structural immunology and crystallography help immunologists see the immune system in action: how t...

12
Critical Review Structural Immunology and Crystallography Help Immunologists See the Immune System in Action: How T and NK Cells Touch Their Ligands Yong Chen 1,2 , Yi Shi 1,2 , Hao Cheng 1,2 , Yun-Qing An 3 and George F. Gao 1,2,3 1 CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology, Chinese Academy of Sciences, Beijing, People’s Republic of China 2 College of Life Sciences, Graduate University, Chinese Academy of Sciences, Beijing, People’s Republic of China 3 Department of Immunology, Capital Medical University, You-an-men, Beijing, People’s Republic of China Summary Host immune system is an important and sophisticated sys- tem, maintaining the balance of host response to ‘‘foreign’’ anti- gens and ignorance to the normal-self. To fulfill this achieve- ment the system manipulates a cell–cell interaction through appropriate interactions between cell-surface receptors and cell- surface ligands, or cell-secreted soluble effector molecules to their ligands/receptors/counter-receptors on the cell surface, triggering further downstream signaling for response effects. T cells and NK cells are important components of the immune system for defending the infections and malignancies and main- taining the proper response against over-reaction to the host. Receptors on the surface of T cells and NK cells include a num- ber of important protein molecules, for example, T cell receptor (TCR), co-receptor CD8 or CD4, co-stimulator CD28, CTLA4, KIR, CD94/NKG2, LILR (ILT/LIR/CD85), Ly49, and so forth. These receptor molecules interact with their ligands on the tar- get cells, including major histocompatibility complex (MHC) (or human leukocyte antigen, HLA), CD80, CD86, and so forth. Detailed understanding of these receptor–ligand pair interac- tions is crucial for our full knowledge of the immune system, ultimately for us to manipulate the T cell and NK cell functions. Accumulations of the receptor–ligand complex crystal struc- tures in the recent years have provided us a unique angel to see how the immune cells interacting with their partner cells. In this review, we discussed binding specificity, plasticity, and flexi- bility of the T cell and NK cell receptor/ligand interaction, fit- ting the structural data with their functions. Structural immu- nology indeed helps us see how T and NK cells ‘‘touch’’ their target cells in our immune system. Ó 2009 IUBMB IUBMB Life, 61(6): 579–590, 2009 Keywords T cell; NK cell; structural immunology; crystallography; immune system; receptor; ligand; interaction; MHC; HLA; KIR; LILR. INTRODUCTION Vertebrates have had not only innate immunity but also are able to mount defense mechanisms that constitute adaptive im- munity to protect the body from foreign or changed self-attack. The vertebrate immune system has a unique ability to mount a highly specific response against almost any foreign antigens, even those never seen before in the course of evolution, and more importantly antigens of self-malignancies (self-changed antigens). Cells involved in the immune system cover a broad range of different types. Among them the cytotoxic lympho- cytes play a crucial role in destroying the foreign or changed self-antigens. The cytotoxic lymphocytes include at least the following three types: T cells, NK (natural killer) cells, and the NKT cells (natural killer T cells). T cells belong to a group of lymphocytes expressing CD3, CD4/CD8, playing important roles in antigen-specific cell-mediated immunity. NK cells are large granular cytotoxic lymphocytes expressing CD16 and CD56, but not CD3, constituting a major component of the innate immune system. NKT cells are a heterogeneous group of T cells that share properties of both T cells and NK cells, rec- ognizing foreign or self lipids and glycolipids presented by non- polymorphic CD1d. Address correspondence to: Prof. George F. Gao, DPhil, CAS Key Laboratory of Pathogenic Microbiology and Immunology, Institute of Microbiology, Chinese Academy of Sciences, Beijing 100101, People’s Republic of China. Tel: 186-10-64807688. Fax: 186-10-64807882. E-mail: [email protected] Received 15 March 2009; accepted 15 March 2009 ISSN 1521-6543 print/ISSN 1521-6551 online DOI: 10.1002/iub.208 IUBMB Life, 61(6): 579–590, June 2009

Upload: yong-chen

Post on 11-Jun-2016

213 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

Critical Review

Structural Immunology and Crystallography HelpImmunologists See the Immune System in Action:How T and NK Cells Touch Their Ligands

Yong Chen1,2, Yi Shi1,2, Hao Cheng1,2, Yun-Qing An3 and George F. Gao1,2,3

1CAS Key Laboratory of Pathogenic Microbiology and Immunology (CASPMI), Institute of Microbiology,Chinese Academy of Sciences, Beijing, People’s Republic of China2College of Life Sciences, Graduate University, Chinese Academy of Sciences, Beijing, People’s Republic of China3Department of Immunology, Capital Medical University, You-an-men, Beijing, People’s Republic of China

Summary

Host immune system is an important and sophisticated sys-tem, maintaining the balance of host response to ‘‘foreign’’ anti-gens and ignorance to the normal-self. To fulfill this achieve-ment the system manipulates a cell–cell interaction throughappropriate interactions between cell-surface receptors and cell-surface ligands, or cell-secreted soluble effector molecules totheir ligands/receptors/counter-receptors on the cell surface,triggering further downstream signaling for response effects. Tcells and NK cells are important components of the immunesystem for defending the infections and malignancies and main-taining the proper response against over-reaction to the host.Receptors on the surface of T cells and NK cells include a num-ber of important protein molecules, for example, T cell receptor(TCR), co-receptor CD8 or CD4, co-stimulator CD28, CTLA4,KIR, CD94/NKG2, LILR (ILT/LIR/CD85), Ly49, and so forth.These receptor molecules interact with their ligands on the tar-get cells, including major histocompatibility complex (MHC)(or human leukocyte antigen, HLA), CD80, CD86, and so forth.Detailed understanding of these receptor–ligand pair interac-tions is crucial for our full knowledge of the immune system,ultimately for us to manipulate the T cell and NK cell functions.Accumulations of the receptor–ligand complex crystal struc-tures in the recent years have provided us a unique angel to seehow the immune cells interacting with their partner cells. Inthis review, we discussed binding specificity, plasticity, and flexi-bility of the T cell and NK cell receptor/ligand interaction, fit-ting the structural data with their functions. Structural immu-

nology indeed helps us see how T and NK cells ‘‘touch’’ theirtarget cells in our immune system. � 2009 IUBMB

IUBMB Life, 61(6): 579–590, 2009

Keywords T cell; NK cell; structural immunology; crystallography;

immune system; receptor; ligand; interaction; MHC;

HLA; KIR; LILR.

INTRODUCTION

Vertebrates have had not only innate immunity but also are

able to mount defense mechanisms that constitute adaptive im-

munity to protect the body from foreign or changed self-attack.

The vertebrate immune system has a unique ability to mount a

highly specific response against almost any foreign antigens,

even those never seen before in the course of evolution, and

more importantly antigens of self-malignancies (self-changed

antigens). Cells involved in the immune system cover a broad

range of different types. Among them the cytotoxic lympho-

cytes play a crucial role in destroying the foreign or changed

self-antigens. The cytotoxic lymphocytes include at least the

following three types: T cells, NK (natural killer) cells, and the

NKT cells (natural killer T cells). T cells belong to a group of

lymphocytes expressing CD3, CD4/CD8, playing important

roles in antigen-specific cell-mediated immunity. NK cells are

large granular cytotoxic lymphocytes expressing CD16 and

CD56, but not CD3, constituting a major component of the

innate immune system. NKT cells are a heterogeneous group of

T cells that share properties of both T cells and NK cells, rec-

ognizing foreign or self lipids and glycolipids presented by non-

polymorphic CD1d.

Address correspondence to: Prof. George F. Gao, DPhil, CAS Key

Laboratory of Pathogenic Microbiology and Immunology, Institute of

Microbiology, Chinese Academy of Sciences, Beijing 100101, People’s

Republic of China. Tel: 186-10-64807688. Fax: 186-10-64807882.

E-mail: [email protected]

Received 15 March 2009; accepted 15 March 2009

ISSN 1521-6543 print/ISSN 1521-6551 online

DOI: 10.1002/iub.208

IUBMB Life, 61(6): 579–590, June 2009

Page 2: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

The recognition molecule on T cells is the membrane-anch-

ored T cell antigen receptor (TCR) which recognize a compos-

ite ligand consisted by small antigen fragments and major histo-

compatibility complex (MHC) (or human lymphocyte antigen,

HLA, in human) (1). T cell receptors are classified as two dif-

ferent heterodimers, abTCR, and cdTCR. Receptors on NK cell

surface are divergent with different members as discussed in the

text later and interact with several groups of ligands, including

peptide MHC (pMHC).

The T cell/NK cell system is quite sophisticated and coordi-

nated each other in the immune system. To escape from killing

of T cells, cells infected by viruses or other pathogens would

have a lowered level of, or that have lost the expression of,

MHC class I molecules. However, these ‘‘missing-self’’ cells

can be recognized by NK cells which can spare normal cells

expressing adequate amounts of MHC class I molecules (2).

Thus, the altered expression of class I antigens, a common

event in tumor transformation or following viral infection with

CTL escape, leads to NK-mediated target cell lysis.

