You are on page 1of 15

RES EARCH

◥ transferred retrogenic T cells expressing re-


RESEARCH ARTICLE SUMMARY versed or canonical docking TCRs of varying
affinities. The inability of T cells expressing
IMMUNOLOGY reversed-docking TCRs to be recruited into the
antiviral immune response demonstrates that
Canonical T cell receptor docking on peptide–MHC TCR–pMHCI docking topology supersedes
TCR–pMHCI affinity as the primary determi-
is essential for T cell signaling nant for effective in vivo immune recruitment.
Using fluorescence lifetime imaging micros-

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
Pirooz Zareie, Christopher Szeto†, Carine Farenc, Sachith D. Gunasinghe, Elizabeth M. Kolawole, copy (FLIM)–Förster resonance energy trans-
Angela Nguyen, Chantelle Blyth, Xavier Y. X. Sng, Jasmine Li, Claerwen M. Jones, Alex J. Fulcher, fer (FRET) analyses, we show that canonical
Jesica R. Jacobs, Qianru Wei, Lukasz Wojciech, Jan Petersen, Nicholas R.J. Gascoigne, TCR–pMHCI docking is essential for the
Brian D. Evavold, Katharina Gaus, Stephanie Gras†‡*, Jamie Rossjohn‡*, Nicole L. La Gruta‡* colocalization of CD8–Lck with CD3z, which
is impaired when the TCR engages pMHCI
with reversed polarity. The requirement for
INTRODUCTION: T cell receptor (TCR) recogni- hypothesized that the underlying driver of canonical TCR–pMHCI docking can be cir-
tion of peptide–major histocompatibility com- the canonical docking polarity is the colocal- cumvented by the removal of the CD8 core-
plexes (pMHCs) is one of the most diverse ization of signaling molecules central to the ceptor or by dissociation of Lck from CD8,
receptor–ligand interactions in biology. Never- TCR signal transduction pathway. suggesting that sequestration of Lck by the
theless, these interactions exhibit a CD8 coreceptor has a dual role: po-
highly conserved, or canonical, TCR– tentiating signaling arising from ca-
TCR-pMHC docking
pMHC docking polarity in both mice nonical TCR–pMHCI interactions and
Canonical Reversed
and humans. Whether this canon- impeding reversed-polarity TCR–
ical docking polarity is driven by APC APC pMHCI signaling.
evolutionarily conserved, germline-
encoded complementarity between CONCLUSION: The inability of reversed-
the TCR and MHC or by signaling polarity TCRs to participate in the im-
constraints imposed by coreceptors pMHC pMHC mune response occurs independently
has been a question of enduring de- CD8 CD8 of TCR–pMHCI binding affinity and
bate. Here, we demonstrate that al- instead is a direct consequence of re-
though reversed-polarity TCR–pMHC versed TCR–pMHCI engagement. Most
recognition is prevalent within a TCR TCR TCR–pMHC complexes that have been
naïve, viral epitope–specific T cell solved to date, upon which the canoni-
repertoire and may exhibit relatively cal TCR–pMHCI docking paradigm
high pMHCI affinity, such TCRs are CD3 CD3
has been established, were derived
unable to support TCR signaling in from expanded immune repertoires.
the presence of CD8 coreceptor be- Thus, we conclude that the highly
cause of mislocalization of Lck. These Lck Lck conserved docking polarity is driven
data support a paradigm in which the predominantly by the structural con-
highly conserved TCR–pMHCI dock- >10 nm straints imposed on TCR signaling and
Signal
ing polarity is driven by structural transduction recruitment into an immune response.
constraints on TCR signaling. CD8+ T cell CD8+ T cell In addition to the well-recognized
augmentation of signaling resulting
RATIONALE: Evidence suggests that The canonical polarity of TCR–pMHC docking is essential for colo- from canonical TCR–pMHCI engage-
the canonical TCR–pMHC docking calization of CD3 and coreceptor-associated Lck and for produc- ment, our findings suggest a role for
polarity is driven by evolutionary hard- tive TCR signaling. Schematic shows how canonical TCR–pMHC coreceptor–Lck association in pre-
wiring of complementary germline- recognition colocalizes Lck and CD3, driving TCR-mediated signaling. venting signaling by noncanonical
encoded motifs at the TCR and MHC By contrast, a reversed TCR–pMHC recognition polarity TCR–pMHC recognition. Such neg-
interface. An alternate model sug- mislocalizes Lck and CD3, impeding signaling. ative regulation would serve to limit
gests that TCR recognition of pMHC the extent of functional TCR cross-
is driven during thymic selection by the need RESULTS: In this study, we demonstrate that reactivity and constrain the number of signaling-
for the CD4 or CD8 coreceptors to bind MHC
and deliver coreceptor-associated Lck to the
reversed TCRs are prevalent in a naïve virus–
specific repertoire but are poorly represented
competent TCR-binding modalities.

CD3 signaling complex. We previously iden- in the immune response after virus challenge. The list of author affiliations is available in the full article online.
tified reversed-polarity TRBV17+ TCRs from We identified antigen-specific TCRab clono- *Corresponding author. Email: nicole.la.gruta@monash.edu
the preimmune influenza A virus (IAV)–specific types that were either poorly recruited or (N.L.L.G.); jamie.rossjohn@monash.edu (J.R.); s.gras@
latrobe.edu.au (S.G.)
repertoire that bound pMHCI (H-2DbNP366) clonally expanded and found an overriding †Present address: Department of Biochemistry and Genetics,
with a moderate affinity but were unable to association between immune prevalence and La Trobe Institute for Molecular Science, La Trobe University,
support robust T cell recruitment. Here, using canonical TCR–pMHCI docking. This was Melbourne, Victoria, Australia.
‡Joint senior authors.
a range of canonical and reversed TCRs spe- irrespective of pMHCI affinity, catch or slip
Cite this article as P. Zareie et al., Science 372, eabe9124
cific for the same cognate pMHCI, we tested bond formation, or TCR clustering, demon- (2021). DOI: 10.1126/science.abe9124
the hypothesis that the TCR–pMHCI dock- strating that a canonical docking polarity is
ing polarity precedes binding strength as required for T cell activation. This finding READ THE FULL ARTICLE AT
a key determinant of T cell activation. We was verified after viral challenge of adoptively https://doi.org/10.1126/science.abe9124

Zareie et al., Science 372, 1056 (2021) 4 June 2021 1 of 1


RES EARCH

◥ taining single–amino acid substitutions at


RESEARCH ARTICLE H-2Db Glu18 and/or Ala89, residues that are
uniquely important for binding the reversed
IMMUNOLOGY B17.R1 TCRa chain (1, 12). Ala89Glu substitu-
tion (H-2DbA89E–NP366) completely abrogated
Canonical T cell receptor docking on peptide–MHC tetramer binding to B17.R1 at high TCR expres-
sion levels without affecting B13.C1 TCR bind-
is essential for T cell signaling ing (Fig. 1, A and B). The loss of B17.R1 TCR
binding was verified by surface plasmon reso-
Pirooz Zareie1, Christopher Szeto1†, Carine Farenc1, Sachith D. Gunasinghe2, Elizabeth M. Kolawole3,

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
nance (SPR) analysis (Fig. 1C and Table 1).
Angela Nguyen1, Chantelle Blyth1, Xavier Y. X. Sng1, Jasmine Li4, Claerwen M. Jones1, We next used comparative staining with
Alex J. Fulcher5, Jesica R. Jacobs3, Qianru Wei6, Lukasz Wojciech6, Jan Petersen1,7, the H-2DbWT–NP366 and H-2DbA89E–NP366 tet-
Nicholas R.J. Gascoigne6, Brian D. Evavold3, Katharina Gaus2, Stephanie Gras1,7†‡*, ramers to determine the proportion of the
Jamie Rossjohn1,7,8‡*, Nicole L. La Gruta1‡* naïve H-2Db–NP366–specific CD8+ T cell pop-
ulation that was affected by this mutation,
T cell receptor (TCR) recognition of peptide–major histocompatibility complexes (pMHCs) is suggestive of a reversed TCR–pMHCI docking
characterized by a highly conserved docking polarity. Whether this polarity is driven by recognition or polarity (Fig. 1, D and E). Although a similar
signaling constraints remains unclear. Using “reversed-docking” TCRb-variable (TRBV) 17+ TCRs from the number of TRBV13+ cells was detected using
naïve mouse CD8+ T cell repertoire that recognizes the H-2Db–NP366 epitope, we demonstrate that their either tetramer (Fig. 1F), the mutant A89E
inability to support T cell activation and in vivo recruitment is a direct consequence of reversed docking tetramer detected only ~48% of TRBV17+
polarity and not TCR–pMHCI binding or clustering characteristics. Canonical TCR–pMHCI docking cells detected by the wild-type (WT) tetramer
optimally localizes CD8/Lck to the CD3 complex, which is prevented by reversed TCR–pMHCI polarity. (Fig. 1G). By contrast, these two tetramers
The requirement for canonical docking was circumvented by dissociating Lck from CD8. Thus, the showed equivalent staining of both TRBV13+
consensus TCR–pMHC docking topology is mandated by T cell signaling constraints. and TRBV17+ T cells in mice infected with
influenza A virus (IAV) (Fig. 1, H to K). Thus,
although TRBV17+ TCRs that bind H-2Db–NP366

T
cell–mediated immunity to pathogens and MHCII a1 helix (1). Evidence suggests that in a reversed orientation are prevalent in the
and cancers requires activation of T cells the conserved TCR–pMHC docking polarity naïve repertoire, they are not recruited into the
through ab T cell antigen receptor (TCR) is “hard-wired” by evolutionarily conserved immune response after IAV infection.
recognition of antigenic peptides pre- amino acid motifs in the germline-encoded
sented by major histocompatibility regions of TCRs and MHC molecules (2–5). An Recruitment into the immune response is
complex class I (MHCI) or class II (MHCII) alternate model suggests that TCR recognition associated with TCR–pMHCI docking topology
molecules. The extreme diversity inherent in of pMHC is driven during thymic selection by independently of TCR–pMHCI affinity
both the TCR repertoire and the array of the need for the CD4 or CD8 coreceptors to To gain a further understanding of TCR-intrinsic
pMHC ligands is reflected in the substantial bind MHC and deliver coreceptor-associated determinants of recruitment, we analyzed
variation at the TCR–pMHC interface (1). De- Lck to the CD3 signaling complex (5). Because TRBV17+ H-2Db–NP366–specific TCRab sequences
spite this variation, nearly all of the TCR–pMHC of the proposed positioning of CD3 in the TCR– from uninfected (13) and infected B6 mice. The
ternary complexes solved to date exhibit a highly pMHC–CD4/CD8 complex, this model posits naïve TRBV17+ TCRab repertoire was diverse,
consistent docking polarity, with the TCRa that only canonical polarity TCR–pMHC inter- comprising a range of TCRa-variable (TRAV)
chain sitting over the MHCI a2 or MHCII actions are conducive to signaling (6–8). The genes with distinct CDR3a and CDR3b se-
b1 helix, and TCRb docking over the MHCI biological significance of the canonical docking quences (Fig. 2A) (13). By contrast, each im-
polarity remains unclear and has not been mune repertoire was characterized by the
1 tested experimentally because of the rarity of dominance of only one or two clones (Fig. 2B).
Infection and Immunity Program and Department of
Biochemistry and Molecular Biology, Biomedicine Discovery TCR–pMHC docking polarities outside of this Thus, the low prevalence of TRBV17+ TCRs in
Institute, Monash University, Clayton, Victoria, Australia. paradigm (1, 9–11). We recently identified CD8+ the H-2Db–NP366–specific immune repertoire
2
European Molecular Biology Laboratory (EMBL) Australia T cells expressing “reversed” TCRs that bind (13) is caused by an inability of most of these
Node in Single Molecule Science and the ARC Centre of
Excellence in Advanced Molecular Imaging, School of Medical their cognate pMHCI in a 180° reversed orien- clones to respond to IAV.
Sciences, University of New South Wales, New South Wales, tation, signaled poorly, and drove a weak To demonstrate that the key criteria for
Australia. 3Department of Pathology, University of Utah antiviral immune response (12). Here, we immune recruitment from the TRBV17 subset
School of Medicine, Salt Lake City, UT, USA. 4Infection and
Immunity Program and Department of Microbiology, investigated the key drivers of the canonical was a canonical docking polarity, we selected
Biomedicine Discovery Institute, Monash University, Clayton, TCR–pMHC docking polarity and its role in three TRBV17+ TCRs from the immune rep-
Victoria, Australia. 5Monash Micro Imaging, Monash T cell recognition and activation. ertoire for further structural and biophysical
University, Clayton, Victoria, Australia. 6Immunology
Translational Research Programme and Department of
analyses. These TCRs were taken from ex-
Microbiology and Immunology, Yong Loo Lin School
Results panded clones (mouse 1: B17.C1; mouse 3:
of Medicine, National University of Singapore, Singapore Unconventional TRBV17+ TCRs are prevalent in B17.C2) and from the single TRAV14+ clone
117545. 7Australian Research Council Centre of Excellence the naïve H-2Db–NP366–specific repertoire but from mouse 3 (B17.R2) (Fig. 2B). Although
in Advanced Molecular Imaging, Monash University,
do not contribute to the immune response the B17.R2 TCR was identified from an in-
Clayton, Victoria, Australia. 8Institute of Infection and
Immunity, Cardiff University School of Medicine, Heath Park, We have previously described two naïve TRBV17+ fected mouse, the sensitive detection method
Cardiff, UK.
TCRs (NP1-B17, hereafter referred to as B17.R1, and apparent lack of clonal expansion means
†Present address: Department of Biochemistry and Genetics,
La Trobe Institute for Molecular Science, La Trobe University, and NP2-B17) that recognize the H-2Db–NP366 that it was likely derived from a naïve T cell.
Melbourne, Victoria, Australia. epitope in a 180° reversed orientation (12). To We performed tetramer staining of 293T cells
‡Joint senior authors. determine whether reversed TCR docking was a expressing these TCRs, along with a TRBV13+
*Corresponding author. Email: nicole.la.gruta@monash.edu
(N.L.L.G.); jamie.rossjohn@monash.edu (J.R.); s.gras@latrobe. general feature of TRBV17+ H-2Db–NP366–specific TCR (B13.C1) known to drive robust immune
edu.au (S.G.) TCRs, we generated H-2Db–NP366 tetramers con- recruitment (12, 13). Those TCRs that were

