ArticlePDF AvailableLiterature Review

Superantigen Recognition and Interactions: Functions, Mechanisms and Applications

Frontiers
Frontiers in Immunology
Authors:

Abstract and Figures

Superantigens are unconventional antigens which recognise immune receptors outside their usual recognition sites e.g. complementary determining regions (CDRs), to elicit a response within the target cell. T-cell superantigens crosslink T-cell receptors and MHC Class II molecules on antigen-presenting cells, leading to lymphocyte recruitment, induction of cytokine storms and T-cell anergy or apoptosis among many other effects. B-cell superantigens, on the other hand, bind immunoglobulins on B-cells, affecting opsonisation, IgG-mediated phagocytosis, and driving apoptosis. Here, through a review of the structural basis for recognition of immune receptors by superantigens, we show that their binding interfaces share specific physicochemical characteristics when compared with other protein-protein interaction complexes. Given that antibody-binding superantigens have been exploited extensively in industrial antibody purification, these observations could facilitate further protein engineering to optimize the use of superantigens in this and other areas of biotechnology.
This content is subject to copyright.
Superantigen Recognition
and Interactions: Functions,
Mechanisms and Applications
Anthony M. Deacy
1
, Samuel Ken-En Gan
2,3
*and Jeremy P. Derrick
1
*
1
School of Biological Sciences, Faculty of Biology, Medicine, and Health, University of Manchester, Manchester,
United Kingdom,
2
Antibody & Product Development Lab, Experimental Drug Development Centre Bioinformatics
Institute (EDDC-BII), Agency for Science Technology and Research (ASTAR), Singapore, Singapore,
3
James Cook
University,Singapore,Singapore
Superantigens are unconventional antigens which recognise immune receptors outside
their usual recognition sites e.g. complementary determining regions (CDRs), to elicit a
response within the target cell. T-cell superantigens crosslink T-cell receptors and MHC
Class II molecules on antigen-presenting cells, leading to lymphocyte recruitment,
induction of cytokine storms and T-cell anergy or apoptosis among many other effects.
B-cell superantigens, on the other hand, bind immunoglobulins on B-cells, affecting
opsonisation, IgG-mediated phagocytosis, and driving apoptosis. Here, through a review
of the structural basis for recognition of immune receptors by superantigens, we show
that their binding interfaces share specic physicochemical characteristics when
compared with other protein-protein interaction complexes. Given that antibody-
binding superantigens have been exploited extensively in industrial antibody purication,
these observations could facilitate further protein engineering to optimize the use of
superantigens in this and other areas of biotechnology.
Keywords: superantigen, T-cell, B-cell, cytokine storm, interface, antibody purication
INTRODUCTION
Superantigens are unconventional antigens in the sense that they elicit a response by binding outside
the complementary determining regions (CDRs) of their target immune receptor macromolecules
(antibodies or T-cell receptors). At their initial description in 1989, superantigens were originally
dened as proteins that hyper-stimulate T-cells via the crosslinking of T-cell receptors (TCRs) and
MHC Class II molecules (1,2). This denition required extension following the discovery of B-cell
superantigens. B-cell superantigens can hyper-stimulate a large population of B-cells without
necessarily having the ability to crosslink TCRs with MHC Class II receptors; they therefore have a
different mechanism and specicity compared to T-cell superantigens (3). B-cell superantigens are
commonly known to (i) stimulate a high proportion of B-cells, and (ii) bind outside of the CDRs (4).
An extended denition of the term superantigenwas suggested to incorporate both functions, as a
molecule which has antigen-receptor mediated interactions with over 5% of the lymphocyte pool
(5). This functional denition is therefore based on the hyper-activity of the target receptor upon
exposure, and we will use the term in this context here.
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 7318451
Edited by:
Ester Boix,
Universitat Autònoma de Barcelona,
Spain
Reviewed by:
Marco Pio La Manna,
University of Palermo, Italy
Chozha V. Rathinam,
University of Maryland, United States
*Correspondence:
Jeremy P. Derrick
jeremy.derrick@manchester.ac.uk
Samuel Ken-En Gan
samgan@apdskeg.com
Specialty section:
This article was submitted to
Microbial Immunology,
a section of the journal
Frontiers in Immunology
Received: 28 June 2021
Accepted: 30 August 2021
Published: 20 September 2021
Citation:
Deacy AM, Gan SK-E
and Derrick JP (2021)
Superantigen Recognition and
Interactions: Functions,
Mechanisms and Applications.
Front. Immunol. 12:731845.
doi: 10.3389/fimmu.2021.731845
REVIEW
published: 20 September 2021
doi: 10.3389/fimmu.2021.731845
Here we review the current understanding of superantigens,
how they directly interact with immune receptors of T and B-
cells, what common features may be identied in recognition
interfaces and how such insights could be adapted to facilitate
further protein engineering of these versatile macromolecules for
therapeutic, diagnostic, and biotechnological applications.
T-CELL SUPERANTIGENS
T-cell superantigens are typically microbial proteins. They were
rst identied from observation of the hyper-stimulation of T-
cells by Staphylococcal Enterotoxin B (SEB). This phenomenon
was caused by the crosslinking of T-cell receptors (TCRs) Vb
with MHC class II a
1
on antigen presenting cells (APC) by SEB
(1,2). By crosslinking MHC Class II to TCR, small amounts of
superantigens can stimulate extensive T-cell proliferation. In a
normal adaptive immune response, only around 0.0001% of T-
cells are activated. In contrast, superantigen exposure can
activate up to 30% of the T-cell pool, leading to severe
pathologies following infection (6,7).
Enterotoxins produced by Staphylococcus aureus and
Streptococcus pyogenes form a common family of T-cell
superantigens. These enterotoxins are small (20-28 kDa), two
domain proteins which are diverse in sequence (15-90%) (8).
Despite this variation, enterotoxins and enterotoxin-like proteins
from both Staphylococcus aureus and Streptococcus pyogenes are
structurally similar (Figure 1), possessing a conserved Greek key
motif at the N-terminus known as an oligonucleotide (OB)-fold
(9). The C-terminus consists of a conserved b-domain capped by
an a-helix (9) with the two b-folds separated by a cluster of a-
helices. Due to their structural similarity, it has been suggested by
others that the enterotoxins from Staphylococcus aureus and
Streptococcus pyogenes shared a common ancestor (8).
Enterotoxins are thermostable, can withstand extreme pH
and are resistant to degradation by proteolytic enzymes such as
pepsin and trypsin (10,11). Some can retain activity after the
cooking and digestive process to cause food poisoning (12):
FIGURE 1 | Comparison of selected Staphylococcal aureus and Streptococcus pyogenes enterotoxin and enterotoxin-like structures. Staphylococcal aureus
enterotoxins include SEA (PDB code: 1ESF), SEB (PDB code: 1SE4) and TSST-1 (PDB code: 2QIL). The Staphylococcal aureus enterotoxin-like protein shown is
SSL4 (PDB code: 4DXF). Two Streptococcal pyogenes enterotoxins are displayed: SpeA1 (PDB code: 1UUP) and SpeC (PDB code: 1KTK). The structures are
shown as a ribbon plot with a-helices, b-strands and loops coloured in red, yellow, and green, respectively.
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 7318452
nearly 25% of food poisoning cases in the USA are attributed
to Staphylococcal enterotoxins (13). In addition, T-cell
superantigens also contribute to the development of systemic
inammatory response syndrome (SIRS) known as sepsis (14),
toxic shock syndrome (15,16), scarlet fever (17) and atopic
dermatitis (18).
CELLULAR RESPONSES TO T-CELL
SUPERANTIGENS
Observations of the cellular responses of T-cells to superantigens
are inconsistent, depending on the type and maturity of the T-
cell populations studied. T-cell superantigens can cause
immature CD4
+
and CD8
+
T-cells to become depleted. Mature
CD4
+
and CD8
+
T-cells on the other hand, proliferate and
produce a cytokine storm (1925) driving mature T-cells into
a state of anergy (26). TCR activation upregulates Fyn signalling,
preventing the protein tyrosine kinase ZAP-70 from associating
with TCRs via CD3, thus inhibiting TCR signalling (27). The
depletion of immature T-cells and anergy of mature T-cells
would potentially allow a pathogen to evade the innate
immune response, increasing pathogen survivability.
TCR binding to the MHC class II receptors on APCs results in
a variety of responses that is dependent on the APC type; the
principal pathways and components are summarized in
Figure 2.Duringinfection,neutrophilsarerecruitedalong
with other effector cells through the release of cytokines (e.g.
IFN-g, IL-17, IL-12) and CXC chemokines produced primarily
by CD4
+
T-cells (28,5052). Counterintuitively, the recruitment
of leukocytes increases the survivability of Staphylococcus aureus,
due to the hyper-stimulation of T-cells, eventually leading to T-
cell anergy and cell death. S. aureus is known to survive within
neutrophils and macrophages in abscesses (29,30).
Alongside TCR/MHC Class II activation, signalling pathways
are co-stimulated by crosslinking CD28 on the T-cell with CD80/
B7-2 on APCs (5356). T-cell superantigens can also crosslink the
a-subunit of laminin, LAMA2, with G-protein coupled receptor
(GPCR), resulting in T-cell stimulation (5759)(Figure 2).
MHC CLASS II BINDING
T-cell superantigens rst bind to MHC Class II receptors and
accumulate on the surface of the APC before binding to the TCR
(9). There are two possible binding sites on MHC Class II: a Zn-
dependent high afnity site (K
d
=10
-7
-10
-8
M) located on MHC
Class II bchain, and a low afnity site (K
d
= ~10
-5
M) located on
MHC Class II achain (60). Most superantigens bind via the Zn-
dependent binding site, forming a complex which is stable for
more than 40 hours (61). The high afnity interface between SEH
and MHC Class II achain is shown in Figure 3A, showing a
hydrophobic pocket surrounded by polar residues. In addition to
H-bonds and salt bridges, a Zn ion contributes to the high
binding afnity by stabilizing the complex through crosslinking
H81 on the MHC Class II b-strand and H206, N208 on the b-
strand 12 on SEH (62). This stabilization allows for the
formation of 4 extra H-bonds due to the proximity of the
chains where the removal of the Zn ion results in a decrease in
binding for SEA, SED, SEE and SEH (63).
FIGURE 2 | Principal components involved in the superantigen activation of T-cells, B-cells, macrophages, and neutrophils. The interactions displayed are based on
material from references (2849). The responses contribute to and escalate the hyper-activation of T-cells and subsequent cytokine storm.
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 7318453
The low afnity binding site is exemplied by a structure
containing the enterotoxin SEB, which forms a complex with
MHC class II (61): the low afnity interface is shown in
Figure 3B. In addition to cross-interface bonds, there is a
hydrophobic patch on SEB comprised of F44, L45 and F47
which inserts into a hydrophobic pocket on MHC Class II
achain.
The enterotoxin SEA can also bind to the low and high afnity
sites to crosslink two MHC Class II molecules (61,64,65).
Staphylococcal Enterotoxin H (SEH) was shown to bind the Zn-
dependent high afnity site on MHC class II (62), as well as to
TCR Vainstead of Vb(66,67). A list of T-cell superantigens and
their site specicities has been previously summarized by Proft
and Fraser (9). T-cell superantigen selectivity for the aor b
chains of the MHC Class II complex is dependent on the
presence of the Zn atom at the C-terminal bdomain. Its
absence leads to the binding of the a-chain of MHC Class II
via a hydrophobic ridge on the N-terminal OB-domain (9).
T-CELL RECEPTOR BINDING
Superantigens bind to the TCR after adhesion to MHC Class II;
there are also two sites on the TCRs in all superantigen
complexes studied to date. Some T-cell superantigens bind to
the achain [SEH (68)], although most recognize the bchain.
Unlike complexes with MHC Class II, both TCR interfaces bind
T-cell superantigens at low afnity (K
d
=10
-4
10
-6
M) (68,69)
and yet both are capable of mediating activation of a cytokine
storm (8,7073). SEB binding to the TCR bchain is shown in
Figure 4A where the interface is located at the TCR binding cleft
between the N-terminal b-barrel and the second a-helix. It is
characterised by several cross-interface bonds, with N23 playing a
crucial role, and a nearby hydrophobic patch formed of V26, Y79
and Y80 on SEH packing against the CDR2 loop of TCR Vb(74).
SEH binding to the TCR achain forms an interface
comprising hydrophobic and hydrophilic patches, with a
notable hydrophilic patch surrounding a Na ion (Figure 4B).
Comparing this to the SEB-TCR bchain interface, there are 7
fewer H-bonds and 2 fewer salt bridges, although the binding
afnities are similar (K
d
=10
-4
10
-6
M) (68). The lack of
contacts between SEH and the TCR achain may be bolstered by
the presence of the Na ion. N16 found on the second a-helix and
the hydrophobic patch (Y79 and Y80) on SEH are well conserved
among T-cell superantigens whether they bind to the TCR aor b
chains (67). The mutation N23A (equivalent to N16 in SEH) in
SEC2 caused the loss of mitogenic activity (75) and the same
mutation in SEB resulted in poorer proliferation of T-cells (76).
BINDING OF B-CELL SUPERANTIGENS
B-cell superantigens bind immunoglobulins outside the CDRs;
proteins which would t this denition of a superantigen were
rst described in the early 1990s (3). Binding to the Fab fragment
drives B-cells into apoptosis by hyper-activation of B-cell
A
B
FIGURE 3 | Binding of SEH and SEB superantigens to -MHC Class II. (A) Left panel: ribbon plot of SEH (blue) bound to the high afnity site on MHC Class II a
Chain (purple) (PDB code: 2XN9). Middle Panel: MHC Class II showing polar (red) and hydrophobic residues (blue). Right Panel: SEH. (B). Left panel: ribbon plot of
SEB (green) bound to the low afnity site on MHC Class II bChain (orange) (PDB code: 1SEB). Middle panel: MHC Class II bChain. Right panel: SEB.
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 7318454
receptors (BCRs). Considering that 20 to 50% of B-cells have
BCRs on their surfaces (77), B-cell superantigens can elicit a
potent immune response. However, B-cell superantigens are
better known for their ability to bind Fc and their applications
as afnity resins for antibody purication.