Initiation of the immune response of the T cells starts with

interaction of TCR with its ligand pMHC. Current set of struc-

tural data of abTCR–pMHC complex reveals that the abTCR–pMHC interaction adopts an ‘‘induced-fit’’ model (3). This means

that the abTCR undergoes large conformational change or rear-

rangement when it engages on the pMHC, adjusting their fit to

the interactive ligands. On the other hand, CD8/MHC and NK

receptors/MHC adopts a ‘‘lock and key’’ model. ‘‘Induced fit’’

represents the adaptive receptor–ligand interaction, whereas ‘‘lock

and key’’ represents the innate receptor–ligand interaction (4).

As NK cells are bipartite and their functions were regulated

by a fine tuning between opposite signals delivered by inhibi-

tory and/or activating receptors (2). Human NK cells express

multiple receptors specific for different alleles of HLA class I

molecules expressed on normal cells. This interaction leads to

an inhibitory signal which prevents normal cell lysis. These

receptors can also be cataloged into two distinct structural fami-

lies: members of the immunoglobulin (Ig) receptor superfamily,

which mainly include the killer-cell Ig-like receptors (KIRs)

and leukocyte Ig-like receptor (LILR/LIR or immunoglobulin-

like transcript, ILT, or CD85). The other receptors are C-lectin

type receptors mainly composed of CD94/NKG2 heterodimer

and Ly49 homodimer.

As the late Professor Don C. Wiley (Harvard, GFG’s postdoc

mentor during 1999-2001) once said, ‘‘I do not know the biol-

ogy until I see it.’’ How can we see the biology? Structural

biology indeed helps the immunologists understand the immune

recognition in a simpler way, ‘‘see’’ the action. Peptide presen-

tation by MHC is a very good example. Don Wiley, Pamela

Bjorkman, and Jack Strominger and their colleagues’ work

made the T cell recognition of the foreign antigen really visible,

as for a long time immunologists did not understand why for-

eign antigens were recognized by the immune system always

correlate with the MHC. The peptide comfortably sitting on the

top of MHC helices’ ‘‘bed’’ gave us a clear picture. We really

see the biology, the immunobiology! Functional data were inter-

preted perfectly by the structural data.

As discussed earlier, the receptor–ligand interactions between

the T and NK cells are quite complicated and a lot of receptors/

ligands are involved in the cell–cell interface. In this review,

our discussion mainly focuses on the structural binding modes

of the T cell receptors and NK receptors. These receptors are

the major interests in our laboratory, such as LILR family and

TCR. Other receptors are mentioned briefly.

SPECIFICITY DETERMINED BY INTERACTIONOF ab T CELL RECEPTORS WITH PEPTIDE-MHC

T cell receptor (TCR) engaging with peptide-major histo-

compatibility complex (pMHC) is a complicated molecule–mol-

ecule interaction which still presents one of the most interesting

and enduring structural puzzles in biology. Recent progress on

TCR-pMHC complex structure (Fig. 1) truly reflects the beauty

of structural biology which vividly reveals the molecular basis

of their interaction: seeing the immunobiology by crystallogra-

phy. Similar to antibody molecules, abTCRs are generated by

recombination of a family of gene segments that form the

diverse TCR repertoire (as high as to 1014 different sequences)

42 Va (5) and 46 Vb (6) genes are used to constitute the first

two variable loops (CDR1 and CDR2), while the third variable

loops (CDR3) are formed by the randomly joining of V and J

(in TCR a), or by V, D, and J (in TCR b). The joining process

itself generates further diversity both by removing nucleotides

and by introducing nongermline nucleotides at each junction.

The diversity of TCR repertoire facilitates the existence of spec-

ificity of T cell response which is determined by recognition of

an antigenic peptide bound to a class I or class II MHC mole-

cule by the antigen-specific TCR.

Up to 20 TCR-pMHC complex structures have been pub-

lished in the past 10 years, but there is still no uniform agree-

ment among structural biologists and immunologists about how

to interpret this structural information. Here, we only want to

discuss the original hypothesis that TCR and MHC molecules

coevolved a ‘‘preference or specificity’’ to interact with one

another (7). To analyze the specificity determined by the inter-

action of abTCRs with pMHC, the binding interface can be

functionally and structurally segregated into four distinct com-

ponents that collectively form composite surfaces (8). In abTCRmolecules, the germline-encoded components (CDR1s and

CDR2s) and the highly diverse components (CDR3s) form the

pMHC-binding site; while in MHC molecules, the germline-

encoded components (a1 and a2 helices) and the highly diverse

components (a array of peptides) form the combining site

‘‘visible’’ to the TCR. Thus, the TCR–pMHC interface can be

segregated into invariant and variant parts based on different

sources in which the most variable CDR3 loops of the TCR are

positioned over the centre of the binding site where they contact

the peptide, whereas the relatively conserved CDR1 and CDR2

loops of the TCR are located on the top region of MHC helices

580 CHEN ET AL.

Page 3: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

Figure 1. Interaction surface between T cell and antigen presenting cell (APC) (representing the known complex crystal structures).

(A) Models of TCR/pMHCI/CD8aa/CD3ed/CD3ec complex, (B) TCR/pMHCII/CD4/CD3ed/CD3ec complex, and (C) co-stimulatory

molecules of CTLA-4/CD80 and CTLA-4/CD86 complexes. The TCR/pMHCI/CD8aa is modeled by superimposing two structures,

HLA-A2/CD8aa complex (PDB 1AKJ) and the TCR A6/HLA-A2/TaxP6A complex (PDB 1QRN); while the TCR/pMHCII/CD4 is

modeled by a series of superimpositions using three structures, CD4/I-Ak/peptide complex (PDB 1JL4), TCR HA1.7/HLA-DR1/

peptide complex (PDB 1FYT) and four-domain CD4 (PDB 1WIO). The positions of CD3ed (PDB 1XIW) and CD3ec (PDB 1SY6)

relative to TCR on the T cell are based on the proposal by Arnett et al. (88). Costimulatory system is modeled by two complex

structures, CTLA-4/CD80 complex (PDB 1I8L) and CTLA-4/CD86 complex (PDB 1I85). In the models, peptides are colored in or-

ange, MHCI heavy chain and MHCII b chain in yellow, MHCI b2m and MHCII a chain in green, CD8aa in red and blue, CD4 in

blue, TCR a chain in purple and TCR b chain in cyan, CD3e in marine, CD3d in magenta, CD3c in olive. The oval drawing in or-

ange color represents the structure-unknown D2 domain of CD86. These images were prepared using the program PYMOL.

Figure 2. Different interaction modes for ab TCRs and cd TCRs binding to their ligands. (A) Overview structure of 2C ab TCR

bound to its ligand H2-Kb-dEV8. MHCI heavy chain is colored in limon, MHCI b2m in marine, peptide in red, TCR a chain in

cyan, TCR b chain in pink. (B) Footprints of ab TCR CDR loops on the pMHC interface. It shows a stereotyped polarity of the

Va and Vb CDR loops on pMHC, in which the Va domain lies mainly over the amino-terminal end of the peptide and the a2 helix

(MHCI) or b1 helix (MHCII), while the Vb domain lies mainly over the carboxy-terminal region of the peptide and the respective

a1 helices. The whole pMHC interface is colored in limon, while the peptide in red. CDR1s and CDR2s marked with semitranspar-

ent cyan oval drawings, which constitute the relatively conserved components of the TCR. CDR3s marked with a semitransparent

magenta oval drawing constitute the high diversity components of the TCR. (C) Overview structure of G8 cd TCR bound to its

ligand MHC Ib T22. MHC Ib heavy chain is colored in yellow, MHC Ib b2m in magenta, TCR c chain in pink, TCR d chain in

blue. G8 interacts with T22 at a titled angle which differs from the ab TCRs. (D) Footprints of cd TCR CDR loops on the MHC

Ib interface. The surface of whole MHC interface is colored in limon. cd TCR G8 predominantly uses its CDR3d loop (marked

with semitransparent red oval drawing) to interact with its ligand T22, and the remaining CDRd and CDR3c (marked with semi-

transparent magenta oval drawing) bind to T22 with degenerate contacts.

581STRUCTURAL IMMUNOLOGY AND CRYSTALLOGRAPHY HELP IMMUNOLOGISTS

Page 4: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

surrounding the central CDR3-peptide region (9, 10) (Figs. 2A

and 2B). This interaction mode suggests that the specificity of

TCR binding to pMHC is determined by two parts: contacts

between the germline-encoded TCR V-gene products and MHC

gene products, and also contacts between the diverse TCR

CDR3 regions and peptides in the MHC groove. Logically, we

can predict that invariant parts of TCR (CDR1 and CDR2) have

evolved to interact with MHC molecule in specific and con-

served way, leaving the CDR3-peptide region aside.

Nevertheless, it is not quite simple as that. Except the speci-

ficity for one MHC, the TCR also has cross-reactivity that it is

cross-reactive with many other MHC molecules (11). In princi-

ple, each abTCR can recognize any MHC allele, as there does

not seem to be class or allotype specificity by particular TCR

germline segments. This cross-reactivity is essential to enable

the TCR to dock and ‘‘scan’’ the peptide contents of many dif-

ferent MHC molecules. Although the usage preferences of the

TCR chains do exist in the T cell response, but it doesn’t mean

that each abTCR is specific to one MHC molecule.