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 1 of 13


RES EARCH | R E S E A R C H A R T I C L E

A B B13.C1 B17.R1 C B17.R1


150 WT
A89V 400 WT KD = 37.5 ± 4.4 M
B13.C1

Response Units (RU)


A89E A89E KD = >200 M
B17.R1
% MFI of WT

100 A89F 300


A89R
E18A
200
E18V
50 E18F

H-2Db –NP366
E18L 100

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
E18W
0
0
0 25 50 100 200
9E

9R
A8 F

8F

E1 L
9V

8V
A8 T

8A

8W
Overlay Concentration (µM)
8
9
W

E1
A8

E1
A8

E1
E1

TCR

D E F G
1.0 1.0

TRBV13 + (fold change)

TRBV17 + (fold change)


H-2DbA89E –NP366
30.58 0
H-2DbWT –NP366

13.21 0
Pre-immune

0.5 0.5

TRBV17
TRBV17

0.0 0.0

57.02 12.40 67.92 18.87 -0.5 -0.5

TCR TRBV13 TCR TRBV13 -1.0 -1.0


H-2DbA89E –NP366 H-2DbA89E –NP366

H I J K
1.0 1.0

TRBV13 + (fold change)

TRBV17 + (fold change)


H-2DbA89E –NP366

0.20 0
H-2DbWT –NP366

0.24 0
Immune

TRBV17
TRBV17

0.5 0.5

0.0 0.0

79.62 20.18 79.14 20.62


-0.5 -0.5
CD44 TRBV13 CD44 TRBV13
-1.0 -1.0
H-2DbA89E –NP366 H-2DbA89E –NP366

Fig. 1. Reverse-docking TRBV17+ T cells are not recruited into the immune TRBV13 (Vb8.3) or TRBV17 (Vb9) TCRb chains isolated from naïve mice
response. (A and B) HEK293T cells were transfected with B13.C1 (black) and [10 mice pooled/data point; (D) to (G)] or immune mice 10 days after
B17.R1 (gold) TCRs, and binding of WT or mutant H-2Db–NP366 tetramers was infection [one mouse per data point; (H) to (K)]. Plots represent the percentage
analyzed 48 hours after transfection. Live GFP+ cells were gated and analyzed for change in TRBV13+ T cells bound by the H-2DbA89E–NP366 tetramer relative to
change in geometric mean fluorescence intensity (geoMFI). Shown is geoMFI as H-2DbWT–NP366 tetramer from naïve mice (F) and immune mice (J), and the
a percentage change from the WT tetramer (A) and representative dot plots percentage change in TRBV17+ T cells bound by the H-2DbA89E–NP366
staggered (top panel) or overlaid (bottom panel) of TCR expression and binding tetramer relative to H-2DbWT–NP366 tetramer binding from naïve mice (G) and
of various mutant tetramers (B). Data shown in (A) are from two independent immune mice (K). Data shown from 10 pooled naïve mice in (D) to (E)
experiments combined. Data shown in (B) are dot plots from one representative represent one sample of n = 3. Summary data shown in (F) and (G) are the
experiment. (C) Binding response of B17.R1 TCR against H-2DbWT–NP366 in black mean and SEM of three independent datasets (each containing 10 pooled
or H-2DbA89E–NP366 in green. Data presented are from a single experiment mice). Data from an individual immune mouse shown in (H) and (I) represent
representative of two independent experiments. (D to K) Representative dot one sample of n = 9. Summary data shown in (J) and (K) are mean and
plots and graphs showing the proportion of H-2DbWT–NP366 binding [(D) and (H), SEM from a representative three samples (each containing one mouse)
black] or H-2DbA89E–NP366 binding [(E) and (I), green] T cells using either collected on one day.

well represented in the immune response, in- cells, we determined TCR affinity by SPR (Table 1). Structural determination of TCR–H-2Db–NP366
cluding B13.C1 (Fig. 2C), B17.C1 (Fig. 2D), and The canonical immune B17.C1 TCR had an docking topologies
B17.C2 (Fig. 2F), all showed equivalent binding extremely weak affinity for H-2Db–NP366 (KD We next determined the crystal structures of
by the H-2DbWT–NP366 and H-2DbA89E–NP366 > 200 mM) (Fig. 2D). By contrast, the minor or B17.R2 and B17.C1 TCRs in complex with H-
tetramers. By contrast, the poorly represented naïve B17.R2 TCR had a substantially higher 2Db–NP366 (Fig. 3 and tables S1 to S4). As
or unrecruited B17.R2 TCR showed significantly affinity (KD = 6.34 mM) and tetramer binding suggested from tetramer binding (Fig. 2), the
reduced binding of the H-2DbA89E–NP366 (Fig. 2E), similar to that of the immunodom- B17.R2 TCR adopted a reversed docking po-
tetramer (Fig. 2E) and a 10-fold reduced af- inant B13.C1 TCR (KD = 4.13 mM) (Fig. 2C). larity over H-2Db–NP366, forming a docking
finity for H-2D bA89E –NP 366 , suggesting a Thus, the prevalence of T cells in the immune angle of 238° and binding in a similar manner
reversed TCR–pMHCI docking polarity. response is primarily associated with canon- to the previously determined B17.R1–H-2Db–
To determine the role of TCR–pMHCI af- ical TCR–pMHC docking polarity, independent NP366 and NP2-B17–H-2Db–NP366 complexes
finity in driving immune recruitment of TRBV17+ of TCR–pMHC affinity. (Fig. 3, A and B, and table S4) (12). By contrast,

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 2 of 13


RES EARCH | R E S E A R C H A R T I C L E

Table 1. TCRs, recruitment characteristics, docking polarity, and KD.

TCR TCRa TCRb Immune recruitment Docking polarity KD (mM)


H-2D bWT
–NP366 H-2DbA89E–NP366
B13.C1 (NP1–B13*) TRAV16; RVSGGSNAKL TRBV13–1; SGGGNTGQL Immunodominant Canonical† 4.13 ± 1.55 ND
............................................................................................................................................................................................................................................................................................................................................
B17.R1 (NP1-B17*) TRAV14; SETSGSWQL TRBV17; SRDLGRDTQ Naïve or poorly recruited Reversed 37.5 ± 4.4 >200
............................................................................................................................................................................................................................................................................................................................................
B17.R2 TRAV14; SETSASWQL TRBV17; SRDLGRDTQ Naïve or poorly recruited Reversed 6.34 ± 1.58 62 ± 30
............................................................................................................................................................................................................................................................................................................................................

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
B17.C1 TRAV4–4; VTGNTGKL TRBV17; SRGTIHSNTEV Clonally expanded Canonical >>200 >200
............................................................................................................................................................................................................................................................................................................................................
B17.C2 TRAV14D; SRRGSAKL TRBV17; SRGGLSYEQ Clonally expanded Canonical† ND ND
............................................................................................................................................................................................................................................................................................................................................

*Previously reported in (12). †Structure undetermined; polarity inferred by MHCI mutational analyses. ND, not determined. SPR values are the mean ± SEM of experiments
performed at a minimum twice in duplicate.

A C 40

Response Units (RU)


WT KD = 4.13 ± 1.55 M
CDR3 CDR3 B13.C1
10 30
9 Pre-Immune
Number of H-2Db–NP366

20
-specific TRBV17+ cells

8
7 10
6
0
5 0 25 50 100
4 Concentration ( M)
3
2 D 40 WT KD = >200 M

Response Units (RU)


B17.C1 A89E KD = >200 M
1 30
0
m1 m2 m3 m4 m5 m6 20

10

B 0
0 25 50 100 200
60 Concentration ( M)
Immune CDR3 CDR3
E
Number of H-2Db–NP366

WT KD = 6.34 ± 1.58 M
-specific TRBV17+ cells

500

Response Units (RU)


B17.R2 A89E KD = 62.0 ± 30 M
400
40
300

200

100
20
0
0 25 50 100 200
Concentration ( M)

0 F
m1 m2 m3 m4 B17.C2 H-2DbWT–NP366
H-2DbA89E–NP366
H-2Db–NP366

TCR

Fig. 2. Prevalence of TCRs in the immune response is unrelated to TCR– or H-2DbA89E–NP366 (green) tetramer staining analyzed by flow cytometry
pMHCI affinity. (A and B) TRBV17 TCRb+ H-2Db–NP366–specific TCR clonotypes 48 hours later. Shown are representative flow cytometry plots of TCRb expression
presented as bar graphs with corresponding CDR3a/CDR3b sequences from and tetramer binding from live GFP+ cells (left) and SPR sensorgrams (right)
six individual naïve (m1 to m6) (13) (A) or four individual immune mice (m1 to of B13.C1 (C), B17.C1 (D), B17.R2 (E), and B17.C2 (F) TCRs. Data shown in (C) to
m4) 10 days after infection with IAV (B). (C to F) HEK293T cells were transfected (F) are from one experiment representative of two (SPR) or three (flow cytometry)
with pMIGII vectors encoding abTCR and CD3gdez and H-2DbWT–NP366 (black) independently performed experiments.

the B17.C1 TCR adopted a canonical docking surface area (BSA)] in the interaction, with compassing the framework region of the
polarity (Fig. 3, C and D, and table S4). In the only the CDR3a loop contributing to binding b-chain (FWb) region (39.2% of the BSA),
B17.R2 TCR–H-2Db–NP366 complex, the TCRa (Fig. 3B and table S4). By contrast, the TCRb the CDR2b loop (27.3% of the BSA), and the
chain played a lesser role [26.4% of the buried chain contributed to 73.6% of the BSA, en- CDR3b loop (7.1% of the BSA) (Fig. 3B and

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 3 of 13


RES EARCH | R E S E A R C H A R T I C L E

A B C D
B17.R2

B17.C1 B17.C1

NP366 238°
33°

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
B17.R2 NP366

CDR2 CDR3 CDR1 CDR1

CDR3

2m FW
H-2Db
H-2Db
CDR3
CDR2
2m
CDR3
Fw

Fig. 3. B17.R2 TCR and B17.C1 TCR in complex with H-2Db–NP366. (A and C) TCR–pMHC complexes of B17.R2 TCR–H-2Db–NP366 (A) and B17.C1 TCR–H-2Db–
NP366 (C). (B and D) TCR atomic footprints on the surface of each corresponding pMHC complex. Spheres represent the center of mass of Va (pink) and Vb (blue).
Pie charts represent the relative contributions of each TCR segment to the pMHCI interaction. Contacts are colored according to the CDR loop involved. The
TCRa chain is colored pink, the TCRb chain blue, H-2Db white, b2m orange, and peptide black/dark gray. CDR1a, CDR2a, and CDR3a are colored in green, teal, and
purple, respectively, and CDR1b, CDR2b, and CDR3b in red, orange, and yellow, respectively. Framework (FW) is colored pink for the FWa and blue for the FWb.

