You are on page 1of 20

Epub ahead of print September 27, 2017 - doi:10.1189/jlb.

4MR0617-227RR

Review

Group A streptococcal pharyngitis: immune


responses involved in bacterial clearance
and GAS-associated immunopathologies
Amelia T. Soderholm,* Timothy C. Barnett,*,† Matthew J. Sweet,‡,1 and Mark J. Walker*,1,2
*School of Chemistry and Molecular Biosciences and ‡Institute for Molecular Bioscience, Institute for Molecular Bioscience Centre
for Inflammation and Disease Research, Australian Infectious Diseases Research Centre, The University of Queensland, Brisbane,
Australia; and †Telethon Kids Institute, Perth, Western Australia, Australia
RECEIVED JUNE 7, 2017; REVISED AUGUST 24, 2017; ACCEPTED AUGUST 25, 2017. DOI: 10.1189/jlb.4MR0617-227RR

ABSTRACT Introduction
Streptococcus pyogenes, the Group A Streptococcus S. pyogenes (also designated GAS) is the most common bacterial
(GAS), is the most common cause of bacterial pharyn- cause of acute pharyngitis [1]. Over 616 million incident cases of
gitis in children and adults. Innate and adaptive host GAS pharyngitis are estimated to occur globally each year,
immune responses are fundamental for defense against making this one of the most prevalent bacterial diseases in
streptococcal pharyngitis and are central to the clinical
humans [2]. Transmission primarily occurs through direct
manifestation of disease. Host immune responses also
human-to-human contact, via respiratory droplets or nasal
contribute to the severe poststreptococcal immune
diseases that constitute the major disease burden for
secretions from infected individuals [3]. Illness predominantly
this organism. However, until recently, little was known occurs in children between 5 and 15 yr of age but can also occur
about the host responses elicited during infection. Cel- in adults [4]. Disease severity of GAS pharyngitis is variable, but
lular mediators of innate immunity used during host in most cases, it is a mild, self-limiting disease that resolves within
defense against GAS include epithelial cells, neutrophils, 7 d. Characteristic symptoms include sudden onset of sore throat,
macrophages, and dendritic cells (DCs), which are fever, headache, abdominal pain, nausea, and vomiting [1]. GAS
reported to secrete a number of soluble inflammatory can also asymptomatically infect the oropharynx for weeks to
mediators, such as antimicrobial peptides (AMPs); ei- months at a time, during which time, little or no immune
cosanoids, including PGE2 and leukotriene B4 (LTB4); response to streptococcal antigens is elicited [5, 6]. Approxi-
chemokines; and proinflammatory cytokines. Th1 and
mately 10–20% of school-age children are asymptomatic carriers
Th17 responses play significant roles in adaptive immu-
of GAS [7, 8].
nity in both murine models of GAS pharyngitis and in
human tonsil tissue. A number of inflammatory compli- Upon etiologic diagnosis of an active infection, usually by rapid
cations are associated with GAS pharyngitis, which can antigen-detection tests and/or throat culture, antibiotic treat-
lead to chronic disease in patients. These include scarlet ment is used to reduce the severity and duration of pharyngitis
fever, tonsillar hypertrophy, and sleep apnea, as well as symptoms, as well as to limit disease spread, suppurative
postinfectious sequelae, such as acute rheumatic fever complications (peritonsillar or retropharyngeal abscess, cervical
(ARF), poststreptococcal glomerulonephritis, and lymphadenitis, sinusitus, otitis media, and mastoiditis), and
guttate psoriasis (GP). This review aims to present the development of ARF [9]. Penicillin is the treatment of choice for
current state of knowledge on innate and adaptive GAS pharyngitis, as a result of its efficacy and low cost [10].
immune responses elicited during GAS pharyngitis, There is no preventative vaccine against GAS currently available,
mechanisms by which GAS evades these responses, the
although multiple vaccine candidates are in clinical and pre-
emerging role of the pharyngeal microbiota, and how the
interplay among these factors can influence the outcome
clinical development [11]. A better understanding of immuno-
of infection and inflammation-related complications. logic responses that are effective against GAS could lead to
J. Leukoc. Biol. 103: 000–000; 2018. host-directed preventative or therapeutic regimes.
Previous studies on GAS-related immune responses have
largely focused on those elicited during invasive GAS infections,
where severe morbidity and mortality may result from failure to
Abbreviations: AMP = antimicrobial peptide, ARF = acute rheumatic fever,
AspA = AgI/II family adhesin, BLIS = bacteriocin-like inhibitory substances,
BMDM = bone marrow-derived macrophage, C5a = complement
1. These authors contributed equally to this work.
component 5a, CLA = cutaneous lymphocyte antigen, CysLT = cysteinyl
leukotriene, DC = dendritic cell, GAS = Group A streptococcus, GP = 2. Correspondence: School of Chemistry and Molecular Biosciences, Austra-
lian Infectious Diseases Research Centre, The University of Queensland,
guttate psoriasis, HBD = human b defensin, IFNAR = IFN-a/b receptor,
Cooper Road, St Lucia, QLD 4072, Australia. E-mail: mark.walker@uq.edu.
(continued on next page) au

0741-5400/18/0103-0001 © Society for Leukocyte Biology Volume 103, February 2018 Journal of Leukocyte Biology 1
Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
Copyright 2017 by The Society for Leukocyte Biology.
control bacterial infection [12]. The findings from such studies INNATE IMMUNE RESPONSE TO
are not necessarily reflective of immune responses elicited GAS PHARYNGITIS
during superficial GAS infections, such as pharyngitis and
As with other infections, the innate immune system plays an
impetigo, and indeed, the molecular and cellular bases of host
essential role in the early detection of GAS and subsequent
immune responses to superficial GAS infections are compara-
initiation of proinflammatory responses. GAS pharyngitis is
tively less well understood. Although GAS is exclusively a human
characterized by bacterial colonization of the palatine tonsil, a
pathogen, several animal models that at least partly mimic
specialized lymphoid tissue that contains a number of cell types
human GAS pharyngitis have been established [13–15]. Murine
with innate immune defense roles, including epithelial cells,
nasopharyngeal infection models are most commonly used, with
neutrophils, macrophages, and DCs (Fig. 1). In response to GAS
mice being inoculated with GAS under each nostril. In this
infection, these cell types secrete a number of inflammatory
model, GAS colonizes the NALT, which is functionally equivalent
mediators (Table 1), including cytokines, chemokines, AMPs,
to the human palatine tonsil [15]. Such models have allowed the
and PGs, as part of a highly coordinated host process. These
analysis of immune responses that are associated with either host
mediators orchestrate the ensuing inflammatory response to GAS
protection or susceptibility. For example, transgenic expression
and also instruct the resulting adaptive immune response. Below,
of the human MHC-II molecules HLA-DR4 and HLA-DQ8 in
we discuss some of the key innate cell types and soluble
mice promoted acute nasopharyngeal GAS infection, suggesting
mediators that have been examined in the context of GAS
that MHC-II molecules contribute to the human-specific tropism
pharyngitis, as well as specific mechanisms by which GAS
of a GAS serotype M18 strain [16]. This was mediated, at least in
counteracts their actions.
part, by the GAS superantigen SpeA, suggesting a possible role
for GAS superantigens during human colonization.
In addition to mouse models, several (usually immortalized) Epithelial cells
cell lines have been used to examine GAS-induced inflammatory The pharyngeal and tonsil epithelia are an important component
responses. In vitro studies have been useful in investigating the of innate immune defense against GAS infection, primarily by
immune response of individual cell types following GAS in- acting as a physical barrier but also by secreting AMPs and
fection; however, the variety of cellular models used, as well as cytokines/chemokines to signal to other immune cell mediators.
the dearth of studies in primary cells, makes the interpretation of GAS not only forms biofilms on the surface of the tonsillar
such findings complex. Much of this work has also been done epithelium but can also invade the epithelium to survive
with nonpharyngeal-derived cell lines, such as HEp-2 (HeLa intracellularly [20, 21]. Primary human tonsil fibroblasts and
derivative epithelial cells from cervix) and HaCaT (skin single-cell suspensions of NALT or human tonsil cells secrete
keratinocytes), which may not respond in the same way as TGF-b1 following exposure to GAS, which in concert with IL-6,
pharyngeal cells to GAS infection. Fortunately, follow-up studies secreted by GAS-infected epithelial cells and macrophages, drives
that have used primary tonsil or pharyngeal-derived cells appear CD4+ Th17 cell differentiation [22, 23]. TGF-b1 also regulates
to be largely in agreement with studies that used nonpharyngeal the expression of extracellular matrix proteins, including
cell lines for a wide number of immune mediators secreted in fibronectin and ab integrins [24, 25], and TGF-b1 signaling has
response to GAS infection, such as IL-6, TNF-a, and IL-8 been shown to enhance GAS invasion of epithelial cells [23],
(Table 1). In addition, work using both human and murine cell suggesting that it may play a role in promoting bacterial
lines has shown that the level of inflammatory mediators colonization. In response to GAS infection, epithelial cells also
produced in response to GAS is often genotype (emm-type)- secrete neutrophil chemoattractants, such as IL-8 [26], with this
dependent [17–19], making results difficult to extrapolate for process likely to be an important protective mechanism elicited
other emm types. In this review, we present the current state of by epithelial cells during GAS pharyngitis [19].
knowledge on innate and adaptive immune responses elicited
during GAS pharyngitis and discuss how these might influence Neutrophils
the outcome of infection. Neutrophils, the most abundant type of leukocyte in the blood,
rapidly infiltrate the NALT in a murine GAS nasopharyngeal
infection model [16]. Neutrophils kill GAS by the release of
AMPs through degranulation, phagocytosis, and phagolysosomal
(continued from previous page) destruction of GAS by reactive oxygen species and the formation
IL-1ra = IL-1 receptor antagonist, LL-37 = cathelicidin antimicrobial peptide, of NETs [27]. Repeated GAS infection or intranasal vaccination
LTB4 = leukotriene B4, MAC = membrane attack complex, MCET = mast cell
stimulates an IgA/IgG antibody and Th17 cellular response in
extracellular trap, MHC-II = major histocompatibility complex class II
molecule, MIF = macrophage migration inhibitory factor, NALT = nasal- mice, which dramatically enhances neutrophil recruitment and
associated lymphoid tissue, NET = neutrophil extracellular trap, Nlrp3 = myeloperoxidase antimicrobial activity during early infection
nucleotide-binding domain leucine-rich repeat containing family pyrin [28]. This is thought to be mediated by Th17 cell secretion of
domain containing 3, OSAS = obstructive sleep apnea syndrome, PAMP =
pathogen-associated molecular pattern, PerR = peroxide regulator, PG =
IL-17A, which stimulates the production of chemokines and
prostaglandin, PKA = protein kinase A, PRR = pattern recognition receptor, cytokines, promoting the recruitment and activation of
RHD = rheumatic heart disease, SCPA = group A streptococcal C5a polymorphonuclear leukocytes, such as neutrophils, as well as
peptidase, SIC = streptococcal inhibitor of complement, Ska = streptokinase,
macrophages [29, 30]. Indeed, a neutralizing antibody against
SlaA = Streptococcal phospholipase A2, SLO = streptolysin O, SLS =
streptolysin S, Spe = streptococcal pyrogenic exotoxin, SpyCEP = Strepto- IL-17A prevented GAS clearance from the NALT [31], likely
coccus pyogenes cell envelope protease, Treg = regulatory T cell because of impaired leukocyte infiltration.