Staphylococcal Protein A (SpA), Streptococcal Protein G (SpG)
and Peptostreptococcal Protein L (PpL) are B-cell superantigens
located on the bacterial cell wall (78,79). SpA was classied as a
superantigen in 1995 due to its observed effect on B-cells (4).
However, SpA was rst isolated in 1940 and identied in 1964 due
to its Fc binding ability (78,80). It comprises a 42 kDa protein
arranged into ve homologous domains (E-D-A-B-C), each
forming a three a-helix bundle fold (Figure 5A)(81,82). The
domains are linked by conserved, exible linkers (82). Native SpA
also includes region X, a 12 x 8-residue repeat sequence which
binds peptidoglycan. All 5 A-E domains can bind both Fc and Fab
fragments (83). The binding afnity for specic immunoglobulins
depends on the isotype and species origin. In humans, SpA binds
strongly to IgG1, IgG2, IgG4 and weakly to IgA1, IgA2 and IgM.
Mutations R435H and F436Y on hIgG3 have been identied as the
reason SpA cannot bind human IgG3 (84). Interestingly,
mutations in CDR2 from the therapeutic antibodies Herceptin
and Pertuzumab were shown to contribute to binding SpA (85).
SpG was rst identied in 1984 by Björck and Kronvall (86)and
subsequently described as a B-cell superantigen. The sequence of SpG
differs depending on the Streptococcus strain of origin (Figure 5B).
SpG from group C Streptococcus sp. contains 2 immunoglobulin
binding domains (B1-B2) whereas group G has 3 (C1-C2-C3) (87
89). Between each immunoglobulin binding domain are spacers,
known as D domains. All SpG immunoglobulin-binding domains
can bind both the Fc and Fab fragments (83). SpG has provided an
alternative to SpA in antibody manufacturing, due it its ability to bind
some antibody isotypes not recognised by SpA. It can strongly bind to
all four human IgG subclasses (IgG1, IgG2, IgG3 and IgG4).
PpL was shown to induce apoptosis in B-cells by binding to the
V
L
region outside of the CDRs of BCRs, fullling the denition of
a B-cell superantigen (90). It was rst isolated in 1985 and
characterised as an immunoglobulin-binding protein capable of
binding to the variable light chain in 1988 (79,91). Of the two
most common strains of Peptostreptococcus magnus, strain 312
produces a 79 kDa, 5 domain (B1-B2-B3-B4-B5) protein whereas
strain 3316 expressesa 106 kDa 4 domain (C1-C2-C3-C4) protein
(Figure 5C)(92). PpL recognizes the light chain exclusively and
cannot bind to the Fc region. This makes it highly suitable for
afnity-purication of non-IgG antibodies (93,94).
All three B-cell superantigens (SpA, SpG and PpL) share
several common features; they form small, stable, multidomain
structures with a beads on a stringtype structure. Kim et al.
compared antibody levels of IgG and V
H
3+ IgM in mice when
infected with SpA mutants with one to 6 domains. The results
showed that the optimal number of immunoglobulin binding
domains to induce the largest B-cell response was 5 (95). This
observation suggests that B-cell superantigens are driven by the
need for multivalency of binding and the consequent improved
cross-linking of BCRs. These results were corroborated by a
similar study with PpL (92). Although SpG and PpL share no
signicant sequence homology (15%), their immunoglobulin
binding domains have similar folds, forming a b-sheet packed
A
B
FIGURE 4 | Binding of SEB and SEH superantigens to -TCRs. (A) Left panel: ribbon plot of SEB (blue) bound to TCR (green) (PDB code: 4C56). Middle panel: TCR
showing polar (red) and hydrophobic residues (blue). Right panel: SEB. (B) Left panel: ribbon plot of SEH (black) bound to TCR (purple) (PDB code: 2XN9). Middle
panel: TCR. Right panel: SEH.
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 7318455
against a single a-helix. A gene transfer event between
Streptococcus aureus and Peptostreptococcus magnus has been
proposed to explain a possible common evolutionary origin of
SpG and PpL (96). All three B-cell superantigens also utilise
regions W and M for crossing the cell membrane, featuring the
common Gram-positive protein anchoring motif LPXTG (97,
98). SpG and PpL also contain albumin binding domains (99,
100), which are absent in SpA.
CELLULAR RESPONSES TO B-CELL
SUPERANTIGENS
B-cell superantigens cross-link BCRs to activate BCR dependent
signalling (101,102). This initial signal transduction leads to the
downregulation of BCRs, and an upregulation of several cluster
of differentiation (CD) receptors (102), resulting in B-cell
capping (summarized schematically in Figure 6). MHC Class
A
B
C
FIGURE 5 | Schematic diagrams of SpA, SpG and PpL domain structures. (A) Left panel: Individual SpA domains including S (sorting peptide), Domains E-D-A-B-C, Region
X and Region M. Right panel: SpA Domain C (PDB code: 4WWI) Each SpA immunoglobulin binding domains consists of 3 a-helices (red). (B) Left panel: Individual SpG
domains including S (sorting peptide), Region E, Albumin Binding Domains A1-A2-A3, immunoglobulin binding domains B1-B2/C1-C2-C3 and Region W. Right panel: SpG
Domain B1 (PDB code: 3GB1) Each SpG immunoglobulin binding domain consists of 1 a-helix (red) and 4 anti-parallel b-strands (yellow). (C) Left panel: Individual PpL
domains including S (sorting peptide), Immunoglobulin Binding Domains B1-B2-B3-B4-B5/C1-C2-C3-C4, Albumin Binding Domains D1 to D4, Region W and M. Right panel:
PpL Domain B1 (PDB code: 1HEZ). Each PpL immunoglobulin binding domain consists of 1 a-helix (red) and 4 anti-parallel b-strands (yellow).
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 7318456
II is also upregulated (102). The upregulation of these receptors
leads to the activation of pro-apoptotic signals, such as Caspase
3, causing mitochondrial permeabilization and apoptosis (5,101,
102). Recently it has been shown that SpA B-cell superantigen
activity is dependent on the presence of the LPXTG anchoring
motif as well as the LysM domainbetween region X and the
LPXTG motif (103). These observations imply SpA must be
bound to peptidoglycan to cause B-cell stimulation.
The precise functional role of microbial B-cell superantigens
binding to Fc is obscure, although it has been shown recently that
soluble IgG is a requirement for the successful activation of BCRs by
SpA (104). The efciency of BCR activation was dependent on the
strength of Fc binding to each IgG subclass (104). The binding of
SpA-IgG complex to BCR is predicted to increase the functional
valency of the complex (104). SpA-IgG is thought to form a lattice
structure around the B-cells by crosslinking BCR Fab with IgG Fc
and other BCR Fab regions promoting a sustained stimulation.
Other potential functions of the B-cell superantigen-
immunoglobulin interaction are the blocking of immunoglobulin
effector functions, opsonization and immunoglobulin-mediated
phagocytosis, antibody-dependent cell mediated cytotoxicity
(ADCC) and complement-dependent cytotoxicity (CDC) (105
107). Expression of B-cell superantigens ultimately leads to B-cell
depletion and evasion of the immune system: in this sense, they can
be considered as virulence factors (108111).
SpA, SpG and PpL bind to BCRs at different sites on the Fab
fragment, although the activation results in similar cellular
responses. SpG binds to the C
H
1domain(112), implying
isotype dependent binding, whereas SpA binds to the V
H
3
family only. A comparison of the conservation of key residues
betweenthesevenV
H
families shows that, although many
residues are conserved, there are several which are key and
which, when mutated, result in the loss of binding for SpA
(113), including in the V
H
-CDR2 (85). PpL domains only bind
the klight chain V
L
region and therefore lacks the ability to bind
lchains. The binding afnity of PpL differs between the families
of klight chain, specically to FW1: it can bind to human VkI,
III and IV, but not II (114,115).
Several T-cell superantigens have the ability to bind BCRs,
although generally in a weak and non-specic manner (4), and
without a B-cell response. Exceptions have been noted, for
example, SEA increased the survival of V
H
3 B-cells (116). SED
has also been shown to increase survival of V
H
4 B-cells (117).
However, the in vivo response is yet to be determined.
Recent research suggests that B-cell superantigens also
enhance immune defences (118). Two superantigens have been
identied from the commensal bacteria Lachnospiraceae sp:
Immunoglobulin Binding Proteins A (IbpA) and B (IbpB). Both
were observed to activate BCRs by binding V
H
3 leading to the
increased secretion of IgA, although this was only shown in vitro.
B-CELL SUPERANTIGEN-FAB
COMPLEX INTERFACES
The crystal structure of the SpA and IgM Fab complex is
illustrated in Figure 7A, showing that the interface occurs at the
V
H
domain (involving residues from b-strands B to E) of the Fab
fragment and a-helices 2 and 3 of SpA (113). The interface is
dominated by polar residues with three negatively charged
residues from SpA and two positively charged residues from Fab
forming electrostatic interactions (113). All SpA domains can bind
FIGURE 6 | Proposed mechanisms for the activation of B-cell receptors by SpA. Activation leads to B-cell capping and B-cell apoptosis.
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 7318457
to the Fab fragment (119), and each domain varies in its afnity
towards V
H
3. The interacting residues form a predominantly
hydrophilic interface forming several cross-interface bonds.
SpG domain C2 co-crystalized with IgG Fab (Figure 7B): the
interface forms an antiparallel alignment between the last b-
strand of the C
H
1 domain and the second b-strand of SpG (120).
The antiparallel complex also results in interactions occurring
between rst b-strand of C
H
1 and the C-terminal end of the a-
helix of SpG (121). The interface is formed by mostly hydrophilic
residues anking a small hydrophobic patch.
The rst of two binding sites of PpL to IgM Fab is shown in
(Figure 7C). The majority of the interface occurs at framework
region 1 (FR1) of the V
L
region, with several contacts occurring
outside of the V
L
region: K107 between the V
L
and C
L
regions,
E143 from the C
L
region and R24 on the b-strand of CDR-L1 of
IgM Fab. The interface includes residues from the a-helix and
second b-strand of PpL domain B1 (122). The interface has a high
afnity (K
d
:110nM)(123), forming a predominantly hydrophilic
interaction characterised by 9 H-bonds, although several residues
have been proposed as hotspots from in silico alanine scanning of
the Fab and PpL (115,123,124). Interestingly, recent evidence
showed distal FWR3 effects on the PpL binding site at the FW1
(125) adding to the considerations for the light chain pairing with
the heavy chain (126,127).
The second binding site is formed from 15 residues at b-
strands A, B, C and D of the V
L
region of IgM Fab, and the a-helix
and third b-strand of PpL (Figure 7D). Although the second
binding site is slightly larger and composedof more cross-interface
contacts, it has a lower binding afnity (3.4 µM) (115). The rst
and second binding sites of PpL share only one common residue
from PpL (R52) but 10 out of 15 residues from IgM Fab.
B-CELL SUPERANTIGEN-FC BINDING
The crystal structure of a single domain from SpA was determined
in complex with IgG Fc: it showed that the protein-protein interface
occurs between a-helix 1 and 2 of SpA domain B and C
H
2andC
H
3
of the Fc (113)(Figure 8A). The residues forming the interface are
generally hydrophilic (128). SpA residues Q9, Q10, D36 and D37,
are conserved in the ve immunoglobulin binding domains of SpA
and are required for Fc binding (129). Mutating residue H435 in
IgG eliminates SpA binding, as this residue is situated on the C-
terminus of the C
H
3 region and protrudes into the C
H
2-C
H
3cleft
forming surface contacts with SpA (84).
The crystal structure of SpG C2 in complex with IgG Fc
showed that it binds at the same site as SpA, with SpG binding to
IgG Fc at the C
H
2-C
H
3 interface (Figure 8B). SpG ts within the
C
H
2-C
H
3 cleft and binds through residues on the a-helix and
third b-strand. As they recognise essentially the same site, SpA
and SpG bind competitively to IgG Fc (130,131). The strong
binding afnity of SpG for IgG Fc is contributed by a
hydrophobic pocket surrounded by hydrophilic residues.
Comparing the binding sites of SpG for Fab and Fc, Fab
binding uses b-strands 1 and 2 as well as the a-helix, whereas
Fc binding uses b-strand 3 as well as a more prominent
contribution of a-helix residues.
PHYSICOCHEMICAL CHARACTERISTICS
OF T AND B CELL SUPERANTIGEN
INTERFACES
The structures of the complexes of T and B-cell superantigens
with immune macromolecules were examined to compare the
nature of the interfaces with all other structures of protein-
protein complexes. A list was compiled from the Protein Data
Bank, extracting specic data on hydrophobicity, number of
hydrogen bonds, salt bridges, interface area, binding afnity,
and charges at the interface. These values were then condensed
onto a two-dimensional plot using t-distributed stochastic
neighbor embedding, such that each point represents a
complex (Figure 9).
Complexes were categorized according to function: most were
well dispersed by functional category, although peptide
complexes tended to predominate in the upper half of the plot
(Figure 8A). The superantigen complexes are grouped in
the central and right side of the plot, indicating that their
binding interfaces share some physicochemical characteristics
(Figure 9B). An explanation for this phenomenon is that T-cell
and B-cell superantigen interactions are transient-type complexes,
as dened by Noreen and Thornton (132). Such complexes tend to
be small and less hydrophobic than obligate, homo oligomeric
complexes. Both T- and B-cell superantigen interfaces form
interface areas less than 1000 Å
2
and range from slightly to
very hydrophilic.
The T-cell superantigens are located on the centre and right-
hand side of the plot and are more scattered than the B-cell
superantigen interfaces (Figure 9B). The interface areas of T-cell
and B-cell superantigens have similar ranges: 436 944 Å
2
and
517 714 Å
2
respectively. Fractional hydrophobicity of the T-cell
superantigen interfaces range from 18 49%, similar to those for
B-cell superantigens (9 40%).
Although B-cell superantigens recognise different binding
sites within the Fab molecule (V
H
,V
L
and C
H
1) they share
very similar interface physicochemical properties, which align
closely to those seen in peptide complexes.