table S4). Similarly, the B17.C1 TCR–H-2Db– CD8+ TCR transductants were stimulated with munodominant B13.C1 TCR, for further inves-
NP366 complex exhibited an unusually high peptide bound to H-2D bWT or to mutant tigation. We then generated retrogenic mice
contribution of the TCRb chain (75% of the H-2DbD227K to assess the contribution of co- expressing the canonical polarity B13.C1 or
BSA), with the interactions dominated by receptor binding to bond strength (19). Both B17.C2 TCRs and the reversed-polarity B17.R1
the CDR3b loop (37.8% of the BSA) and the of the canonical docking TCRs, the high- or B17.R2 TCRs (23). We were unsuccessful
CDR2b loop (23.2% of the BSA) (Fig. 3D and affinity B13.C1 TCR and the low-affinity B17.C1 in expressing the B17.C1 TCR in vivo despite
table S4). Moreover, this complex structure TCR, were able to form catch bonds, peaking at validating construct fidelity and instead gen-
presents an unusually low number of con- ~10 pN (fig. S1, B and C). CD8 binding con- erated TCR-retrogenic mice expressing the
tacts between the TCR and the MHCI (table tributed significantly to bond lifetime only for B17.C2 TCR, which exhibited similar prop-
S2), as well as a poor shape complementarity the low-affinity B17.C1 TCR (fig. S1, B and C). By erties. That is, it expressed TRBV17, had a
(table S4), and was consistent with the contrast, reversed-polarity TCRs (B17.R1 and moderate to low avidity for H-2Db–NP366,
low affinity of this interaction (Table 1 and B17.R2) showed the formation of slip bonds bound H-2Db–NP366 independently of Ala89
table S2). with pMHCI, with a loss of bond lifetime with (and thus likely docked in a canonical orien-
increasing force (fig. S1, D and E). However, at tation) (Fig. 2F), and was expanded in the
CD8+ T cell recruitment does not correspond to least for the high-affinity B17.R2 TCR, the immune repertoire (Table 1). Consistent with
two-dimensional TCR–pMHCI affinities nor bond bond lifetime generated at ~10 pN, an approx- our previously published data (12), adoptive
duration under force imation of the physiological force on a TCR transfer of retrogenic B13.C1+ and B17.R1+
To test the possibility that T cell recruitment (20–22), was similar to that observed at the T cells, either alone or in combination, fol-
correlated with two-dimensional (2D) TCR– peak of the catch bond formation (fig. S1F). lowed by IAV challenge (Fig. 4A), resulted in
H-2Db–NP366 affinities, we measured the rel- Thus, although the reversed TCR–H-2Db–NP366 the effective recruitment and expansion of
ative 2D affinity of B13.C1, B17.R1, B17.R2, and interaction is characterized by slip-bond for- canonical B13.C1+ T cells but not reversed B17.
B17.C1 TCRs for H-2Db–NP366 using the 2D mation, it exhibits relatively high bond life- R1+ T cells (Fig. 4, B to N, and fig. S2A). Failure
micropipette adhesion frequency assay (2D– times at the physiological force of 10 pN. to recruit B17.R1+ T cells was not caused by
MP) (14–18). Although the reversed B17.R2 green fluorescent protein (GFP) expression be-
TCR had the second highest 2D affinity after Only canonical docking TCRs can support cause the same experiment performed with
the B13.C1 TCR (fig. S1A), it was not condu- immune recruitment irrespective of GFP+ B13.C1+ T cells showed similar recruit-
cive to robust immune recruitment (Fig. 2B). TCR–H-2Db–NP366 affinity ment profiles as those coexpressing mCherry
By contrast, the canonical docking B17.C1 TCR To confirm our earlier observations (Fig. 1, D (fig. S2, B to E).
with a lower 2D affinity (fig. S1A) was expanded to K) that T cell recruitment into the immune To distinguish the impact of TCR–pMHCI
in the immune repertoire (Fig. 2B). Thus, response was primarily dependent on a ca- affinity and docking polarity on recruitment,
the recruitment of the H-2Db–NP366–specific nonical TCR–pMHCI docking polarity indepen- we adoptively transferred T cells expressing the
TRBV17+ T cells occurs independently of TCR– dent of TCR–pMHCI binding strength, we high-affinity reversed B17.R2 TCR either alone
pMHCI affinity. selected B17.R1, which binds with low to mod- or with B13.C1+ T cells before IAV challenge.
We next measured the TCR–H-2Db–NP366 erate affinity, and the B17.R2 TCR, which binds The B17.R2 T cells were not detectable in the
bond lifetime under conditions of force (14–17). with high affinity similar to that of the im- immune response after either single transfer

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 4 of 13


RES EARCH | R E S E A R C H A R T I C L E

Individual
A CD45.2+ Co-transfers
TCR retrogenic donors transfers

B13.C1 / B17.R1 / 25 25 50 25
B17.C2 B17.R2 C F I L

% of CD8+ T cells
% of CD8+ T cells
20 20 40 20
15

BAL
15 30 15
Adoptive transfer of
CD8+ T cells (4000) 10 10 20 10
5 5 10 5
CD45.1+ host mice

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
0 0 0

15 15 20 15
D G J M

% of CD8+ T cells
+ T cells
15
10

Spleen
10 10
Intranasal influenza A virus
10

% of CD8
infection (1x10E4
PFU HKx31)
5 5 5
5
10 days

0 0 0
Harvest BAL, spleen and mLN
5 5 5 5
B E H K N

% of CD8+ T cells
4 4
CD8+ T cells
4 4
mLN

3 3 3 3
2 2 2 2
% of

1 1 1 1
0 0 0 0
1

2
1

2
C

R
C

C
3.

7.

7.

7.

3.

7.
3.

7.

7.

7.
B1

B1

B1

B1

B1

B1
B1

B1

B1

B1
Fig. 4. TCR–pMHCI docking orientation is a primary determinant of in mice that received cotransfers of retrogenic T cells. (C to N) Percentage
vivo T cell activation and recruitment. (A) Schematic diagram of the retrogenic CD8+ T cells of total CD8+ T cells isolated from the BAL [(C), (F),
experimental protocol. Retrogenic CD45.2+ GFP/mCherry+ CD4− CD8+ T cells (I), (L)], spleen [(D), (G), (J), (M)], or mLNs [(E), (H), (K), (N)] from single
were sorted from B13.C1 (mCherry), B17.R1 (GFP), B17.R2 (GFP), and adoptive transfers [(C) to (E)] or cotransfers [(F) to (N)] at day 10 after
B17.C2 (mCherry) retrogenic mice, and 4 × 103 cells were transferred infection. Each point represents data from an individual mouse (n = 2 to 8),
individually or cotransferred into naïve C57BL/6 mice that were infected and the combined dataset was collected over 4 separate days. Each sample
intranasally with 1 × 104 PFUs of HKx31 IAV the following day. Mice were testing cotransferred retrogenic T cells was paired with individual transfers as
euthanized for analysis 10 days after infection. (B) Dot plots from the BAL of experimental controls.

(Fig. 4, C to E) or cotransfers (Fig. 4, B and I to A and B) for analysis of TCR clustering by Using our current structural understanding
K). Finally, we adoptively transferred the low- dSTORM (Fig. 5A). For both unstimulated of the interactions between TCR–pMHCI (1),
to-moderate avidity, canonical B17.C2+ T cells and stimulated T cells, TCRs exhibited a non- MHCI, and CD8 (26) and TCRab and the CD3
and the high-affinity, reversed B17.R2+ T cells random clustered spatial distribution on the chains (27, 28), we modeled the quaternary
into B6 mice, which were then challenged with cell membrane, as indicated by a significantly TCR–pMHC–CD8–CD3 structure for canonical-
IAV. Retrogenic B17.C2+ T cells were readily larger L(r)-r value relative to complete spatial polarity B17.C1 TCR–H-2Db–NP366 (Fig. 5D)
recovered from bronchoalveolar lavage (BAL) randomness (Fig. 5B). The peak of molecular and reversed-docking B17.R1 TCR–H-2Db–
(Fig. 4, B, C, and L), spleen (Fig. 4, D and M), TCR clustering [max L(r)-r], was higher after NP366 interactions (Fig. 5E). The 180° re-
and mediastinal lymph nodes (mLNs) (Fig. 4, E stimulation (ICAM + pMHCI) compared with versal of the B17.R1 TCR over the H-2Db–NP366
and N). By contrast, the B17.R2 TCR did not unstimulated T cells (ICAM), indicative of substantially altered the position of CD8
support detectable immune expansion into any antigen-driven TCR clustering (Fig. 5, B and C). relative to CD3.
tissue (Fig. 4, L to N). Thus, TCR–pMHCI dock- This antigen-driven TCR clustering was similar To experimentally determine whether re-
ing topology supersedes TCR–pMHCI affinity for both the canonical (B13.C1) and reversed versed TCR–pMHCI docking affected the local-
as the primary determinant for effective in vivo (B17.R2) docking TCRs. Thus, reversed TCR– ization of CD8-associated Lck to the CD3
immune recruitment. pMHCI docking does not impede the forma- complex, we used fluorescence lifetime imag-
tion of multimeric TCR–CD3 structures. ing microscopy (FLIM)–Förster resonance
Reversed-polarity TCRs do not prevent energy transfer (FRET) microscopy to deter-
TCR clustering Reversed TCR recognition of H-2Db–NP366 mine the close (<10 nm) molecular associa-
To determine whether the reversed TCR– impedes the localization of CD3 and CD8 tion between CD8b–mCherry and CD3z-GFP
pMHCI docking prevents the formation of Assuming that a similar arch-like structure is fusion proteins in live TCR+ hybridoma cells
signaling-competent multimers (10), 5KC formed after pMHCI recognition by the TCR/ after epitope-specific stimulation (29–32) because
T cell hybridoma cells (TCRab−CD4−CD8−) CD3 and CD8 as has been observed for TCR– this was not readily feasible in primary T cells.
(24) expressing either the B13.C1 or B17.R2 pMHCII–CD4 (25), the canonical TCR–pMHC We used the FLIM–FRET approach over con-
TCRs were placed on a supported lipid bi- docking polarity may be essential for coreceptor- ventional superresolution microscopy (20-
layer (SLB) containing ICAM-1 (unstimulated) associated Lck to be situated proximally to CD3 to 30-nm resolution) to allow us to resolve
or ICAM-1 and H-2Db–NP366 (stimulated) (Fig. 5, for the initiation of signal transduction (25). protein–protein interactions (<10 nm).

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 5 of 13


RES EARCH | R E S E A R C H A R T I C L E

B13.C1 B17.R2
A
dSTORM images

ry
F .4
) er
C2 FP Ch sit
e
(D -G -m po
V 3 8 m IM
CT CD CD Co FL

B13.C1
TCR Cluster map

B17.R1

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
B17.R2
B B13.C1 B17.R2 C B13.C1 B17.R2
1200 1200 ns

B17.C1
1000 ICAM-1 + pMHCI 1000 ns
ICAM-1 + pMHCI

Max. L(r) - r
800 800
L(r) - r

600 600

B17.C2
400
400
ICAM-1 200
ICAM-1
200
Random distribution Random distribution 0
0

2
C

R
0 100 200 300 400 500 0 100 200 300 400 500

3.

7.

3.

7.
G
B1

B1

B1

B1
r (nm) r (nm)
ICAM-1 ICAM-1 + pMHCI

D B17.C1-H-2Db–NP366 E B17.R1-H-2Db–NP366 5 B13.C1


B17.R1

AvAmp (%)
2m 0
B17.R2
CD8
-5 B17.C1
MHC-I
CD8 B17.C2
2m
-10
MHC-I Syn
TCR
TCR
-15
*** *** ***
-20
NP366-374 - + - + - + - + - +
CD3 CD3
CD3 CD3
CD3 CD3

Fig. 5. Reversed TCR–pMHCI docking does not impede TCR clustering but CD8 coreceptor (PDB 3DMM), and ternary complex of B17.C1 TCR–H-2Db–NP366 (D)
mislocalizes the CD8 coreceptor and CD3 complex. (A) Single-molecule and B17.R1 TCR–H2Db–NP366 (PDB 5SWZ) (E). The different chains are shown in
images of 5KC TCR transductants expressing canonical docking B13.C1 or B17.R2 black for CD3zz, red for the CD3ge, pink for the CD3de, and blue for the TCRab
TCRs on supported lipid bilayers decorated with either ICAM-1 only (ICAM-1) or chains. The MHC is shown in white, and the CD8 coreceptor is shown in orange.
ICAM-1 + H-2Db–NP366 (ICAM-1 + pMHC) at 5 min. Scale bar, 5 mm. Close-up view (F) DC2.4 cells labeled with CTV were pulsed with 10 mM NP366 peptide for 1 hour
of single-molecule localization microscopy image (2 × 2 mm) as TCR cluster maps before coculture with 5KCzGFP.CD8bmCherry–expressing TCRs as indicated. T cell
(lower panels) from representative regions (boxed, top panel), with TCRb hybrids interacting with a DC2.4 cell were imaged up to 20 min after coculture
molecules in clusters shown in green and molecules outside clusters shown in blue. by confocal microscopy and subsequently analyzed by FLIM to measure GFP
Cluster contours are highlighted in red lines. (B) Ripley’s K analysis of TCR lifetime 10 to 20 s later (fig. S3). Scale bar, 10 mm. (G) Amplitude weighted lifetime
clustering [L(r)-r] against radii (r). Complete spatial randomness is shown as a of GFP (TavAMP) of B13.C1, B17.R1, B17.R2, B17.C1, and B17.C2 TCR+ T cells (±NP366
solid gray line where L(r)-r = 0. Positive L(r)-r values indicate molecular clustering peptide) measured as percentage change at the synapse versus nonsynapse
relative to the random distribution, shown as the mean (solid line) ± 95% (SynDTavAMP). Cells were observed on three different days. Statistical analysis was
confidence interval (dashed lines) for TCRs under each condition. Dotted lines performed by two-way ANOVA to examine the effect of stimulation and the day on
indicate ± SEM. (C) The maximum L(r)-r value derived from the peak of the graph which each cell was observed on the SynDTavAMP for each cell line. Significant
in (B) corresponds to the spatial scale (r) at which the highest degree of clustering effects on the SynDTavAMP by peptide stimulation are indicated by ***P ≤ 0.001 as
of localizations is being observed (n = 20). Statistical analysis performed by indicated. For each cell line, no significant effect on the SynDTavAMP was found for
one-way ANOVA. (D and E) Representation of the TCR–CD3 complex (PDB 6JXR), the day each cell was observed.