2 Journal of Leukocyte Biology Volume 103, February 2018 www.jleukbio.org


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
TABLE 1. Host factors implicated in GAS pharyngitis

Inflammatory mediator Class Function Source Evidence References

www.jleukbio.org
Azurocidin AMP Chemotactic for monocytes, Macaque throat swab mRNA [94]
antimicrobial activity
HBD1 AMP Antimicrobial HaCaT, primary human tonsil tissue mRNA [116]
HBD2 AMP Antimicrobial HaCaT, primary human tonsil tissue, mRNA [88, 116]
human foreskin keratinocytes
LL-37/cathelicidin AMP Antimicrobial, functions in cell HaCaT, primary human tonsil tissue mRNA [116]
chemotaxis, immune mediator
induction, and inflammatory
response regulation
RNASE7 AMP Broad-spectrum antimicrobial HaCaT mRNA [19]
CCL19/MIP-3b Chemokine Chemotactic for T and B cells PBMC mRNA [54]
CCL2/MCP-1 Chemokine Chemotactic for monocytes and Murine nasal turbinate, PBMC mRNA, protein [16, 54]
basophils
CCL20/MIP-3a Chemokine Chemotactic for lymphocytes and HaCaT, PBMC mRNA, protein [19, 54]
dendritic cells and can repress
proliferation of myeloid progenitors
CCL3/MIP-1a Chemokine Plays role in inflammatory responses Murine nasal turbinate, PBMC mRNA, protein [16, 54]
CCL4/MIP-1b Chemokine Chemotactic and inflammatory functions Murine nasal turbinate Protein [16]
CCL5/RANTES Chemokine Chemotactic for blood monocytes, HEp-2, PBMC, murine nasal mRNA, protein [16, 26, 54]
memory Th cells, and eosinophils; turbinate
causes histamine release from
basophils and activates eosinophils
CCL7/MCP-3 Chemokine Chemotactic for macrophages PBMC mRNA, protein [54]
CXCL1/GROa Chemokine Plays a role in inflammation, Murine nasal turbinate, HaCaT mRNA, protein [16, 19]
chemotactic for neutrophils
CXCL10/IP-10 Chemokine Stimulation of monocytes and NK Detroit 562, murine nasal turbinate, mRNA, protein [16, 54, 96]
and T cell migration, modulation PBMC, tonsil fluid from GAS
of adhesion molecule expression pharyngitis patients
CXCL11/I-TAC Chemokine Chemotactic for activated T cells Detroit 562 Protein [96]
CXCL2/GROb Chemokine Suppresses hematopoietic progenitor HEp-2 mRNA [89]
cell proliferation
CXCL3/GROg Chemokine Plays a role in inflammation and HEp-2, HaCaT mRNA [19, 89]
chemotactic for neutrophils
CXCL9/MIG Chemokine/ Chemotactic for lymphocytes, Detroit 562, THP-1, murine nasal mRNA, protein [16, 54, 96, 97]
cytokine antimicrobial against GAS turbinate, tonsil fluid from GAS
pharyngitis patients, PBMC
IL-8/CXCL8 Chemokine Major mediators of the inflammatory HEp-2, HaCaT, THP-1, PBMC, tonsil mRNA, protein [18, 19, 26, 54, 89,
response, functions as explant tissue 91, 98, 136]
chemoattractant, and is also a
potent angiogenic factor

Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
GM-CSF/CSF-2 Cytokine Controls the production, HaCaT mRNA [19, 188]
differentiation, and function of
granulocytes and macrophages
IFN-a Cytokine Antiviral activity PBMC Protein [104]
(continued on next page)

Volume 103, February 2018 Journal of Leukocyte Biology


Soderholm et al. Group A streptococcal pharyngitis and host immune responses

3
4
TABLE 1. (continued)

Inflammatory mediator Class Function Source Evidence References

IFN-b Cytokine Antiviral factor that also induces BMDM, murine DCs, and Protein [17, 52, 53, 74]
transcription of inflammatory neutrophils
cytokines and chemokines, controls
IL-1b levels
IFN-g Cytokine Triggers cellular response to viral and HEp-2, murine CD4+ T cells, PBMC, Protein [18, 89, 135, 136,
microbial infections human tonsillar T cells, tonsil 141, 142, 189, 190]
explant tissue
IL-1F9/IL-36g Cytokine Keratinocyte inflammatory response HaCaT mRNA [19]
IL-10 Cytokine Has pleiotropic effects in Tonsil explant tissue Protein [136]
immunoregulation and

Journal of Leukocyte Biology


inflammation; enhances B cell
survival, proliferation, and antibody
production; down-regulates
expression of Th1 cytokines, MHC-II
antigens, and costimulatory
molecules in macrophages
IL-11 Cytokine Stimulates hematopoietic stem cell HaCaT mRNA [19]
proliferation, stimulates T cell-
dependent development of Ig-
producing B cells

Volume 103, February 2018


IL-12a (p35) Cytokine Growth factor for activated T cells HEp-2 mRNA [188]
and NK cells, stimulates the
production of IFN-g by PBMC;
forms heterodimer with p40 to
generate IL-12p70, which drives
Th1 development
IL-12b (p40) Cytokine Growth factor for activated T cells HEp-2, murine nasal turbinate mRNA, protein [16, 188]
and NK cells, stimulates the
production of IFN-g by PBMC;
forms heterodimer with p35 to
generate IL-12p70, which drives
Th1 development
IL-13 Cytokine Involved in B cell maturation and PBMC Protein [135]
differentiation, down-regulates
macrophage activity and
inflammatory cytokine/chemokine
production
IL-17A Cytokine Positively regulates proinflammatory Murine nasal turbinate, HEp-2, A549, Protein, mRNA [16, 22, 135,
cytokine production primary human tonsillar T cells, 141–143, 189]
murine CD4+ T cells, PBMC

Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
IL-1Ra Cytokine Inhibits activities of IL-1a and IL-1b HaCaT Protein [19]
and modulates IL-1-related immune
and inflammatory responses
IL-1a Cytokine Pleiotropic cytokine involved in HaCaT, HEp-2, tonsil explant tissue mRNA, protein [19, 26, 91, 136]
inflammatory processes and
hematopoiesis
(continued on next page)

www.jleukbio.org
TABLE 1. (continued)

Inflammatory mediator Class Function Source Evidence References

www.jleukbio.org
IL-1b Cytokine Important mediator of the Murine nasal turbinate, HaCaT, mRNA, protein [16, 19, 26, 91, 104,
inflammatory response, involved in HEp-2, PBMC, BMDM, tonsil 136, 188]
cell proliferation, differentiation, explant tissue
and apoptosis; induces
cyclooxygenase-2; promotes Th17
differentiation of T cells
IL-2 Cytokine Induces proliferation of T and B cells Murine nasal turbinate, HEp-2, protein, mRNA [16, 136, 188, 190]
human tonsillar CD4+ T cells,
tonsil explant tissue
IL-20 Cytokine Regulates proliferation and HaCaT mRNA [19]
differentiation of epithelial cells
during inflammation
IL-23 Cytokine Forms a heterodimer with IL-12b, HEp-2, A549 mRNA [143]
stimulates the production of IFN-g
IL-24 Cytokine May contribute to terminal cell HaCaT mRNA [19]
differentiation
IL-4 Cytokine Activates B cells Human tonsillar CD4+ T cells Protein [190]
IL-5 Cytokine Acts as a growth and differentiation PBMCs, human tonsillar CD4+ T cells Protein [135, 190]
factor for B cells and eosinophils
IL-6 Cytokine Functions in inflammation and Murine nasal turbinate, HEp-2, mRNA, protein [16–18, 26, 89, 91, 98,
maturation of B cells, endogenous HaCaT, BMDM, PBMC, THP-1, 103, 104, 136, 190]
pyrogen capable of inducing fever human tonsillar CD4+ T cells,
tonsil explant tissue
LIF Cytokine Involved in the induction of Murine nasal turbinate Protein [16]
hematopoietic differentiation
MIF Cytokine Lymphokine involved in cell- HaCaT Protein [19]
mediated immunity,
immunoregulation, and
inflammation; regulates
macrophage function
TNF-a Cytokine Multifunctional proinflammatory Murine nasal turbinate, PBMC, Protein [16–18, 89, 104, 135,
cytokine, involved in a wide BMDM, HEp-2, tonsil explant 136]
spectrum of inflammatory processes, tissue
including cell proliferation,
apoptosis, and differentiation
TNF-b/ Cytokine Mediates inflammatory, HEp-2, tonsil explant tissue mRNA, protein [136, 188]
lymphotoxin-a immunostimulatory, and antiviral
responses
CysLT Eicosanoid Proinflammatory and Human tonsil mononuclear cells Protein [177]
immunoregulatory roles

Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
PGE2 Eicosanoid Proinflammatory and immunoregulatory HaCaT, BMDM Protein [57, 91]
PTGS2 Eicosanoid pathway Key enzyme in PG biosynthesis HEp-2 mRNA [89]
CTGF Growth factor Important for wound healing, plays a HaCaT mRNA [19]
role in chondrocyte proliferation
and differentiation and cell adhesion
HBEGF Growth factor May be involved in macrophage- HaCaT mRNA [19]

Volume 103, February 2018 Journal of Leukocyte Biology


mediated cellular proliferation
Soderholm et al. Group A streptococcal pharyngitis and host immune responses

5
(continued on next page)
6
TABLE 1. (continued)

Inflammatory mediator Class Function Source Evidence References

IGF-II Growth factor Potent mitogen for cultured cells HEp-2 mRNA [188]
TGF-a Growth factor Activates cell proliferation, HaCaT mRNA [19]
differentiation, and development
TGF-b1 Growth factor Required for Th17 differentiation; Primary human tonsil cells, NALT Protein [22]
regulates cell proliferation,
differentiation, and growth
VEGFA Growth factor Induces permeability of blood vessels HaCaT mRNA [19]
CD147/EMMPRIN Receptor Regulatory of leukocyte migration HaCaT Protein [19]
IL-1RL1/ST2 Receptor Receptor induced by HaCaT mRNA [19]
proinflammatory stimuli, involved

