer used for cDNA Uhberg et al, 1997 KIR2DL1(R)R CAGGAAACAGCTATGACCGTCACTGGGAGCTGACAC SSP primer used for cDNA Uhberg et al, 1997 3DL13D0F TGTAAAACGACGGCCAGTCTTCTGGGCACTGGGAGT KIR3DL1 exon 3 Norman et al, 2007 3DL13D0R CAGGAAACAGCTATGACCACAGTGAGAAGCCCAGACG KIR3DL1 exon 3 Norman et al, 2007 3DL14D1F TGTAAAACGACGGCCAGTAGGAGAGAGACAGACACG KIR3DL1 exon 4 Norman et al, 2007 3DL14D1R CAGGAAACAGCTATGACCTGTCCCAGTGACAATGAGAAC KIR3DL1 exon 4 Norman et al, 2007 3DL15D2F TGTAAAACGACGGCCAGTAAAGGTAGAAGGAGGAAACAGAT KIR3DL1 exon 5 Norman et al, 2007 3DL15D2R CAGGAAACAGCTATGACCGGAAGCTCCTTAGCTAAGGATT KIR3DL1 exon 5 Norman et al, 2007 3DL17-9F TGTAAAACGACGGCCAGTCTTGTCCGAAAGAGATGCTGTAA KIR3DL1 exons 7-9 Norman et al, 2007 3DL17-9R CAGGAAACAGCTATGACCAAGCAAGAGAGAGGCACCA KIR3DL1 exons 7-9 Norman et al, 2007 M13F TGTAAAACGACGGCCAG sequence tags In house M13R CAGGAAACAGCTATGAC sequence tags In house HGHI CAGTGCCTTCCCAACCATTCCCTTA internal primers for KIR SSP In house HGHII ATCCACTCACGGATTTCTGTTGTGTTTC internal primers for KIR SSP In house

Appendices

166