Expression of the FRET pair constructs (fig. S3A) S3, B to D). Stimulation of B13.C1 TCR+ cells expressing the canonical B17.C1 and B17.C2
did not negatively affect TCR signaling because resulted in FRET, as measured by a substantial TCRs (Fig. 5, F and G). However, stimulation
phosphorylated extracellular signal-regulated reduction in the amplitude weighted lifetime of of cells expressing the reversed B17.R1 and
kinase (pERK) could be detected after stimula- the donor (GFP) at the synapse (fig. S3, E and B17.R2 TCRs resulted in negligible FRET (Fig. 5,
tion of the B13.C1 TCR+ 5KCCD3zGFP.CD8abmCherry F), indicating colocalization of the CD8bmCherry F and G). Thus, a reversed TCR–pMHCI dock-
cells, similar to B13.C1 TCR+ 5KC T cells ex- and CD3zGFP molecules (Fig. 5, F and G). We ing topology results in improper localization
pressing WT CD8ab and CD3z (Fig. 6F and fig. also observed FRET after stimulation of cells of CD8bmCherry and CD3zGFP in a manner that

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 6 of 13


RES EARCH | R E S E A R C H A R T I C L E

A CD8WT
B CD8NULL
C CD8CxC F CD8WT G CD8NULL H CD8CxC
100
B13.C1 180 180 180
B13.C1

pERK MFI (% of control)


80 B17.R2
160 160 160
60
140 140 140 B17.R2
40

20

0 120 120 120


3 3 4 5 3 3 4 5 3 3 4 5
-10 0 10 10 10 -10 0 10 10 10 -10 0 10 10 10
TCR

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
100 100 100

0 20 40 60 0 20 40 60 0 20 40 60
Min
CD8

4000 4000 4000

pERK signal magnitude


**
CD8 3000 3000 3000

(AUC)
2000 2000 2000
D B13.C1 E B17.R2
1000 1000 1000
IP : CD8α
IB : Lck
0 0 0
IP : CD8α

tim

tim

tim
6

6
36

36

36
IP : CD8α

P
ns

ns

ns
N

N
U

U
LL

LL
xC

xC
T

T
U

60 60 60
8W

8W
8N

8N
8C

8C

[IL-2] (% of Ab stim)
D

D
D

D
D

D
C

C
C

*
C

40 * 40 40
*

20 20 20

0 0 0
1 2 10 1 2 10 1 2 10
tim

tim

tim
ns

ns

ns
U

U
[Peptide] ( M)

Fig. 6. Signaling constraints that mandate TCR–pMHCI docking orientation stimulation. Samples were tested in duplicate (n = 3), and the mean and SEM of
are driven by Lck sequestration and localization by the CD8 coreceptor. all datasets is shown. Statistical analyses were performed using a paired-samples
(A to C) Representative histograms of TCRb expression (top panel) and dot plots t test to pair by dataset. Statistical significance is indicated by **P ≤ 0.01 as
of CD8ab expression (bottom panel) by 5KC TCR transductants expressing indicated. IL-2 secretion (bottom panels) into the supernatant by CD8WT (F),
B13.C1 (black) and B17.R2 (red). (D and E) Immunoblots of CD8-associated Lck CD8NULL (G), and CD8CxC (H) transductants was measured by enzyme-linked
in CD8WT CD8NULL and CD8CxC transductants expressing B13.C1 (D) or B17.R2 (E) immunosorbent assay (ELISA) after 16 hours of coincubation with peptide-pulsed
TCRs. (F to H) Time-resolved pERK induction (top panels) up to 60 min after DC2.4 cells and is presented as a percentage of the plate-bound anti-CD3
coincubation with peptide-pulsed DC2.4 cells, presented as a percent change antibody stimulation controls. Samples were tested in duplicate (n = 3), and the
from a no-peptide control (measured at corresponding time point) using cells mean and SEM of all datasets is shown. Statistical analyses were performed
that express CD8WT (F), CD8NULL (G), and CD8CxC (H). Middle panel: pERK signal using a paired-samples t test to pair by dataset. Statistical significance is
magnitude analyzed by area under the curve (AUC) analysis of 0 to 60 min of indicated by *P ≤ 0.05 as indicated.

is independent of the strength of TCR–pMHCI on CD8 for binding to H-2Db–NP366 (fig. S1, The CD8WT B13.C1 TCR+ transductants me-
binding. B, E, and F). Each TCR was expressed (i) with diated robust signaling throughout the time
WT CD8ab (CD8WT) (Fig. 6A), (ii) in the ab- course, inducing a significantly higher pERK
The CD8 coreceptor inhibits TCR signaling by sence of CD8 (CD8NULL) (Fig. 6B), or (iii) with signal magnitude and IL-2 secretion com-
reversed-polarity TCRs mutant CD8ab containing C227A and C229A pared with B17.R2 (Fig. 6F). For B13.C1 pERK
We hypothesized that when TCR–pMHCI po- substitutions in the cytoplasmic tail of was induced as early as 10 min and main-
larity is reversed, the association of Lck with CD8a to abrogate Lck binding (CD8 CxC ) tained for 60 min after stimulation, and there
CD8 prevents, rather than promotes, effective (33) (Fig. 6, C to E). All cell lines showed a was substantial IL-2 production at 16 hours
Lck localization to CD3. To test this, 5KC similar sensitivity to phorbol 12-myristate (Fig. 6F). By contrast, the high-affinity re-
T cells were transduced with either the high- 13-acetate–ionomycin stimulation or anti- versed B17.R2 TCR showed negligible signal
affinity canonical B13.C1 TCR or the high- body-mediated polyclonal stimulation, as transduction when coexpressed with CD8WT,
affinity reversed B17.R2 TCR (Fig. 6, A to C) measured by pERK or interleukin-2 (IL-2) as evidenced by minimal pERK and no de-
because neither of these TCRs is dependent production (fig. S4). tectable IL-2 (Fig. 6F). Whereas the loss of

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 7 of 13


RES EARCH | R E S E A R C H A R T I C L E

CD8 (CD8NULL) severely attenuated the sig- Our current work provides clear support for preferentially sequestered away from the TCR–
nal transduction capacity of the B13.C1 TCR, this model by demonstrating that the inability CD3 complex to impair TCR signaling in the
detectable, low-level pERK and IL-2 secre- of reversed TCRs to signal (i) occurs indepen- absence of MHC ligands. In the presence of
tion was evident after stimulation of B17.R2 dently of binding strength, (ii) is dependent on MHC ligands, the coreceptor delivers Lck and
TCR+ cells (Fig. 6G) and was statistically in- CD8 binding of Lck, and (iii) is characterized promotes MHC-restricted TCR signaling. This
distinguishable from B13.C1. To distinguish by an inability to colocalize CD8 and CD3 after may be exacerbated in the case of CD4, which
the contribution of CD8 to MHCI binding antigen stimulation. Moreover, our demon- binds Lck with higher affinity than CD8 (42, 43).
versus Lck delivery, we stimulated cells ex- stration of robust TCR–CD3 cluster formation Although small amounts of residual free Lck
pressing the mutant CD8CxC. Again, both the suggest that unusual docking topologies do not may be able to initiate some early phosphory-
B13.C1 TCR+ and B17.R2 TCR+ cells transduced

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
preclude signaling by inhibiting TCR multi- lation events (32), it is insufficient to support full
a signal of similar kinetics and magnitude, merization (10, 34), which is consistent with activation.
with increased pERK compared with that ob- previous work showing that dense TCR–CD3 Although a useful tool for determining the
served in the absence of CD8 (Fig. 6H). This cluster formation can occur independently mechanism constraining signaling-competent
signaling again corresponded to detectable of signaling and better reflects TCR binding modalities of TCR recognition, the drivers of
and equivalent levels of IL-2 production. (35). Our observation that the reversed TCR– reversed TCR docking in this instance are
Similar findings were made upon analysis pMHCI interaction formed slip bonds and yet unclear. It has been suggested that a reversed
of 5KCzGFP.CD8bmCherry T cells (fig S3, B to D) remained inherently capable of signal trans- TCR–pMHC orientation may be a consequence
and independently generated B13.C1, B17.R1, duction, supports the notion that catch bonds of positively charged residues and/or proline
and B17.R2 TCR+ cells (fig. S5A). These find- are not essential (although likely still opti- within the CDR3b loop, preventing interaction
ings were also in agreement with TCRb down- mal) for signaling. with a conserved cluster of positively charged
regulation analysis after stimulation (fig. S5B). The observation that CD8 was an impedi- residues on the CDR3b contact regions of
Signaling mediated by canonical TCR– ment to signaling by reversed TCR–pMHCI MHCI a2 helices (44). A comparison of the
pMHCI docking is augmented slightly by recognition through binding of Lck demon- CDR3b loops of naïve TRBV17+ (enriched for
CD8 binding and substantially by CD8 de- strated that this polarity resulted in a mis- reversed TCRs) and immune H-2Db–NP366–
livery of Lck. However, reversed-polarity localization of coreceptor-associated Lck and specific repertoires (13) (enriched for canon-
TCR–pMHCI recognition prevents signal- CD3 after pMHCI ligation. Previous studies ical TCRs) revealed a similar frequency of His,
ing caused by CD8 sequestration and mis- have shown that preventing coreceptor seques- Lys, Arg, and Pro usage. However, given that
localization of Lck. We present evidence that tration of Lck in vivo, either by deletion of the TRBV17 gene element encodes an Arg at
the highly conserved TCR–pMHCI docking coreceptors (36) or by mutation of corecep- position 108, all but one of the naïve TRBV17+
polarity is mandated not by binding require- tor-binding sites on Lck (37), facilitates TCR TCRs contained at least one of these residues
ments, but instead by the need to colocal- signaling after recognition of non-MHC ligands. but they were present in only ~40% of the
ize key signaling molecules to enable signal Thus, the association of Lck with the CD8 immune CDR3b sequences. Thus, the relevance
transduction. coreceptor, in addition to dictating the MHC of these residues in driving noncanonical dock-
ligand (5), also dictates the manner in which ing requires further investigation.
Discussion the MHC ligand must be recognized. The inability of the reversed TCRs to sup-
Although a topic of much speculation, there has Other deviations from the typical TCR– port signaling would appear to preclude their
been no definitive demonstration of whether pMHC docking angle exist. Most notably, two ability to support thymic selection. It is possi-
the canonical TCR–pMHC docking polarity po- identified human induced regulatory T cell ble, given the reduced threshold for thymic
tentiates effective TCR binding or signaling, nor TCRs were found to dock on pMHCII in a selection compared with peripheral activation
of the mechanism by which it does so. Although reversed orientation (9). Although broadly (45–48), that an attenuated signal may be
evolutionarily conserved pairwise interactions maintaining the canonical docking polar- sufficient for positive selection. Alternatively,
between TCR and MHC molecules can pre- ity, extreme docking angles over the pMHC such TCRs may mediate selection through
dispose TCRs to MHC recognition in a canonical have been observed in autoreactive human canonical TCR–pMHC recognition and exhibit
orientation (5), our data revealed that the in- CD4+ T cells (38, 39) and in a nonsignaling unconventional pMHC recognition only in the
ability of reversed TCR–pMHC recognition mouse H-2Ld–restricted TCR (10). The cur- periphery.
to support T cell activation was unrelated rent study provides a potential mechanism by In summary, the current study demonstrates
to binding affinity. Instead, these findings which such unconventional pMHC-docking a dual role for coreceptor association of Lck:
support a paradigm in which the polarity polarities may diminish TCR signaling to augmenting signaling mediated by canonical
of TCR recognition of pMHCI is a primary prevent negative selection or abrogate TCR- TCR–pMHC interactions and preventing sig-
determinant of T cell signaling through the mediated signaling. Such exceptions to the naling by unconventional modes of recogni-
colocalization of molecules critical for TCR canonical TCR–pMHC docking “rule” should tion. We hypothesize that, in this way, excessive
signal transduction. be explored to further advance our understand- TCR cross-reactivity is constrained by the num-
Our data align with current knowledge of ing of T cell signaling requirements. ber of signaling-competent binding modalities,
the structural organization of TCR-signaling Although Lck association with coreceptors thereby enhancing the exquisite functional spe-
molecules. The complete ternary structure of a can have a marked effect on TCR signaling, cificity of the TCR–pMHC interaction.
TCR–pMHCII–CD4 complex (25) revealed the some intracellular Lck untethered to corecep-
formation of a 70-Å wide “arch” between the tors is present, highly active (40, 41), and able Materials and Methods
TCR and the CD4 coreceptor, within which to support in vivo signaling (30, 36, 37). Why, TCR transfection of HEK293T cells
the asymmetrically arranged CD3 signaling then, does “free” Lck not allow for signaling by Human embryonic kidney (HEK) 293T cells
complex (28) is positioned. It was postulated reversed TCRs? Critically, Lck can be found (ATCC, #CRL-3216) were maintained in a
that extreme docking polarities (such as a associated with the TCR–CD3 complex in mice humidified incubator at 37°C and 10% CO2.
reversed polarity) would place the bulk of the lacking CD4 and CD8 coreceptors but not in HEK293T cells were plated at 3.5 × 105 cells/
CD3 complex outside of the arch, impeding mice expressing coreceptors (36). We propose well of a six-well plate in 3.5 ml of complete
optimal Lck delivery (25) and T cell signaling. that when coreceptors are expressed, Lck is medium [Dulbecco’s modified Eagle’s medium