The fact that superantigens are promiscuous and capable of
recognition of different binding partners indicates there is scope for
improving binding afnity and extending specicity for specic
targets. The observation that B-cell superantigens, and more
specically superantigen-Fab complexes, are physicochemically
similar may allow for development of engineering strategies
which makes use of this facility. Nonetheless, just as we expanded
the denition of superantigens in this review to include B-cell
activation based on new ndings, we are also aware of novel
superantigen-like behaviours by non-proteins e.g., nickel (133)
that may in time be included as superantigens in the future.
SUPERANTIGEN APPLICATIONS
Superantigens have been employed in multiple applications, both
clinically and industrially. Though many improvements have
been made, there is room to engineer and expand their scope
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 7318458
and applications. Understanding the biochemistry of the
superantigen-antibody interfaces provides an information
resource for the development of novel biotechnological and
pharmaceutical applications.
INDUSTRIAL
Since the approval of the rst therapeutic monoclonal antibody in
1986 (Muromonab-CD3), the use of antibody-based drugs has
A
B
D
C
FIGURE 7 | Binding of SpA, SpG and PpL to antibody Fab fragments: (A) Left panel: ribbon plot of SpA (Red) bound to V
H
3 domain of IgM Fab (Green) (PDB code:
1DEE). Middle panel: IgM Fab showing polar (red) and hydrophobic residues (blue). Right panel: SpA. (B) Left panel: ribbon plot of SpG (Orange) bound to C
H
1
domain of IgG Fab (Cyan) (PDB code: 1QKZ). Middle panel: IgG Fab. Right panel: SpG. (C) Left panel: ribbon plot of PpL Domain B
1
(Purple) interface 1 bound to
IgM Fab (Green) at the V
L
domain (PDB code: 1HEZ). Middle panel: IgM Fab. Left panel: PpL. (D) Left panel: ribbon plot of PpL Domain B
1
(Purple) interface 2
bound to IgM Fab (Blue) at the V
L
domain (PDB code: 1HEZ). Middle panel: IgM Fab. Right panel: PpL.
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 7318459
expanded signicantly with technological developments such as scFv,
antibody-drug conjugates (ADCs) and bispecics. In 2019, 70% of all
biopharmaceutical products sold were monoclonal antibodies in a
market worth over $150 billion. Antibody-based drugs continue to
increase their market share, with current estimates predicting global
revenue to increase to over $300 billion by 2025 (134). The expansion
of antibody-based drugs has therefore created a need for improved
manufacturing and purication processes.
The most prominent industrial application of B-cell
superantigens is their use as afnity resins for the purication of
antibodies, allowing highly efcient separation of antibodies for
clinical and research applications. Improvement of these afnity
resins has allowed pharmaceutical companies to develop cost-
effective antibody purication techniques, increasing the
feasibility of large-scale manufacturing of antibodies, resulting in
the expansion of the industry. There are, however, some limitations;
80% of the downstream processing cost occurs at the capture and
purication phase (135), and there is no single resin which can bind
all antibody isotypes from all species of interest. Furthermore,
antibodies are eluted from the afnity resins at low pH values,
frequently causing aggregation.
Some investigators have engineered superantigens to optimize
their application in antibody purication (136,137). For example
Domain Zwas developed in 1987- a mutant of Domain B with
two mutations, A1V and G29A (138), which resulted in SpA
losing the ability to bind V
H
3-Fab while retaining Fc afnity (139);
this innovation allows for the selective purication of the Fc
fragment after pepsin digestion. One such Z domain afnity
resin is Cytivas mAb Select SuRe (140). When testing mutant
N23T, the stability of the SpA Z domain resin increased (141).
Recently a new SpA resin has been developed: AviPure. This resin
is formed of two B domains with two cysteine and histidine
residues at the C-terminus to the steric hindrance, increasing
binding capacity and increasing its resistance to pHs extremes,
while retaining high binding afnity (135). Afnity resins undergo
cleaning in place (CIP) procedures commonly using 0.5 M NaOH;
therefore, afnity resins with high alkaline stability are desirable.
To address the issue with CIP procedures, SpA was shown to have
higher alkaline stability with a single mutation at position 29, with
G29W being the most stable (142). Two further mutations N23T
and F30A to the SpA Z domain resulted in a higher alkaline
resistance when compared to wild type (143). SpA Z domain was
also engineered to include six glycine residues on the second loop,
which resulted in an increase in the elution pH (143). Wild type
SpA is less susceptible to extreme alkaline conditions, with a half-
life of 16 h (141) compared to SpG, which has a half-life of under
10 mins (144). Asn residues were identied as the most susceptible
to deamination: mutation of all three Asn residues of SpG (N8T,
N35A and N37A) improved alkaline stability by 8-fold (145). SpG
was further demonstrated to have higher alkaline stability when
three other mutations Y3F, T16I and T1I were introduced (145).
By increasing alkaline stability, the lifespan of afnity resins can be
increased, lowering the overall cost of antibody production. It has
also been recently demonstrated that adding an additional alkaline
A
B
FIGURE 8 | Binding of SpA and SpG superantigens to -IgG Fc. (A) Left panel: ribbon plot of SpA Domain B (Orange) bound to IgG Fc (Pink) at the C
H
2-C
H
3
interface (PDB code: 5U4Y). Middle panel: IgG Fc showing polar (red) and hydrophobic residues (blue). Right panel: SpA. (B) Left panel: ribbon plot of SpG Domain
C2 (Blue) bound to IgG Fc (Orange) at the C
H
2-C
H
3 interface (PDB code: 1FCC). Middle panel: IgG Fc. Right panel: SpG.
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 73184510
wash step after the antibody capture step results in a decrease in
antibody aggregation, lower impurity levels and an increase in
antibody yield (146).
With the development of new formats such as single chain
variable fragments (scFvs), strong Fab binding is required.
Unfortunately, SpA and SpG have lower binding afnities for
the Fab fragment compared to Fc. PpL has the advantage over
other B-cell superantigens of binding strongly to the Vkof scFv,
(K
d
= 4.5nM) (147). The scFv structural arrangement consists of
the V
H
and V
L
domains connected by a short linker. scFv
molecules have the advantage of retaining the CDRs while
being signicantly smaller than whole antibodies. SpA can also
bind scFv but at a lower binding afnity than PpL, whereas SpG
is unable to bind scFv. The most signicant downside for the use
of PpL as an afnity resin is its inability to bind llight chains.
Therefore, in human antibody production, roughly 34% of the
antibodies will not bind to the resin, suggesting that engineering
PpL to bind llight chains could be valuable.
CLINICAL USAGE
Diagnostics Potential
Superantigens are used to detect IgG in serum (148), making use
of their immunoglobulin binding specicity. On the contrary,
superantigens recognized by IgGs allow for the detection of
Staphylococcus aureus (149,150) in disease states.
Engineering of superantigens to be specic to regions of TCRs
or antibody V-region families or isotypes for the development of
diagnostic kits could be applied to the quantication of disease-
associated proteins e.g. IgE in allergy. The ability to specically
bind antibodies can also allow its development in easy-to-use, non-
technical point-of-care testing home-use devices (151), as recently
applied during the COVID-19 pandemic. Such superantigen-based
diagnostics can be coupled with colorimetric, home-made devices
[e.g. mobile spectrophotometers (152,153)]. Given the increasing
association of antibody V
H
families with certain diseases e.g. [V
H
5
in nickel allergy (85)], superantigens that can differentiate antibody
V
H
families have clear potential in diagnostic kit development.
Therapeutic Potential
The role of superantigens in sepsis, a leading cause of death listed
by the WHO, makes them an important target for toxic-shock
syndrome (154). Several short peptide regions (~40 residues)
from SEA and SPEA have been identied as causes of vasodilation
(155), suggesting an application in the development of
antihypertension drugs.
Superantigens can also be used as a target for an anti-
Staphylococcus aureus vaccine. There have been several attempts
atproducinga vaccineagainstS. aureus,withoutsuccess,althoughit
A
B
FIGURE 9 | t-SNE plot of protein-protein interactions highlighting superantigen-antibody complexes. The position of each complex was determined using the following
parameters: Buried Surface Area (A
2
),Buried Surface Area Hydrophobicity (A
2
),Number of Interface Residues,H-Bonds,Salt Bridges,Category,Total Positive
Charge at Interface (A
2
),Total Negative Charge at Interface (A
2
)with a perplexity of 30. (A) Distribution of general protein-protein interactions categorized by function
(T-cell superantigen complex, B-cell superantigen complex, enzyme PPI, immune PPI, inhibitory PPI, peptide PPI, receptor PPI, signaling PPI, structural PPI, toxin-
antitoxin PPI and transport PPI). (B) The same plot as (A), but coloured for T-cell superantigen complexes (green) and B-cell superantigen complexes (red).
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 73184511
has been shown that the use of anti-SpA antibodies leads to the
promotion of opsonophagocytic clearance of Staphylococcus aureus
(156,157).
Superantigens have also shown promise in the treatment of
cancer through a synergistic effect with antibodies in the
recruitment of T-cells (158). The ability of SEB to hyper-
stimulate and proliferate CAR T-cells led to a more effective
antitumour response when used in combination (159). PpL has
been shown to induce apoptosis in malignant k
+
Bcell
lymphomas in humans and mice (160), demonstrating the
potential use of superantigens as anti-cancer drugs, particularly
when sagaciously paired with a suitable V
k
light chain (94). A
range of potential T-cell superantigen-based anticancer drugs
have been recently reviewed (161), including SEB, demonstrating
the ability to inhibit metastasis and tumour growth (162). Several
Fab-superantigen fusion proteins show promising Phase I/II
clinical trial results. A major drawback with using
superantigens is their potential to elicit a toxic response. One
way to prevent this is to reduce the over-stimulation of T-cells.
SEA was split into two functionally inactive domains and
attached to a scFv. When used in combination, the two SEA
fragments reassemble, forming a functionally active superantigen
and resulting in the selective activation of T-cells (163). Another
way to avoid superantigen toxicity is to utilize superantigen-like
proteins which, as mentioned previously, are very similar in
structure and function, although they do not result in emesis.
They have been shown to inhibit tumour growth by 30% without
signicant toxicity (164).
The importance of understanding superantigens goes beyond
bacterial sepsis to viruses, where for example, SARS-CoV2 spike
protein displays superantigen properties (165,166)causing
multisystem inammatory syndrome in children through its
unspecic activation of T-cells (167).
SUMMARY
With the development of new clinical therapeutics, B-cell
superantigen engineering presents an opportunity to develop
novel applications, as well as improving current superantigen-
based technology, such as purication resins. Structural
information on B-cell superantigen interfaces has been useful in
providing a basis for the engineering of binding characteristics.
The application of protein engineering principles offers
considerable scope for directed modication of superantigen
binding properties and harnessing for applications in medicine
and the pharmaceutical industry.
AUTHOR CONTRIBUTIONS
AD wrote the manuscript, with critical revisions from SG and JD.
AD and JD prepared the gures. All authors contributed to the
article and approved the submitted version.
FUNDING
AD is a PhD student funded by The University of Manchester,
UK and the Agency for Science, Technology and Research
(A*STAR), Singapore. This review was partially funded by the
National Research Foundation (NRF) Singapore grant to
Experimental Drug Development Centre (EDDC), ASTAR.
REFERENCES
1. White J, Herman A, Pullen AM, Kubo R, Kappler JW, Marrack P. The V
Beta-Specic Superantigen Staphylococcal Enterotoxin B: Stimulation of
Mature T Cells and Clonal Deletion in Neonatal Mice. Cell (1989) 56:2735.
doi: 10.1016/0092-8674(89)90980-x
2. Seth A, Stern LJ, Ottenhoff TH, Engel I, Owen MJ, Lamb JR, et al. Binary and
Ternary Complexes Between T-Cell Receptor, Class II MHC and
Superantigen In Vitro.Nature (1994) 369:3247. doi: 10.1038/369324a0
3. Pascual V, Capra JD. B-Cell Superantigens? Curr Biol (1991) 1:3157.
doi: 10.1016/0960-9822(91)90097-g
4. Levinson AI, Kozlowski L, Zheng Y, Wheatley L. B-Cell Superantigens:
Denition and Potential Impact on the Immune Response. J Clin Immunol
(1995) 15:26S36S. doi: 10.1007/BF01540891
5. Silverman GJ, Goodyear CS. Confounding B-Cell Defences: Lessons From a
Staphylococcal Superantigen. Nat Rev Immunol (2006) 6:46575.
doi: 10.1038/nri1853
6. Choi Y, Lafferty JA, Clements JR, Todd JK, Gelfand EW, Kappler J, et al.
Selective Expansion of T Cells Expressing V Beta 2 in Toxic Shock
Syndrome. J Exp Med (1990) 172:9814. doi: 10.1084/jem.172.3.981
7. Kotzin BL, Leung DY, Kappler J, Marrack P. Superantigens and Their
Potential Role in Human Disease. Adv Immunol (1993) 54:99166.
doi: 10.1016/s0065-2776(08)60534-9
8. Proft T, Fraser JD. Bacterial Superantigens. Clin Exp Immunol (2003)
133:299306. doi: 10.1046/j.1365-2249.2003.02203.x
9. Fraser JD, Proft T. The Bacterial Superantigen and Superantigen-Like Proteins.
Immunol Rev (2008) 225:22643. doi: 10.1111/j.1600-065X.2008.00681.x
10. Thomas D, Chou S, Dauwalder O, Lina G. Diversity in Staphylococcus Aureus
Enterotoxins. ChemImmunol Allergy (2007)93:2441. doi: 10.1159/000100856
11. Li SJ, Hu DL, Maina EK, Shinagawa K, Omoe K, Nakane A. Superantigenic
Activity of Toxic Shock Syndrome Toxin-1 Is Resistant to Heating and
Digestive Enzymes. J Appl Microbiol (2011) 110:72936. doi: 10.1111/j.1365-
2672.2010.04927.x
12. Argudin MA, Mendoza MC, Rodicio MR. Food Poisoning and
Staphylococcus Aureus Enterotoxins. Toxins (Basel) (2010) 2:175173.
doi: 10.3390/toxins2071751
13. Herman A, Kappler JW, Marrack P, Pullen AM. Superantigens: Mechanism
of T-Cell Stimulation and Role in Immune Responses. Annu Rev Immunol
(1991) 9:74572. doi: 10.1146/annurev.iy.09.040191.003525
14. Martin GS, Mannino DM, Eaton S, Moss M. The Epidemiology of Sepsis in
the United States From 1979 Through 2000. N Engl J Med (2003) 348:1546
54. doi: 10.1056/NEJMoa022139
15. Bergdoll MS, Crass BA, Reiser RF, Robbins RN, Davis JP. A New
Staphylococcal Enterotoxin, Enterotoxin F, Associated With Toxic-Shock-
Syndrome Staphylococcus Aureus Isolates. Lancet (1981) 1:101721.
doi: 10.1016/s0140-6736(81)92186-3
16. Schlievert PM, Shands KN, Dan BB, Schmid GP, Nishimura RD.
Identication and Characterization of an Exotoxin From Staphylococcus
Aureus Associated With Toxic-Shock Syndrome. J Infect Dis (1981)
143:50916. doi: 10.1093/infdis/143.4.509
17. Stevens DL, Tanner MH, Winship J, Swarts R, Ries KM, Schlievert PM, et al.
Severe Group A Streptococcal Infections Associated With a Toxic Shock-
Like Syndrome and Scarlet Fever Toxin a. N Engl J Med (1989) 321:17.
doi: 10.1056/NEJM198907063210101
18.KimJ,KimBE,AhnK,LeungDYM.InteractionsBetweenAtopic
Dermatitis and Staphylococcus Aureus Infection: Clinical Implications.