Recent study focusing on the interaction of one particular

Vb segment (Vb8.2) with different I-A MHC alleles has dem-

onstrated the conservation of specific TCR-MHC interfacial

contacts (12, 13). A ‘‘codon hypothesis’’ which suggests that

each TCR variable-region gene product engages each type of

MHC through a ‘‘menu’’ of structurally coded recognition

motifs that have arisen through coevolution has been proposed

to clarify the existence of ‘‘TCR-MHC bias’’ or ‘‘germline-

encoded recognition’’ of MHC. Here, ‘‘bias’’ is simply a euphe-

mism for specificity, and cases of true cross-reactivity are more

correctly called ‘‘polyspecificity’’ (14). ‘‘Induce-fit’’ interaction

mode facilitates polyspecificity to occur which is usually seen

in the immune system of specific recognition by one protein of

a spectrum of diverse ligands.

Returning the issue of specificity determined by interaction

of abTCR with pMHC, it is reasonable that each TCR engages

on each pMHC molecule using distinct interaction codon which

is derived from the coordination of the germline-encoded recog-

nition bias and peptide-mediated editing of germline-encoded

recognition. And this distinct interaction codon determines the

specificity of TCR binding to pMHC.

HELP OF CO-RECEPTORS CD8/CD4 TO THEPEPTIDE-MHC/TCR BINDING

The interaction of the abTCR with pMHC is enhanced by

the recognition of the co-receptor CD8 or CD4 to the same

pMHC in the immune response when T cells ‘‘touch’’ the anti-

gen presenting cells (APC). Binding dynamics and complex

structures have shed light on this complex interactions for a sin-

gle ligand (pMHC) recognized simultaneously by two receptors,

both abTCR and co-receptor CD8 or CD4 (15–21).

CD8 (two forms of aa homodimer and ab heterodimer) acts

as a co-receptor in the function of cytotoxic T lymphocytes

(CTLs), whereas CD4 acts as a co-receptor in T helper (Th)

cells. Thus, CD8 interacts with class I pMHC (pMHCI), while

CD4 interacts with class II pMHC (pMHCII). In the current

model, both CD8 and CD4 bind to the same pMHC (class I or

class II, respectively) as the ab TCR and are thus called T cell

co-receptor [reviewed in (22)]. Crystal structures of both human

and murine CD8aa-pMHCI complexes (15, 17), muring

CD8ab-pMHC (23), and TL antigen-CD8aa complex (24) have

shown that the Ig-like domain is involved in the binding to

pMHC in a similar way to antibody-antigen binding as pre-

dicted in the earlier functional studies (Fig. 1A). The binding of

CD8aa and CD8ab to pMHC mainly involves the a3 domain

with some hydrogen bonds in the a2 and b2m domains away

from the peptide interface. CD4 molecule consisted of four Ig-

like domains and the complex structure involving human CD4

(the two N-terminal domains) and murine pMHC (I-Ak)

revealed that the N-terminal Ig-like V domain is the only one

of the four Ig-like domains directly binding to the two mem-

brane-proximal domains (a2 and b2 domains) (Fig. 1B) (18).

Although the binding modes of CD8aa-pMHCI and CD4-

pMHCII are completely different, but in both cases the binding

conformations ensure that the specificity for pMHC is deter-

mined by the antigen-specific TCR, whereas CD8 or CD4 act as

a universal co-receptor for any polymorphic pMHCs, confirming

their true ‘‘help’’ function as co-receptors.

Biophysically both CD8 and CD4 bind pMHC with a lower

affinity, faster kinetics and independently from the abTCRensuring that binding is dominated by the TCR rather than the

‘‘assistant’’ co-receptors. Furthermore, the BIAcore binding

experiments involving soluble reagents of pMHC, TCR and

CD8 or CD4 do not indicate any direct binding of the TCR

with CD8 or CD4, or enhancement by CD4 or CD8 to the bind-

ing of TCR-pMHC (19–21). However, one early study demon-

strated some enhancement of TCR-pMHC binding in the pres-

ence of soluble CD8 using BIAcore analysis (16). In the future,

a triple complex crystal structure involving the TCR-pMHCI-

CD8 or the TCR-pMHCII-CD4 complex will help us to under-

stand the nature of these crucial interactions that lie at the heart

of antigen recognition and T-cell activation. In both human and

mouse, the functions of CD8 involved in immune responses

have been extensively studied (25, 26).

Simian immunodeficiency virus (SIV) infection of rhesus

macaque (macaca mulatta) is widely used as an animal model

for human immunodeficiency virus (HIV) infection (27–29) as

well as other human diseases. However, for macaque monkeys,

little is known on the structures of the CTL-related molecules

(e.g., TCR, pMHC and CD8). Recently, we presented the crys-

tal structures of Mamu-A*01 complexed with two immunodom-

inant SIV epitopes: the nonamer CM9 of group specific antigen

(Gag, 181-189; CTPYDINQM) and the octamer TL8 of tran-

scription activator (Tat, 28-35; TTPESANL) for the rational

design of Mamu-A*01-restricted CTL epitopes with improved

binding, as a step towards development of AIDS vaccines (30).

Furthermore, recently we also present the crystal structure of

rhesus macaque CD8aa (rCD8aa) homodimer and found that

582 CHEN ET AL.

Page 5: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

with two Thr43 residues in C-C’ loop, rCD8aa shows a unique

extra hydrogen bond in the homodimeric interface indicating a

tighter homodimeric interaction (31).

SECONDARY SIGNALS OF T CELL ACTIVATION

Specific immune responses are strictly regulated by cell–cell

interactions. When pMHC-peptide complex on APC cells is rec-

ognized by, TCR, it is called the first signal, the fully activation

of the T cell needs the secondary signal which occurs after the

co-receptor interactions (32).

The B7 family is such one kind of co-stimulatory molecules

including CD28, CTLA4 (CD152) and their ligands B7-1

(CD80), B7-2 (CD86), ICOS and its ligands ICOSL, PD-1 and

its ligands PD-L1, PD-L2, and so on, which are playing a cru-

cial role in regulating the activation, differentiation, and toler-

ance of T lymphocytes. All of these costimulatory molecules

belong to the Ig (immunoglobulin-like) superfamily, as stimula-

tory or inhibitory functions. Stimulatory receptor CD28 is con-

stitutively expressed on naıve T cells, when engaged by B7-1

and B7-2 on APCs, positive signal will provide to initiate and

sustain T cell responses (33, 34). ICOS is not expressed consti-

tutively on naive T cells but can be induced rapidly on T cells

after TCR engagement (35), taking the T-cell proliferation and

differentiation as its major function (36). On the other hand,

CTLA4 is induced and expressed after T cell activation, which

can competitively binds B7-1 and B7-2 to down regulate or

even terminate the responses by providing negative signal (37).

Recently, PD-1 was also found to generates negative signal

expressed on T cell after binding PD-L1 and PD-L2 (38, 39).

Until now, the extracellular region structure of CTLA4-B7-1

complex (40) and CTLA4-B7-2 (only IgV domain) (41) have

been resolved. As CD28 may be hard for crystallization alone,

we only know the structure of CD28 extracellular region com-

plexed with the Fab fragment of a mitogenic antibody (42). PD-

1 and its ligands, another hot spot of B7 family, are still in pro-

gress. The structure of murine PD-1 (43), murine PD-1 and

human PD-L1 hybrid complex (44), murine PD-1 and murine

PD-L2 (45) complex have been reported. However, human PD-

1 and its complex still remains a mystery. Other important B7

family members as well as other T cell co-receptor molecules

are waiting for us to investigate.

INTERACTION OF cd T CELL RECEPTORSWITH THEIR LIGANDS

cd T cell receptors (TCRs) are one of the three lineages of

somatically recombined antigen receptors which also include

abTCRs and antibodies. Previous studies demonstrated that aband cd T cells have different functional roles in the immune

system (46), yet the identity of endogenous ligands for cd T

cells is still unclear. Compared with antibodies and abTCRs,cdTCRs have the highest potential CDR3 diversity (1018) gener-

ated by VDJ recombination, but limited diversity derived from

pairing of germline-encoded V regions, due to small numbers of

Vc and Vd segments (8). CDR3 length distribution in cdTCRsis more similar to antibodies than to abTCRs, as the CDR3dloops are long and variable, while the CDR3c loops are short

and constrained (47). However, cd T cell reactivity seems to

correlate with V gene usage, suggesting that CDR3 diversity

has little role on the recognition of specific ligands (48).