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 8 of 13


RES EARCH | R E S E A R C H A R T I C L E

(DMEM; Invitrogen, #11960), 10% fetal bovine into two matched 50-ml conical centrifuge Adoptive transfer of retrogenic CD8+ T cells for
serum (FBS), Hepes, L-glut, and penicillin- tubes. Each experimental sample was first IAV challenge
streptomycin (PenStrep)]. The following day, blocked using Fc block (2.4G2 supernatant CD45.1−CD45.2+CD4−CD8+ GFP/mCherry+
4.2 ml of FuGene 6 HD (Promega) was added to + 1% normal mouse serum + 1% normal rat T cells were isolated by FACS from female
171 ml of OptiMEM (Invitrogen) in an Eppendorf serum). Each pair of tubes was labeled with TCR-retrogenic mice using a BD FACSAria III
tube and incubated for 10 min at room tem- either H-2DbWT–NP366 or H-2DbA89E–NP366 Fusion or BD Influx cell sorter (BD Bioscien-
perature (RT). The FuGene:OptiMEM mixture tetramers conjugated to allophycocyanin (APC) ces). Retrogenic T cells (4 × 103) from each
was then added dropwise to 700 ng of pMIGII for 1 hour at RT. Tetramer-labeled cells were line were resuspended in 200 ml of PBS + 2%
encoding an abTCR sequence and 700 ng of then incubated with anti-APC microbeads FBS and injected intravenously into naïve
pMIGII encoding CD3gde and z subunits and female B6.SJptprca (CD45.1) mice. The next

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
(Miltenyi Biotec) for 30 min at 4°C. Tetramer-
incubated for a further 15 min at RT. The bound cells were positively enriched by passage day, mice were infected with 1 × 104 PFUs of
FuGene-OptiMEM-DNA mixture was then through an LS column (Miltenyi Biotec) HKx31 IAV as described above. At the peak of
added dropwise to each well of a six-well plate mounted on a QuadroMACS (Miltenyi Biotec) the CD8 T cell response (10 days after infection),
and swirled to mix gently before returning magnetic separator. Enriched samples were mice were euthanized and BAL, spleen, and
to the incubator. After 48 hours, the culture then labeled with antibodies (as listed in mLNs were harvested for flow cytometry analy-
medium was aspirated and cells were de- table S5) against cell surface antigens, includ- sis. The gating strategy for identifying donor-
tached from the plate by repeated washing ing Vb9 (TRBV17) and Vb8.3 (TRBV13-1), and derived retrogenic T cells is described in fig. S3.
with fluorescence-activated cell sorting (FACS) stained for viability before analyzing en-
buffer [phosphate-buffered saline (PBS) + tire samples on a Fortessa X20 (BD Bio- In vitro TCR expression by
0.1% bovine serum albumin]. Transfected sciences) or Symphony A3 (BD Biosciences) retroviral transduction
cells were labeled with indicated tetramers for flow cytometer. TCRnull 5KC cells (TCRab−CD4−CD8−) (gift from
1 hour at RT, followed by staining for TCRb P. Marrack) were maintained in a humidified
and viability. Tetramer binding was analyzed Single-cell TCR sequencing and T cell cloning of incubator at 37°C and 10% CO2. 5KC cells were
by flow cytometry using a Fortessa X20 (BD immune TRBV17+ H-2Db–NP366–specific T cells sorted for loss of CD4 to establish a CD4−CD8−
Biosciences). Tetramer-bound cells were enriched and iso- TCR− cell line. HEK293T cells were plated 1 ×
lated as described above for immune reper- 106 cells/dish in a 15-cm tissue culture dish
Mice and influenza A virus infection toires. Samples enriched for tetramer-bound (Sarstedt) in 10 ml of complete DMEM (cDMEM)
Female C57BL/6J (CD45.2) mice were bred cells were run on a FACSAria III (BD Bio- containing DMEM, 10% FBS, Hepes, L-glut, and
and housed at the Monash Animal Research sciences) cell sorter, and live CD19−CD4− PenStrep). The following day, 30 ml of FuGene
Platform (MARP; Monash University, Victoria, CD8+TCRb+CD44+Vb9+ H-2Db–NP366–specific 6 HD (Promega, #E2691) was added to 470 ml
Australia). B6.SJptprca (CD45.1) and Rag1−/− T cells were single-cell sorted using a FACS of OptiMEM (Invitrogen, #31985) in a micro-
(CD45.2) mice were purchased from the Walter AriaIII Fusion (BD Biosciences) into 96-well centrifuge tube and incubated for 10 min at
and Eliza Hall Institute and housed at MARP. polymerase chain reaction (PCR) plates (Eppendorf) RT. The FuGene:OptiMEM mixture was then
Naïve female C57BL/6J mice aged 6 to and stored at −80°C until use. Single-cell multi- added dropwise to 4 mg of pMIGII encoding
10 weeks were briefly anesthetized by iso- plex reverse transcription (RT)–PCR of abTCR an abTCR sequence, along with 4 mg of pPAM-E
flurane inhalation and infected intranasally was performed as previously described (13). and 2 mg of pVSVg, and incubated for a further
with 1 × 104 plaque-forming units (PFUs) of PCR product was sequenced by Sanger se- 15 min at RT. The FuGene-OptiMEM-DNA
HKx31 (H3N2) influenza A virus in 30 ml of quencing at the Monash Micromon Genomics mixture was then added dropwise to the
saline. All animal experimentation was reviewed Facility (Monash University, Victoria, Australia). HEK293T cell culture and swirled to mix
and approved by the Monash University Animal Antigen-specific, P2A-linked TCRab gene con- gently before returning to the incubator.
Ethics Committee (AEC8585, AEC14182, and structs were custom ordered from Genscript The next day, medium containing FuGene:
AEC17693). and cloned into pMIGII (RRID: Addgene_52107; OptiMEM:DNA was replaced with fresh cDMEM
a gift from D.A.A. Vignali) vector and sequenced and incubated for 12 hours. Supernatant was
Tetramer-based magnetic enrichment for to confirm the correct TCR sequence. Plasmids removed approximately every 12 hours five to six
H-2Db–NP366–specific T cells encoding antigen specific P2A-linked TCRab times and filtered through a 0.45-mm syringe-
Tetramer-based magnetic enrichment of epitope- were prepared and propagated for retroviral driven filter. Polybrene (Sigma-Aldrich, #H9268)
specific T cells was performed largely as de- transduction using 10-beta competent Escherichia (6 mg/ml) was added to the supernatant before
scribed previously (49, 50). Spleens and all coli (E. coli) (New England Biolabs, #CR019H), resuspending 5KC cells in filtered, retrovirus-
easily dissected lymph nodes were harvested and plasmids were isolated using the EndoFree containing supernatant. After five to six virus
from 10 naïve female C57BL/6J mice (pre- Maxi Prep Kit (Qiagen, #12362). transfers, 5KC cells were grown to confluency
immune repertoire) or individual mice 10 days in fresh cDMEM and sorted for similar TCRab
after infection with HKx31 IAV intranasally Generation of TCR-retrogenic mice and CD8ab expression. For CD8 transductions,
(immune repertoire). One female C57BL/6J Plasmids encoding TCRa and TCRb genes of only cells expressing an endogenous ratio of
mouse infected with HKx31 between 10 and interest linked by the P2A peptide were or- CD8a:CD8b were sorted.
60 days after infection was also harvested to dered from Genscript and cloned into the
be used as a positive control for tetramer pMIGII or pMIC vector expressing GFP or pERK detection by phospho-flow cytometry
staining of naïve samples. For analysis of mCherry, respectively (RRID: Addgene_52107, For a positive control, 96-well U-bottom plates
the preimmune repertoire, single-cell sus- RRID: Addgene_52114; a gift from D.A.A. were coated overnight with 100 ml of anti-
pensions from 10 naïve mice were pooled Vignali). TCR-retrogenic mice were generated mouse CD3 antibody diluted to 10 mg/ml in
and then evenly divided into eight separate as previously described (12, 23) but with the use PBS overnight. DC2.4 cells (gift from K. Rock)
50-ml conical centrifuge tubes, each for en- of congenically distinct female Rag1−/− (CD45.2) were cultured in cDMEM and maintained in a
richment through an LS column (Miltenyi mice as bone marrow donors and female humidified incubator at 37°C and 10% CO2.
Biotec). For analysis of immune repertoires, B6.SJptprca (CD45.1) as recipients to aid the DC2.4 cells were stained with Aqua Blue Fixable
samples from individual mice were split evenly identification of donor-derived cells. viability stain (Life Technologies), seeded at 1 ×

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 9 of 13


RES EARCH | R E S E A R C H A R T I C L E

105 cells/well in 50 ml of a 96-well plate (Nunc), Lysates were centrifuged at 13,000g for 20 min, counting using a PicoHarp 300 (PicoQuant)
and allowed to adhere for 1 hour in the incu- and 30 ml of supernatant was kept as whole-cell running Symphotime 64 (PicoQuant) fitted
bator. Transduced 5KC cell lines were labeled lysate/input. The remaining lysate was pre- to an Olympus FV1000 laser scanning con-
with Aqua Blue Fixable viability stain (Life Tech- cleared with 20 ml washed Protein G Sepharose focal microscope. A 485-nm pulsed laser was
nologies), plated at 50,000 cells/well, and allowed for 60 min at 4°C. Protein G Sepharose (20 ml) used. Time-correlated single-photon counting
to equilibrate in the incubator for at 1 hour. NP366 was incubated with 10 mg of anti-CD8a (53-6.7) decay curves were fitted to a biexponential
peptide (Genscript) was then added to the DC2.4 for 60 min at 4°C. Antibody-conjugated Protein reconvolution decay model in SymphoTime
cultures at the indicated concentrations and G Sepharose was washed three times with 64 to determine donor (GFP) lifetime. A good
incubated for a further 1 hour. 5KC cells (5 × Pierce® IP Lysis/Wash Buffer. Immunoprecipi- biexponential reconvolution decay curve fit
104) were added to peptide-pulsed DC2.4 cul- was characterized by c2 values close to 1. c2