Journal of Leukocyte Biology


in Th cell function
F2RL1 Receptor Modulates inflammatory responses, HaCaT mRNA [19]
activation dilates blood vessels and
lowers blood pressure
IFIT1 ssRNA-binding Type I IFN signaling pathway, HEp-2 mRNA [89]
protein antiviral activity
IFIT2 ssRNA-binding Type I IFN signaling pathway, antiviral HEp-2 mRNA [89]
protein activity, can promote apoptosis
IFIT4 ssRNA-binding Type I IFN signaling pathway, HEp-2 mRNA [89]
protein antiviral activity

Volume 103, February 2018


IgG1/IgG2/IgG3 Antibody Opsonizes microbes to enhance PBMC, blood plasma Protein [135]
phagocytosis, activates the
complement system
HSP70B Chaperone Implicated in a wide variety of cellular HEp-2 mRNA [89]
processes, such as protection of the
proteome from stress
TNFAIP3 DNA-binding Induced by TNF, involved in cytokine- HaCaT, HEp-2 mRNA [19, 89]
protein mediated immune and
inflammatory responses
ISG20 Exonuclease Antiviral defense HEp-2 mRNA [89]
HAS2 Glycosyltransferase Essential for hyaluronic acid HaCaT mRNA [19]
synthesis, which is produced during
wound healing and tissue repair;
hyaluronic acid interacts with the
leukocyte receptor CD44 and is
important in tissue-specific homing
by leukocytes
HLA-DPB1 MHC-II molecule Plays a central role in the immune HEp-2 mRNA [89]
system by presenting peptides
derived from extracellular proteins

Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
ISG15 Ubiquitin-like Chemotactic factor for neutrophils, HEp-2 mRNA [89]
protein antiviral activity
GROa, growth-regulated oncogene a; IP-10, IFN-g-inducible protein 10; I-TAC, IFN-inducible T-cell a chemoattractant; MIG, monokine induced by IFN-g; IL-1F9, IL-1 family, member 9;
PTGS2, PG-endoperoxide synthase 2; CTGF, connective tissue growth factor; HBEGF, heparin-binding epidermal growth factor-like growth factor; IGF-II, insulin-like growth factor II; VEGFA,
vascular endothelial growth factor A; EMMPRIN, extracellular matrix metalloproteinase inducer; IL-1RL1, IL-1 receptor-like 1; F2RL1, proteinase-activated receptor 2; IFIT, IFN-induced protein
with tetratricopeptide repeats; HSP70B, heat shock protein 70B; TNFAIP3, TNF-a-induced protein 3; ISG, IFN-stimulated gene; HAS2, hyaluronan synthase 2; DPB1, DPb1.

www.jleukbio.org
Soderholm et al. Group A streptococcal pharyngitis and host immune responses

Figure 1. Interplay between immune cells in the


clearance of GAS within the tonsil, as well as GAS
subversion mechanisms. Upon infection, GAS
(red) colonizes and invades the tonsillar epithe-
lium. Epithelial cells secrete bactericidal AMPs
and neutrophil chemotactic factors, such as IL-8,
whereas tissue resident macrophages phagocytose
colonizing bacteria and produce TNF-a, which in
turn, activates neutrophils and monocytes. Upon
GAS recognition, DCs promote Th1 maturation by
the secretion of Th1-polarizing cytokines, such as
IL-12, whereas epithelial cells and macrophages
contribute to Th17 maturation by the secretion of
the Th17-polarizing cytokines IL-6, TGF-b-1, and
IL-1b. Th17 cells secrete IL-17A, which promotes
epithelial cell-mediated recruitment and activation
of neutrophils and macrophages. Th1 cells pro-
duce IFN-g, which activates neutrophils and regu-
lates neutrophil recruitment to the infection site
but also contributes to bacterial carriage in the
NALT. A number of GAS virulence factors have
been characterized using in vivo models of GAS
pharyngitis, which target soluble and cellular
immune mediators to subvert infection (red text).
SpyCEP cleaves IL-8, abrogating its chemotactic
activity, whereas PerR is a peroxide response
regulator that provides resistance to macrophage
phagocytosis. The adhesin AspA is an anti-
phagocytic factor that inhibits macrophage and
neutrophil phagocytosis, and SpeB cleaves pro-
IL-1b to produce mature IL-1b during infection,
which enhances neutrophil recruitment. SIC
binds to and inhibits secreted AMPs.

Several GAS virulence factors have been characterized to protected mice against nasopharyngeal infection [37]. A GAS
impair neutrophil recruitment and activity during infection. The knockout mutant lacking SpyCEP had significantly reduced
bacteriophage-encoded GAS DNase (Sda1) protects GAS from survival compared with wild-type GAS in human neutrophil
neutrophil killing by degrading NETs, which are released by killing assays [38], and SpyCEP expression also enhanced
neutrophils to entrap and kill invading bacteria [32]. M1T1 is a bacterial dissemination from the NALT to the lungs in mice [39].
globally disseminated clone of GAS that is frequently recovered SpyCEP expression by the commensal Lactococcus lactis enabled
from cases of streptococcal pharyngitis [33], and an M1T1 Sda1 its survival in the nasopharynx, and whereas GAS expression of
mutant showed reduced survival compared with wild-type strains SpyCEP also contributed to its survival in the NALT, it was not
in human neutrophil killing assays [34]. A DNase inhibitor also essential, suggesting that other virulence factors expressed by
increased GAS sensitivity to neutrophil killing in vitro and in a GAS are able to compensate for SpyCEP [39]. In a murine soft-
murine model of necrotizing fasciitis [32], indicating that Sda1 is tissue model of GAS infection, the infiltration of polymorpho-
essential for GAS resistance to neutrophils. SCPA cleaves murine nuclear leukocytes to the infection site was impaired [40], with
and human polymorphonuclear leukocyte chemotactic factor this effect being attributed to SpyCEP-mediated degradation of
C5a, and intranasal vaccination against SCPA generated serum the polymorphonuclear leukocyte-attracting chemokine IL-8. In
IgG/IgA responses and prevented GAS nasopharyngeal coloni- mice infected with SpyCEP-deficient GAS, polymorphonuclear
zation of mice [35]. In a murine air-sac model, SCPA mutant infiltration was increased, and the infection was controlled.
GAS were cleared more efficiently than wild-type GAS and However, this strain caused a lethal infection in polymorpho-
induced the infiltration of significantly higher numbers of nuclear leukocyte-depleted mice, thus underscoring the impor-
polymorphonuclear leukocytes [36], suggesting that subversion tance of neutrophils in the innate immune response to GAS
of leukocyte recruitment is critical for GAS to establish an infection.
infection. Immunization with SpyCEP (or ScpC), which cleaves The hemolysins SLS and SLO are pore-forming toxins that
chemokines that attract phagocytes, including neutrophils, induce osmotic lysis of a broad spectrum of host cells, including

www.jleukbio.org Volume 103, February 2018 Journal of Leukocyte Biology 7


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
neutrophils [41]. GAS production of SLO is associated with destruction of infected tissues [52]. Another study showed that
human neutrophil lysis in vitro, with a reduction in neutrophil subcutaneous infection of Ifnar12/2 mice with GAS was
lysis observed following incubation with SLO mutant GAS [42]. accompanied by elevated systemic levels of the proinflammatory
SLS has also been shown to induce apoptosis of human cytokine IL-1b that exceeded the protective range [53].
neutrophils, with SLS mutant GAS inducing significantly lower Exogenous IFN-b suppressed IL-1b gene expression and
levels of neutrophil apoptosis [43]. Elimination of neutrophils secretion from wild-type but not Ifnar12/2 BMDMs, suggesting
that encounter GAS, by the expression of cytotoxic virulence that type I IFN signaling is required to control Mf-expressed
factors, such as SLO and SLS, is a mechanism the bacterium may IL-1b to enable protective effects, rather than pathologic ones,
use during pharyngeal infection to evade host innate immune during GAS infection. Supernatants from GAS-infected macro-
responses. phages (containing CXCL9 and CXCL10) stimulated the
migration of Th1 cells in vitro [54], suggesting that macrophages
Mast cells are important for Th1 cell recruitment during infection. GAS
Mast cells participate in the early recognition and clearance of infection of human macrophages also activated the Nlrp3
pathogens and contribute to pathology in allergic diseases, inflammasome (see below), leading to the secretion of the
rapidly degranulating upon activation to release preformed proinflammatory cytokine IL-1b [55]. This cytokine has a well-
mediators, including histamine and TNF-a [44]. Mast cells characterized role in promoting neutrophil recruitment; how-
appear to play an important role in GAS skin-infection models ever, its function in the context of pharyngitis infection is not yet
[45, 46]; however, studies have yet to investigate the role of mast known.
cells during streptococcal pharyngitis. Mast cells have been Eicosanoids, such as PGE2 and leukotrienes, are lipid
reported to kill GAS by extracellular structures similar to NETs mediators that play both proinflammatory and immunoregula-
(MCETs), and disruption of MCETs reduced the antimicrobial tory roles in the innate immune response [56] and are known to
activity of mast cells against GAS in vitro, suggesting that MCETs regulate the capacity of macrophages to respond to GAS during
are important for the inhibition of bacterial growth [47]. Mast infection. PGE2 was produced by macrophages following GAS
cell-deficient mice were also more susceptible to GAS dissemi- infection and suppressed the bactericidal activity of BMDMs in
nation in a skin-infection model, which was attributed to a loss of vitro by preventing the generation of reactive oxygen species
the mast cell-derived AMP cathelicidin (LL-37), a key component [57]. This was attributed to PGE2-mediated accumulation of
of MCETs [45]. SLO activates primary murine bone marrow- intracellular cAMP and activation of PKA, an endogenous
derived mast cells to produce a number of cytokines and suppressor of phagocytosis and reactive oxygen intermediate
chemokines, including TNF-a [48], which is critical for the early generation [58]. Treatment of BMDMs with PGE2 or the PKA
recruitment of neutrophils in mouse models of bacterial activator (S)-p-8-(-4-chlorophenylthio) adenosine-39,59-cAMP en-
infection. One study reported that the cysteine protease SpeB hanced bacterial survival, whereas treatment with the PGE2
stimulates histamine release by human mast cells in vitro, receptor EP2 inhibitor AH6809 or the PKA inhibitor (PKI12–22)
although its role in GAS infection of mucosal membranes is not restored BMDM-mediated killing of GAS [57]. In contrast, LTB4
yet known [49]. is a potent stimulator of macrophage phagocytosis of GAS, and
treatment of human macrophages with LTB4 before GAS
Macrophages infection reduced intracellular cAMP levels and significantly
Tissue resident macrophages contribute to control of GAS increased macrophage phagocytic activity and intracellular
infection by phagocytosis and the production of reactive oxygen bacterial killing [59].
species, while stimulating the recruitment of inflammatory Several mechanisms of GAS resistance to macrophage killing
monocytes and their differentiation to inflammatory macro- have been described in the literature. SLS and SLO induced
phages at the site of infection. Macrophages express a number of oncotic cell death of BMDMs, with strains deficient in both SLS
cytokines in response to GAS, including IL-6, TNF-a, IFN-b, and and SLO activity significantly more attenuated for cytotoxicity
IL-8 [17, 50]. IL-6 contributes to the differentiation of Th17 cells than single-mutant strains [60]. The peroxide response regulator
from naive T cells during GAS infection, whereas TNF-a recruits PerR, which is thought to enhance GAS virulence by modulating
and activates many cell types, including neutrophils and bacterial responses in oxidative environments, appears to play a
monocytes. In a murine soft-tissue model of GAS infection, TNF- critical role in GAS resistance to macrophage phagocytosis, as
a secretion by macrophages was required for monocyte re- well as virulence during pharyngeal infection [61]. Deletion of
cruitment to the site of infection [51]. TNF-a-deficient mice were PerR was associated with reduced resistance to phagocytic killing
impaired in monocyte infiltration, whereas systemic depletion of by murine macrophages and to human blood leukocytes in vitro
monocytes, using clodronate-containing liposomes, rendered and attenuated GAS colonization in a baboon model of GAS
mice sensitive to GAS infection, thus suggesting that monocytes pharyngitis [61]. The streptococcal AspA, which is required for
and/or macrophages play an important role in resistance to GAS GAS colonization of the nasopharynx by mediating adhesion to
infection. The function of type I IFN signaling during GAS pharyngeal epithelial cells, is also an anti-phagocytic factor
infection has currently only been investigated in invasive required for resistance to murine macrophage phagocytosis and
infection, where it plays an immunomodulatory role. Mice neutrophil killing in vitro. Deletion of AspA results in increased
deficient for the Ifnar12/2 had decreased survival following susceptibility to macrophage phagocytic killing and failure of
subcutaneous GAS infection, which was accompanied by in- GAS (serotypes M2 and M28) to establish infection following
creased neutrophil recruitment, as well as exaggerated intranasal challenge in mice [62], suggesting that AspA plays an