Allergy Asthma Immunol Res (2019) 11:593603. doi: 10.4168/aair.2019.
11.5.593
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 73184512
19. Jenkinson EJ, Kingston R, Owen JJ. Newly Generated Thymocytes Are Not
Refractory to Deletion When the Alpha/Beta Component of the T Cell
Receptor is Engaged by the Superantigen Staphylococcal Enterotoxin B. Eur
J Immunol (1990) 20:251720. doi: 10.1002/eji.1830201125
20. Coppola MA, Blackman MA. Bacterial Superantigens Reactivate Antigen-
SpecicCD8+MemoryTCells.Int Immunol (1997) 9:1393403.
doi: 10.1093/intimm/9.9.1393
21. Murphy KM, Heimberger AB, Loh DY. Induction by Antigen of Intrathymic
Apoptosis of CD4+CD8+TCRlo Thymocytes In Vivo.Science (1990)
250:17203. doi: 10.1126/science.2125367
22. Lin YS, Lei HY, Low TL, Shen CL, Chou LJ, Jan MS. In Vivo Induction of
Apoptosis in Immature Thymocytes by Staphylococcal Enterotoxin B.
J Immunol (1992) 149:115663.
23. Meilleur CE, Wardell CM, Mele TS, Dikeakos JD, Bennink JR, Mu HH, et al.
Bacterial Superantigens Expand and Activate, Rather Than Delete or
Incapacitate, Preexisting Antigen-Specic Memory CD8+ T Cells. J Infect
Dis (2019) 219:130717. doi: 10.1093/infdis/jiy647
24. Xu R, Shears RK, Sharma R, Krishna M, Webb C, Ali R, et al. IL-35 is Critical
in Suppressing Superantigenic Staphylococcus Aureus-Driven Inammatory
Th17 Responses in Human Nasopharynx-Associated Lymphoid Tissue.
Mucosal Immunol (2020) 13:46070. doi: 10.1038/s41385-019-0246-1
25. Florquin S, Amraoui Z, Abramowicz D, Goldman M. Systemic Release and
Protective Role of IL-10 in Staphylococcal Enterotoxin B-Induced Shock in
Mice. J Immunol (1994) 153:261823.
26. MacDonald HR, Baschieri S, Lees RK. Clonal Expansion Precedes Anergy
and Death of V Beta 8+ Peripheral T Cells Responding to Staphylococcal
Enterotoxin B In Vivo.Eur J Immunol (1991) 21:19636. doi: 10.1002/
eji.1830210827
27. Watson AR, Janik DK, Lee WT. Superantigen-Induced CD4 Memory T Cell
Anergy. I. Staphylococcal Enterotoxin B Induces Fyn-Mediated Negative
Signaling. Cell Immunol (2012) 276:1625. doi: 10.1016/j.cellimm.2012.02.003
28. Xu SX, Gilmore KJ, Szabo PA, Zeppa JJ, Baroja ML, Haeryfar SM, et al.
Superantigens Subvert the Neutrophil Response to Promote Abscess
Formation and Enhance Staphylococcus Aureus Survival In Vivo.Infect
Immun (2014) 82:358898. doi: 10.1128/IAI.02110-14
29. Gresham HD, Lowrance JH, Caver TE, Wilson BS, Cheung AL, Lindberg FP.
Survival of Staphylococcus Aureus Inside Neutrophils Contributes to
Infection. J Immunol (2000) 164:371322. doi: 10.4049/jimmunol.164.7.3713
30. Pidwill GR, Gibson JF, Cole J, Renshaw SA, Foster SJ. The Role of
Macrophages in Staphylococcus Aureus Infection. Front Immunol (2020)
11:620339. doi: 10.3389/mmu.2020.620339
31. Zeng X, Moore TA, Newstead MW, Deng JC, Lukacs NW, Standiford TJ. IP-
10 Mediates Selective Mononuclear Cell Accumulation and Activation in
Response to Intrapulmonary Transgenic Expression and During
Adenovirus-Induced Pulmonary Inammation. J Interferon Cytokine Res
(2005) 25:10312. doi: 10.1089/jir.2005.25.103
32. Lei L, Altstaedt J, von der Ohe M, Proft T, Gross U, Rink L. Induction of
Interleukin-8 in Human Neutrophils After MHC Class II Cross-Linking
With Superantigens. J Leukoc Biol (2001) 70:806. doi: 10.1189/jlb.70.1.80
33. Gainet J, Dang PM, Chollet-Martin S, Brion M, Sixou M, Hakim J, et al.
Neutrophil Dysfunctions, IL-8, and Soluble L-Selectin Plasma Levels in
Rapidly Progressive Versus Adult and Localized Juvenile Periodontitis:
Variations According to Disease Severity and Microbial Flora. J Immunol
(1999) 163:50139.
34. Tuffs SW, James DBA, Bestebroer J, Richards AC, Goncheva MI, OShea M,
et al. The Staphylococcus Aureus Superantigen SElX Is a Bifunctional Toxin
That Inhibits Neutrophil Function. PloS Pathog (2017) 13:e1006461.
doi: 10.1371/journal.ppat.1006461
35. Gonzalez CD, Ledo C, Giai C, Garofalo A, Gomez MI. The Sbi Protein
Contributes to Staphylococcus Aureus Inammatory Response During
Systemic Infection. PloS One (2015) 10:e0131879. doi: 10.1371/journal.
pone.0131879
36. Pliyev BK, Dimitrieva TV, Savchenko VG. Cytokine-Mediated Induction
of MHC Class II in Human Neutrophils Is Dependent on NADPH
Oxidase Activity. Eur J Cell Biol (2015) 94:6770. doi: 10.1016/j.ejcb.
2014.11.001
37. Noli Truant S, De Marzi MC, Sarratea MB, Antonoglou MB, Meo AP,
Iannantuono Lopez LV, et al. Egc SuperantigensImpairMonocytes/
Macrophages Inducing Cell Death and Inefcient Activation. Front
Immunol (2019) 10:3008. doi: 10.3389/mmu.2019.03008
38. Chen CL, Wu YY, Lin CF, Kuo CF, Han CL, Wang S, et al. Streptococcal
Pyrogenic Exotoxin B Inhibits Apoptotic Cell Clearance by Macrophages
Through Protein S Cleavage. Sci Rep (2016) 6:26026. doi: 10.1038/srep26026
39. Watanabe I, Ichiki M, Shiratsuchi A, Nakanishi Y. TLR2-Mediated Survival
of Staphylococcus Aureus in Macrophages: A Novel Bacterial Strategy
Against Host Innate Immunity. J Immunol (2007) 178:491725.
doi: 10.4049/jimmunol.178.8.4917
40. Nabavi N, Freeman GJ, Gault A, Godfrey D, Nadler LM, Glimcher LH.
Signalling Through the MHC Class II Cytoplasmic Domain is Required for
Antigen Presentation and Induces B7 Expression. Nature (1992) 360:2668.
doi: 10.1038/360266a0
41. Mourad W, Scholl P, Diaz A, Geha R, Chatila T. The Staphylococcal Toxic
Shock Syndrome Toxin 1 Triggers B Cell Proliferation and Differentiation
via Major Histocompatibility Complex-Unrestricted Cognate T/B Cell
Interaction. J Exp Med (1989) 170:201122. doi: 10.1084/jem.170.6.2011
42. Bekeredjian-Ding I, Inamura S, Giese T, Moll H, Endres S, Sing A, et al.
Staphylococcus Aureus Protein A Triggers T Cell-Independent B Cell
Proliferation by Sensitizing B Cells for TLR2 Ligands. J Immunol (2007)
178:280312. doi: 10.4049/jimmunol.178.5.2803
43. Parcina M, Miranda-Garcia MA, Durlanik S, Ziegler S, Over B, Georg P,
et al. Pathogen-Triggered Activation of Plasmacytoid Dendritic Cells
Induces IL-10-Producing B Cells in Response to Staphylococcus Aureus.
J Immunol (2013) 190:1591602. doi: 10.4049/jimmunol.1201222
44. Narita K, Hu DL, Asano K, Nakane A. Interleukin-10 (IL-10) Produced by
Mutant Toxic Shock Syndrome Toxin 1 Vaccine-Induced Memory T Cells
Downregulates IL-17 Production and Abrogates the Protective Effect
Against Staphylococcus Aureus Infection. Infect Immun (2019) 87:112.
doi: 10.1128/IAI.00494-19
45. Keener AB, Thurlow LT, Kang S, Spidale NA, Clarke SH, Cunnion KM, et al.
Staphylococcus Aureus Protein A Disrupts Immunity Mediated by Long-
Lived Plasma Cells. J Immunol (2017) 198:126373. doi: 10.4049/
jimmunol.1600093
46. Parcina M, Wendt C, Goetz F, Zawatzky R, Zahringer U, Heeg K, et al.
Staphylococcus Aureus-Induced Plasmacytoid Dendritic Cell Activation Is
Based on an IgG-Mediated Memory Response. J Immunol (2008) 181:3823
33. doi: 10.4049/jimmunol.181.6.3823
47. Bhardwaj N, Friedman SM, Cole BC, Nisanian AJ. Dendritic Cells are Potent
Antigen-Presenting Cells for Microbial Superantigens. J Exp Med (1992)
175:26773. doi: 10.1084/jem.175.1.267
48. Coutant KD, de Fraissinette AB, Cordier A, Ulrich P. Modulation of the
Activity of Human Monocyte-Derived Dendritic Cells by Chemical Haptens,
a Metal Allergen, and a Staphylococcal Superantigen. Toxicol Sci (1999)
52:18998. doi: 10.1093/toxsci/52.2.189
49. Wu X, Xu F. Dendritic Cells During Staphylococcus Aureus Infection:
Subsets and Roles. J Transl Med (2014) 12:358. doi: 10.1186/s12967-014-
0358-z
50. Laan M, Cui ZH, Hoshino H, Lotvall J, Sjostrand M, Gruenert DC, et al.
Neutrophil Recruitment by Human IL-17 via C-X-C Chemokine Release in
the Airways. J Immunol (1999) 162:234752.
51. McLoughlin RM, Solinga RM, Rich J, Zaleski KJ, Cocchiaro JL, Risley A,
et al. CD4+ T Cells and CXC Chemokines Modulate the Pathogenesis of
Staphylococcus Aureus Wound Infections. Proc Natl Acad Sci USA (2006)
103:1040813. doi: 10.1073/pnas.0508961103
52. McLoughlin RM, Lee JC, Kasper DL, Tzianabos AO. IFN-Gamma Regulated
Chemokine Production Determines the Outcome of Staphylococcus Aureus
Infection. J Immunol (2008) 181:132332. doi: 10.4049/jimmunol.181.2.1323
53. Arad G, Levy R, Nasie I, Hillman D, Rotfogel Z, Barash U, et al. Binding of
Superantigen Toxins Into the CD28 Homodimer Interface is Essential for
Induction of Cytokine Genes That Mediate Lethal Shock. PloS Biol (2011) 9:
e1001149. doi: 10.1371/journal.pbio.1001149
54. Levy R, Rotfogel Z, Hillman D, Popugailo A, Arad G, Supper E, et al.
Superantigens Hyperinduce Inammatory Cytokines by Enhancing the B7-
2/CD28 Costimulatory Receptor Interaction. Proc Natl Acad Sci USA (2016)
113:E643746. doi: 10.1073/pnas.1603321113
55. Kaempfer R, Popugailo A, Levy R, Arad G, Hillman D, Rotfogel Z. Bacterial
Superantigen Toxins Induce a Lethal Cytokine Storm by Enhancing B7-2/CD28
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 73184513
Costimulatory Receptor Engagement, a Critical Immune Checkpoint. Receptors
Clin Investig (2017) 4:19. doi: 10.14800/rci.1500
56. Popugailo A, Rotfogel Z, Supper E, Hillman D, Kaempfer R. Staphylococcal
and Streptococcal Superantigens Trigger B7/CD28 Costimulatory Receptor
Engagement to Hyperinduce Inammatory Cytokines. Front Immunol
(2019) 10:942. doi: 10.3389/mmu.2019.00942
57. Yamasaki S, Tachibana M, Shinohara N, Iwashima M. Lck-Independent
Triggering of T-Cell Antigen Receptor Signal Transduction by Staphylococcal
Enterotoxins. JBiolChem(1997) 272:1478791. doi: 10.1074/jbc.272.