The molecular basis of the recognition of ligand by cdTCRsis not well understood. As the first co-crystal structure of

cdTCR bound to its ligand was reported (49), we can have a

glimpse of the molecular basis of the recognition of ligand by

cdTCRs. The structure shows that G8 cdTCR predominantly

use its CDR3d loop to bind the MHC class Ib molecule T22 at

a tilted angle with degenerate contacts for the remaining CDRdand CDR3c loops, differing from the straight-on approach

which abTCRs adopt to interact with pMHC (Figs. 2A, 2C, and

2D). Furthermore, the residues involved in the interface are

derived mainly from the germline-encoded Dd segments, and it

shows that G8 cdTCR has a germline-encoded basis for T22

recognition. Recent study has revealed that an autonomous

CDR3d is sufficient for the recognition of the ligands T10 and

T22 (50). The interaction mode of G8 cdTCR binding to T10/

T22 shows that the conserved amino acid motif in the CDR3dloop can have its ligand affinity ‘‘tuned’’ by different surround-

ing contexts of CDR3d sequence. All the data suggest that the

G8 cdTCR may function in the immune system with a hybrid

of innate and adaptive recognition strategies.

INTERACTION OF KIR WITH PEPTIDE-HLA

In contrast to TCR-MHC ‘‘induce-fit’’ mode, many of NK

cell receptors interact with MHC class I molecules as ‘‘lock and

key’’ and modulate cells activity. As shown in Fig. 3, KIR and

CD94/NKG2 dock on the a1 helix and a2 helix including pep-

tide (51–53) while Ly49 and LILR interacts with pMHC a3 do-

main and b2m (54–57).

The human KIR gene family contains closely linked 15

genes and two pseudogenes in the chromosome 19q13.4.

Human KIRs are type I transmembrane proteins containing ei-

ther two (KIR2D) or three (KIR3D) immunoglobulin-like

domains that recognize polymorphic HLA-C and HLA-B mole-

cules, respectively (4). The Ig-like domains have been desig-

nated D0, D1, and D2, or D1 and D2, from N to C-terminal in

KIR3D/KIR2D proteins. A short stalk region separates the Ig-

like domains from the transmembrane segment, and the cyto-

plasmic domains are variable in length. Some receptors possess

long (L) cytoplasmic domains with one or two ITIM sequences,

and other receptors have short (S) cytoplasmic domains without

signaling motifs.

To date, the crystal structures of three free-state KIRs

(KIR2DL1, 2DL2 and 2DL3) and two KIR/pMHC complexes

(KIR2DL2/HLA-Cw3, KIR2DL1/HLA-Cw4) have been

reported (2), while no crystal structures available for KIR3D

receptors. As shown in Fig. 4, for example, KIR2DL2 bind

583STRUCTURAL IMMUNOLOGY AND CRYSTALLOGRAPHY HELP IMMUNOLOGISTS

Page 6: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

HLA-Cw3 ligands through the a1 and a2 helices and the C-ter-

minal portion of the peptide (52). The KIR D1D2 axis is

approximately orthogonal to the axis of the peptide with D1

mainly binds to the HLA a1 helix and D2 to a2. The interface

is highly charged, and shows electron complementary that are

critical for binding (Fig. 3B). A comparison of the KIR2DL2/

HLA-Cw3 and KIR2DL1/HLA-Cw4 complexes revealed that

due to side chain rearrangements, many conserved residues in

KIR2D and HLA-C mediate different interactions in the two

structures, explaining the allelic specificity of KIR2DLs/HLA

complexes (58). For example, different alleles of HLA-C have

either Ser 77/Asn80 or Asn77/Lys80, and these are recognized

by different isoforms of KIR2DL.

Furthermore, peptide preferences have been documented in

KIR2D binding to HLA-C molecules (59), although the function

of peptide selectivity is not very clear in NK cell receptors. As

shown in Fig. 4B, the KIR/HLA interaction results in the sensi-

tivity to substitutions at peptide positions 7 and 8. Gln71 of

KIR2DL2 is hydrogen bonded to the main-chain nitrogen atom

of P8 Ala (60). This hydrogen bond may restrict the size of the

side chain accommodated at this position, consistent with the

observed preference for Ala or Ser at the P8 position. By com-

parison, TCRs, which exhibit much greater peptide selectivity

than KIRs, generally focus on the central portion of the MHC-

bound peptide, at and around the P5 position (Fig. 2) (61).

Although the specificity of the inhibitory KIR have been

extensively characterized, very little is known about the ligands

for the KIR2DS and KIR3DS molecules. The similarity between

several pairs of the activating and inhibitory KIR suggests they

arose by gene duplication. However, in all cases studied, the

activating KIR either does not bind HLA class I, or binds with

an affinity much weaker than that of the paired inhibitory KIR.

INTERACTION OF LILR (ILT/LIR/CD85)WITH PEPTIDE-HLA

Leukocyte immunoglobulin-like receptors (LILRs/LIRs), also

called Immunoglobulin-like transcripts (ILTs) (62, 63) or CD85

(64), including 13 members (with two of them as pseudogenes)

of either activating receptors (LILRA) or inhibitory receptors

(LILRB), regulating a broad range of cells in the immune

responses. Since LILR was first discovered by David Cosman

and Marco Colonna in 1997 (62, 63, 65), lots of studies have

been performed on its ligands and structures.

According to the amino acid similarity responsible for the

interaction between LILRB1 (ILT2/LIR-1/CD85j) and HLA-A2/

b2m, LILR can be divided into two groups (56): Group 1 mem-

bers include the inhibitory receptors LILRB1 and LILRB2

(ILT4/LIR-2/CD85d), the activating receptors LILRA1 (LIR6/

CD85i) and LILRA2 (ILT1/LIR-7/CD85h), and the soluble re-

ceptor LILRA3 (ILT6/LIR-4/CD85e). Group 2 members mainly

comprise of LILR A4/A5/A6 and B3/B4/B5 (ILT 7/11/8 and

ILT 5/3/LIR8), showing less than 60% sequence identity with

Group 1 members.

Most of the LILR family members contain four Ig-like

domains in their extracellular domains (designated D1, D2, D3,

and D4 from N to C terminus). Current available crystal struc-

tures of LILR B1/B2/A2/A5 only include their D1 and D2

domains (D1D2) (55, 56, 66–69). Though the instability and

precipitation of wild-type LILRA2 (ILT1/LIR-7/CD85h) D1D2

has been documented (70), a cysteine-introduced mutation

(R142C) was developed to form a disulfide bond with the spare

Cys132, which stabilizes the protein and enhances the recombi-

nant production of the LILRA2 D1D2 protein (71).

As inhibitory receptors with ITIM-motifs in their cytoplas-

mic region, LILRB1 and LILRB2 have been reported to bind a

broad range of classical or nonclassical pMHCIs and MHC-like

molecules (UL18) and differentiate or impair many lymphocyte

cells (72–74). The complex crystal structures of the LILRB1-

Figure 3. Overview structure of NK receptors binding to a

pMHCI molecule (representing the known complex crystal

structures). NK receptors were marked by different circles with

corresponding colors as following: NKG2A (PDB 3CDG) in

light blue; CD94 (PDB 3CDG) in pink; KIR (PDB 1EFX) in

yellow; Ly49 (PDB 1P4L) in limon; LILRB1/B2 (PDB 1P7Q/

2DYP) in brown. MHC molecule (green and blue) (HLA-Cw3

as an example, PDB 1EFX) with peptide (red, GAVDPLLAL)

was located at the center.

584 CHEN ET AL.

Page 7: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

HLA-A2 (56) (Fig. 5A), the LILRB2-HLA-G (55), and

LILRB1-UL18 (69) were also reported.

One of the most important questions we concerned about is

the binding specificity of LILR-pMHC. LILRB1/B2 D1D2

binds pMHCIs through a3 domain and b2m, respectively (Fig.

5A). This orientation differs largely from the docking mode of

KIR onto pMHC as we have shown earlier. The size of the

LILRB1-HLA-A2 interface (�1,700 A2) is larger than those of

KIR-pMHC interfaces (�1,500 A2) and similar to those of TCR-

pMHC complexes (1,700�1,900 A2) (9). About 70% of the

LILRB1-HLA-A2 binding surface is involved in contacts with

14 b2m residues. This relative dominance of b2m is unprece-

dented among HLA-binding receptors, and is partly responsible

for the broad recognition to both classical and nonclassical

pMHCIs of LILRB1.

Comparison of the binding pattern of LILRB1-HLA-A2 with

CD8aa-HLA-A2 demonstrated that steric hindrance exists

between CD8aa and LILRB1 consistent with functional data

(72), even though the footprints of these two molecules onto

HLA-A2 show little overlapping (Fig. 5B).

However, the effects, mechanisms, and structures of many

LILR activating receptors remain unknown. Some LILR activat-

ing receptors with short cytoplasmic tail were proved to recruit

the c-chain of FceRI with charged Arg residue in transmem-

brane region to activate the immune system via ITAM motifs of

FceRIc (75–77). Shiroishi et al. demonstrated the crystal struc-

ture of LILRA5 (ILT11/LIR-9/CD85f) in 2006 and found its

nonbinding with several classical and non-classical pMHCI

molecules (66). As a member of ‘‘Group 1’’ activating recep-

tors, LILRA2, shows more than 80% sequence identity to LILR

B1/B2, but also proved to be a nonbinder of pMHCIs (77). The

exact mechanism for this nonbinding is yet elusive.