8 Journal of Leukocyte Biology Volume 103, February 2018 www.jleukbio.org


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
Soderholm et al. Group A streptococcal pharyngitis and host immune responses

important role in GAS colonization and persistence in the conserved cellular pathway that targets intracellular bacteria for
pharynx. SpeB is known to facilitate evasion of autophagy and lysosomal degradation. Of interest in this regard, autophagy plays
enhance GAS replication intracellularly by degrading host a role in the defense of nonphagocytic cells against GAS in vitro
autophagy adaptor proteins [63] and has been shown to [79], and the suppression of the autophagy pathway has been
contribute to GAS survival within human macrophages from soft implicated in the success of the globally disseminated GAS M1T1
tissue infection biopsies [64]. SpeB-deficient GAS show de- clone [63]. Mice deficient in TLR2 or Unc93b1 (a trafficking
creased viability following infection of human macrophages in protein required for endosomal TLR function) showed an
vitro [64], suggesting that SpeB could contribute to immune increased susceptibility to GAS infection upon subcutaneous
evasion and bacterial persistence during GAS pharyngitis. challenge, suggesting that both TLR2 and TLR13 are required
for protective defense against GAS in mice [50].
DCs GAS has also been shown to trigger non-TLR PRRs during
DCs are abundant in the pharynx, and these cells have key infection by activating the Nlrp3 inflammasome. In response to
functions in priming T cell responses during GAS infection. The endogenous and microbial stimuli, this signaling complex
interaction of GAS with murine and human DCs in vitro induced initiates caspase-1 activation, which drives the proteolytic
DC maturation and the production of Th1-polarizing cytokines, processing of inflammatory cytokines, such as IL-1b and IL-18
such as IL-12 [65, 66]. Selective depletion of CD11c+ DCs in mice [80]. In response to live GAS expressing the virulence factor
abolished GAS-induced IL-12 production and enhanced dissem- SLO, murine macrophages activate caspase-1 and secrete the
ination of GAS into the lymph nodes and liver from a localized proinflammatory cytokine IL-1b [81]. In contrast to these
subcutaneous infection [65]. IL-12 is known to stimulate NK cells findings, extracellular NAD+-glycohydrolase, a SLO-coexpressed
to generate IFN-g [67], which is a classic macrophage-activating toxin, is reported to decrease inflammasome-dependent IL-1b
cytokine [68]. Whereas NK cells are implicated in the patho- release from infected macrophages [82], suggesting that GAS is
genesis of GAS-invasive infections, where they are major sources able to manipulate host cell secretion of IL-1b by multiple
of IFN-g, IL-6, and IL-12 [69], the role of NK cells during GAS mechanisms. With the use of macrophages deficient in inflam-
pharyngitis and superficial infections is currently not known. In a masome components, it was shown that both Nlrp3 and
nasopharyngeal infection model, a significant decrease in apoptosis-associated speck-like protein containing a caspase
CD11c+ leukocytes was observed, which the authors hypothesized activation and recruitment domain were crucial for caspase-1
to be mediated by superantigen-mediated cytokine responses activation and IL-1b secretion during GAS infection. The
that induced immunosuppression, allowing GAS to escape membrane-bound GAS virulence factor SpyA, an ADP-
myeloid cell-mediated killing [16]. As with findings in macro- ribosyltransferase, has also been shown to stimulate caspase-1-
phages (see above), type I IFN signaling also appears to dependent inflammatory responses and IL-1b production in
negatively regulate inducible IL-1b expression in DCs, thus murine macrophages [83], whereas the GAS virulence factor
limiting IL-1b-associated immunopathology [52, 53]. SpeB has been proposed to function as an inflammatory caspase
mimic to cleave pro-IL-1b to generate mature IL-1b [84]. This
PRRs and signaling pathways linked to GAS-initiated suggests that GAS can generate active IL-1b via multiple
inflammatory responses mechanisms. Another study using human macrophages showed
The host uses families of PRRs, such as the TLRs, to recognize that following stimulation with live or inactivated M1 GAS,
and respond to highly conserved PAMPs that are displayed by inflammasome activation mediates the maturation of pro-IL-1b
invading microorganisms. Early studies demonstrated that the into mature-IL-1b [55]. Consistent with the inflammasome
TLR adaptor protein MyD88 is required for GAS-mediated playing a protective role during GAS infection, BMDMs from
production of key cytokines [70]; however, the identification of caspase-12/2 mice showed decreased survival and impaired
the TLRs engaged upstream of MyD88 has presented a killing of GAS [83].
significant challenge to the field [71, 72]. Innate immune cells Downstream of PRRs, particularly the TLRs, activation of the
from mice deficient in TLR1, TLR2, TLR4, or TLR9, TLR2/6 MAPK signaling pathways (ERK, JNK, and p38), as well as the
double-deficient mice, and TLR2/4/9 triple-deficient mice were proinflammatory transcription factors NF-kB and AP-1, mediate
still able to produce inflammatory cytokines following GAS proinflammatory gene expression during GAS pharyngitis [19,
infection [73, 74]. More recently, however, TLR2 and TLR13 26, 85–87]. For example, mRNA expression of the AMP HBD2 by
were identified as GAS sensors, with knockout of both receptors primary human keratinocytes requires the activity of the MAPKs
required for abrogation of macrophage-mediated inflammatory p38 and JNK, as well as NF-kB; inhibitors of these components
responses to GAS in vitro [50]. TLR13, which is not present in blocked GAS-inducible expression of this AMP [88]. A tran-
humans, recognizes bacterial rRNA [75], thus raising questions scriptome study of GAS-infected HEp-2 epithelial cells detected
about possible species differences in innate immune recognition differential expression of the AP-1 transcription factor family
of GAS. However, another study showed that in human members c-JUN and Fluoro-Jade B murine osteosarcoma viral
monocyte-derived macrophages, GAS RNA is recognized by oncogene homolog B, as well as activating transcription factor 3
TLR8 [76], a functional equivalent of TLR13 [77]. TLR8 is [89], which interacts with AP-1 [90]. This further implicates the
located in endosomes of myeloid cells, such as monocytes, MAPK–AP-1 signaling axis as a key mediator of GAS-regulated
macrophages, and myeloid DCs, and its activation triggers the inflammatory gene expression. These signaling pathways lead to
release of IL-6, TNF-a, IFN-b, and LL-37 [76, 78]. Interestingly, the production of numerous inflammatory mediators, the roles
TLR8 signaling has also been shown to induce autophagy [78], a of which in GAS pharyngitis are discussed below.