23.14787
58. Bueno C, Lemke CD, Criado G, Baroja ML, Ferguson SS, Rahman AK, et al.
Bacterial Superantigens Bypass Lck-Dependent T Cell Receptor Signaling by
Activating a Galpha11-Dependent, PLC-Beta-Mediated Pathway. Immunity
(2006) 25:6778. doi: 10.1016/j.immuni.2006.04.012
59. Li Z, Zeppa JJ, Hancock MA, McCormick JK, Doherty TM, Hendy GN, et al.
Staphylococcal Superantigens Use LAMA2 as a Coreceptor To Activate T
Cells. J Immunol (2018) 200:14719. doi: 10.4049/jimmunol.1701212
60. Li Y, Li H,Dimasi N, McCormickJK, Martin R, Schuck P, et al.Crystal Structure
of a Superantigen Bound to the High-Afnity, Zinc-Dependent Site on MHC
Class II. Immunity (2001) 14:93104. doi: 10.1016/s1074-7613(01)00092-9
61. PlessDD, Ruthel G, ReinkeEK, Ulrich RG,Bavari S. Persistenceof Zinc-Binding
Bacterial Superantigens at the Surface of Antigen-PresentingCells Contributes
to the Extreme Potency of These Superantigens as T-Cell Activators. Infect
Immun (2005) 73:535866. doi: 10.1128/IAI.73.9.5358-5366.2005
62. Petersson K, Hakansson M, Nilsson H, Forsberg G, Svensson LA, Liljas A,
et al. Crystal Structure of a Superantigen Bound to MHC Class II Displays
Zinc and Peptide Dependence. EMBO J (2001) 20:330612. doi: 10.1093/
emboj/20.13.3306
63. Nilsson H, Bjork P, Dohlsten M, Antonsson P. Staphylococcal Enterotoxin
H Displays Unique MHC Class II-Binding Properties. J Immunol (1999)
163:668693.
64. Hudson KR, Tiedemann RE, Urban RG, Lowe SC, Strominger JL, Fraser JD.
Staphylococcal Enterotoxin A has Two Cooperative Binding Sites on Major
Histocompatibility Complex Class II. JExpMed(1995) 182:71120.
doi: 10.1084/jem.182.3.711
65. Tiedemann RE, Fraser JD. Cross-Linking of MHC Class II Molecules by
Staphylococcal Enterotoxin A Is Essential for Antigen-Presenting Cell and T
Cell Activation. J Immunol (1996) 157:395866.
66. Petersson K, Pettersson H, Skartved NJ, Walse B, Forsberg G. Staphylococcal
Enterotoxin H Induces V Alpha-Specic Expansion of T Cells. J Immunol
(2003) 170:414854. doi: 10.4049/jimmunol.170.8.4148
67. Saline M, Rodstrom KE, Fischer G, Orekhov VY, Karlsson BG, Lindkvist-
Petersson K. The Structure of Superantigen Complexed With TCR and
MHC Reveals Novel Insights Into Superantigenic T Cell Activation. Nat
Commun (2010) 1:119. doi: 10.1038/ncomms1117
68. Pumphrey N, Vuidepot A, Jakobsen B, Forsberg G, Walse B, Lindkvist-Petersson
K. Cutting Edge: Evidence of Direct TCR Alpha-Chain Interaction With
Superantigen. J Immunol (2007) 179:27004. doi: 10.4049/jimmunol.179.5.2700
69. Krakauer T. Update on Staphylococcal Superantigen-Induced Signaling
Pathways and Therapeutic Interventions. Toxins (Basel) (2013) 5:162954.
doi: 10.3390/toxins5091629
70. Krakauer T. Differential Inhibitory Effects of Interleukin-10, Interleukin-4,
and Dexamethasone on Staphylococcal Enterotoxin-Induced Cytokine
Production and T Cell Activation. J Leukoc Biol (1995) 57:4504.
doi: 10.1002/jlb.57.3.450
71. Bohach GA, Fast DJ, Nelson RD, Schlievert PM. Staphylococcal and Streptococcal
Pyrogenic Toxins Involved in Toxic Shock Syndrome and Related Illnesses.
Crit Rev Microbiol (1990) 17:25172. doi: 10.3109/10408419009105728
72. Marrack P, Kappler J. The Staphylococcal Enterotoxins and Their Relatives.
Science (1990) 248:1066. doi: 10.1126/science.2343314
73. Miethke T, Wahl C, Heeg K, Echtenacher B, Krammer PH, Wagner H. T
Cell-Mediated Lethal Shock Triggered in Mice by the Superantigen
Staphylococcal Enterotoxin B: Critical Role of Tumor Necrosis Factor.
J Exp Med (1992) 175:918. doi: 10.1084/jem.175.1.91
74. Rodstrom KE, Elbing K, Lindkvist-Petersson K. Structure of the
Superantigen Staphylococcal Enterotoxin B in Complex With TCR and
Peptide-MHC Demonstrates Absence of TCR-Peptide Contacts. J Immunol
(2014) 193:19982004. doi: 10.4049/jimmunol.1401268
75. Leder L, Llera A, Lavoie PM, Lebedeva MI, Li H, Sekaly RP, et al. A
Mutational Analysis of the Binding of Staphylococcal Enterotoxins B and C3
to the T Cell Receptor Beta Chain and Major Histocompatibility Complex
Class II. J Exp Med (1998) 187:82333. doi: 10.1084/jem.187.6.823
76. Kappler JW, Herman A, Clements J, Marrack P. Mutations Dening
Functional Regions of the Superantigen Staphylococcal Enterotoxin B.
J Exp Med (1992) 175:38796. doi: 10.1084/jem.175.2.387
77. Silverman GJ, Sasano M, Wormsley SB. Age-Associated Changes in Binding
of Human B Lymphocytes to a VH3-Restricted Unconventional Bacterial
Antigen. J Immunol (1993) 151:584055.
78. Verwey WF. A Type-Specic Antigenic Protein Derived From the
Staphylococcus. J Exp Med (1940) 71:63544. doi: 10.1084/jem.71.5.635
79. Erntell M, Myhre EB, Kronvall G. Non-Immune IgG F(Ab)2 Binding to
Group C and G Streptococci is Mediated by Structures on Gamma Chains.
Scand J Immunol (1985) 21:1517. doi: 10.1111/j.1365-3083.1985.tb01414.x
80. Oeding P, Grov A, Myklestad B. Immunochemical Studies on Antigen
Preparations From Staphylococcus Aureus. 2. Precipitating and Erythrocyte-
Sensitizing Properties of Protein a (Antigen a) and Related Substances. Acta
Pathol Microbiol Scand (1964) 62:11727. doi: 10.1111/apm.1964.62.1.117
81. Myers JK, Oas TG. Preorganized Secondary Structure as an Important
Determinant of Fast Protein Folding. NatStructBiol(2001) 8:5528.
doi: 10.1038/88626
82. Deis LN, Pemble C, Qi Y, Hagarman A, Richardson DC, Richardson JS, et al.
Multiscale Conformational Heterogeneity in Staphylococcal Protein a:
Possible Determinant of Functional Plasticity. Structure (2014) 22:1467
77. doi: 10.1016/j.str.2014.08.014
83. Moks T, Abrahmsen L, Nilsson B, Hellman U, Sjoquist J, Uhlen M.
Staphylococcal Protein A Consists of Five IgG-Binding Domains.
Eur J Biochem (1986) 156:63743. doi: 10.1111/j.1432-1033.1986.tb09625.x
84. Jendeberg L, Nilsson P, Larsson A, Denker P, Uhlen M, Nilsson B, et al.
Engineering of Fc(1) and Fc(3) From Human Immunoglobulin G to Analyse
Subclass Specicity for Staphylococcal Protein a. J Immunol Methods (1997)
201:2534. doi: 10.1016/s0022-1759(96)00215-3
85. Lua WH, Su CT, Yeo JY, Poh JJ, Ling WL, Phua SX, et al. Role of the IgE
Variable Heavy Chain in FcepsilonRIalpha and Superantigen Binding in
Allergy and Immunotherapy. J Allergy Clin Immunol (2019) 144:51423.e5.
doi: 10.1016/j.jaci.2019.03.028
86. Bjorck L, Kronvall G. Purication and Some Properties of Streptococcal
Protein G, a Novel IgG-Binding Reagent. J Immunol (1984) 133:96974.
87. Guss B, Eliasson M, Olsson A, Uhlen M, Frej AK, Jornvall H, et al. Structure
of the IgG-Binding Regions of Streptococcal Protein G. EMBO J (1986)
5:156775. doi: 10.1002/j.1460-2075.1986.tb04398.x
88. Sjobring U, Bjorck L, Kastern W, Streptococcal protein G. Gene Structure
and Protein Binding Properties. J Biol Chem (1991) 266:399405. doi:
10.1016/S0021-9258(18)52448-0
89. Nilvebrant J, Hober S. The Albumin-Binding Domain as a Scaffold for
Protein Engineering. Comput Struct Biotechnol J (2013) 6:e201303009.
doi: 10.5936/csbj.201303009
90. Goodyear CS, Narita M, Silverman GJ. In Vivo VL-Targeted Activation-
Induced Apoptotic Supraclonal Deletion by a Microbial B Cell Toxin.
J Immunol (2004) 172:2870710. doi: 10.4049/jimmunol.172.5.2870
91. Bjorck L, Protein L. A Novel Bacterial Cell Wall Protein With Afnity for Ig
L Chains. J Immunol (1988) 140:11947.
92. Kastern W, Sjobring U, Bjorck L. Structure of Peptostreptococcal Protein L
and Identication of a Repeated Immunoglobulin Light Chain-Binding
Domain. J Biol Chem (1992) 267:128205. doi: 10.1016/S0021-9258(18)
42349-6
93. Lua WH, Ling WL, Yeo JY, Poh JJ, Lane DP, Gan SK. The Effects of
Antibody Engineering CH and CL in Trastuzumab and Pertuzumab
Recombinant Models: Impact on Antibody Production and Antigen-
Binding. Sci Rep (2018) 8:718. doi: 10.1038/s41598-017-18892-9
94. Ling WL, Lua WH, Gan SK. Sagacity in Antibody Humanization for
Therapeutics, Diagnostics and Research Purposes: Considerations of
Antibody Elements and Their Roles. Antib Ther (2020) 3:717910.
95. Kim HK, Falugi F, Missiakas DM, Schneewind O. Peptidoglycan-Linked
Protein A Promotes T Cell-Dependent Antibody Expansion During
Staphylococcus Aureus Infection. Proc Natl Acad Sci USA (2016)
113:571823. doi: 10.1073/pnas.1524267113
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 73184514
96. Goward CR, Scawen MD, Murphy JP, Atkinson T. Molecular Evolution of
Bacterial Cell-Surface Proteins. Trends Biochem Sci (1993) 18:13640.
doi: 10.1016/0968-0004(93)90021-e
97. Schneewind O, Model P, Fischetti VA. Sorting of Protein A to the Staphylococcal
Cell Wall. Cell (1992) 70:26781. doi: 10.1016/0092-8674(92)90101-h
98. Navarre WW, Schneewind O. Surface Proteins of Gram-Positive Bacteria
and Mechanisms of Their Targeting to the Cell Wall Envelope. Microbiol
Mol Biol Rev (1999) 63:174229. doi: 10.1128/MMBR.63.1.174-229.1999
99. Akerstrom B, Nielsen E, Bjorck L. Denition of IgG- and Albumin-Binding
Regions of Streptococcal Protein G. J Biol Chem (1987) 262:1338891.
doi: 10.1016/S0021-9258(19)76438-2
100. Murphy JP, Duggleby CJ, Atkinson MA, Trowern AR, Atkinson T, Goward
CR. The Functional Units of a Peptostreptococcal Protein L. Mol Microbiol
(1994) 12:91120. doi: 10.1111/j.1365-2958.1994.tb01079.x
101. Broker BM, Holtfreter S, Bekeredjian-Ding I. ImmuneControl of Staphylococcus
Aureus - Regulation and Counter-Regulation of the Adaptive Immune Response.
Int J Med Microbiol (2014) 304:20414. doi: 10.1016/j.ijmm.2013.11.008
102. Goodyear CS, Silverman GJ. Death by a B Cell Superantigen: In Vivo VH-
Targeted Apoptotic Supraclonal B Cell Deletion by a Staphylococcal Toxin.
J Exp Med (2003) 197:112539. doi: 10.1084/jem.20020552
103. Shi M, Willing SE, Kim HK, Schneewind O, Missiakas D. Peptidoglycan
Contribution to the B Cell Superantigen Activity of Staphylococcal Protein a.
mBio (2021) 12:113. doi: 10.1128/mBio.00039-21
104. Ulloa-Morales AJ, Goodyear CS, Silverman GJ. Essential Domain-
Dependent Roles Within Soluble IgG for In Vivo Superantigen Properties
of Staphylococcal Protein A: Resolving the B-Cell Superantigen Paradox.
Front Immunol (2018) 9:2011. doi: 10.3389/mmu.2018.02011
105. Horstmann RD, Sievertsen HJ, Knobloch J, Fischetti VA. Antiphagocytic
Activity of Streptococcal M Protein: Selective Binding of Complement
Control Protein Factor H. Proc Natl Acad Sci USA (1988) 85:165761.
doi: 10.1073/pnas.85.5.1657
106. Kihlberg BM, Collin M, Olsen A, Bjorck L. Protein H, an Antiphagocytic
Surface Protein in Streptococcus Pyogenes. Infect Immun (1999) 67:170814.
doi: 10.1128/IAI.67.4.1708-1714.1999
107. Tuffs SW, Haeryfar SMM, McCormick JK. Manipulation of Innate and
Adaptive Immunity by Staphylococcal Superantigens. Pathogens (2018) 7:1
23. doi: 10.3390/pathogens7020053
108. Patella V, Casolaro V, Bjorck L, Marone G, Protein L. A Bacterial Ig-Binding
Protein That Activates Human Basophils and Mast Cells. J Immunol (1990)
145:305461.
109. Genovese A, Bouvet JP, Florio G, Lamparter-Schummert B, Bjorck L,
Marone G. Bacterial Immunoglobulin Superantigen Proteins A and L
Activate Human Heart Mast Cells by Interacting With Immunoglobulin E.