Recently, we report LILRA2 extracellular D1D2 domain

crystal structure at 2.6 A and the structure reveals structural

shifts of the corresponding pMHC-binding amino acid residues

in comparison with LILR B1/B2, explaining its non-binding

property to pMHCI molecules (78). We also identify the transi-

tion of 310 in LILRB1/B2 to b-strand in LILRA2 with great

influences on the local structure which only exist in the pMHC-

binding receptors. Moreover, in the structure, LILRA2 forms a

domain swapped dimer. Further mutational work of these key

swapping residues yields a monomeric form, confirming that the

domain-swapping is amino acid sequence-specific. The structure

described by our group not only supports the dimer conforma-

tion in solution observed earlier but also implies a stress

induced regulation by dimerization, consistent with its particular

heat shock promoter.

INTERACTION OF CD94/NKG2 WITH HLA

CD94 and NKG2 encode type II transmembrane proteins of

the C-type lectin-like family. CD94 can be expressed on the

cell surface as a disulfide-linked homodimer or as a disulfide-

linked heterodimer with NKG2A or NKG2C (4). NKG2A has

an ITIM in its cytoplasmic domain and CD94/NKG2A hetero-

dimers function as inhibitory receptors while CD94/NKG2C

heterodimers serve as activating receptors.

In contrast to the promiscuity of most NK receptors, the

human CD94/NKG2 family solely recognizes a nonclassical

MHC molecule, HLA-E (79). The members of the NKG2 fam-

ily, including NKG2A, -2B, -2C, and -2E, dimerize with CD94

in vitro (4) with different binding affinity to HLA-E, such as

the CD94/NKG2A binds with HLA-E more tightly than CD94/

Figure 4. Detailed binding region between KIR2DL2 and HLA-Cw3. (A) Zoom-in of the interface of KIR2DL2 D1D2 and HLA-

Cw3 (PDB 1EFX). (B) Down-view of HLA-Cw3 a1/a2/peptide binding with the molecular surface of KIR2DL2 colored by electro-

static potential (positive: blue, negative: red).

585STRUCTURAL IMMUNOLOGY AND CRYSTALLOGRAPHY HELP IMMUNOLOGISTS

Page 8: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

NKG2C. To fully understand the mode of CD94/NKG2A inter-

action with HLA-E, the structural investigation is essential (79).

The crystal structure of CD94/NKG2A-HLA-E was shown in

Fig. 6. CD94 mainly binds to HLA-E a2 helix while NKG2A

binds to a1 helix with this orientation similar to the docking

mode of TCRs/KIRs onto pMHC, but is completely distinct

from those of LILR or Ly49 NK receptors. Analysis of the elec-

trostatic surfaces of HLA-E and CD94/NKG2A highlighted a

role for charge complementarity at the CD94/NKG2A-HLA-E

interface (Fig. 6B). The buried surface area (BSA) of the inter-

face was about 2,100 A2, which is marginally larger than TCR/

pMHC complex, with CD94 and NKG2A contributing �69 and

�31%, respectively. The peptide contributed 23% of the BSA

at the HLA-E interface, in which CD94 subunit played a much

more marked role (80%) in interacting with the peptide com-

pared with the NKG2A chain (20%).

As we discussed earlier, classical and nonclassical pMHCIs

molecules can be recognized by a diverse set of NK receptors.

However, NK cell recognition of MHC-like ligands, (i.e.,

MICA/MICB and ULBP1-4 in humans and H60, RAE-1 pro-

teins, and MULT1 in mice) is conserved across species and

mediated predominantly through the homodimeric NKG2D

receptors (80).

LY49 AND OTHER NK CELL RECEPTORS

In humans, classical pMHC molecules are recognized by the

Ig superfamily of NK receptors, such as KIR and LILR,

whereas in mice they are recognized by structurally divergent

NK receptors of the C-type lectin superfamily, termed Ly49

(81). Ly49 receptors are transmembrane glycoproteins expressed

on murine NK cells and on a subset of cytotoxic T-cells.

The best characterized Ly49/MHCIs interaction is that

between the inhibitory Ly49A receptor and its H-2Dd ligand

(57). Ly49A is a homodimer on the NK cell surface, with an

interchain disulfide in the stalk region. In the crystal structure

of the Ly49A/H-2Dd complex, the Ly49A homodimer contacts

H-2Dd through two distinct interfaces, however, the site-directed

mutants of both H-2Dd and Ly49A designed on the basis of the

crystal structure of the complex identified that the primary bind-

ing site for Ly49A on MHCIs formed by the a2 and a3 domains

of H-2Dd and b2-microglobulin (b2m) (Fig. 3) (82). It is one

side of the pMHCI binding platform, away from the peptide

antigen. This concave region partially overlaps the binding site

for CD8 (15) with the total buried surface of approximately

3400A2.

Nevertheless, there are other groups of NK cell receptors,

such as NKR-P1, LAIR-1, Siglec-7/9, NCR, and so forth. Natu-

ral cytotoxicity receptors (NCR) is a set of Ig superfamily

receptors mainly including NKp46, NKp30, and NKp44

involved in the triggering of natural cytotoxicity (83). Recent

studies have unveiled the structures of NCR and identified the

ligands of NKp46 and NKp44 as the hemagglutinin (HA) of

influenza virus and the hemagglutinin-neuraminidase (HN) of

parainfluenza virus (84, 85). These findings indicate how NCR-

expressing NK cells recognize target cells infected by influenza

or parainfluenza without the decreased expression of target-cell

MHCI protein.

CONCLUSIVE REMARKS AND PERSPECTIVES

Immunologists always want to know what and how were the

related cells involved in immune responses. Although more and

more functional data presented for the cell–cell interaction, the

Figure 5. Detailed binding region between LILRB1 and HLA-A2, in comparison with CD8 foot print. (A) Zoom-in the interface

of LILRB1 D1D2 and HLA-A2 (PDB 1P7Q). (B) Footprints of LILRB1 and CD8aa binding to the molecular surface of HLA-A2

(Side-view) (CD8/HLA-A2 PDB 1AKJ).

586 CHEN ET AL.

Page 9: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

molecular basis under these processes is in the spotlight for us

to further understand the immune system in action. In this arti-

cle, we reviewed the crystal structures of some MHC-centered

molecules and protein complexes as well as recent progress on

the T and NK cell recognition, especially the ligand-receptor

binding (or touch) modes. We also try to fit the functional data

with the complex structures, explaining the touch of the

immune cell (T and NK cells) of the APCs.

So far, from the comparatively limited number of TCR/

pMHC structures, we can conclude that the TCR binding to

pMHC with an interaction codon which is determined by the

germline-encoded recognition bias and the peptide-mediated

editing of germline-encoded bias. However, there are still some

cases of TCR-pMHC complexes that have minimal contacts

with germline-encoded residues yet still maintain the stereotypi-

cal polarity (86, 87). More structures of TCR-pMHC complexes

are in need to refine the codon concept. Despite the TCR-

pMHC interaction, the structure of TCR-pMHC-CD4 (CD8)-

CD3 four-molecule complex which represent the intact immu-

nological synapse should be determined in the future, if we want

to know how the signaling process work at molecular level.

Many NK cell receptors bind to MHC molecules, some of

which follow the similar docking mode as TCR-pMHC. Emerg-

ing evidence suggests that NK cells, previously considered an

‘‘ancient’’ immune effector cell type, have more likely

coevolved with T cells, given that both of these lymphocytes

are focused on recognition of classical and non-classical MHC

molecules. Nevertheless, while the term ‘‘NK receptor’’ has

been used to describe molecules that were first discovered on

NK cells, the majority of these NK receptors are expressed on

at least a subset of T lymphocytes, in particular on cd TCR1 T

cells and on activated CD81 T cells, and more importantly on

the surface of other cell types, for example, dendritic cells or

monocytes or other myeloid cells.

As we know, NK cell function depends on the delicate bal-

ance of triggering and inhibitory signals. Previous extensive

functional and structural studies on inhibitory receptors on NK

cells provide the fundamental analysis for the ‘‘suspension’’ of

immune responses. However, more and more evidence shows

up indicating that NK cells can be activated to lyse target even

though the inhibitory signal exists or the amount of MHC mole-

cules on the target cells remains normal. On the other hand, for

example, although human red blood cells do not express

MHCIs, NK cells do not attack them; therefore, these cells may

lack ligands capable of engaging the activating NK cell recep-

tors. Until now, biological rationale for many NK activating

receptors has remained an enigma, including both function and

structure. That should be one of our major tasks in the future.

To ‘‘see’’ the immune system in action needs both functional

and structural approaches to answer the ‘‘what and how’’

questions.

REFERENCES1. Zinkernagel, R. M. and Doherty, P. C. (1974) Restriction of in vitro T

cell-mediated cytotoxicity in lymphocytic choriomeningitis within a

syngeneic or semiallogeneic system. Nature 248, 701–702.

2. Sawicki, M. W., Dimasi, N., Natarajan, K., Wang, J., Margulies, D. H.,

and Mariuzza, R. A. (2001) Structural basis of MHC class I recognition

by natural killer cell receptors. Immunol. Rev. 181, 52–65.