www.jleukbio.org Volume 103, February 2018 Journal of Leukocyte Biology 9


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
PGE2 and leukotrienes appeared to be largely dependent on GAS superantigens, as a
Several studies have suggested a role for the eicosanoid PGE2 GAS-deletion strain for all 6 superantigen genes (encoding SpeA,
during GAS pharyngitis [57, 89, 91]. This lipid mediator is SpeC, SpeG, SpeL, SpeM, and SmeZ) induced much lower levels
secreted by many cell types in response to cytokines, such as TNF- of inflammatory chemokine secretion and only mild neutrophil
a, IL-1, and IL-6, or bacterial PAMPs, such as LPS. PGE2 has both infiltration.
proinflammatory and immunosuppressive effects on innate Given the key role of chemokines in orchestrating leukocyte
immune cells, including macrophages, neutrophils, NK cells, and recruitment and host-protective immune responses, it is not
DCs [92]. PGE2 is synthesized from arachidonic acid via the surprising that GAS can combat their activities. GAS secretes
actions of the cyclooxygenase 1/2 enzymes and downstream several proteases that specifically cleave and inactivate various
enzymes and signals through four known G protein-coupled chemokines. SpyCEP cleaves and abrogates the activities of
receptors (EP1, EP2, EP3, and EP4). Several bacterial pathogens, CXCL1, CXCL2, CXCL6, and IL-8 [39], thus impairing
including Salmonella spp., Escherichia coli, mycobacteria, and GAS, neutrophil recruitment and function [99]. During GAS lower
are known to induce PGE2 production, which has inhibitory respiratory tract infection in mice, an inverse relationship
effects on immune cells, aiding in bacterial colonization of between the bacterial load and CXCL1 levels was observed, which
mucosal surfaces [93]. In response to GAS infection, HaCaT was attributed to the protease activity of SpyCEP [39]. SpeB also
epithelial cells promote the secretion of PGE2 [91]. BMDMs also cleaves a number of chemokines, including CXCL1, CXCL2,
secrete PGE2 in response to GAS infection [57], which is CXCL3, CXCL10, and CXCL11, and abolishes the chemotactic
associated with decreased macrophage bactericidal activity, likely but not antimicrobial activity of CXCL9 [100]. The activity of
facilitating GAS colonization and survival during the early stages SpeB is required for GAS proliferation in human saliva [101], as
of infection. well as the dampening of the early host immune responses to
Leukotrienes—another family of eicosanoid inflammatory establish skin infections [102]. As a result of SpeB cleavage of
mediators derived from arachidonic acid—have also been host chemokines that attract macrophages and neutrophils to the
associated with GAS pharyngitis. LTB4 has been reported to infection site, SpeB activity may potentially reduce the re-
enhance proinflammatory responses to several pathogens, in- cruitment of specific cell types to the infection site and thus,
cluding GAS, by suppressing intracellular cAMP production in enhance GAS survival and immune evasion during GAS
differentiated THP-1 macrophages, thus stimulating macrophage pharyngitis.
phagocytosis and intracellular killing [59]. A transcriptome
analysis of pharyngeal infection in cynomolgus macaques de- Proinflammatory cytokines
tected a positive correlation between expression of the unchar- Cytokines secreted during GAS pharyngitis signal in an
acterized GAS transaldolase mipB and two genes encoding host autocrine and/or paracrine manner to instruct specific in-
leukotriene receptors, LTB4 receptor (LTB4R) and CysLT flammatory responses during infection, with multiple studies
receptor 2 (CYSLTR2) [94]. Transaldolase is a key enzyme investigating those cytokines released from different cell types.
involved in carbohydrate metabolism, which is a cue for virulence GAS infection of HEp-2 epithelial cells induced the secretion of
gene expression in GAS [95], suggesting that leukotriene IL-6 and TNF-a [18, 89, 103] and increased mRNA expression
signaling may enhance the proinflammatory responses to GAS of RANTES, IL-1a, and IL-1b [26]. One study showed that M49
virulence factors during infection. The role of leukotrienes GAS infection of HEp-2 epithelial cells induced a significant
during GAS pharyngitis now requires functional studies to proinflammatory response, with the authors speculating that
investigate the cellular mediators and proinflammatory pheno- this would lead to tissue destruction in vivo, thus allowing GAS
types that leukotrienes orchestrate during infection. to disseminate deeper into host tissues [89]. In response to
GAS, HaCaT keratinocytes increased expression of mRNA for
the cytokines IL-1a, IL-1b, and IL-6 [91], and HaCaT cells
Chemokines incubated with M1 protein released MIF, IL-1a, and IL-1Ra
GAS has been reported to induce a plethora of chemokines from [19]. BMDMs are reported to secrete IL-6, TNF-a, and IFN-b in
the pharyngeal-derived epithelial cell line Detroit 562 [96, 97], response to GAS [17], and PBMCs secrete IL-1b, IL-6, TNF-a,
nonpharyngeal-derived epithelial cell lines HEp-2 [18, 26, 89] and IFN-a [104]. Blood samples from patients with acute GAS
and HaCaT [19, 26, 91], primary human macrophages [54], and pharyngitis showed high levels of the proinflammatory cyto-
the monocyte-like cell line THP-1 [97, 98]. GAS-induced kines TNF-a and IL-6 and reduced levels of the Th2 signature
secretion of IL-8 from HEp-2 cells promoted monocyte (U937) cytokines IL-4 and IL-10, suggesting that immune responses are
transmigration, suggesting that epithelial cell-secreted IL-8 is an skewed away from a Th2 phenotype [105]. In a murine
important mediator for chemoattraction of monocytes during nasopharyngeal GAS infection model, analysis of nasal turbi-
GAS infection [26]. Supernatants from GAS-stimulated human nates revealed an early Th17-skewed response, with enhanced
macrophages induce the migration of Th1 cells, suggesting that secretion of TNF-a, IL-6, and IL-17A [16]. Other cytokines
macrophages may be involved in the recruitment of Th1 cells to secreted included IL-1a, IL-1b, IL-2, LIF, and M-CSF. Despite
the infection site [54]. In HLA-DR4 and HLA-DQ8 mice (a these findings on individual cytokines produced during GAS
murine nasopharyngeal infection model), the profiling of nasal infection, in many cases, their specific functions in host
turbinates showed that a robust chemokine response was induced protection vs. susceptibility during GAS pharyngitis are still not
by the NALT [16]. The secretion of inflammatory chemokines well understood, as their roles have mainly been investigated
was accompanied by the marked infiltration of neutrophils and using GAS-invasive disease models.

10 Journal of Leukocyte Biology Volume 103, February 2018 www.jleukbio.org


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
Soderholm et al. Group A streptococcal pharyngitis and host immune responses

Multiple studies have identified IL-1b as a GAS-inducible LL-37 is important for controlling GAS infection at the epithelial
cytokine [81, 106, 107]. Pro-IL-1b must be proteolytically surface [122]. Mechanisms of GAS resistance to LL-37 have also
processed to generate biologically active IL-1b that is secreted been reported, where subinhibitory concentrations of LL-37
from cells, and IL-1b is critical in host defense against GAS- induced the GAS CsrRS (also designated CovRS) two-component
invasive infection [3], which may be mediated by the ability of regulatory system that up-regulates the expression of virulence
IL-1b to enhance the production of neutrophil chemo- factors SLO and hyaluronic acid capsule, significantly reducing
attractants [108]. In this respect, a recent study by LaRock GAS killing by human oropharyngeal keratinocytes, neutrophils,
et al. [84] found that the cysteine protease SpeB, which is and macrophages [123]. Μ1 protein has also been demonstrated
secreted by the majority of disease-causing GAS strains, to promote resistance to LL-37 that is present in the extracellular
including the M1T1 strain 5448 [109, 110], is able to generate traps of human neutrophils and the human mast cell line 1; loss of
mature and active IL-1b by proteolytic cleavage. This effect M1 protein increased GAS killing by neutrophil NETs and mast cell
was not observed for another M1 GAS strain, which may MCETs [124]. M1 protein appears to act by enabling GAS to evade
reflect strain variation differences [82]. During invasive LL-37 killing by sequestering and neutralizing LL-37 antimicrobial
infection, GAS isolates often acquire mutations that down- activity, as well as binding to the LL-37 precursor human cationic
regulate SpeB expression and therefore, reduce the capacity antimicrobial protein 18 and preventing its maturation [125].
of such strains to cleave IL-1b. This presumably decreases
IL-1b-mediated host responses. LaRock et al. [84] also The complement system
hypothesized that the increased rate of invasive GAS disease The complement system contributes to the opsonization of
observed in patients who receive the IL-1b inhibitor drug GAS for subsequent phagocytic killing, and a number of GAS
anakinra is therefore a result of a loss of host immunity in virulence factors are reported to target components of the
response to IL-1b, which would normally protect against the complement system during infection as an immune-evasion
development of invasive GAS infection. strategy. SCPA cleaves the complement component C5a, a
potent polymorphonuclear leukocyte chemotactic factor,
AMPs and intranasal vaccination against SCPA prevented GAS
AMPs are an important component of host innate defense nasopharyngeal colonization in mice [35]. SIC inhibits
against GAS infection, through both direct antimicrobial action complement-mediated formation of the MAC by blocking the
at mucosal surfaces and chemokine-like immunomodulatory membrane-insertion site on C5b-7 [126], and inactivation of
activities [111]. The bactericidal activity of AMPs against GAS has SIC impaired M1 GAS colonization of the mouse oropharynx
been investigated extensively. Primary human epithelia produce [121]. GAS, like all Gram-positive bacteria, is also resistant to
the HBDs, HBD2 and HBD3, which show bactericidal activity MAC-dependent lysis [127]. The mechanism is thought to
against GAS [112]. Human and mouse cathelicidins (LL-37 and involve the thick peptidoglycan layer present in the cell wall of
cathelin-related antimicrobial peptide, respectively) are Gram-positive bacteria that prevents incorporation of the
expressed in the skin and potently inhibit GAS bacterial growth complex into the membrane and subsequent lysis. Whereas
[113]. The immunomodulatory activity of LL-37 has been widely MAC ring structures are reported to form on the cell wall of
studied in vitro, where it has been shown to stimulate IL-8 GAS, the role of MAC in immune defense against GAS is
production by monocytes, as well as activate the AKT, CREB, and unclear [110]. GAS M protein also plays a role in resistance to
NF-kB signaling pathways, leading to the expression of a number complement-mediated opsonophagocytosis by binding to
of cytokines and chemokines [114]. complement-inhibitory proteins present in human serum,
AMPs are often down-regulated by pathogenic bacteria to such as C4b-binding protein [128], factor H [129], and factor
establish an infection [115]. Indeed, GAS infection of HaCaT H-like protein 1 [130]. M Protein also binds to other host
keratinocytes reduced mRNA expression of genes encoding the proteins, such as fibrinogen, which inhibits complement C3
AMPs HBD1 and HBD2 [116]. Primary epidermal keratinocytes convertase deposition and provides resistance to phagocytosis
did not express HBD2 in response to GAS; however, HBD2 mRNA by human blood leukocytes [131] and plasmin, which prevents
expression was restored in response to a CS101 GAS mutant deposition of the opsonin C3b and inhibits neutrophil
lacking hyaluronic acid capsule expression (has mutant) [88]. phagocytosis of GAS [132].
GAS also circumvents the antimicrobial activity of host AMPs by
secreting the protein SIC and through the action of the cell wall-
anchored M1 protein, which binds AMPs (including LL-37,
ADAPTIVE IMMUNITY DURING
human a-defensin-1, HBD2, HBD3, lysozyme, and secretory
GAS INFECTION
leukocyte protease inhibitor) and prevents them from accessing
the bacterial membrane [117–120]. Inactivation of SIC resulted A number of studies have investigated the role of acquired
in decreased GAS proliferation in human saliva and significantly immunity in the clearance or protection against GAS pharyngitis.
impaired colonization of the mouse oropharynx [101, 121]. GAS Patients lacking the innate signaling components MyD88 (a TLR
also secretes the plasminogen activator Ska, which activates host adaptor) or the serine/threonine kinase IL-1 receptor-associated
plasmin that can bind to the GAS surface, resulting in kinase 4 are susceptible to invasive and noninvasive pyogenic
degradation of host LL-37 [122]. Mutagenesis of the ska gene or infections as children but become less susceptible with age [133,
proteolytic inhibition of plasmin reduced the virulence of GAS in 134]. This suggests that the adaptive immune response, which
a murine skin lesion-infection model, implying that the activity of develops throughout life, can compensate for innate immune