Infect Immun (2000) 68:551724. doi: 10.1128/iai.68.10.5517-5524.2000
110. Genovese A, Borgia G, Bjorck L, Petraroli A, de Paulis A, Piazza M, et al.
Immunoglobulin Superantigen Protein L Induces IL-4 and IL-13 Secretion
From Human Fc Epsilon RI+ Cells Through Interaction With the Kappa
Light Chains of IgE. J Immunol (2003) 170:185461. doi: 10.4049/
jimmunol.170.4.1854
111. Patel AH, Nowlan P, Weavers ED, Foster T. Virulence of Protein A-Decient
and Alpha-Toxin-Decient Mutants of Staphylococcus Aureus Isolated by
Allele Replacement. Infect Immun (1987) 55:310310. doi: 10.1128/
IAI.55.12.3103-3110.1987
112. Derrick JP, Wigley DB. Crystal Structure of a Streptococcal Protein G Domain
Bound to an Fab Fragment. Nature (1992) 359:7524. doi: 10.1038/359752a0
113. Graille M, Stura EA, Corper AL, Sutton BJ, Taussig MJ, Charbonnier JB, et al.
Crystal Structure of a Staphylococcus Aureus Protein A Domain Complexed
With the Fab Fragment of a Human IgM Antibody: Structural Basis for
Recognition of B-Cell Receptors and Superantigen Activity. Proc Natl Acad
Sci USA (2000) 97:5399404. doi: 10.1073/pnas.97.10.5399
114. Nilson BH, Solomon A, Bjorck L, Akerstrom B. Protein L From
Peptostreptococcus Magnus Binds to the Kappa Light Chain Variable
Domain.JBiolChem(1992)267:22349. doi:10.1016/S0021-9258(18)45867-X
115. Housden NG, Harrison S, Housden HR, Thomas KA, Beckingham JA,
Roberts SE, et al. Observation and Characterization of the Interaction
Between a Single Immunoglobulin Binding Domain of Protein L and Two
Equivalents of Human Kappa Light Chains. J Biol Chem (2004) 279:93708.
doi: 10.1074/jbc.M312938200
116. Domiati-Saad R, Lipsky PE. Staphylococcal Enterotoxin A Induces Survival
of VH3-Expressing Human B Cells by Binding to the VH Region With Low
Afnity. J Immunol (1998) 161:125766.
117. Domiati-Saad R, Attrep JF, Brezinschek HP, Cherrie AH, Karp DR, Lipsky
PE. Staphylococcal Enterotoxin D Functions as a Human B Cell
Superantigen by Rescuing VH4-Expressing B Cells From Apoptosis.
J Immunol (1996) 156:360820.
118. Bunker JJ, Drees C, Watson AR, Plunkett CH, Nagler CR, Schneewind O,
et al. B Cell Superantigens in the Human Intestinal Microbiota. Sci Transl
Med (2019) 11:127. doi: 10.1126/scitranslmed.aau9356
119. Jansson B, Uhlen M, Nygren PA. All Individual Domains of Staphylococcal
Protein A Show Fab Binding. FEMS Immunol Med Microbiol (1998) 20:69
78. doi: 10.1111/j.1574-695X.1998.tb01112.x
120. Derrick JP, Maiden MC, Feavers IM. Crystal Structure of an Fab Fragment in
Complex With a Meningococcal Serosubtype Antigen and a Protein G
Domain. J Mol Biol (1999) 293:8191. doi: 10.1006/jmbi.1999.3144
121. Derrick JP, Wigley DB. The Third IgG-Binding Domain From Streptococcal
Protein G. An Analysis by X-Ray Crystallography of the Structure Alone and
in a Complex With Fab. JMolBiol(1994) 243:90618. doi: 10.1006/
jmbi.1994.1691
122. Graille M, Stura EA, Housden NG, Beckingham JA, Bottomley SP, Beale D, et al.
Complex Between Peptostreptococcus Magnus Protein L and a Human Antibody
Reveals Structural Convergence in the Interaction Modes of Fab Binding
Proteins. Structure (2001) 9:67987. doi: 10.1016/s0969-2126(01)00630-x
123. Beckingham JA, Housden NG, Muir NM, Bottomley SP, Gore MG. Studies
on a Single Immunoglobulin-Binding Domain of Protein L From
Peptostreptococcus Magnus: The Role of Tyrosine-53 in the Reaction With
Human IgG. Biochem J (2001) 353:395401. doi: 10.1042/0264-6021:3530395
124. Svensson HG, Wedemeyer WJ, Ekstrom JL, Callender DR, Kortemme T,
Kim DE, et al. Contributions of Amino Acid Side Chains to the Kinetics and
Thermodynamics of the Bivalent Binding of Protein L to Ig Kappa Light
Chain. Biochemistry (2004) 43:244557. doi: 10.1021/bi034873s
125. Su CT, Ling WL, Lua WH, Poh JJ, Gan SK. The Role of Antibody Vkappa
Framework 3 Region Towards Antigen Binding: Effects on Recombinant
Production and Protein L Binding. Sci Rep (2017) 7:3766. doi: 10.1038/
s41598-017-02756-3
126. Ling WL, Lua WH, Poh JJ, Yeo JY, Lane DP, Gan SK. Effect of VH-VL
Families in Pertuzumab and Trastuzumab Recombinant Production, Her2
and FcgammaIIA Binding. Front Immunol (2018) 9:469. doi: 10.3389/
mmu.2018.00469
127. Ling WL, Su CT, Lua WH, Poh JJ, Ng YL, Wipat A, et al. Essentially Leading
AntibodyProduction: An Investigation of Amino Acids, Myeloma, and Natural
V-Region Signal Peptides in Producing Pertuzumab and Trastuzumab
Variants. Front Immunol (2020) 11:604318. doi: 10.3389/mmu.2020.604318
128. Salvalaglio M, Zamolo L, Busini V, Moscatelli D, Cavallotti C. Molecular
Modeling of Protein A Afnity Chromatography. J Chromatogr A (2009)
1216:867886. doi: 10.1016/j.chroma.2009.04.035
129. Kim HK, Cheng AG, Kim HY, Missiakas DM, Schneewind O. Nontoxigenic
ProteinA Vaccine forMethicillin-ResistantStaphylococcusAureus Infections in
Mice. J Exp Med (2010) 207:186370. doi: 10.1084/jem.20092514
130. Stone GC, Sjobring U, Bjorck L, Sjoquist J, Barber CV, Nardella FA. The Fc
Binding Site for Streptococcal Protein G Is in the C Gamma 2-C Gamma 3
Interface Region of IgG and is Related to the Sites That Bind Staphylococcal
Protein A and Human Rheumatoid Factors. J Immunol (1989) 143:56570.
131. Kato K, Lian LY, Barsukov IL, Derrick JP, Kim H, Tanaka R, et al. Model for
the Complex Between Protein G and an Antibody Fc Fragment in Solution.
Structure (1995) 3:7985. doi: 10.1016/s0969-2126(01)00136-8
132. Nooren IM, Thornton JM. Diversity of Protein-Protein Interactions. EMBO J
(2003) 22:348692. doi: 10.1093/emboj/cdg359
133. Chinh W-HL, Su T, Poh J-J,Ling W-L, Yeo JY, Gan SK. Molecular Insights of
Nickel Binding to Therapeutic Antibodies as a Possible New Antibody
Superantigen. FrontImmunol (2021) 12:111. doi: 10.3389/mmu.2021.676048
134. Grilo AL, Mantalaris A. The Increasingly Human and Protable Monoclonal
Antibody Market. Trends Biotechnol (2019) 37:916. doi: 10.1016/j.tibtech.
2018.05.014
135. Kangwa M, Yelemane V, Ponnurangam A, Fernandez-Lahore M. An
Engineered Staphylococcal Protein A Based Ligand: Production,
Characterization and Potential Application for the Capture of
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 73184515
Immunoglobulin and Fc-Fusion Proteins. Protein Expr Purif (2019) 155:27
34. doi: 10.1016/j.pep.2018.11.003
136. Inganas M. Comparison of Mechanisms of Interaction Between Protein A
From Staphylococcus Aureus and Human Monoclonal IgG, IgA and IgM in
Relation to the Classical FC Gamma and the Alternative F(Ab)2 Epsilon
Protein A Interactions. Scand J Immunol (1981) 13:34352. doi: 10.1111/
j.1365-3083.1981.tb00143.x
137. Ey PL, Prowse SJ, Jenkin CR. Isolation of Pure IgG1, IgG2a and IgG2b
Immunoglobulins From Mouse Serum Using Protein A-Sepharose.
Immunochemistry (1978) 15:42936. doi: 10.1016/0161-5890(78)90070-6
138. Nilsson B, Moks T, Jansson B, Abrahmsen L, Elmblad A, Holmgren E, et al.
A Synthetic IgG-Binding Domain Based on Staphylococcal Protein a. Protein
Eng (1987) 1:10713. doi: 10.1093/protein/1.2.107
139. Starovasnik MA, OConnell MP, Fairbrother WJ, Kelley RF. Antibody
Variable Region Binding by Staphylococcal Protein A: Thermodynamic
Analysis and Location of the Fv Binding Site on E-Domain. Protein Sci
(1999) 8:142331. doi: 10.1110/ps.8.7.1423
140. Seldon TA, Hughes KE, Munster DJ, Chin DY, Jones ML. Improved Protein-
A Separation of V(H)3 Fab From Fc After Papain Digestion of Antibodies.
J Biomol Tech (2011) 22:502.
141. Linhult M, Gulich S, Graslund T, Simon A, Karlsson M, Sjoberg A, et al.
Improving the Tolerance of a Protein a Analogue to Repeated Alkaline
Exposures Using a Bypass Mutagenesis Approach. Proteins (2004) 55:407
16. doi: 10.1002/prot.10616
142. Minakuchi K, Murata D, Okubo Y, Nakano Y, Yoshida S. Remarkable
Alkaline Stability of an Engineered Protein A as Immunoglobulin Afnity
Ligand: C Domain Having Only One Amino Acid Substitution. Protein Sci
(2013) 22:12308. doi: 10.1002/pro.2310
143. Xia HF, Liang ZD, Wang SL, Wu PQ, Jin XH. Molecular Modication of
Protein A to Improve the Elution pH and Alkali Resistance in Afnity
Chromatography. Appl Biochem Biotechnol (2014) 172:400212.
doi: 10.1007/s12010-014-0818-1
144. Gulich S, Linhult M, Stahl S, Hober S. Engineering Streptococcal Protein G
for Increased Alkaline Stability. Protein Eng (2002) 15:83542. doi: 10.1093/
protein/15.10.835
145. Palmer B, Angus K, Taylor L, Warwicker J, Derrick JP. Design of Stability at
Extreme Alkaline pH in Streptococcal Protein G. JBiotechnol(2008)
134:22230. doi: 10.1016/j.jbiotec.2007.12.009
146. Imura Y, Tagawa T, Miyamoto Y, Nonoyama S, Sumichika H, Fujino Y, et al.
Washing With Alkaline Solutions in Protein A Purication Improves
Physicochemical Properties of Monoclonal Antibodies. Sci Rep (2021)
11:1827. doi: 10.1038/s41598-021-81366-6
147. Akerstrom B, Nilson BH, Hoogenboom HR, Bjorck L. On the Interaction
Between Single Chain Fv Antibodies and Bacterial Immunoglobulin-Binding
Proteins. J Immunol Methods (1994) 177:15163. doi: 10.1016/0022-1759
(94)90152-x
148. Nielsen K, Smith P, Yu W, Nicoletti P, Elzer P, Vigliocco A, et al. Enzyme
Immunoassay for the Diagnosis of Brucellosis: Chimeric Protein A-Protein G
as a Common EnzymeLabeled Detection Reagent for Sera for Different Animal
Species. Vet Microbiol (2004) 101:1239. doi: 10.1016/j.vetmic.2004.02.014
149. Urmann K, Reich P, Walter JG, Beckmann D, Segal E, Scheper T. Rapid and
Label-Free Detection of Protein a by Aptamer-Tethered Porous Silicon
Nanostructures. JBiotechnol(2017) 257:1717. doi: 10.1016/j.jbiotec.2017.01.005
150. Yue H, Zhou Y, Wang P, Wang X, Wang Z, Wang L, et al. A Facile Label-
Free Electrochemiluminescent Biosensor for Specic Detection of
Staphylococcus Aureus Utilizing the Binding Between Immunoglobulin G
and Protein a. Talanta (2016) 153:4016. doi: 10.1016/j.talanta.2016.03.043
151. Ling WL, Ng YL, Wipat A, Lane DP, Gan SK. The Quantication of
Antibody Elements and Receptors Subunit Expression Using qPCR: The
Design of VH, VL, CH, CL, FcR Subunits Primers for a More Holistic View
of the Immune System. J Immunol Methods (2020) 476:112683. doi: 10.1016/
j.jim.2019.112683
152. Ng K, Liang W, Liew A, Yeo Y-H, Lua J, Qian W-H, et al. Republication
APD SpectBT: Arduino-Based Mobile Vis-Spectrophotometer. Sci Phone
Apps Mobile Device (2019) 5:14. doi: 10.30943/2019/s23122019
153. Poh J-JG, Tan N, Gan S, Ken-En S. Spectrophotometer on-the-Go: The
Development of a 2-in-1 UVVis Portable Arduino-Based Spectrophotometer.
Sensors Actuators A: Phys (2021) 325:18. doi: 10.1016/j.sna.2021.112698
154. Salgado-Pabon W, Breshears L, Spaulding AR, Merriman JA, Stach CS,
Horswill AR, et al. Superantigens are Critical for Staphylococcus Aureus
Infective Endocarditis, Sepsis, and Acute Kidney Injury. mBio (2013) 4:19.
doi: 10.1128/mBio.00494-13
155. Bashraheel SS, AlQahtani AD, RashidiFB, Al-Sulaiti H, Domling A, Orie NN,
et al. Studies on Vascular Response to Full Superantigens and Superantigen
Derived Peptides: Possible Production of Novel Superantigen Variants With
Less Vasodilation Effect for Tolerable Cancer Immunotherapy. BioMed
Pharmacother (2019) 115:108905. doi: 10.1016/j.biopha.2019.108905
156. Yang Y, Qian M, Yi S, Liu S, Li B, Yu R, et al. Monoclonal Antibody
Targeting Staphylococcus Aureus Surface Protein A (SasA) Protect Against
Staphylococcus Aureus Sepsis and Peritonitis in Mice. PloS One (2016) 11:
e0149460. doi: 10.1371/journal.pone.0149460
157. Varshney AK, Kuzmicheva GA, Lin J, Sunley KM, Bowling RA Jr, Kwan TY,
et al. A Natural Human Monoclonal Antibody Targeting Staphylococcus
Protein A Protects Against Staphylococcus Aureus Bacteremia. PloS One
(2018) 13:e0190537. doi: 10.1371/journal.pone.0190537
158. Forsberg G, Ohlsson L, Brodin T, Bjork P, Lando PA, Shaw D, et al. Therapy
of Human Non-Small-Cell Lung Carcinoma Using Antibody Targeting of a
Modied Superantigen. Br J Cancer (2001) 85:12936. doi: 10.1054/
bjoc.2001.1891
159. von Scheidt B, Wang M, Oliver AJ, Chan JD, Jana MK, Ali AI, et al.