3. Rudolph, M. G., Stanfield, R. L., and Wilson, I. A. (2006) How TCRs

bind MHCs, peptides, and coreceptors. Annu. Rev. Immunol. 24, 419–

466.

4. Lanier, L. L. (2005) NK cell recognition. Annu. Rev. Immunol. 23, 225–

274.

5. Arden, B., Clark, S. P., Kabelitz, D., and Mak, T. W. (1995) Human T-

cell receptor variable gene segment families. Immunogenetics 42, 455–

500.

6. Rowen, L., Koop, B. F., and Hood, L. (1996) The complete 685-kilo-

base DNA sequence of the human beta T cell receptor locus. Science272, 1755–1762.

Figure 6. Detailed binding region between CD94/NKG2A and HLA-E. (A) Zoom-in of the interface of CD94/NKG2A and HLA-E

(PDB 3CDG). (B) Down-view of HLA-E a1/a2/peptide binding with the molecular surface of CD94/NKG2A colored by electro-

static potential (positive: blue, negative: red).

587STRUCTURAL IMMUNOLOGY AND CRYSTALLOGRAPHY HELP IMMUNOLOGISTS

Page 10: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

7. Jerne, N. K. (1971) The somatic generation of immune recognition.

Eur. J. Immunol. 1, 1–9.

8. Davis, M. M. and Bjorkman, P. J. (1988) T-cell antigen receptor genes

and T-cell recognition. Nature 334, 395–402.

9. Garboczi, D. N., Ghosh, P., Utz, U., Fan, Q. R., Biddison, W. E., and

Wiley, D. C. (1996) Structure of the complex between human T-cell

receptor, viral peptide and HLA-A2. Nature 384, 134–141.

10. Garcia, K. C., Degano, M., Stanfield, R. L., Brunmark, A., Jackson, M.

R., Peterson, P. A., Teyton, L., and Wilson, I. A. (1996) An alphabeta

T cell receptor structure at 2.5 A and its orientation in the TCR-MHC

complex. Science 274, 209–219.

11. Zerrahn, J., Held, W., and Raulet, D. H. (1997) The MHC reactivity of

the T cell repertoire prior to positive and negative selection. Cell 88,

627–636.

12. Feng, D., Bond, C. J., Ely, L. K., Maynard, J., and Garcia, K. C. (2007)

Structural evidence for a germline-encoded T cell receptor-major histo-

compatibility complex interaction ’codon’. Nat. Immunol. 8, 975–983.

13. Dai, S., Huseby, E. S., Rubtsova, K., Scott-Browne, J., Crawford, F.,

Macdonald, W. A., Marrack, P., and Kappler, J. W. (2008) Crossreac-

tive T Cells spotlight the germline rules for alphabeta T cell-receptor

interactions with MHC molecules. Immunity 28, 324–334.

14. Wucherpfennig, K. W., Allen, P. M., Celada, F., Cohen, I. R., De Boer,

R., Garcia, K. C., Goldstein, B., Greenspan, R., Hafler, D., Hodgkin, P.,

Huseby, E. S., Krakauer, D. C., Nemazee, D., Perelson, A. S., Pinilla,

C., Strong, R. K., and Sercarz, E. E. (2007) Polyspecificity of T cell

and B cell receptor recognition. Semin. Immunol. 19, 216–224.

15. Gao, G. F., Tormo, J., Gerth, U. C., Wyer, J. R., McMichael, A. J.,

Stuart, D. I., Bell, J. I., Jones, E. Y., and Jakobsen, B. K. (1997) Crystal

structure of the complex between human CD8alpha(alpha) and HLA-

A2. Nature 387, 630–634.

16. Garcia, K. C., Scott, C. A., Brunmark, A., Carbone, F. R., Peterson, P. A.,

Wilson, I. A., and Teyton, L. (1996) CD8 enhances formation of stable T-cell

receptor/MHC class I molecule complexes. Nature 384, 577–581.

17. Kern, P. S., Teng, M. K., Smolyar, A., Liu, J. H., Liu, J., Hussey, R. E.,

Spoerl, R., Chang, H. C., Reinherz, E. L., and Wang, J. H. (1998) Struc-

tural basis of CD8 coreceptor function revealed by crystallographic anal-

ysis of a murine CD8alphaalpha ectodomain fragment in complex with

H-2Kb. Immunity 9, 519–530.

18. Wang, J. H., Meijers, R., Xiong, Y., Liu, J. H., Sakihama, T., Zhang, R.,

Joachimiak, A., and Reinherz, E. L. (2001) Crystal structure of the

human CD4 N-terminal two-domain fragment complexed to a class II

MHC molecule. Proc. Natl. Acad. Sci. USA 98, 10799–10804.

19. Wyer, J. R., Willcox, B. E., Gao, G. F., Gerth, U. C., Davis, S. J., Bell,

J. I., van der Merwe, P. A., and Jakobsen, B. K. (1999) T cell receptor

and coreceptor CD8 alphaalpha bind peptide-MHC independently and

with distinct kinetics. Immunity 10, 219–225.

20. Xiong, Y., Kern, P., Chang, H., and Reinherz, E. (2001) T Cell Recep-

tor Binding to a pMHCII Ligand Is Kinetically Distinct from and Inde-

pendent of CD4. J. Biol. Chem. 276, 5659–5667.

21. Arcaro, A., Gregoire, C., Bakker, T. R., Baldi, L., Jordan, M., Goffin,

L., Boucheron, N., Wurm, F., van der Merwe, P. A., Malissen, B., and

Luescher, I. F. (2001) CD8beta endows CD8 with efficient coreceptor

function by coupling T cell receptor/CD3 to raft-associated CD8/

p56(lck) complexes. J. Exp. Med. 194, 1485–1495.

22. Janeway, C. A., Jr. (1992) The T cell receptor as a multicomponent sig-

nalling machine: CD4/CD8 coreceptors and CD45 in T cell activation.

Annu. Rev. Immunol. 10, 645–674.

23. Chang, H. C., Tan, K., Ouyang, J., Parisini, E., Liu, J. H., Le, Y.,

Wang, X., Reinherz, E. L., and Wang, J. H. (2005) Structural and muta-

tional analyses of a CD8alphabeta heterodimer and comparison with the

CD8alphaalpha homodimer. Immunity 23, 661–671.

24. Liu, Y., Xiong, Y., Naidenko, O. V., Liu, J. H., Zhang, R., Joachimiak,

A., Kronenberg, M., Cheroutre, H., Reinherz, E. L., and Wang, J. H.

(2003) The crystal structure of a TL/CD8alphaalpha complex at 2.1 A

resolution: implications for modulation of T cell activation and memory.

Immunity 18, 205–215.

25. Gao, G. F. and Jakobsen, B. K. (2000) Molecular interactions of core-

ceptor CD8 and MHC class I: the molecular basis for functional coordi-

nation with the T-cell receptor. Immunol. Today. 21, 630–636.

26. Gao, G. F., Rao, Z., and Bell, J. I. (2002) Molecular coordination of

alphabeta T-cell receptors and coreceptors CD8 and CD4 in their recog-

nition of peptide-MHC ligands. Trends. Immunol. 23, 408–413.

27. Allen, T. M., Sidney, J., del Guercio, M. F., Glickman, R. L., Len-

smeyer, G. L., Wiebe, D. A., DeMars, R., Pauza, C. D., Johnson, R. P.,

Sette, A., and Watkins, D. I. (1998) Characterization of the peptide

binding motif of a rhesus MHC class I molecule (Mamu-A*01) that

binds an immunodominant CTL epitope from simian immunodeficiency

virus. J. Immunol. 160, 6062–6071.

28. Matano, T., Shibata, R., Siemon, C., Connors, M., Lane, H. C., and Martin,

M. A. (1998) Administration of an anti-CD8 monoclonal antibody inter-

feres with the clearance of chimeric simian/human immunodeficiency virus

during primary infections of rhesus macaques. J. Virol. 72, 164–169.

29. Schmitz, J. E., Kuroda, M. J., Santra, S., Sasseville, V. G., Simon, M. A.,

Lifton, M. A., Racz, P., Tenner-Racz, K., Dalesandro, M., Scallon, B. J.,

Ghrayeb, J., Forman, M. A., Montefiori, D. C., Rieber, E. P., Letvin,

N. L., and Reimann, K. A. (1999) Control of viremia in simian immuno-

deficiency virus infection by CD81 lymphocytes. Science, 283, 857–860.

30. Chu, F., Lou, Z., Chen, Y. W., Liu, Y., Gao, B., Zong, L., Khan, A. H.,

Bell, J. I., Rao, Z., and Gao, G. F. (2007) First glimpse of the peptide

presentation by rhesus macaque MHC class I: crystal structures of

Mamu-A*01 complexed with two immunogenic SIV epitopes and

insights into CTL escape. J. Immunol. 178, 944–952.

31. Zong, L., Chen, Y., Peng, H., Gao, F., Iwamoto, A., and Gao, G. F.

(2009) Rhesus macaque: a tight homodimeric CD8alphaalpha. Proteins75, 241–244.