www.jleukbio.org Volume 103, February 2018 Journal of Leukocyte Biology 11


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
deficiencies, and its role in protection against GAS infection is the infection site. This raises the intriguing possibility that an
likely underestimated. Indeed, the majority of GAS pharyngitis imbalance between IL-17A- and IFN-g-producing CD4 T cells
cases occurs in children, whereas infection in adults is less could contribute to GAS carriage in humans. Th17 cells and
common [1]. A recent study comparing the humoral and cellular IL-17A have been implicated in murine models of various GAS
adaptive immune responses of children and adults with etiologies, such as chronic arthritis, acute sepsis, and puerperal
conserved GAS antigens revealed that strong Th1 and Th17 sepsis. In a murine puerperal sepsis model, neutrophil re-
cellular memory responses, IFN-g release, and an IgG1/IgG3- cruitment and clearance of GAS were significantly attenuated in
dominated humoral immune response develop in children IL-17A2/2 mice [144], suggesting that IL-17A contributes to the
following exposure to GAS [135]. Children showed significantly control of GAS mucosal colonization. During acute sepsis,
lower levels of IFN-g and IgG3 than in adults, which may enhanced levels of IL-17A were detected in the serum of both
contribute to the higher GAS-infection rates that are observed in sham- and GAS-infected mice within a short time of inoculation,
children. Thus, Th1- and/or Th17-biased responses are likely to indicating the speed and potency of the IL-17A response, which
play an important role in the development and preservation of can be stimulated by trauma alone [145]. In a murine model of
GAS immunity over time (Fig. 1). Tonsil explants from tonsillar chronic arthritis, IL-17 receptor-deficient mice showed reduced
hypertrophy and recurrent tonsillitis patients stimulated with joint destruction [146]. Mice deficient for IL-1b, which is
heat-killed GAS also developed a Th1 cytokine response, with required for the differentiation of Th17 cells, were fully
IL-1b, IFN-g, and TNF-b as the dominate cytokines secreted protected against joint destruction during late stages of disease,
[136]. IFN-g has been suggested to prevent the local dissemina- implicating Th17 cells in host-destructive inflammatory responses
tion of GAS in a murine NALT model [137] and is critical for during GAS infection. It is important to note that gd T cells are
protection against lethal skin infections in mice [138]. A study by also a major source of IL-17A and are present in large numbers
Fan et al. [31] suggested that the role of B cells may be less in the surface epithelium of tonsils from patients with recurrent
important for clearance of, or protection against, GAS infection. tonsillitis [147]. As gd T cells differentiate quickly upon
In this study, immunized B cell-deficient mice cleared GAS stimulation and can respond to different antigens independently
intranasal challenges as efficiently as wild-type mice, and B cell- of classic MHC-mediated presentation of peptide epitopes, these
adoptive transfer from immunized to naı̈ve mice did not provide cells are likely to play an important role in bridging the innate
protection against infection. and adaptive immune responses during GAS infection [148].
Antigen presentation to CD4+ T cells in the presence of the Several studies have shown that Tregs and Th2 responses are
cytokines IL-6 and TGF-b1 promotes Th17 differentiation [23, not typically generated during murine intranasal GAS infection
139], with the IL-12 family member, IL-23, also contributing to [22, 149]. In contrast, Tregs, which dampen immune responses to
this phenotype [23]. Not surprisingly, several studies have self-antigens and to foreign antigens, are induced by super-
implicated IL-17A-producing Th17 cells, which are particularly antigens during systemic GAS infection [150], and in vitro, GAS
important in conferring host protection against extracellular M proteins (M5 and M22) have been shown to induce the
pathogens at epithelial surfaces [140], as playing a dominant role differentiation of human CD4+ lymphocytes into Tregs [151].
in the clearance of GAS in a murine infection model of During systemic infection, Tregs can be induced by superantigens
pharyngitis [22, 141, 142]. In a murine pharyngitis model, Th17 or M protein, triggering the cross-linking of the TCR and CD46
cells had a protective role against GAS infection. For example, on naı̈ve T cells [151]. GAS may exploit the Treg signaling
transfer of wild-type CD4+ T cells from immunized IL-17A+/+ pathway as a strategy to induce an immunosuppressive/
mice enabled naı̈ve mice to clear a GAS infection, whereas this immunomodulatory T cell phenotype, which may facilitate GAS
protective effect was not apparent when IL-17A2/2 CD4+ T cells immune evasion, as well as contribute to pathogenesis. Mucosa-
from immunized mice were used [22]. During GAS infections, associated invariant T cells, a subclass of innate-like T cells, have
TGF-b1, produced by monocytes and DCs, was shown to be an recently been identified as a major source of IFN-g and
essential driver of Th17 cell differentiation [141]. In addition, proinflammatory cytokines (TNF-a, IL-2, but not IL-17A) after
IL-6, secreted by a specialized CD301b+ DC subset from cervical exposure to superantigen staphylococcal enterotoxin B [152]
lymph nodes [142], as well as epithelial-derived IL-6 and IL-23, and therefore, may contribute to the cytokine storm and
also contributed to the development of this T cell response subsequent immunosuppression observed during systemic
[143]. Although the precise mechanisms by which Th17 cells superantigen-mediated GAS infections.
control GAS infections are yet to be fully elucidated, IL-17A is
well known to mediate the recruitment and expansion of
neutrophils and macrophages [141], key cell types that contrib-
PROTECTIVE ROLES OF PHARYNGEAL
ute to the detection and clearance of GAS.
MICROBIOTA AGAINST GAS INFECTION
Interestingly, Th17-skewed CD4 T cell responses to GAS are
dependent on intranasal inoculation, whereas intravenous or The normal microbiota that colonizes mucosal layers is an
subcutaneous inoculation generated a Th1 response, with a important component of first-line defense against invading
characteristic IFN-g signature [142]. In IL-62/2 mice, IFN-g+ pathogens, with commensal and probiotic bacteria competing
CD4 T cells were expanded during GAS infection, and these with foreign microbes for niche and nutrients. Furthermore,
mice were unable to clear the infection from the NALT and initial studies showed that normal throat flora contains bacterial
became long-term carriers [142]. This is likely because the IFN-g+ species (nonhemolytic streptococci, viridans streptococci, and
CD4 T cells are unable to attract host-protective neutrophils to Neisseria spp.) that are both inhibitory and bactericidal to GAS

12 Journal of Leukocyte Biology Volume 103, February 2018 www.jleukbio.org


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
Soderholm et al. Group A streptococcal pharyngitis and host immune responses

and can protect against GAS colonization in children [153, 154]. ARF and RHD
The microbiota can also regulate host immunity and contribute ARF and RHD are serious autoimmune diseases that can develop
to host defense by modulating the cytokines and AMPs that host following GAS pharyngitis. These GAS-associated conditions have
cells secrete [155]. For example, probiotic Lactobacillus species been reviewed extensively [166–168]. The pathogenesis of GAS
commonly colonize mucosal membranes of the oral tract, and infection-induced, nonsuppurative pathologies remain elusive;
certain strains can skew immune responses into a regulatory or however, they appear to be the result of an exaggerated immune
tolerant mode by suppressing proinflammatory responses (IL-6, response to specific GAS epitopes in susceptible individuals
IL-12, TNF-a, IL-17A/IL-23, and Th2 cytokines) [143, 156, 157], [169]. Clinical features of ARF include arthritis, carditis, and
modulating expression and/or function of PRRs that detect GAS fever, and ARF episodes can occur recurrently [170]. RHD
components [143], and inducing the production of regulatory develops as a result of ARF and refers to the long-term damage of
cytokines, such as IL-10 [156]. Strains of Lactobacillus (L. rhamosus heart valves [166]. Prevention of these autoimmune conditions is
Kx151A1 and L. reuteri PTA-5289) have also been reported to best achieved through timely identification and antibiotic
attenuate the expression of SLS at the transcriptional level and treatment of GAS pharyngitis [171].
inhibit the hemolytic activity of GAS in vitro [158], which may
help protect the pharyngeal mucosa from GAS colonization
Scarlet fever
during early infection.
Scarlet fever is a skin rash that can be present during GAS
In light of the evidence above, microbiota-mediated sup-
pharyngitis. The rash typically appears within the first few days of
pression of inflammation could potentially inhibit GAS growth
GAS infection, spreading from the trunk to the extremities of the
and colonization of the tonsils. Proinflammatory cytokine
body [165]. The pathogenesis of scarlet fever is not well
production is often accompanied by increased epithelial
characterized; however, GAS secreted pyrogenic exotoxins have
permeability, as a result of perturbation of tight junctions,
been implicated in evoking a rash in naı̈ve subjects. No single
which GAS can exploit to gain access to host metabolites,
toxin has been consistently associated with scarlet fever, but
adhesive factors, and underlying tissues [159]. Therefore, the
combinations of the exotoxins SpeA, SpeC, and streptococcal
ability of microbiota to dampen host proinflammatory re-
superantigen are often seen in GAS strains causing scarlet fever
sponses is predicted to be beneficial in the control of GAS
outbreaks [172–175]. The mechanisms by which these exotoxins
infections. Conversely, the anti-inflammatory effects of micro-
may contribute to the symptoms of scarlet fever are not well
biota could also contribute to asymptomatic and/or chronic
studied, but they have been found to alter IgG production and
bacterial carriage in children, where immune tolerance
reticuloendothelial clearance function, induce T cell prolifera-
prevents the clearance of GAS. In addition, the ability of
tion, and enhance skin reactivity [176].
Lactobacilli to reduce SLS production suggests that microbiota
may also play a role in regulating GAS virulence factor
production to control bacterial colonization. Adenotonsillar hypertrophy and pediatric sleep apnea
The oral microbiota can also protect against pathogenic GAS pharyngitis is associated with pediatric adenotonsillar
bacterial infection by the secretion of bactericidal compounds, hypertrophy (abnormal enlargement of the tonsil) and has
and BLIS K12 is a commercially available oral cavity probiotic recently been implicated in pediatric OSAS, which is primarily
that protects against GAS pharyngitis [160]. BLIS K12 is characterized by enlarged tonsils [177]. Tonsillar hypertrophy is
developed from Streptococcus salivarius K12, which is a bacterial caused by the proliferation of immune cells in the follicles and
species found in the oral microbiota of healthy humans. BLIS interfollicular areas of the tonsils in response to infection, and
K12 antagonizes GAS growth by secreting the bacteriocins GAS was present in the tonsillar-reticulated crypt epithelium of
salivaricin A2 and B and has been shown to reduce GAS 37% of children who had tonsillectomy for adenotonsillar
pharyngitis and/or tonsillitis episodes in children [161–163] and hypertrophy [8]. The composition of immune cells in tonsils
adults [164] who experience recurrent GAS infections. Given the from recurrent GAS tonsillitis patients and from tonsillar
widespread interest in microbiota-mediated immune regulation hypertrophy patients is similar, and upon stimulation with GAS,
and control of disease outcomes, this will undoubtedly be an area both produce a Th1 cytokine response [136, 147]. A study by
of burgeoning interest in the future. Viciani et al. [177] reported an association between OSAS and
GAS tonsil colonization. These authors showed that the GAS
toxin SLO induced production of CysLTs by a TLR4-mediated
pathway that involved the TLR adaptors MyD88 and Toll/IL-1R
COMPLICATIONS ARISING FROM
domain-containing adapter-inducing IFN-b and p38 MAPK.
GAS PHARYNGITIS
CysLTs activated primary T and B cell proliferation in vitro,
GAS pharyngitis has been associated with the development of which might mediate the development of tonsil hypertrophy and
several disease pathologies, including autoimmune diseases. The thus, pediatric OSAS. A second study by the same authors
pharynx is the primary site of infection of GAS, and from there, identified the GAS virulence factor SlaA, which is reported to
local complications can occur, such as peritonsillar or retro- target the CysLT production pathway to also be associated with
pharyngeal abscess formation, as well as tonsillar hypertrophy. OSAS [178]. The slaA gene was found in 45.5% of GAS strains
Autoinflammatory complications that may accompany pharyngi- isolated from OSAS patients. SlaA cleaves the fatty acids palmitic
tis infection can include scarlet fever, whereas postinfectious and oleic acids to release arachidonic acid, which is then
sequelae include ARF and GP [165]. converted to leukotrienes and CysLTs by the 5-lipoxygenase