Enterotoxins Can Support CAR T Cells Against Solid Tumors. Proc Natl
Acad Sci USA (2019) 116:2522935. doi: 10.1073/pnas.1904618116
160. Lorenzo D, Duarte A, Mundinano J, Berguer P, Nepomnaschy I, Piazzon I. A
B-Cell Superantigen Induces the Apoptosis of Murine and Human
Malignant B Cells. PloS One (2016) 11:e0162456. doi: 10.1371/journal.
pone.0162456
161. Chen JY. Superantigens, Superantigen-Like Proteins and Superantigen
Derivatives for Cancer Treatment. Eur Rev Med Pharmacol Sci (2021)
25:162230. doi: 10.26355/eurrev_202102_24873
162. Heidary MF, Mahmoodzadeh Hosseini H, Mehdizadeh Aghdam E, Nourani
MR, Ranjbar R, Mirnejad R, et al. Overexpression of Metastatic Related
MicroRNAs, Mir-335 and Mir-10b, by Staphylococcal Enterotoxin B in the
Metastatic Breast Cancer Cell Line. Adv Pharm Bull (2015) 5:2559.
doi: 10.15171/apb.2015.035
163. Golob-Urbanc A, Rajcevic U, Strmsek Z, Jerala R. Design of Split
Superantigen Fusion Proteins for Cancer Immunotherapy. J Biol Chem
(2019) 294:6294305. doi: 10.1074/jbc.RA118.006742
164. He Y, Sun Y, Ren Y, Qiao L, Guo R, Du J, et al. The T Cell Activating Properties
and Antitumour Activity of Staphylococcal Enterotoxin-Like Q. Med Microbiol
Immunol (2019) 208:78192. doi: 10.1007/s00430-019-00614-9
165. Brown M, Bhardwaj N. Super(antigen) Target for SARS-CoV-2. Nat Rev
Immunol (2021) 21:72. doi: 10.1038/s41577-021-00502-5
166. Cheng MH, Zhang S, Porritt RA, Noval Rivas M, Paschold L, Willscher E,
et al. Superantigenic Character of an Insert Unique to SARS-CoV-2 Spike
Supported by Skewed TCR Repertoire in Patients With Hyperinammation.
Proc Natl Acad Sci USA (2020) 117:2525462. doi: 10.1073/pnas.2010722117
167. Kouo T, Chaisawangwong W. SARS-CoV-2 as a Superantigen in
Multisystem Inammatory Syndrome in Children. J Clin Invest (2021)
131. doi: 10.1172/JCI149327
Conict of Interest: The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be construed as a
potential conict of interest.
Publishers Note: All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated organizations, or those of
the publisher, the editors and the reviewers. Any product that may be evaluated in
this article, or claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
Copyright © 2021 Deacy, Gan and Derrick. This is an open-access article distributed
under the terms of the Creative Commons Attribution License (CC BY). The
use, distribution or reproduction in other forums is permitted, provided the original
author(s) and the copyright owner(s) are credited and that the original publication in
this journal is cited, in accordance with accepted academic practice. No use,
distribution or reproduction is permitted which does not comply with these terms.
Deacy et al. Superantigen Recognition and Interactions
Frontiers in Immunology | www.frontiersin.org September 2021 | Volume 12 | Article 73184516
... While the impact of HIV on the immune system is not yet fully understood, the direct interaction of envelope glycoprotein gp120 and CD4 renders primed CD4+ T helper cells susceptible to activation-induced apoptosis while promoting Treg subset survival. Superantigens such as staphylococcal enterotoxin B (SEB) are potent T-cell mitogens that can non-specifically stimulate polyclonal T cells, resulting in massive systemic activation of T cells and the release of pro-inflammatory cytokines [62]. The enterotoxins produced by Staphylococcus aureus and Streptococcus pyogenes can directly bind T cell receptors (TCRs), bypassing antigen processing and presentation. ...
... The enterotoxins produced by Staphylococcus aureus and Streptococcus pyogenes can directly bind T cell receptors (TCRs), bypassing antigen processing and presentation. When stimulated with superantigens, mature CD4+ and CD8+ T lymphocytes proliferate and overwhelm the surrounding environment with pro-inflammatory cytokines, resulting in severe systemic pathologies [62]. In conclusion, a small number of pathogens can directly attack immune cells and cause immunological dysfunction by activating and depleting immune cells, as well as changing their functions, resulting in a variety of diseases. ...
Article
Full-text available
The optimal immune response eliminates invading pathogens, restoring immune equilibrium without inflicting undue harm to the host. However, when a cascade of immunological reactions is triggered, the immune response can sometimes go into overdrive, potentially leading to harmful long-term effects or even death. The immune system is triggered mostly by infections, allergens, or medical interventions such as vaccination. This review examines how these immune triggers differ and why certain infections may dysregulate immune homeostasis, leading to inflammatory or allergic pathology and exacerbation of pre-existing conditions. However, many vaccines generate an optimal immune response and protect against the consequences of pathogen-induced immunological aggressiveness, and from a small number of unrelated pathogens and autoimmune diseases. Here, we propose an “immuno-wave” model describing a vaccine-induced “Goldilocks immunity”, which leaves fine imprints of both pro-inflammatory and anti-inflammatory milieus, derived from both the innate and the adaptive arms of the immune system, in the body. The resulting balanced, ‘quiet alert’ state of the immune system may provide a jump-start in the defense against pathogens and any associated pathological inflammatory or allergic responses, allowing vaccines to go above and beyond their call of duty. In closing, we recommend formally investigating and reaping many of the secondary benefits of vaccines with appropriate clinical studies.
... MHC-II and TCR are two major receptors for SEB, and the interactions between these molecules are crucial as they form the basis of the ternary complex that is pivotal in the pathogenic mechanism of SEB. 24 Therefore, we investigated whether Hm0487, upon binding to SEB, could affect the binding of SEB to these receptors. As shown in Figure 3d,e, the BLI response signal clearly showed the interactions between SEB and both receptors. ...
Article
Full-text available
Staphylococcal Enterotoxin B (SEB), produced by Staphylococcus aureus (S. aureus), is a powerful superantigen that induces severe immune disruption and toxic shock syndrome (TSS) upon binding to MHC-II and TCR. Despite its significant impact on the pathogenesis of S. aureus, there are currently no specific therapeutic interventions available to counteract the mechanism of action exerted by this toxin. In this study, we have identified a human monoclonal antibody, named Hm0487, that specifically targets SEB by single-cell sequencing using PBMCs isolated from volunteers enrolled in a phase I clinical trial of the five-antigen S. aureus vaccine. X-ray crystallography studies revealed that Hm0487 exhibits high affinity for a linear B cell epitope in SEB (SEB138–147), which is located distantly from the site involved in the formation of the MHC-SEB-TCR ternary complex. Furthermore, in vitro studies demonstrated that Hm0487 significantly impacts the interaction of SEB with both receptors and the binding to immune cells, probably due to an allosteric effect on SEB rather than competing with receptors for binding sites. Moreover, both in vitro and in vivo studies validated that Hm0487 displayed efficient neutralizing efficacy in models of lethal shock and sepsis induced by either SEB or bacterial challenge. Our findings unveil an alternative mechanism for neutralizing the pathogenesis of SEB by Hm0487, and this antibody provides a novel strategy for mitigating both SEB-induced toxicity and S. aureus infection.
... histocompatibility complex molecules, show the way to release of massive mediators of T cell (Akshay, 2019;Anthony et al., 2021), depending on virulence and sequalae of bacteria post infection can categorized into two classes: Class I strains cause suppurative inflammation such as meningitis and abscess in both brain and liver while class II strains cause non suppurative inflammation like acute glomerulonephritis and rheumatic fever (Niluni et al., 2021;Patience et al., 2023). ...
Article
Background Concanavalin-A is a plant lectin neither antibodies, nor enzymes which has four binding sites for glucose and act as an antigen-independent mitogen, frequently used to stimulate proliferation of T-cells and activate the immune response. Aims The present study conducted for showing effect of Concanavalin-A, whole killed lyophilized Streptococcus pyogenes antigen(WKLA) on immune response of rat. Methods To achieve this purpose, 20 Albino male rats were divided equally into four groups and immunized subcutaneously(s/c), 1st group was immunized with (100 mg/ml) of S. pyogenes antigen with added equal volume of Freund’s adjuvant, 2 doses, 14 days intervals, 2nd group was immunized s/c with Concanavalin-A Con-A (20 mg/ml), 3rd group was immunized with mixed S. pyogenes Ag. and Con-A, 2 doses, 14 days intervals, and 4th group was given Phosphate buffer saline (PBS) as negative control group. Results Analysis of cellular and humeral immunity recorded higher concentration of Immunoglobulin G IgG, Tumer Necrosis Factor alpha TNF-a and Interleukin 10 IL-10 were revealed in 3rd immunized group (40.91 ±0.26, 953.03 ±4.82 and 1223.96 ±10.44) then decline in 2nd and 1st groups as (32.65 ±1.07, 858.42±9.26 and 1223.96±10.44) ( 21.07 ±0.36, 769.38 ±3.64 and 986.73 ±13.48) respectively. Histopathological findings revealed marked lymphoid hyperplasia with obvious perivascular Mono Nuclear Cells MNCs aggregation composed of lymphocyte and macrophage mainly in hepatic tissue of 3rd group with evidence of reactive lymphoid hyperplasia in splenic tissue of 2nd group and periarteriolar, pericortical lymphoid hyperplasia were reported in spleen of 1st group immunized with Streptococcus pyogenes lyophilized antigen. Conclusion This study provide we can conclude that jointly of concanavalin-A and S.pyogenes antigen capable to enhance potential immune response butter than antigen alone.
... There is some literature on the mechanisms of the MHCII and TCR interactions with SEs and their application to the diagnosis and treatment of SE poisoning. There is also some literature on the use of T cell receptor structural domains for capturing the ELISA detection of SEs [11,12]. In this paper, we designed two capture molecules aiming to replace the traditional antibodies used in the LC-MS/MS detection of SEs, thereby making detection more efficient. ...
Article
Full-text available
Food poisoning caused by Staphylococcal enterotoxins (SEs) is prevalent globally, making efficient detection of these toxins very important. Traditionally, liquid chromatography–mass spectrometry required immunosorbent enrichment by magnetic bead-coupled antibodies obtained by animal-specific immunization. However, this method is time-consuming and costly. In this study, two recombinant protein capture molecules were designed based on the principle of toxins binding to Major Histocompatibility Complex (MHCII) and T cell receptor (TCR) molecules. The two capture molecules are called MHCII and MHCII-D10. The design of the MHCII and TCR-D10 was achieved through searching for the binding site protein sequence of Staphylococcal enterotoxins in the relevant literature, and MHCII-D10 was to link MHCII sequence with TCR-D10 sequence using linker (G4S)3 linking peptide. These capture molecules were shown to effectively bind to seven types of toxins and to capture SEs in various matrices. The digestion time, ratio, and temperature were further optimized, reducing the overall digestion time to just 2 h. The specificity, linearity, sensitivity, precision (RSD%), and recovery of the two methods were verified by liquid chromatography–mass spectrometry. When the MHCII and MHCII-D10 captured the toxins, the limit of quantification (LOD) in the 1 × PBS, plasma, and milk matrices ranged from 1.5625 to 100 fmol/µL, with the recovery rate ranging from 18.4% to 96%. The design of these capture molecules eliminates the need for animal-specific immunization, simplifying the pre-detection process and avoiding ethical concerns. This development holds significant promise for clinical diagnosis and reference.
... Superantigens are typically produced by bacteria, viruses, and other microorganisms. They broadly activate T cells by bridging MHC-II and TCR, overwriting antigen restriction between an antigen presenting cell and a T cell 23 . MAM is a superantigen with a unique binding mode to the MHC-II (Protein Data Bank (PDB) ID: 2ICW 22 ) featuring an L-shaped structure consisting of two domains. ...
Preprint
Full-text available
Class-II major histocompatibility complexes (MHC-IIs) are central to the communications between CD4+ T cells and antigen presenting cells (APCs), but intrinsic structural features associated with MHC-II make it difficult to develop a general targeting system with high affinity and antigen specificity. Here, we introduce a protein platform, Targeted Recognition of Antigen-MHC Complex Reporter for MHC-II (TRACeR-II), to enable the rapid development of peptide-specific MHC-II binders. TRACeR-II has a small helical bundle scaffold and uses an unconventional mechanism to recognize antigens via a single loop. This unique antigen-recognition mechanism renders this platform highly versatile and amenable to direct structural modeling of the interactions with the antigen. We demonstrate that TRACeR-II binders can be rapidly evolved across multiple alleles, while computational protein design can produce specific binding sequences for a SARS-CoV-2 peptide of unknown complex structure. TRACeR-II sheds light on a simple and straightforward approach to address the MHC peptide targeting challenge, without relying on combinatorial selection on complementarity determining region (CDR) loops. It presents a promising basis for further exploration in immune response modulation as well as a broad range of theragnostic applications.
... Some researchers have suggested that this region may have super antigenic properties, which could potentially contribute to the severe immune response observed in some COVID-19 patients, leading to cytokine storms and multiorgan failure [35][36][37]. Superantigen-mediated T cell expansion occurs through a less specific interaction with TCRs, leading to the activation of a large proportion of the T-cell population [38]. Although the super antigenic character of the S673-699 region is still being investigated, it is possible that the efficacy of the mRNA COVID-19 vaccines in preventing severe disease and death is also due to reduced exposure to the super antigenic viral determinant, through neutralizing antibodies or reduced viral replication. ...