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
tation was performed by addition of precleared
tures at a final culture volume of 100 ml, and lysate to antibody-conjugated Protein G Sepharose values that deviated by ±1 were uncommon in
briefly centrifuged at 600g for 1 min to en- and incubation for 3 hours at 4°C. Samples were our dataset but were excluded from the analysis.
courage contact. At the indicated time points, centrifuged, washed five times in Pierce IP Lysis/ To detect FRET, the amplitude weighted donor
100 ml of prewarmed, 2× Lyse/Fix buffer (BD Wash Buffer and once in 50 mM Tris-HCl (pH average lifetime (tAvAmp) was used because this
Biosciences, #558049) was added to each well, 7.4). Elution was performed by boiling in 3× reflects the quenching of the donor caused by
and the mixture was incubated at 37°C for Laemmli buffer with 50 mM dithiothreitol the FRET process. To determine FRET between
10 min. Fixed cells were washed twice in 200 ml (95°C, 5 min). Samples were centrifuged and CD8b-mCherry and CD3z-GFP, we measured
of PBS. Fixed cell pellets were permeabilized by supernatant containing immunoprecipitated tAvAmp at the immunological synapse where the
the addition of 100 ml of −20°C Perm Buffer III CD8 was collected. Samples were resolved on T cell interacted with a dendritic cell by selecting
(BD Biosciences) and then stored overnight at 10 to 14% SDS–polyacrylamide gel electropho- a region of interest in the FLIM image. Non-
−20°C. The following day, fixed and permeabi- resis under reducing conditions at 100 V for 3 to synaptic tAvAmp was also measured from the
lized cells were washed with 200 ml of FACS 4 hours. Proteins were wet transferred onto same cell as an internal control and was used to
buffer and stained with cell surface anti- polyvinylidene difluoride membranes at con- calculate the percentage change in tAvAmp of
bodies and a rabbit anti-phospho p44 MAPK stant 300 mA for 2 hours. Membranes were GFP at the synapse (SynDtAvAmp). For analysis,
(Cell Signaling Technology) for 1 hour on ice. blocked for 60 min at RT in 5% skim milk (w/v) n-exponential reconvolution using the n = 2
Cells were washed with FACS buffer and then in 0.1% (v/v) TBS-Tween before incubation model parameter was used for donor curve
stained with an anti-rabbit PE F(ab′)2 frag- with specific antibody (1:3000) overnight at fitting.
ment (Cell Signaling Technology) for 30 min. 4°C. Membranes were washed three times in
Cells were washed twice in FACS buffer 0.1% (v/v) TBS-Tween and probed with rel- 2D micropipette adhesion frequency
before running the samples on a BD Fortessa evant horseradish peroxidase–conjugated sec- assay (2D-MP)
X20 or BD Symphony A3 (BD Biosciences) flow ondary antibody as indicated for 60 min at RT. The relative 2D affinity of the H-2Db–restricted
cytometer. After three washes in 0.1% (v/v) TBS-Tween, nucleoprotein epitope (NP366; ASNENMETM)
enhanced chemiluminesence substrate was TCRs expressed in 5KC hybridoma cell lines was
IL-2 ELISA added to membranes for 2 min. Blots were measured by the previously described 2D-MP
DC2.4 cells were seeded at 1 × 105 cells/well visualized on a ChemiDoc XRS+ (Bio-Rad (14–18). In short, human red blood cells (hRBCs)
in 100 ml of a flat-bottom 96-well plate (Nunc) Laboratories). coated with Biotin-LC-NHS (BioVision) strepta-
and allowed to adhere for 1 hour in the in- vidin (Thermo Fisher Scientific), coated with
cubator. Transduced 5KC cell lines were plated Confocal and FLIM for analysis of FRET biotinylated H-2Db–NP366D227K, and mounted
at a concentration of 1 × 106 cells/ml and al- DC2.4 cells were maintained in phenol-free on a glass micropipette. 5KC hybridomas ex-
lowed to equilibrate in the incubator for at cDMEM (Invitrogen 31053), seeded in 35-mm pressing B13.C1, B17.R1, B17.R2, or B17.C1 TCRs
least 1 hour. NP366 peptide was then added to Fluorodish (World Precision Instruments) cell were mounted on opposing glass micropipettes.
the DC2.4 cultures at the indicated concen- culture dishes at 1 × 105 cells/dish in 1 ml of The adhesion frequency between the TCR of
trations and incubated for a further 1 hour. culture, and incubated overnight at 37°C and interest and pMHC aspirated on opposing
5KC cells (5 × 104) were added to peptide- 10% CO2. The following day, DC2.4 cells were micropipettes was observed using an inverted
pulsed DC2.4 cultures or to the anti-CD3–coated washed three times with prewarmed PBS and microscope. An electronically controlled piezo-
wells at a final culture volume of 200 ml and then stained with 5 mM CellTrace Violet (CTV; electric actuator brought the opposing cells into
briefly centrifuged at 600g for 1 min to en- Invitrogen C34557) for 30 min. CTV was then contact and repeated a T cell contact and
courage contact. After 16 hours of coincuba- aspirated from the dish and the labeled cells separation cycle with the pMHC-coated RBCs
tion, plates were centrifuged at 935g for 3 min were washed three times with prewarmed, 50 times while keeping the contact area (Ac)
to pellet cells and supernatant was aspirated, phenol-free cDMEM. Labeled DC2.4 cells were and time (t) constant. Upon retraction of the
transferred to a new plate, and stored at −20°C incubated with 10 mM NP366 peptide for 1 hour. T cell, adhesion (binding of TCR–pMHC) was
until required. IL-2 secreted in the supernatant 5KC hybridoma cells expressing TCR and the observed as a distention of the RBC membrane,
was measured using the BD IL-2 mouse ELISA FRET pairs CD3z-GFP and CD8b-mCherry allowing for quantification of the adhesion
kit (BD Biosciences, #555148) according to the were plated at a density of 1 × 106 cells/ml in a frequency (Pa) at equilibrium. Surface pMHC
manufacturer’s instructions. Absorbance was six-well dish and equilibrated in the incuba- (ml) and TCRb (mr) densities were determined
measured at 450 nm using a CLARIOstar plate tor for at least 1 hour before use. For imaging, by flow cytometry using an anti-TCRb PE anti-
reader (BMG LabTech). DC2.4 cells were brought into focus and then body (BD Biosciences, #H57-597) and an anti-
100 ml of T cells (~1 × 105 cells) was added to H2Db antibody (clone 28-14-8; eBioscience),
Immunoprecipitation and immunoblotting the culture dish containing labeled DC2.4 cells both at saturating concentrations, along with
5KC T cells (1.5 to 2 × 107) were lysed for and imaged by confocal microscopy up to BD QuantiBRITE PE beads for standardization
60 min at 4°C in 300 ml of Pierce IP Lysis/Wash 20 min later using an Olympus FV1000 run- (BD Biosciences). The calculation of molecules
Buffer (25 mM Tris-HCl pH 7.4, 150 mM NaCl, ning Fluoview software (Olympus). The fluo- per area was determined by dividing the num-
1% NP-40, 1 mM EDTA, and 5% glycerol) and rescent lifetime of the donor molecule GFP was ber of TCRs and pMHCs per cell by the respective
1× Halt Protease Inhibitor Cocktail (Pierce). measured by time-correlated single-photon surface areas. The relative 2D affinities were

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 10 of 13


RES EARCH | R E S E A R C H A R T I C L E

calculated using the following equation: AcKa = N-(cap biotinyl) (sodium salt)), and 0.5% TetraSpeck microspheres (Invitrogen) were
–ln[1 – Pa(1)]/mrml. PEG5000-PE (1,2-distearoyl-sn-glycero-3- embedded onto the lipid bilayer.
phosphoethanolamine-N-[methoxy(poly-
Biomembrane force probe assay ethylene glycol)-5000] (ammonium salt) Single-molecule localization microscopy
Bond lifetime measurements under force were (mol%; all available from Avanti Polar Lipids with dSTORM
captured using the biomembrane force probe (DOPC, 850375C), (DGS-NTA(Ni), 790404C), For single-molecule imaging, an imaging buf-
(BFP) assay. Procedures for coupling pMHC to (Biotinyl-Cap-PE, 870273C), (PEG5000-PE, fer consisting of TN buffer [50 mM Tris-HCl
glass beads have been described previously 880220C) was created by vesicle extrusion, as pH 8.0, 10 mM NaCl), oxygen scavenger system
(21). Briefly, hRBCs were first biotinylated described previously (35). Extruded liposomes GLOX [0.5 mg/ml glucose oxidase (Sigma-
Aldrich, #G2133); 40 mg/ml catalase (Sigma-

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
with EZ-link NHS-PEG-Biotin (Thermo Fisher were added to eight-well chambers at a ratio
Scientific) and then reacted to streptavidin. of 1:5 with Milli-Q water (with 10 mM of Aldrich, #C-100); and 10% w/v glucose], and
Borosilicate beads were cleaned, silanized, and CaCl2) and incubated for 30 min at RT before 10 mM 2-aminoethanethiol (MEA; Sigma-
reacted to streptavidin–maleimide (Sigma- gently rinsing with PBS repeatedly. During Aldrich, #M6500) was used. dSTORM image
Aldrich, St. Louis, MO). Streptavidin beads washing steps, the disruption of the lipid sequences were acquired on a total internal
were then coated with H-2Db–NP366 or H- bilayer was minimized by retaining ~200 ml reflection fluorescence microscope (commer-
2Db–D227K-NP366. pMHC monomer–coated of PBS in each well. Lateral mobility of the cial setup, Zeiss Elyra) with a 100× oil-immersion
beads (which serve as a force probe) were freshly prepared SLB was confirmed by adding objective (numerical aperture = 1.46). For Alexa
then placed on the apex of an hRBC that was 10 mg/ml of fluorescently labeled streptavidin Fluor 647, 633 nm (15 mW) laser illumination
aspirated onto a glass micropipette. The posi- (Invitrogen, #S11223) and monitoring fluores- was used, along with a 405-nm activator laser
tion of the edge of the bead was tracked by a cence recovery after photobleaching (FRAP) as (15 mW) for imaging. Time series of 10,000 frames
high-resolution camera (1600 frames/s) with described elsewhere (35). Excess Ca2+ ions in were acquired per sample with a cooled, electron-
< 3-nm displacement precision. The position the system were removed by adding 0.5 mM of multiplying charge-coupled device camera (iXon
of the edge of the bead was tracked by a high- EDTA, followed by gently rinsing with Milli-Q DU-897D; Andor) with an exposure time of
resolution camera (1600 frames/s) with <3 nm water. The NTA groups in DGS-NTA(Ni) lip- 50 ms. Image processing, including fiducial
displacement precision using a Zeiss micro- ids were then recharged by adding 1 mM of marker–based drift correction and generation
scope. The cell was brought into contact with NiCl2 for 15 min and gently rinsed with PBS of x–y particle coordinates for each molecule
the pMHC-coated bead:RBC; the cell was then repeatedly. Finally, SLB was blocked with detected in the acquisition, were performed
retracted and held at the desired force by the 5% bovine serum albumin in PBS for 15 min using Zeiss Zen software (Zen Black 2012
computer-controlled piezoelectric actuator until at RT. version).
bond dissociation occurred. If adhesion was
present, it was detected by tensile force caused T cell activation on SLB Expression, refolding, purification,
by stretching of the hRBC and tracked by To decorate the SLB with biotinylated pMHC crystallization, and structure determination
displacement of the pMHC-coated bead. The and His-tagged ICAM-1, 100 mg/ml of strepta- DNA fragments encoding the TRAV and TRBV
bond lifetime was measured from the time the vidin (Life Technologies) was incubated for segments of the B17.C1 TCR were purchased,
desired force was reached to the time it took 20 min and rinsed with PBS. Afterward, 500 ng/ml codon optimized (Genscript), amplified, and
the cell to disengage with the bead, which was of biotinylated H-2Db–NP366 (pMHCI) (12) and cloned separately into a previously reported
visualized as the RBC retracted and the bead 200 ng/ml of His-tagged ICAM-1 (Sino Biological) expression vector fused to human Ca and Cb,
returned to its starting position before the prepared in 5% bovine serum albumin in PBS was respectively (12). The B17.R2 TCR was gener-
start of the next cycle. Repeated cycles (known added to the lipid bilayer and incubated for 1 hour ated by mutagenesis of the B17.R1 TCR. The
as force-clamp cycles) could be performed. at RT. SLB was gently rinsed with PBS for several B13.C1 TCRab construct was purchased, codon
Multiple forces were collected for each ligand times to remove excess unbound proteins. Before optimized for mammalian cell expression
(pMHC-coated beads) and were shown in five adding T cell hybridomas, SLB was incubated with (Genscript), and cloned into a pHLsec vector.
to eight bins as the mean ± SEM. For an op- warm DMEM culture medium (37°C) for 30 min. Each TCR is a chimeric construct of mouse
timal response to antigen, bond lifetimes in- T cell hybridomas were then allowed to activate on variable and human constant domain. The
crease with increasing force before reaching a the lipid bilayer for 5 min at 37°C, followed by plasmids constructs were confirmed by DNA
peak and then decrease, which is referred to as immediate cell fixation with 4% paraformaldehyde sequencing. The B13.C1 TCR was produced in
a catch bond. When increasing force leads (vol/vol) in PBS for 15 min at RT. Excess fixatives HEK293S cells as a soluble protein and pu-
to decreasing bond lifetime, this is called a were removed by rinsing with PBS. rified through its His tag over an affinity
slip bond. column and size exclusion chromatography.
Immunostaining of 5KC T cells Soluble H-2Db WT or mutant heavy chain
SLB preparation Before immunostaining 5KC T cells were per- (generated by site-directed mutagenesis), the
Glass coverslips of a 0.17 mm thickness were meabilized with 0.1% Triton X-100 (vol/vol) human and mouse b2m, the B17.R1, B17.R2,
first cleaned with 1 M KOH for 10 min and (Sigma-Aldrich) for 15 min and then rinsed and the B17.C1 TCRs a and b chains were ex-
then rinsed with Milli-Q water and placed in with PBS. Cells were then blocked with 5% pressed separately in E. coli (Novagen, #70236)
100% ethanol for 20 min. These glass cover- bovine serum albumin in PBS for 1 hour. T cells as inclusion bodies, then subsequently solubi-
slips were then plasma cleaned for 5 min. were stained with primary antibodies reactive lized, refolded, and purified as previously
Afterward, the coverslips were adhered to against the TCRb subunit and conjugated to reported (12).
eight-well silicon chambers (ibidi, #80841). A Alexa Fluor 647 fluorophore (BioLegend). Cells Crystals of the ternary B17.R2–H-2Db–NP366
liposome solution of 1 mg/ml with a lipid were probed with primary antibodies for 1 hour complex were grown by the hanging-drop,
ratio of 96.5% DOPC (1,2-dioleoyl-sn-glycero-3- at RT. After antibody staining, samples were vapor-diffusion method at 20°C with a protein/
phosphocholine), 2% DGS-NTA(Ni) (1,2-dioleoyl-sn- repeatedly rinsed with PBS to remove excess reservoir drop ratio of 1:1 at 3 mg/ml in 10 mM
glycero-3-[(N-(5-amino-1-carboxypentyl)iminodiacetic unbound antibodies and fluorophores. Postfixa- Tris-HCl, pH 8.0, 150 mM NaCl using 20% PEG
acid)succinyl] (nickel salt)), 1% Biotinyl-Cap-PE tion was performed using 4% paraformaldehyde 3350, 0.2 M K/Na/tartrate, and 0.1 M Bis-tris-
(1,2-dioleoyl-sn-glycero-3-phosphoethanolamine- (vol/vol) in PBS for 15 min. Before imaging, 0.1-mm propane buffer, pH 6.5. Crystals of the ternary