www.jleukbio.org Volume 103, February 2018 Journal of Leukocyte Biology 13


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
pathway and may contribute to tonsil hypertrophy and thus, microbiota in immunity is an emerging area of research, and the
OSAS [179]. initial studies describing microbiota-mediated regulation of
inflammation during GAS infection will likely gain increasing
GP interest into the future. An improved understanding of the
GP is an abnormal immune reaction characterized by the sudden cellular and humoral responses that mediate immunity to GAS
onset of small, rounded skin lesions, caused by abnormal should enable the development of better therapeutic and
reactivity of specific T lymphocytes in the skin, leading to the preventative strategies to combat GAS infection and related
infiltration of inflammatory cells and proliferation of keratino- immunopathologies.
cytes [180]. GAS pharyngitis is often considered as a trigger of
GP, and in 56–97% of GP cases, GAS pharyngitis precedes lesion AUTHORSHIP
formation [181]. The immunologic mechanisms that cause
lesion formation are not well understood; however, Th17 A.T.S. performed the literature review and drafted the paper.
responses are thought to play an important role [182]. Super- T.C.B., M.J.S., and M.J.W. assisted in drafting, contributed to
antigens from GAS have been demonstrated to enhance T cell revision of the article, and suggested additional references for
expression of CLA+, which is a carbohydrate-moiety expressed by inclusion in the manuscript.
80% of T cells in the skin, and the abundance of CLA+ CD8+
T cells in the blood of patients with psoriasis correlates with ACKNOWLEDGMENTS
disease severity [183]. CLA+ T cells in the tonsils and GP lesions
of the same patient show identical TCRb rearrangement (Vb2+), A.T.S., T.C.B, M.J.S., M.J.W. A.T.S. is supported by a Research
suggesting that migration of these cells occurs from the tonsils to Training Program Scholarship from the Australian government.
the skin [184]. Once in the skin, the CLA+ T cells interact with T.C.B. is supported by a Career Development Fellowship from the
lesional epidermal cells and induce a dominant Th17 response National Health and Medical Research Council (NHMRC)-funded
[185]. Some studies have reported that tonsillectomy results in Improving Health Outcomes in the Tropical North: A Multidisci-
improvement of psoriasis in the majority of cases and is plinary Collaboration (Hot North; APP1131932). M.J.S. is
associated with a reduction in the frequency of skin-homing supported by an NHMRC Senior Research Fellowship
T cells in the blood that were reactive to peptides derived from (APP1107914). M.J.W. is supported by an NHMRC Principal
human epidermal keratins and streptococcal M proteins [185, Research Fellowship (APP1102621). The authors acknowledge
186]. The actual events that drive T cells to migrate from the support from the NHMRC research-granting system.
tonsil to the skin and cause GP are not known. As GAS has been
detected in the skin but not tonsils of patients with GP, it has
been proposed that a transient presence of intracutaneous GAS DISCLOSURES
during GAS pharyngitis could trigger autoimmunity against local The authors declare no conflicts of interest.
skin proteins by molecular mimicry [187].

REFERENCES
1. Bisno, A. L. (2001) Acute pharyngitis. N. Engl. J. Med. 344, 205–211.
CONCLUSIONS 2. Ralph, A. P., Carapetis, J. R. (2013) Group a streptococcal diseases and
their global burden. Curr. Top. Microbiol. Immunol. 368, 1–27.
GAS pharyngitis and resulting postimmune sequelae continue to 3. Walker, M. J., Barnett, T. C., McArthur, J. D., Cole, J. N., Gillen, C. M.,
Henningham, A., Sriprakash, K. S., Sanderson-Smith, M. L., Nizet, V.
cause a significant burden on human health globally, and an (2014) Disease manifestations and pathogenic mechanisms of Group A
improved understanding of how the host immune system Streptococcus. Clin. Microbiol. Rev. 27, 264–301.
4. Shulman, S. T., Bisno, A. L., Clegg, H. W., Gerber, M. A., Kaplan, E. L.,
responds to GAS is important in developing better strategies to Lee, G., Martin, J. M., Van Beneden, C. (2012) Clinical practice
combat infection and disease. An abundance of in vitro data and guideline for the diagnosis and management of group A streptococcal
murine infection studies demonstrates that both the innate and pharyngitis: 2012 update by the Infectious Diseases Society of America.
Clin. Infect. Dis. 55, 1279–1282.
adaptive immune systems play a key role in protection against 5. Martin, J. (2016) The Streptococcus pyogenes carrier state. In Streptococcus
GAS infection. However, our knowledge of the specific signaling pyogenes: Basic Biology to Clinical Manifestations (J. J., Ferretti, D. L.,
Stevens, V. A., Fischetti, eds.), University of Oklahoma Health Sciences
pathways activated during human infection and how innate and Center, Oklahoma City, OK.
adaptive immunity cross communicate with each other is still 6. Kaplan, E. L. (1980) The group A streptococcal upper respiratory tract
carrier state: an enigma. J. Pediatr. 97, 337–345.
quite limited. Key aspects of innate immunity that are particularly 7. Tanz, R. R., Shulman, S. T. (1998) Streptococcal pharyngitis: the carrier
important in defense against GAS include the secretion of state, definition, and management. Pediatr. Ann. 27, 281–285.
8. Roberts, A. L., Connolly, K. L., Kirse, D. J., Evans, A. K., Poehling, K. A.,
proinflammatory mediators by epithelial cells and the roles of Peters, T. R., Reid, S. D. (2012) Detection of group A Streptococcus in
neutrophils and macrophages in the control and clearance of tonsils from pediatric patients reveals high rate of asymptomatic
GAS. Current evidence suggests that Th17 cells are central streptococcal carriage. BMC Pediatr. 12, 3.
9. Anjos, L. M., Marcondes, M. B., Lima, M. F., Mondelli, A. L., Okoshi,
players during GAS pharyngitis, contributing to both protection M. P. (2014) Streptococcal acute pharyngitis. Rev. Soc. Bras. Med. Trop.
against infection as well as immune-related pathologies. Areas 47, 409–413.
10. Hayes, C. S., Williamson, H., Jr. (2001) Management of Group A beta-
that require more extensive exploration in the context of GAS hemolytic streptococcal pharyngitis. Am. Fam. Physician 63, 1557–1564.
pharyngitis include the roles of mast cells and innate lymphoid 11. Steer, A. C., Carapetis, J. R., Dale, J. B., Fraser, J. D., Good, M. F.,
Guilherme, L., Moreland, N. J., Mulholland, E. K., Schodel, F.,
cells, as well as the contribution of the inflammasome pathway to Smeesters, P. R. (2016) Status of research and development of vaccines
inflammation and bacterial clearance in the pharynx. The role of for Streptococcus pyogenes. Vaccine 34, 2953–2958.

14 Journal of Leukocyte Biology Volume 103, February 2018 www.jleukbio.org


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
Soderholm et al. Group A streptococcal pharyngitis and host immune responses