Article
Full-text available
Background The evolving variants of SARS-CoV-2 may escape immunity from prior infections or vaccinations. It’s vital to understand how immunity adapts to these changes. Both infection and mRNA vaccination induce T cells that target the Spike protein. These T cells can recognize multiple variants, such as Delta and Omicron, even if neutralizing antibodies are weakened. However, the degree of recognition can vary among people, affecting vaccine efficacy. Previous studies demonstrated the capability of T-cell receptor (TCR) repertoire analysis to identify conserved and immunodominant peptides with cross-reactive potential among variant of concerns. However, there is a need to extend the analysis of the TCR repertoire to different clinical scenarios. The aim of this study was to examine the Spike-specific TCR repertoire profiles in natural infections and those with combined natural and vaccine immunity. Methods A T-cell enrichment approach and bioinformatic tools were used to investigate the Spike-specific TCRβ repertoire in peripheral blood mononuclear cells of previously vaccinated (n = 8) or unvaccinated (n = 6) COVID-19 patients. Results Diversity and clonality of the TCRβ repertoire showed no significant differences between vaccinated and unvaccinated groups. When comparing the TCRβ data to public databases, 692 unique TCRβ sequences linked to S epitopes were found in the vaccinated group and 670 in the unvaccinated group. TCRβ clonotypes related to spike regions S135-177, S264-276, S319-350, and S448-472 appear notably more prevalent in the vaccinated group. In contrast, the S673-699 epitope, believed to have super antigenic properties, is observed more frequently in the unvaccinated group. In-silico analyses suggest that mutations in epitopes, relative to the main SARS-CoV-2 variants of concern, don’t hinder their cross-reactive recognition by associated TCRβ clonotypes. Conclusions Our findings reveal distinct TCRβ signatures in vaccinated and unvaccinated individuals with COVID-19. These differences might be associated with disease severity and could influence clinical outcomes. Trial registration: FESR/FSE 2014–2020 DDRC n. 585, Action 10.5.12, noCOVID19@UMG.
... By binding to specific Vβ chains of the T-cell receptor, superantigens can activate up to 30% of the mature T-cell pool and induce an extensive proliferation that ends in apoptosis of the stimulated cells and to a state of anergy of the remaining cells (for review [22]). It has been shown later that superantigens also stimulate B cells by binding to complementary determining regions [23]. ...
Article
Full-text available
Background: The neuro-endocrine regulation of immune functions is based on a complex network of interactions. As part of this series of articles, we refer here to immune-sympathetic interactions that are triggered by different types of immune challenge. Summary: We mention the initial hypothesis that led to the proposal that the sympathetic nervous system (SNS) is involved in immunoregulation. We next refer mainly to our initial work performed at a time when most immunologist were concentrated in clarifying aspects of the immune system that are essential for its regulation from within. The first approach was to explore whether immune responses to innocuous antigens and superantigens can elicit changes in the activity of the SNS, and their potential relevance for the regulation of the activity of the immune system. The following step was to explore whether comparable immune-SNS interactions are detected in different models of diseases with immune components, such as parasitic and viral infections and autoimmune pathologies. Key Messages: We pose some general considerations that may at least partially explain seemly discrepant findings, and remark the importance of interpreting immunoregulatory effects of the SNS together with other neuro-endocrine inputs that simultaneously occur when the activity of the immune system changes. Finally, we provide some arguments to re-consider the use of the expression “reflex” in immunology.
Article
Full-text available
Aplastic anemia (AA) and hypoplastic myelodysplastic syndrome are paradigms of autoimmune hematopoietic failure (AHF). Myelodysplastic syndrome and acute myeloid leukemia are unequivocal myeloid neoplasms (MNs). Currently, AA is also known to be a clonal hematological disease. Genetic aberrations typically observed in MNs are detected in approximately one-third of AA patients. In AA patients harboring MN-related genetic aberrations, a poor response to immunosuppressive therapy (IST) and an increased risk of transformation to MNs occurring either naturally or after IST are predicted. Approximately 10%–15% of patients with severe AA transform the disease phenotype to MNs following IST, and in some patients, leukemic transformation emerges during or shortly after IST. Phenotypic transformations between AHF and MNs can occur reciprocally. A fraction of advanced MN patients experience an aplastic crisis during which leukemic blasts are repressed. The switch that shapes the disease phenotype is a change in the strength of extramedullary inflammation. Both AHF and MNs have an immune-active bone marrow (BM) environment (BME). In AHF patients, an inflamed BME can be evoked by infiltrated immune cells targeting neoplastic molecules, which contributes to the BM-specific autoimmune impairment. Autoimmune responses in AHF may represent an antileukemic mechanism, and inflammatory stressors strengthen antileukemic immunity, at least in a significant proportion of patients who have MN-related genetic aberrations. During active inflammatory episodes, normal and leukemic hematopoieses are suppressed, which leads to the occurrence of aplastic cytopenia and leukemic cell regression. The successful treatment of underlying infections mitigates inflammatory stress-related antileukemic activities and promotes the penetration of leukemic hematopoiesis. The effect of IST is similar to that of treating underlying infections. Investigating inflammatory stress-powered antileukemic immunity is highly important in theoretical studies and clinical practice, especially given the wide application of immune-activating agents and immune checkpoint inhibitors in the treatment of hematological neoplasms.
Article
Full-text available
The binding of nickel by immune proteins can manifest as Type IV contact dermatitis (Ni-specific T cells mediated) and less frequently as Type I hypersensitivity with both mechanisms remaining unknown to date. Since there are reports of patients co-manifesting the two hypersensitivities, a common mechanism may underlie both the TCR and IgE nickel binding. Focusing on Trastuzumab and Pertuzumab IgE variants as serendipitous investigation models, we found Ni-NTA interactions independent of Her2 binding to be due to glutamine stretches. These stretches are both Ni-inducible and in fixed pockets at the antibody complementarity-determining regions (CDRs) and framework regions (FWRs) of both the antibody heavy and light chains with influence from the heavy chain constant region. Comparisons with TCRs structures revealed similar interactions, demonstrating the possible underlying mechanism in selecting for Ni-binding IgEs and TCRs respectively. With the elucidation of the interaction, future therapeutic antibodies could also be sagaciously engineered to utilize such nickel binding for biotechnological purposes.
Article
Full-text available
Staphylococcus aureus causes reiterative and chronic persistent infections. This can be explained by the formidable ability of this pathogen to escape immune surveillance mechanisms. Cells of S. aureus display the abundant staphylococcal protein A (SpA). SpA binds to immunoglobulin (Ig) molecules and coats the bacterial surface to prevent phagocytic uptake. SpA also binds and cross-links variable heavy 3 (V H 3) idiotype (IgM) B cell receptors, promoting B cell expansion and the secretion of nonspecific V H 3-IgM via a mechanism requiring CD4 ⁺ T cell help. SpA binding to antibodies is mediated by the N-terminal Ig-binding domains (IgBDs). The so-called region X, uncharacterized LysM domain, and C-terminal LPXTG sorting signal for peptidoglycan attachment complete the linear structure of the protein. Here, we report that both the LysM domain and the LPXTG motif sorting signal are required for the B cell superantigen activity of SpA in a mouse model of infection. SpA molecules purified from staphylococcal cultures are sufficient to exert B cell superantigen activity and promote immunoglobulin secretion as long as they carry intact LysM and LPXTG motif domains with bound peptidoglycan fragments. The LysM domain binds the glycan chains of peptidoglycan fragments, whereas the LPXTG motif is covalently linked to wall peptides lacking glycan. These findings emphasize the complexity of SpA interactions with B cell receptors. IMPORTANCE The LysM domain is found in all kingdoms of life. While their function in mammals is not known, LysM domains of bacteria and their phage parasites are associated with enzymes that cleave or remodel peptidoglycan. Plants recognize microbe-associated molecular patterns such as chitin via receptors endowed with LysM-containing ectodomains. In plants, such receptors play equally important roles in defense and symbiosis signaling. SpA of S. aureus carries a LysM domain that binds glycan strands of peptidoglycan to influence defined B cell responses that divert pathogen-specific adaptive immune responses.
Article
Full-text available
Objective: Bacterial superantigens (SAgs) are proteins produced by few types of bacteria that have been linked to several human diseases. Due to their potent in vitro and in vivo tumoricidal effects, they are extensively investigated for oncological applications either alone or in combination with classical anticancer drugs. However, the intrinsic toxicity of natural SAgs stimulated the development of more effective and less toxic SAg-based immunotherapy. This review summarizes our current knowledge on SAg-based immunotherapy including SAg-like proteins and SAg derivatives, as well as their potential alone or with other therapeutic modalities including chemotherapy and radiotherapy.
Article
Full-text available
Protein A affinity chromatography has been widely used for both laboratory scale purification and commercial manufacturing of monoclonal antibodies and Fc-fusion proteins. Protein A purification is specific and efficient. However, there still remain several issues to be addressed, such as incomplete clearance of impurities including host cell proteins, DNA, aggregates, etc. In addition, the effects of wash buffers in protein A purification on the physicochemical characteristics of antibodies have yet to be fully understood. Here we found a new purification protocol for monoclonal antibodies that can improve physicochemical properties of monoclonal antibodies simply by inserting an additional wash step with a basic buffer after the capture step to the conventional protein A purification. The effects of the alkaline wash on monoclonal antibodies were investigated in terms of physicochemical characteristics, yields, and impurity clearance. The simple insertion of an alkaline wash step resulted in protection of antibodies from irreversible aggregation, reduction in free thiols and impurities, an improvement in colloidal and storage stability, and enhanced yields. This new procedure is widely applicable to protein A affinity chromatography of monoclonal antibodies.
Article
Full-text available
Staphylococcus aureus is a member of the human commensal microflora that exists, apparently benignly, at multiple sites on the host. However, as an opportunist pathogen it can also cause a range of serious diseases. This requires an ability to circumvent the innate immune system to establish an infection. Professional phagocytes, primarily macrophages and neutrophils, are key innate immune cells which interact with S. aureus, acting as gatekeepers to contain and resolve infection. Recent studies have highlighted the important roles of macrophages during S. aureus infections, using a wide array of killing mechanisms. In defense, S. aureus has evolved multiple strategies to survive within, manipulate and escape from macrophages, allowing them to not only subvert but also exploit this key element of our immune system. Macrophage-S. aureus interactions are multifaceted and have direct roles in infection outcome. In depth understanding of these host-pathogen interactions may be useful for future therapeutic developments. This review examines macrophage interactions with S. aureus throughout all stages of infection, with special emphasis on mechanisms that determine infection outcome.
Article
Full-text available
Boosting the production of recombinant therapeutic antibodies is crucial in both academic and industry settings. In this work, we investigated the usage of varying signal peptides by antibody V-genes and their roles in recombinant transient production, systematically comparing myeloma and the native signal peptides of both heavy and light chains in 168 antibody permutation variants. We found that amino acids count and types (essential or non-essential) were important factors in a logistic regression equation model for predicting transient co-transfection protein production rates. Deeper analysis revealed that the culture media were often incomplete and that the supplementation of essential amino acids can improve the recombinant protein yield. While these findings are derived from transient HEK293 expression, they also provide insights to the usage of the large repertoire of antibody signal peptides, where by varying the number of specific amino acids in the signal peptides attached to the variable regions, bottlenecks in amino acid availability can be mitigated.
Article
Staphylococcal enterotoxin B (SEB) is a bacterial superantigen that induces the production of several pro-inflammatory cytokines, leading to a self-limited shock. In the present study, we show that SEB also triggers the systemic release of IL-10, an anti-inflammatory and immunosuppressive cytokine. Serum IL-10 was undetectable (< 1000 pg/ml) in control BALB/c mice and rose to 8500 +/- 2850 pg/ml (mean +/- SEM) 4 h after injection of 100 micrograms SEB. Cell depletion experiments and analysis of IL-10 mRNA expression indicated that CD4+ cells played a major role in SEB-induced IL-10 production. Pretreatment of mice with neutralizing anti-IL-10 mAb before SEB challenge did not modify the release of TNF but led to increased and sustained IL-2 and IFN-gamma serum levels. Furthermore, although no lethality occurred in mice injected with SEB and control mAb, injection of anti-IL-10 mAb before SEB resulted in a 30% lethality (p < 0.05). This lethality was completely prevented by anti-IFN-gamma mAb injection, indicating that IFN-gamma plays a crucial role in the increased toxicity of SEB in anti-IL-10 mAb-injected mice. We conclude that SEB induces the production of IL-10 by CD4+ cells in vivo and that endogenous IL-10 plays an important immunoregulatory role in this model by down-regulating IL-2 and IFN-gamma production.
Article
Multisystem Inflammatory Syndrome in Children (MIS-C) is a rare but deadly new disease in children that rapidly progresses to hyperinflammation, shock, and can lead to multiple organ failure if unrecognized. It has been found to be temporally associated with the COVID-19 pandemic and is often associated with SARS-CoV-2 exposure in children. In this issue of the JCI, Porritt, Paschold, and Rivas et al. identify restricted T cell receptor (TCR) β-chain variable domain (Vβ) usage in patients with severe MIS-C indicating a potential role for SARS-CoV-2 as a superantigen. These findings suggest that a blood test that determines the presence of specific TCR beta variable gene segments (TRBV) may identify patients at risk for severe MIS-C.
Article
The spectrophotometer is an important common analytical equipment that measures the concentration of analytes via their absorbance of light at specific wavelengths. However, its cost, lack of portability and requirement of constant power can limit its widespread use and, in some cases, affect the accuracy of its measurements in time-sensitive field tests. Here we describe the development of a portable and low-cost Arduino-based 2-in-1 UV-Vis spectrophotometer built from off-the-shelf electronics and 3D printing. The assembled device has an overall dimension of 105 × 90 × 140 mm and has photodiodes capable of detecting light wavelengths at a range of 190 – 1100 nm, transmitting the readings to its corresponding SpectBT UV-Vis Pro smartphone Android app via Bluetooth®. The device sensitivity and limit of detection were compared with a commercial research grade spectrophotometer on the quantification of proteins and nucleic acids. The assembled spectrophotometer – ‘APD UV-Vis SpectBT’ performed better for A595 measurements while retaining comparable performance for A280 UV measurements. Together, these findings support the assembly of portable self-assembled laboratory equipment for greater accessibility and freedom of mobility.
Article
This preprint further characterizes a superantigen motif identified in SARS-CoV-2 spike protein and evaluates a monoclonal antibody targeting this region that can neutralize live virus.