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 11 of 13


RES EARCH | R E S E A R C H A R T I C L E

B17.C1–H-2Db–NP366 complex were grown by independent experiments were conducted (n = 33, 437–441 (2012). doi: 10.1016/j.it.2012.05.006;
the hanging-drop, vapor-diffusion method at 2) in duplicate. GraphPad Prism software was pmid: 22771139
6. S. Rangarajan, R. A. Mariuzza, T cell receptor bias for MHC: Co-
20°C with a protein/reservoir drop ratio of 1:1 used for data analysis with the 1:1 Langmuir evolution or co-receptors? Cell. Mol. Life Sci. 71, 3059–3068
at 8 mg/ml in 10 mM Tris-HCl, pH 8.0, 150 mM binding model. (2014). doi: 10.1007/s00018-014-1600-9; pmid: 24633202
NaCl using 16% PEG 3350, 0.2 M potassium 7. N. L. La Gruta, S. Gras, S. R. Daley, P. G. Thomas, J. Rossjohn,

thiocyanide, 4% ethylene glycol, and 0.1 M Bis- Statistical analyses Understanding the drivers of MHC restriction of T cell
receptors. Nat. Rev. Immunol. 18, 467–478 (2018).
tris-propane buffer, pH 7.6. The crystals were Statistical analysis was performed with one-way doi: 10.1038/s41577-018-0007-5; pmid: 29636542
soaked in a cryoprotectant solution containing ANOVA or two-way ANOVA when comparing 8. P. Zareie, C. Farenc, N. L. La Gruta, MHC restriction: Where are
we now? Viral Immunol. 33, 179–187 (2020). doi: 10.1089/
the mother liquor solution with the PEG con- multiple groups as indicated. For data obtained vim.2019.0195; pmid: 32286180

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
centration increased to 30% (w/v) and then over multiple days, we considered the possibility 9. D. X. Beringer et al., T cell receptor reversed polarity
flash-frozen in liquid nitrogen. of day-to-day variation, which we accounted for recognition of a self-antigen major histocompatibility complex.
For the B17.C1–H-2Db–NP366 structure, de- as a “nuisance factor” in the two-way ANOVA. Nat. Immunol. 16, 1153–1161 (2015). doi: 10.1038/ni.3271;
pmid: 26437244
spite successfully reproducing and testing nu- We did not find a statistically significant effect 10. J. J. Adams et al., T cell receptor signaling is limited by docking
merous crystals, only a single crystal gave a of day-to-day variation in our analysis. Where geometry to peptide-major histocompatibility complex.
diffraction at high resolution (i.e., <5 Å) and appropriate, we also performed paired-samples Immunity 35, 681–693 (2011). doi: 10.1016/j.
immuni.2011.09.013; pmid: 22101157
was rapidly destroyed by radiation damage. t tests, as indicated in the figure legends, which 11. B. C. Sim, L. Zerva, M. I. Greene, N. R. J. Gascoigne, Control of
Datasets were collected on the MX1 (51) and pair the dataset by the day in which they were MHC restriction by TCR Valpha CDR1 and CDR2. Science 273,
MX2 (52) beamline at the Australian Synchro- obtained. In the figures, P-values are denoted as 963–966 (1996). doi: 10.1126/science.273.5277.963;
pmid: 8688082
tron (Clayton, Victoria, Australia), processed *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, and ****P ≤ 12. S. Gras et al., Reversed T cell receptor docking on a major
using XDS software (53), and scaled using 0.0001. Ripley’s K analysis was used to quantify histocompatibility class I complex limits involvement in the
Aimless software (54) from the CCP4 suite the degree of clustering in a population of immune response. Immunity 45, 749–760 (2016).
doi: 10.1016/j.immuni.2016.09.007; pmid: 27717799
(55). The data cutoff used was CC1/2 > 0.5% molecules compared with a complete spatial 13. T. Cukalac et al., Paired TCRab analysis of virus-specific CD8
and I/s(I) > 1.5 (56). The structures were randomness (62). For each molecule registered (+) T cells exposes diversity in a previously defined ‘narrow’
determined by molecular replacement using as a localization event, Ripley’s (K) calculates repertoire. Immunol. Cell Biol. 93, 804–814 (2015).
doi: 10.1038/icb.2015.44; pmid: 25804828
the PHASER program (57), with the B17.R1 the number of neighboring localizations with- 14. J. Huang et al., The kinetics of two-dimensional TCR and
TCR from the previous B17.R1–H-2Db–NP366 in a given radius (r) corrected by the total pMHC interactions determine T-cell responsiveness.
complex used as the search model for the density of localizations, providing information Nature 464, 932–936 (2010). doi: 10.1038/nature08944;
pmid: 20357766
TCR [PDB accession code 5SWZ (12)]. Manual on the degree of spatial clustering of molecules
15. J. J. Sabatino Jr.., J. Huang, C. Zhu, B. D. Evavold, High
model building was conducted using Coot soft- within a region of interest. In this study, we prevalence of low affinity peptide-MHC II tetramer-negative
ware (58), followed by maximum-likelihood performed Ripley’s K analysis on single-molecule effectors during polyclonal CD4+ T cell responses. J. Exp. Med.
refinement with the Buster program (59). images using a previously published algorithm 208, 81–90 (2011). doi: 10.1084/jem.20101574;
pmid: 21220453
The final model has been validated using the (27). To determine the average clustering value 16. R. J. Martinez, R. Andargachew, H. A. Martinez, B. D. Evavold,
PDB validation website, and the final refine- within a ROI, this algorithm used a linearized Low-affinity CD4+ T cells are major responders in the primary
ment statistics are summarized in table S1. form of Ripley’s (K), the L(r)−r. Here, r is defined immune response. Nat. Commun. 7, 13848 (2016).
doi: 10.1038/ncomms13848; pmid: 27976744
The electron density at the interface was well as the spatial scale radius. In a complete spatial 17. E. M. Kolawole, R. Andargachew, B. Liu, J. R. Jacobs,
defined despite slightly above average R factors. randomness, L(r)−r value = 0. Similarly, a posi- B. D. Evavold, 2D kinetic analysis of TCR and CD8 coreceptor
The high R factors are caused by poor electron tive value for L(r)−r at a given r radius indicated for LCMV GP33 epitopes. Front. Immunol. 9, 2348 (2018).
doi: 10.3389/fimmu.2018.02348; pmid: 30374353
density for some parts of H2Db a3 domain, the a clustering of localization events, whereas a 18. R. Andargachew, R. J. Martinez, E. M. Kolawole, B. D. Evavold,
b2m, as well as the C terminus of TCRa con- negative value represented a dispersed spatial CD4 T cell affinity diversity is equally maintained during acute
stant domain. However, these regions are distal organization (negative clustering). The start and chronic infection. J. Immunol. 201, 19–30 (2018).
doi: 10.4049/jimmunol.1800295; pmid: 29777029
from the ligand interface. All molecular graphics (0 nm), end (500 nm), and step size (10 nm) 19. T. A. Potter, T. V. Rajan, R. F. Dick 2nd, J. A. Bluestone,
representations were created using PyMol (The for r in the algorithm were user defined. The Substitution at residue 227 of H-2 class I molecules abrogates
PyMOL Molecular Graphics System, version 2.0; maximum L(r)−r value derived from L(r)−r recognition by CD8-dependent, but not CD8-independent,
Schrödinger, LLC). The structures have been versus r graph corresponded to the spatial cytotoxic T lymphocytes. Nature 337, 73–75 (1989).
doi: 10.1038/337073a0; pmid: 2462676
deposited into the PDB database (B17.R2 TCR– scale (r) at which the highest degree of clus- 20. L. Limozin et al., TCR-pMHC kinetics under force in a cell-free
H-2Db–NP366, PDB 7JWI; B17.C1 TCR–H-2Db– tering of localizations was observed. system show no intrinsic catch bond, but a minimal encounter
NP366, PDB 7JWJ). duration before binding. Proc. Natl. Acad. Sci. U.S.A. 116,
16943–16948 (2019). doi: 10.1073/pnas.1902141116;
pmid: 31315981
SPR experiments RE FERENCES AND NOTES
21. B. Liu, W. Chen, B. D. Evavold, C. Zhu, Accumulation of
1. J. Rossjohn et al., T cell antigen receptor recognition of
SPR experiments were conducted at 25°C on antigen-presenting molecules. Annu. Rev. Immunol. 33,
dynamic catch bonds between TCR and agonist peptide-MHC
triggers T cell signaling. Cell 157, 357–368 (2014).
the BIAcore T200 and BIAcore 3000 instru- 169–200 (2015). doi: 10.1146/annurev-immunol-032414-
doi: 10.1016/j.cell.2014.02.053; pmid: 24725404
ment (GE Healthcare, Buckinghamshire, UK) 112334; pmid: 25493333
2. S. Dai et al., Crossreactive T cells spotlight the germline rules 22. J. Hong et al., Force-regulated in situ TCR–peptide-bound MHC
with 10 mM Tris-HCl, pH 8, 150 mM NaCl, for ab T cell-receptor interactions with MHC molecules. class II kinetics determine functions of CD4+ T cells.
0.005% surfactant P20, and 0.5% bovine serum Immunity 28, 324–334 (2008). doi: 10.1016/ J. Immunol. 195, 3557–3564 (2015). doi: 10.4049/
j.immuni.2008.01.008; pmid: 18308592 jimmunol.1501407; pmid: 26336148
albumin buffer. The 12H8 antibody was bound
3. D. Feng, C. J. Bond, L. K. Ely, J. Maynard, K. C. Garcia, 23. J. Holst et al., Generation of T-cell receptor retrogenic mice.
to all flow cells of a CM5 sensor chip through Structural evidence for a germline-encoded T cell receptor- Nat. Protoc. 1, 406–417 (2006). doi: 10.1038/nprot.2006.61;
amine coupling (60), and all TCRs subsequently major histocompatibility complex interaction ‘codon’. pmid: 17406263
bound to the antibody. A negative control (LC13 Nat. Immunol. 8, 975–983 (2007). doi: 10.1038/ni1502; 24. J. White, A. Pullen, K. Choi, P. Marrack, J. W. Kappler, Antigen
pmid: 17694060 recognition properties of mutant V beta 3+ T cell receptors are
TCR) (61) was used on each SPR chip bound to consistent with an immunoglobulin-like structure for the
4. B. D. Stadinski et al., A role for differential variable gene pairing
flow cell 1. Each cycle of TCR injection and in creating T cell receptors specific for unique major receptor. J. Exp. Med. 177, 119–125 (1993). doi: 10.1084/
pMHC injection was regenerated with Actisep histocompatibility ligands. Immunity 35, 694–704 (2011). jem.177.1.119; pmid: 8380294
doi: 10.1016/j.immuni.2011.10.012; pmid: 22101158 25. Y. Yin, X. X. Wang, R. A. Mariuzza, Crystal structure of a
(Sterogene). pMHC was flowed over the surface
5. F. Van Laethem, A. N. Tikhonova, A. Singer, MHC restriction is complete ternary complex of T-cell receptor, peptide-MHC, and
with a concentration range of 0.78 to 200 mM at imposed on a diverse T cell receptor repertoire by CD4 and CD4. Proc. Natl. Acad. Sci. U.S.A. 109, 5405–5410 (2012).
a flow rate of 5 or 30 ml/min. A minimum of two CD8 co-receptors during thymic selection. Trends Immunol. doi: 10.1073/pnas.1118801109; pmid: 22431638

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 12 of 13


RES EARCH | R E S E A R C H A R T I C L E

26. R. Wang, K. Natarajan, D. H. Margulies, Structural basis of the 1504–1514.e7 (2020). doi: 10.1016/j.celrep.2020.01.008; 58. P. Emsley, B. Lohkamp, W. G. Scott, K. Cowtan, Features and
CD8 a b/MHC class I interaction: Focused recognition orients pmid: 32023465 development of Coot. Acta Crystallogr. D Biol. Crystallogr. 66,
CD8 b to a T cell proximal position. J. Immunol. 183, 42. D. L. Wiest et al., Regulation of T cell receptor expression in 486–501 (2010). doi: 10.1107/S0907444910007493;
2554–2564 (2009). doi: 10.4049/jimmunol.0901276; immature CD4+CD8+ thymocytes by p56lck tyrosine kinase: pmid: 20383002
pmid: 19625641 Basis for differential signaling by CD4 and CD8 in immature 59. G. Bricogne et al., “BUSTER version 1.10.0” (The Debian
27. S. V. Pageon, P. R. Nicovich, M. Mollazade, T. Tabarin, K. Gaus, thymocytes expressing both coreceptor molecules. J. Exp. Project, 2011).
Clus-DoC: A combined cluster detection and colocalization Med. 178, 1701–1712 (1993). doi: 10.1084/jem.178.5.1701; 60. N. A. Borg et al., The CDR3 regions of an immunodominant
analysis for single-molecule localization microscopy data. pmid: 8228817 T cell receptor dictate the ‘energetic landscape’ of peptide-
Mol. Biol. Cell 27, 3627–3636 (2016). doi: 10.1091/mbc.e16-07- 43. H. Sanabria et al., Resolving dynamics and function of transient MHC recognition. Nat. Immunol. 6, 171–180 (2005).
0478; pmid: 27582387 states in single enzyme molecules. Nat. Commun. 11, 1231 doi: 10.1038/ni1155; pmid: 15640805
28. D. Dong et al., Structural basis of assembly of the human T cell (2020). doi: 10.1038/s41467-020-14886-w; pmid: 32144241 61. L. Kjer-Nielsen et al., A structural basis for the selection of
receptor-CD3 complex. Nature 573, 546–552 (2019). 44. J. Lu et al., Molecular constraints on CDR3 for thymic selection dominant alphabeta T cell receptors in antiviral immunity.