32. Bretscher, P. and Cohn, M. (1970) A theory of self-nonself discrimina-

tion. Science 169, 1042–1049.

33. Gross, J. A., Callas, E., and Allison, J. P. (1992) Identification and dis-

tribution of the costimulatory receptor CD28 in the mouse. J. Immunol.

149, 380–388.

34. Hathcock, K. S., Laszlo, G., Pucillo, C., Linsley, P., and Hodes, R. J.

(1994) Comparative analysis of B7–1 and B7–2 costimulatory ligands:

expression and function. J. Exp. Med. 180, 631–640.

35. Coyle, A. J., Lehar, S., Lloyd, C., Tian, J., Delaney, T., Manning, S.,

Nguyen, T., Burwell, T., Schneider, H., Gonzalo, J. A., Gosselin, M.,

Owen, L. R., Rudd, C. E., and Gutierrez-Ramos, J. C. (2000) The

CD28-related molecule ICOS is required for effective T cell-

dependent immune responses. Immunity 13, 95–105.

36. Dong, C., Juedes, A. E., Temann, U. A., Shresta, S., Allison, J. P., Rud-

dle, N. H., and Flavell, R. A. (2001) ICOS co-stimulatory receptor is

essential for T-cell activation and function. Nature 409, 97–101.

37. Brunet, J. F., Denizot, F., Luciani, M. F., Roux-Dosseto, M., Suzan, M.,

Mattei, M. G., and Golstein, P. (1987) A new member of the immuno-

globulin superfamily--CTLA-4. Nature 328, 267–270.

38. Freeman, G. J., Long, A. J., Iwai, Y., Bourque, K., Chernova, T., Nishi-

mura, H., Fitz, L. J., Malenkovich, N., Okazaki, T., Byrne, M. C., Horton,

H. F., Fouser, L., Carter, L., Ling, V., Bowman, M. R., Carreno, B. M.,

Collins, M., Wood, C. R., and Honjo, T. (2000) Engagement of the PD-1

immunoinhibitory receptor by a novel B7 family member leads to nega-

tive regulation of lymphocyte activation. J. Exp. Med. 192, 1027–1034.

39. Latchman, Y., Wood, C. R., Chernova, T., Chaudhary, D., Borde, M.,

Chernova, I., Iwai, Y., Long, A. J., Brown, J. A., Nunes, R., Greenfield,

E. A., Bourque, K., Boussiotis, V. A., Carter, L. L., Carreno, B. M.,

Malenkovich, N., Nishimura, H., Okazaki, T., Honjo, T., Sharpe, A. H.

and Freeman, G. J. (2001) PD-L2 is a second ligand for PD-1 and

inhibits T cell activation. Nat. Immunol. 2, 261–268.

40. Stamper, C. C., Zhang, Y., Tobin, J. F., Erbe, D. V., Ikemizu, S., Davis,

S. J., Stahl, M. L., Seehra, J., Somers, W. S., and Mosyak, L. (2001)

588 CHEN ET AL.

Page 11: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

Crystal structure of the B7–1/CTLA-4 complex that inhibits human

immune responses. Nature 410, 608–611.

41. Schwartz, J. C., Zhang, X., Fedorov, A. A., Nathenson, S. G., and

Almo, S. C. (2001) Structural basis for co-stimulation by the human

CTLA-4/B7–2 complex. Nature 410, 604–608.

42. Evans, E. J., Esnouf, R. M., Manso-Sancho, R., Gilbert, R. J., James, J. R.,

Yu, C., Fennelly, J. A., Vowles, C., Hanke, T., Walse, B., Hunig, T.,

Sorensen, P., Stuart, D. I., and Davis, S. J. (2005) Crystal structure of a

soluble CD28-Fab complex. Nat. Immunol. 6, 271–279.

43. Zhang, X., Schwartz, J. C., Guo, X., Bhatia, S., Cao, E., Lorenz, M.,

Cammer, M., Chen, L., Zhang, Z. Y., Edidin, M. A., Nathenson, S. G.

and Almo, S. C. (2004) Structural and functional analysis of the costi-

mulatory receptor programmed death-1. Immunity 20, 337–347.

44. Lin, D. Y., Tanaka, Y., Iwasaki, M., Gittis, A. G., Su, H. P., Mikami,

B., Okazaki, T., Honjo, T., Minato, N., and Garboczi, D. N. (2008) The

PD-1/PD-L1 complex resembles the antigen-binding Fv domains of anti-

bodies and T cell receptors. Proc. Natl. Acad. Sci. USA 105, 3011–

3016.

45. Lazar-Molnar, E., Yan, Q., Cao, E., Ramagopal, U., Nathenson, S. G.,

and Almo, S. C. (2008) Crystal structure of the complex between pro-

grammed death-1 (PD-1) and its ligand PD-L2. Proc. Natl. Acad. Sci.

USA 105, 10483–10488.

46. Hayday, A. C. (2000) cd cells: a right time and a right place for a con-

served third way of protection. Annu. Rev. Immunol. 18, 975–1026.

47. Rock, E. P., Sibbald, P. R., Davis, M. M., and Chien, Y. H. (1994)

CDR3 length in antigen-specific immune receptors. J. Exp. Med. 179,

323–328.

48. Morita, C. T., Mariuzza, R. A., and Brenner, M. B. (2000) Antigen rec-

ognition by human gamma delta T cells: pattern recognition by

the adaptive immune system. Springer Semin. Immunopathol. 22, 191–

217.

49. Adams, E. J., Chien, Y. H., and Garcia, K. C. (2005) Structure of a

gammadelta T cell receptor in complex with the nonclassical MHC

T22. Science 308, 227–231.

50. Adams, E. J., Strop, P., Shin, S., Chien, Y. H., and Garcia, K. C.

(2008) An autonomous CDR3delta is sufficient for recognition of the

nonclassical MHC class I molecules T10 and T22 by gammadelta T

cells. Nat. Immunol. 9, 777–784.

51. 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, 452–460.

52. Snyder, G. A., Brooks, A. G., and Sun, P. D. (1999) Crystal structure of

the HLA-Cw3 allotype-specific killer cell inhibitory receptor KIR2DL2.

Proc. Natl. Acad. Sci. USA 96, 3864–3869.

53. Petrie, E. J., Clements, C. S., Lin, J., Sullivan, L. C., Johnson, D., Huy-

ton, T., Heroux, A., Hoare, H. L., Beddoe, T., Reid, H. H., Wilce,

M. C., Brooks, A. G., and Rossjohn, J. (2008) CD94-NKG2A recogni-

tion of human leukocyte antigen (HLA)-E bound to an HLA class I

leader sequence. J. Exp. Med. 205, 725–735.

54. Deng, L., Cho, S., Malchiodi, E. L., Kerzic, M. C., Dam, J., and Mar-

iuzza, R. A. (2008) Molecular architecture of the major histocompatibil-

ity complex class I-binding site of Ly49 natural killer cell receptors. J.Biol. Chem. 283, 16840–16849.

55. Shiroishi, M., Kuroki, K., Rasubala, L., Tsumoto, K., Kumagai, I., Kuri-

moto, E., Kato, K., Kohda, D., and Maenaka, K. (2006) Structural basis

for recognition of the nonclassical MHC molecule HLA-G by the leuko-

cyte Ig-like receptor B2 (LILRB2/LIR2/ILT4/CD85d). Proc. Natl. Acad.

Sci. USA 103, 16412–16417.

56. Willcox, B. E., Thomas, L. M., and Bjorkman, P. J. (2003) Crystal

structure of HLA-A2 bound to LIR-1, a host and viral major histocom-

patibility complex receptor. Nat. Immunol. 4, 913–919.

57. Tormo, J., Natarajan, K., Margulies, D. H., and Mariuzza, R. A. (1999)

Crystal structure of a lectin-like natural killer cell receptor bound to its

MHC class I ligand. Nature 402, 623–631.

58. 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, 159–166.

59. Rajagopalan, S. and Long, E. O. (1997) The direct binding of a p58 killer

cell inhibitory receptor to human histocompatibility leukocyte antigen

(HLA)-Cw4 exhibits peptide selectivity. J. Exp. Med. 185, 1523–1528.

60. Boyington, J. C., Motyka, S. A., Schuck, P., Brooks, A. G., and Sun,

P. D. (2000) Crystal structure of an NK cell immunoglobulin-like recep-

tor in complex with its class I MHC ligand. Nature 405, 537–543.

61. Rudolph, M. G., Luz, J. G., and Wilson, I. A. (2002) Structural and

thermodynamic correlates of T cell signaling. Annu. Rev. Biophys. Bio-

mol. Struct. 31, 121–149.

62. Colonna, M., Navarro, F., Bellon, T., Llano, M., Garcia, P., Samaridis,

J., Angman, L., Cella, M., and Lopez-Botet, M. (1997) A common

inhibitory receptor for major histocompatibility complex class I

molecules on human lymphoid and myelomonocytic cells. J. Exp. Med.

186, 1809–1818.