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
doi: 10.1038/s41586-019-1537-0; pmid: 31461748 of MHC-restricted TCRs from a random pre-selection Immunity 18, 53–64 (2003). doi: 10.1016/S1074-7613(02)
29. T. Żal, M. A. Żal, N. R. J. Gascoigne, Inhibition of T cell repertoire. Nat. Commun. 10, 1019 (2019). doi: 10.1038/ 00513-7; pmid: 12530975
receptor-coreceptor interactions by antagonist ligands s41467-019-08906-7; pmid: 30833553 62. B. D. Ripley, Tests of “randomness” for spatial point patterns.
visualized by live FRET imaging of the T-hybridoma 45. G. M. Davey et al., Preselection thymocytes are more sensitive J. R. Stat. Soc. B 41, 368–374 (1979). doi: 10.1111/j.2517-
immunological synapse. Immunity 16, 521–534 (2002). to T cell receptor stimulation than mature T cells. J. Exp. Med. 6161.1979.tb01091.x
doi: 10.1016/S1074-7613(02)00301-1; pmid: 11970876 188, 1867–1874 (1998). doi: 10.1084/jem.188.10.1867;
pmid: 9815264 AC KNOWLED GME NTS
30. A. N. Tikhonova et al., ab T cell receptors that do not undergo
46. K. A. Hogquist et al., T cell receptor antagonist peptides induce We wish to dedicate this work to the memory of our coauthor
major histocompatibility complex-specific thymic selection
positive selection. Cell 76, 17–27 (1994). doi: 10.1016/0092- and close colleague, Professor Katharina Gaus. We thank
possess antibody-like recognition specificities. Immunity 36,
8674(94)90169-4; pmid: 8287475 P. Marrack (National Jewish Health, Colorado) and D. Vignali
79–91 (2012). doi: 10.1016/j.immuni.2011.11.013;
47. K. A. Hogquist, S. C. Jameson, The self-obsession of T cells: (University of Pittsburgh) for provision of reagents;
pmid: 22209676
How TCR signaling thresholds affect fate ‘decisions’ and D. Jayasinghe and F. Weide for technical assistance; P. Harrison
31. P. P. Yachi, J. Ampudia, N. R. J. Gascoigne, T. Zal,
effector function. Nat. Immunol. 15, 815–823 (2014). from the Monash Bioinformatics Platform for assistance with
Nonstimulatory peptides contribute to antigen-induced CD8-T
doi: 10.1038/ni.2938; pmid: 25137456 statistical analyses; staff at Monash FlowCore, Monash Animal
cell receptor interaction at the immunological synapse.
48. B. Lucas, I. Stefanová, K. Yasutomo, N. Dautigny, Resource Platform, Monash Macromolecular Crystallization
Nat. Immunol. 6, 785–792 (2005). doi: 10.1038/ni1220;
R. N. Germain, Divergent changes in the sensitivity of maturing Facility, Monash Micro Imaging Platforms; and the ANSTO
pmid: 15980863
T cells to structurally related ligands underlies formation of a Australian Synchrotron MX1/MX2 beamline scientists.
32. J. Casas et al., Ligand-engaged TCR is triggered by Lck not
useful T cell repertoire. Immunity 10, 367–376 (1999). Funding: This work was supported by the Australian Research
associated with CD8 coreceptor. Nat. Commun. 5, 5624 (2014).
doi: 10.1016/S1074-7613(00)80036-9; pmid: 10204492 Council (ARC) (DP170103631 to N.L.L. and S.G., DP201102776 to
doi: 10.1038/ncomms6624; pmid: 25427562
33. J. M. Turner et al., Interaction of the unique N-terminal region 49. N. L. La Gruta et al., Primary CTL response magnitude in mice N.L.L., CE140100011 to J.R. and K.G.); the National Health and
of tyrosine kinase p56lck with cytoplasmic domains of CD4 is determined by the extent of naive T cell recruitment and Medical Research Council of Australia (NHMRC) (APP1182086
and CD8 is mediated by cysteine motifs. Cell 60, 755–765 subsequent clonal expansion. J. Clin. Invest. 120, 1885–1894 to N.L.L. and APP1155162 to K.G.); the NSW Cancer Council
(1990). doi: 10.1016/0092-8674(90)90090-2; pmid: 2107025 (2010). doi: 10.1172/JCI41538; pmid: 20440073 (APP1128488 to K.G.); the Singapore Ministry of Health’s
34. M. E. Birnbaum et al., Molecular architecture of the ab T cell 50. J. J. Moon et al., Naive CD4(+) T cell frequency varies for National Medical Research Council (CBRG/0097/2015 to
receptor-CD3 complex. Proc. Natl. Acad. Sci. U.S.A. 111, different epitopes and predicts repertoire diversity and N.R.J.G.); and the Singapore Ministry of Education (2014–T2–1–
17576–17581 (2014). doi: 10.1073/pnas.1420936111; response magnitude. Immunity 27, 203–213 (2007). 136 to N.R.J.G.). N.L.L. is supported by an ARC Future
pmid: 25422432 doi: 10.1016/j.immuni.2007.07.007; pmid: 17707129 Fellowship, S.G. by an NHMRC Senior Research Fellowship, and
35. S. V. Pageon et al., Functional role of T-cell receptor 51. N. P. Cowieson et al., MX1: A bending-magnet crystallography J.R. by an ARC Laureate Fellowship. Author contributions:
nanoclusters in signal initiation and antigen discrimination. beamline serving both chemical and macromolecular Conceptual and experimental design: N.L.L., P.Z., J.R., S.G.,
Proc. Natl. Acad. Sci. U.S.A. 113, E5454–E5463 (2016). crystallography communities at the Australian Synchrotron. K.G., B.D.E., and N.R.J.G.; recombinant protein expression,
doi: 10.1073/pnas.1607436113; pmid: 27573839 J. Synchrotron Radiat. 22, 187–190 (2015). doi: 10.1107/ structure determination, and SPR analyses: C.S., C.F., J.P.,
36. F. Van Laethem et al., Deletion of CD4 and CD8 coreceptors S1600577514021717; pmid: 25537608 S.G., and J.R.; generation of in vitro cell lines, retrogenic mice,
permits generation of alphabetaT cells that recognize antigens 52. D. Aragão et al., MX2: A high-flux undulator microfocus FLIM-FRET imaging, and assays of T cell function: P.Z. with help
independently of the MHC. Immunity 27, 735–750 (2007). beamline serving both the chemical and macromolecular from C.M.J., Q.W., L.W., X.Y.X.S., A.N., C.B., and A.J.F.; 2D
doi: 10.1016/j.immuni.2007.10.007; pmid: 18023370 crystallography communities at the Australian Synchrotron. measures of binding: J.R.J., E.M.K., and B.D.E.; SMLM: S.D.G.
37. F. Van Laethem et al., Lck availability during thymic selection J. Synchrotron Radiat. 25, 885–891 (2018). doi: 10.1107/ and K.G.; data analysis: all authors; manuscript writing: P.Z.,
determines the recognition specificity of the T cell repertoire. S1600577518003120; pmid: 29714201 N.L.L., S.G., and J.R. with contributions from all other authors.
Cell 154, 1326–1341 (2013). doi: 10.1016/j.cell.2013.08.009; 53. W. Kabsch, XDS. Acta Crystallogr. D Biol. Crystallogr. 66, Competing interests: The authors declare no competing
pmid: 24034254 125–132 (2010). doi: 10.1107/S0907444909047337; interests. Data and materials availability: Structures for the
pmid: 20124692 B17.R2 TCR–H-2Db–NP366 and B17.C1 TCR–H-2Db–NP366
38. M. Hahn, M. J. Nicholson, J. Pyrdol, K. W. Wucherpfennig,
complexes have been deposited into the PDB database (B17.R2
Unconventional topology of self peptide-major 54. P. R. Evans, G. N. Murshudov, How good are my data and what
TCR–H-2Db–NP366, PDB 7JWI; B17.C1 TCR–H-2Db–NP366, PDB
histocompatibility complex binding by a human autoimmune is the resolution? Acta Crystallogr. D Biol. Crystallogr. 69,
7JWJ). All other data are available in the main text or the
T cell receptor. Nat. Immunol. 6, 490–496 (2005). 1204–1214 (2013). doi: 10.1107/S0907444913000061;
supplementary materials.
doi: 10.1038/ni1187; pmid: 15821740 pmid: 23793146
39. D. K. Sethi et al., A highly tilted binding mode by a self-reactive 55. M. D. Winn et al., Overview of the CCP4 suite and current
SUPPLEMENTARY MATERIALS
T cell receptor results in altered engagement of peptide and developments. Acta Crystallogr. D Biol. Crystallogr. 67,
MHC. J. Exp. Med. 208, 91–102 (2011). doi: 10.1084/ 235–242 (2011). doi: 10.1107/S0907444910045749; science.sciencemag.org/content/372/6546/eabe9124/suppl/DC1
jem.20100725; pmid: 21199956 pmid: 21460441 Figs. S1 to S5
Tables S1 to S5
40. Q. Wei et al., Lck bound to coreceptor is less active than free 56. P. A. Karplus, K. Diederichs, Linking crystallographic model and
MDAR Reproducibility Checklist
Lck. Proc. Natl. Acad. Sci. U.S.A. 117, 15809–15817 (2020). data quality. Science 336, 1030–1033 (2012). doi: 10.1126/
doi: 10.1073/pnas.1913334117; pmid: 32571924 science.1218231; pmid: 22628654 View/request a protocol for this paper from Bio-protocol.
41. V. Horkova et al., Dynamics of the Coreceptor-LCK 57. A. J. McCoy et al., Phaser crystallographic software. J. Appl.
Interactions during T Cell Development Shape the Self- Crystallogr. 40, 658–674 (2007). doi: 10.1107/ 22 September 2020; accepted 23 April 2021
Reactivity of Peripheral CD4 and CD8 T Cells. Cell Rep. 30, S0021889807021206; pmid: 19461840 10.1126/science.abe9124

Zareie et al., Science 372, eabe9124 (2021) 4 June 2021 13 of 13


Canonical T cell receptor docking on peptide–MHC is essential for T cell signaling
Pirooz ZareieChristopher SzetoCarine FarencSachith D. GunasingheElizabeth M. KolawoleAngela NguyenChantelle

Downloaded from https://www.science.org at Centro Andaluz de Biologia Molecular Y Medicina Regenerativa on November 12, 2021
BlythXavier Y. X. SngJasmine LiClaerwen M. JonesAlex J. FulcherJesica R. JacobsQianru WeiLukasz WojciechJan
PetersenNicholas R.J. GascoigneBrian D. EvavoldKatharina GausStephanie GrasJamie RossjohnNicole L. La Gruta

Science, 372 (6546), eabe9124. • DOI: 10.1126/science.abe9124

Making sense of TCR–pMHC topology


Most T cells use a T cell receptor (TCR) that recognizes major histocompatibility complex molecules bound to
peptides (pMHCs) derived from both self- and foreign antigens. Although there is great variability in the interface
because of the diversity of both partners, this interaction displays a canonical docking topology for reasons that remain
contested. Zareie et al. tested an assortment of both canonical and reversed-polarity TCRs that were all specific for
the same cognate pMHC-I bearing a peptide derived from influenza A virus (IAV) (see the Perspective by Horkova
and Stepanek). The authors determined that docking topology was the primary driver of in vivo T cell activation and
recruitment when mice were infected with IAV. The canonical topology was required for the formation of a functional
signaling complex, suggesting that T cell signaling constraints dictate how TCR and pMHC meet.
Science, abe9124, this issue p. eabe9124; see also abj2937, p. 1038

View the article online


https://www.science.org/doi/10.1126/science.abe9124
Permissions
https://www.science.org/help/reprints-and-permissions

Use of think article is subject to the Terms of service

Science (ISSN 1095-9203) is published by the American Association for the Advancement of Science. 1200 New York Avenue NW,
Washington, DC 20005. The title Science is a registered trademark of AAAS.
Copyright © 2021 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim
to original U.S. Government Works

You might also like