63. Cosman, D., Fanger, N., Borges, L., Kubin, M., Chin, W., Peterson, L.,

and Hsu, M. L. (1997) A novel immunoglobulin superfamily receptor

for cellular and viral MHC class I molecules. Immunity 7, 273–282.

64. Saverino, D., Fabbi, M., Ghiotto, F., Merlo, A., Bruno, S., Zarcone, D.,

Tenca, C., Tiso, M., Santoro, G., Anastasi, G., Cosman, D., Grossi,

C. E., and Ciccone, E. (2000) The CD85/LIR-1/ILT2 inhibitory recep-

tor is expressed by all human T lymphocytes and down-regulates their

functions. J. Immunol. 165, 3742–3755.

65. Cella, M., Dohring, C., Samaridis, J., Dessing, M., Brockhaus, M., Lan-

zavecchia, A., and Colonna, M. (1997) A novel inhibitory receptor

(ILT3) expressed on monocytes, macrophages, and dendritic cells

involved in antigen processing. J. Exp. Med. 185, 1743–1751.

66. Shiroishi, M., Kajikawa, M., Kuroki, K., Ose, T., Kohda, D., and Mae-

naka, K. (2006) Crystal structure of the human monocyte-activating

receptor, ‘‘Group 2’’ leukocyte Ig-like receptor A5 (LILRA5/LIR9/

ILT11). J. Biol. Chem. 281, 19536–19544.

67. Willcox, B. E., Thomas, L. M., Chapman, T. L., Heikema, A. P., West,

A. P., Jr., and Bjorkman, P. J. (2002) Crystal structure of LIR-2 (ILT4)

at 1.8 A: differences from LIR-1 (ILT2) in regions implicated in the

binding of the Human Cytomegalovirus class I MHC homolog UL18.

BMC. Struct. Biol., 2, 6–15.

68. Chapman, T. L., Heikema, A. P., West, A. P., Jr., and Bjorkman, P. J.

(2000) Crystal structure and ligand binding properties of the D1D2

region of the inhibitory receptor LIR-1 (ILT2). Immunity 13, 727–736.

69. Yang, Z. and Bjorkman, P. J. (2008) Structure of UL18, a peptide-bind-

ing viral MHC mimic, bound to a host inhibitory receptor. Proc. Natl.

Acad. Sci. USA 105, 10095–10100.

70. Garner, L. I., Salim, M., Mohammed, F., and Willcox, B. E. (2006)

Expression, purification, and refolding of the myeloid inhibitory recep-

tor leukocyte immunoglobulin-like receptor-5 for structural and ligand

identification studies. Protein Expr. Purif. 47, 490–497.

71. Chen, Y., Chu, F., Gao, F., Zhou, B., and Gao, G. F. (2007) Stability

engineering, biophysical, and biological characterization of the myeloid

activating receptor immunoglobulin-like transcript 1 (ILT1/LIR-7/

LILRA2). Protein Expr. Purif. 56, 253–260.

72. Shiroishi, M., Tsumoto, K., Amano, K., Shirakihara, Y., Colonna, M.,

Braud, V. M., Allan, D. S., Makadzange, A., Rowland-Jones, S., Will-

cox, B., Jones, E. Y., van der Merwe, P. A., Kumagai, I., and Maenaka,

K. (2003) Human inhibitory receptors Ig-like transcript 2 (ILT2) and

ILT4 compete with CD8 for MHC class I binding and bind preferen-

tially to HLA-G. Proc. Natl. Acad. Sci. USA, 100, 8856–8861.

73. Young, N. T., Waller, E. C., Patel, R., Roghanian, A., Austyn, J. M.,

and Trowsdale, J. (2008) The inhibitory receptor LILRB1 modulates the

differentiation and regulatory potential of human dendritic cells. Blood

111, 3090–3096.

74. Lichterfeld, M., Kavanagh, D. G., Williams, K. L., Moza, B., Mui, S. K.,

Miura, T., Sivamurthy, R., Allgaier, R., Pereyra, F., Trocha, A., Feeney,

589STRUCTURAL IMMUNOLOGY AND CRYSTALLOGRAPHY HELP IMMUNOLOGISTS

Page 12: Structural immunology and crystallography help immunologists see the immune system in action: How T and NK cells touch their ligands

M., Gandhi, R. T., Rosenberg, E. S., Altfeld, M., Allen, T. M., Allen, R.,

Walker, B. D., Sundberg, E. J., and Yu, X. G. (2007) A viral CTL

escape mutation leading to immunoglobulin-like transcript 4-mediated

functional inhibition of myelomonocytic cells. J. Exp. Med. 204, 2813–

2824.

75. Tedla, N., Bandeira-Melo, C., Tassinari, P., Sloane, D. E., Samplaski,

M., Cosman, D., Borges, L., Weller, P. F., and Arm, J. P. (2003) Acti-

vation of human eosinophils through leukocyte immunoglobulin-like re-

ceptor 7. Proc. Natl. Acad. Sci. USA 100, 1174–1179.

76. Sloane, D. E., Tedla, N., Awoniyi, M., Macglashan, D. W., Jr., Borges,

L., Austen, K. F., and Arm, J. P. (2004) Leukocyte immunoglobulin-

like receptors: novel innate receptors for human basophil activation and

inhibition. Blood 104, 2832–2839.

77. Nakajima, H., Samaridis, J., Angman, L., and Colonna, M. (1999)

Human myeloid cells express an activating ILT receptor (ILT1) that

associates with Fc receptor gamma-chain. J. Immunol. 162, 5–8.

78. Chen, Y., Gao, F., Chu, F., Peng, H., Zong, L., Liu, Y., Tien, P., and Gao,

G. F. (2009) Crystal structure of myeloid cell activating receptor leukocyte

Ig-like receptor A2 (LILRA2/ILT1/LIR-7) domain swapped dimer: molecu-

lar basis for its non-binding to MHC complexes. J. Mol. Biol. 386, 841–853.

79. Braud, V. M., Allan, D. S., O’Callaghan, C. A., Soderstrom, K.,

D’Andrea, A., Ogg, G. S., Lazetic, S., Young, N. T., Bell, J. I., Phillips,

J. H., Lanier, L. L., and McMichael, A. J. (1998) HLA-E binds to natu-

ral killer cell receptors CD94/NKG2A, B and C. Nature 391, 795–799.

80. Gasser, S. and Raulet, D. H. (2006) Activation and self-tolerance of

natural killer cells. Immunol. Rev. 214, 130–142.

81. Nakamura, M. C. and Seaman, W. E. (2001) Ligand interactions by

activating and inhibitory Ly-49 receptors. Immunol. Rev. 181, 138–148.

82. Matsumoto, N., Mitsuki, M., Tajima, K., Yokoyama, W. M., and Yama-

moto, K. (2001) The functional binding site for the C-type lectin-like

natural killer cell receptor Ly49A spans three domains of its major his-

tocompatibility complex class I ligand. J. Exp. Med. 193, 147–158.

83. Moretta, A., Bottino, C., Vitale, M., Pende, D., Cantoni, C., Mingari,

M. C., Biassoni, R., and Moretta, L. (2001) Activating receptors and

coreceptors involved in human natural killer cell-mediated cytolysis.

Annu. Rev. Immunol. 19, 197–223.

84. Ho, J. W., Hershkovitz, O., Peiris, M., Zilka, A., Bar-Ilan, A., Nal, B.,

Chu, K., Kudelko, M., Kam, Y. W., Achdout, H., Mandelboim, M., Alt-

meyer, R., Mandelboim, O., Bruzzone, R., and Porgador, A. (2008) H5-

type influenza virus hemagglutinin is functionally recognized by the nat-

ural killer-activating receptor NKp44. J. Virol. 82, 2028–2032.

85. Mandelboim, O., Lieberman, N., Lev, M., Paul, L., Arnon, T. I., Bush-

kin, Y., Davis, D. M., Strominger, J. L., Yewdell, J. W., and Porgador,

A. (2001) Recognition of haemagglutinins on virus-infected cells by

NKp46 activates lysis by human NK cells. Nature 409, 1055–1060.

86. Hahn, M., Nicholson, M. J., Pyrdol, J., and Wucherpfennig, K. W.

(2005) Unconventional topology of self peptide-major histocompatibility

complex binding by a human autoimmune T cell receptor. Nat. Immu-nol. 6, 490–496.

87. Tynan, F. E., Burrows, S. R., Buckle, A. M., Clements, C. S., Borg, N. A.,

Miles, J. J., Beddoe, T., Whisstock, J. C., Wilce, M. C., Silins, S. L., Bur-

rows, J. M., Kjer-Nielsen, L., Kostenko, L., Purcell, A. W., McCluskey, J.,

and Rossjohn, J. (2005) T cell receptor recognition of a ’super-bulged’

major histocompatibility complex class I-bound peptide. Nat. Immunol. 6,

1114–1122.

88. Arnett, K. L., Harrison, S. C., and Wiley, D. C. (2004) Crystal structure

of a human CD3-epsilon/delta dimer in complex with a UCHT1 single-

chain antibody fragment. Proc. Natl. Acad. Sci. USA 101, 16268–16273.

590 CHEN ET AL.