12. Wilkening, R. V., Federle, M. J. (2017) Evolutionary constraints 34. Walker, M. J., Hollands, A., Sanderson-Smith, M. L., Cole, J. N., Kirk,
shaping Streptococcus pyogenes-host interactions. Trends Microbiol. 25, J. K., Henningham, A., McArthur, J. D., Dinkla, K., Aziz, R. K., Kansal,
562–572. R. G., Simpson, A. J., Buchanan, J. T., Chhatwal, G. S., Kotb, M., Nizet, V.
13. Hollingshead, S. K., Simecka, J. W., Michalek, S. M. (1993) Role of M (2007) DNase Sda1 provides selection pressure for a switch to invasive
protein in pharyngeal colonization by group A streptococci in rats. group A streptococcal infection. Nat. Med. 13, 981–985.
Infect. Immun. 61, 2277–2283. 35. Ji, Y., Carlson, B., Kondagunta, A., Cleary, P. P. (1997) Intranasal
14. Sumby, P., Tart, A. H., Musser, J. M. (2008) A non-human primate immunization with C5a peptidase prevents nasopharyngeal
model of acute group a Streptococcus pharyngitis. Methods Mol. Biol. 431, colonization of mice by the group A Streptococcus. Infect. Immun. 65,
255–267. 2080–2087.
15. Park, H. S., Francis, K. P., Yu, J., Cleary, P. P. (2003) Membranous 36. Ji, Y., McLandsborough, L., Kondagunta, A., Cleary, P. P. (1996) C5a
cells in nasal-associated lymphoid tissue: a portal of entry for the peptidase alters clearance and trafficking of group A streptococci by
respiratory mucosal pathogen group A streptococcus. J. Immunol. 171, infected mice. Infect. Immun. 64, 503–510.
2532–2537. 37. Alam, F. M., Bateman, C., Turner, C. E., Wiles, S., Sriskandan, S. (2013)
16. Kasper, K. J., Zeppa, J. J., Wakabayashi, A. T., Xu, S. X., Mazzuca, D. M., Non-invasive monitoring of Streptococcus pyogenes vaccine efficacy using
Welch, I., Baroja, M. L., Kotb, M., Cairns, E., Cleary, P. P., Haeryfar, biophotonic imaging. PLoS One 8, e82123.
S. M., McCormick, J. K. (2014) Bacterial superantigens promote acute 38. Zinkernagel, A. S., Timmer, A. M., Pence, M. A., Locke, J. B.,
nasopharyngeal infection by Streptococcus pyogenes in a human MHC class Buchanan, J. T., Turner, C. E., Mishalian, I., Sriskandan, S., Hanski,
II-dependent manner. PLoS Pathog. 10, e1004155. E., Nizet, V. (2008) The IL-8 protease SpyCEP/ScpC of group A
17. Dinis, M., Plainvert, C., Kovarik, P., Longo, M., Fouet, A., Poyart, C. Streptococcus promotes resistance to neutrophil killing. Cell Host
(2014) The innate immune response elicited by Group A Streptococcus is Microbe 4, 170–178.
highly variable among clinical isolates and correlates with the emm type. 39. Kurupati, P., Turner, C. E., Tziona, I., Lawrenson, R. A., Alam, F. M.,
PLoS One 9, e101464. Nohadani, M., Stamp, G. W., Zinkernagel, A. S., Nizet, V., Edwards,
18. Klenk, M., Nakata, M., Podbielski, A., Skupin, B., Schroten, H., R. J., Sriskandan, S. (2010) Chemokine-cleaving Streptococcus pyogenes
Kreikemeyer, B. (2007) Streptococcus pyogenes serotype-dependent and protease SpyCEP is necessary and sufficient for bacterial
independent changes in infected HEp-2 epithelial cells. ISME J. 1, dissemination within soft tissues and the respiratory tract. Mol.
678–692. Microbiol. 76, 1387–1397.
19. Persson, S. T., Wilk, L., Mörgelin, M., Herwald, H. (2015) Vigilant 40. Hidalgo-Grass, C., Mishalian, I., Dan-Goor, M., Belotserkovsky, I., Eran,
keratinocytes trigger pathogen-associated molecular pattern Y., Nizet, V., Peled, A., Hanski, E. (2006) A streptococcal protease that
signaling in response to streptococcal M1 protein. Infect. Immun. 83, degrades CXC chemokines and impairs bacterial clearance from
4673–4681. infected tissues. EMBO J. 25, 4628–4637.
20. Osterlund, A., Popa, R., Nikkilä, T., Scheynius, A., Engstrand, L. 41. Barnett, T. C., Cole, J. N., Rivera-Hernandez, T., Henningham, A.,
(1997) Intracellular reservoir of Streptococcus pyogenes in vivo: a Paton, J. C., Nizet, V., Walker, M. J. (2015) Streptococcal toxins: role in
possible explanation for recurrent pharyngotonsillitis. Laryngoscope pathogenesis and disease. Cell. Microbiol. 17, 1721–1741.
107, 640–647. 42. Sierig, G., Cywes, C., Wessels, M. R., Ashbaugh, C. D. (2003) Cytotoxic
21. Osterlund, A., Engstrand, L. (1997) An intracellular sanctuary for effects of streptolysin o and streptolysin S enhance the virulence of
Streptococcus pyogenes in human tonsillar epithelium–studies of poorly encapsulated group a streptococci. Infect. Immun. 71, 446–455.
asymptomatic carriers and in vitro cultured biopsies. Acta Otolaryngol. 43. Miyoshi-Akiyama, T., Takamatsu, D., Koyanagi, M., Zhao, J., Imanishi,
117, 883–888. K., Uchiyama, T. (2005) Cytocidal effect of Streptococcus pyogenes on
22. Wang, B., Dileepan, T., Briscoe, S., Hyland, K. A., Kang, J., Khoruts, A., mouse neutrophils in vivo and the critical role of streptolysin S. J. Infect.
Cleary, P. P. (2010) Induction of TGF-beta1 and TGF-beta1-dependent Dis. 192, 107–116.
predominant Th17 differentiation by group A streptococcal infection. 44. Urb, M., Sheppard, D. C. (2012) The role of mast cells in the defence
Proc. Natl. Acad. Sci. USA 107, 5937–5942. against pathogens. PLoS Pathog. 8, e1002619.
23. Wang, B., Li, S., Southern, P. J., Cleary, P. P. (2006) Streptococcal 45. Di Nardo, A., Yamasaki, K., Dorschner, R. A., Lai, Y., Gallo, R. L. (2008)
modulation of cellular invasion via TGF-beta1 signaling. Proc. Natl. Acad. Mast cell cathelicidin antimicrobial peptide prevents invasive group A
Sci. USA 103, 2380–2385. Streptococcus infection of the skin. J. Immunol. 180, 7565–7573.
24. Ignotz, R. A., Massagué, J. (1986) Transforming growth factor-beta 46. Matsui, H., Sekiya, Y., Takahashi, T., Nakamura, M., Imanishi, K.,
stimulates the expression of fibronectin and collagen and their Yoshida, H., Murayama, S. Y., Takahashi, T., Tsuchimoto, K., Uchiyama,
incorporation into the extracellular matrix. J. Biol. Chem. 261, T., Ubukata, K. (2011) Dermal mast cells reduce progressive tissue
4337–4345. necrosis caused by subcutaneous infection with Streptococcus pyogenes in
25. Zambruno, G., Marchisio, P. C., Marconi, A., Vaschieri, C., Melchiori, mice. J. Med. Microbiol. 60, 128–134.
A., Giannetti, A., De Luca, M. (1995) Transforming growth factor-beta 1 47. Von Köckritz-Blickwede, M., Goldmann, O., Thulin, P., Heinemann, K.,
modulates beta 1 and beta 5 integrin receptors and induces the de novo Norrby-Teglund, A., Rohde, M., Medina, E. (2008) Phagocytosis-
expression of the alpha v beta 6 heterodimer in normal human independent antimicrobial activity of mast cells by means of
keratinocytes: implications for wound healing. J. Cell Biol. 129, 853–865. extracellular trap formation. Blood 111, 3070–3080.
26. Tsai, P. J., Chen, Y. H., Hsueh, C. H., Hsieh, H. C., Liu, Y. H., Wu, J. J., 48. Stassen, M., Müller, C., Richter, C., Neudörfl, C., Hültner, L., Bhakdi, S.,
Tsou, C. C. (2006) Streptococcus pyogenes induces epithelial inflammatory Walev, I., Schmitt, E. (2003) The streptococcal exotoxin streptolysin O
responses through NF-kappaB/MAPK signaling pathways. Microbes Infect. activates mast cells to produce tumor necrosis factor alpha by p38
8, 1440–1449. mitogen-activated protein kinase- and protein kinase C-dependent
27. Döhrmann, S., Cole, J. N., Nizet, V. (2016) Conquering neutrophils. pathways. Infect. Immun. 71, 6171–6177.
PLoS Pathog. 12, e1005682. 49. Watanabe, Y., Todome, Y., Ohkuni, H., Sakurada, S., Ishikawa, T.,
28. Chen, X., Li, N., Bi, S., Wang, X., Wang, B. (2016) Co-activation of Th17 Yutsudo, T., Fischetti, V. A., Zabriskie, J. B. (2002) Cysteine protease
and antibody responses provides efficient protection against mucosal activity and histamine release from the human mast cell line HMC-1
infection by Group A Streptococcus. PLoS One 11, e0168861. stimulated by recombinant streptococcal pyrogenic exotoxin B/
29. Laan, M., Prause, O., Miyamoto, M., Sjöstrand, M., Hytönen, A. M., streptococcal cysteine protease. Infect. Immun. 70, 3944–3947.
Kaneko, T., Lötvall, J., Lindén, A. (2003) A role of GM-CSF in the 50. Fieber, C., Janos, M., Koestler, T., Gratz, N., Li, X. D., Castiglia, V.,
accumulation of neutrophils in the airways caused by IL-17 and TNF- Aberle, M., Sauert, M., Wegner, M., Alexopoulou, L., Kirschning, C. J.,
alpha. Eur. Respir. J. 21, 387–393. Chen, Z. J., von Haeseler, A., Kovarik, P. (2015) Innate immune
30. Bozinovski, S., Seow, H. J., Chan, S. P., Anthony, D., McQualter, J., response to Streptococcus pyogenes depends on the combined activation of
Hansen, M., Jenkins, B. J., Anderson, G. P., Vlahos, R. (2015) Innate TLR13 and TLR2. PLoS One 10, e0119727.
cellular sources of interleukin-17A regulate macrophage accumulation 51. Mishalian, I., Ordan, M., Peled, A., Maly, A., Eichenbaum, M. B., Ravins,
in cigarette- smoke-induced lung inflammation in mice. Clin. Sci. 129, M., Aychek, T., Jung, S., Hanski, E. (2011) Recruited macrophages
785–796. control dissemination of group A Streptococcus from infected soft tissues.
31. Fan, X., Wang, X., Li, N., Cui, H., Hou, B., Gao, B., Cleary, P. P., Wang, J. Immunol. 187, 6022–6031.
B. (2014) Sortase A induces Th17-mediated and antibody-independent 52. Gratz, N., Hartweger, H., Matt, U., Kratochvill, F., Janos, M., Sigel, S.,
immunity to heterologous serotypes of group A streptococci. PLoS One Drobits, B., Li, X. D., Knapp, S., Kovarik, P. (2011) Type I interferon
9, e107638. production induced by Streptococcus pyogenes-derived nucleic acids is
32. Buchanan, J. T., Simpson, A. J., Aziz, R. K., Liu, G. Y., Kristian, S. A., required for host protection. PLoS Pathog. 7, e1001345.
Kotb, M., Feramisco, J., Nizet, V. (2006) DNase expression allows the 53. Castiglia, V., Piersigilli, A., Ebner, F., Janos, M., Goldmann, O.,
pathogen group A Streptococcus to escape killing in neutrophil Damböck, U., Kröger, A., Weiss, S., Knapp, S., Jamieson, A. M.,
extracellular traps. Curr. Biol. 16, 396–400. Kirschning, C., Kalinke, U., Strobl, B., Müller, M., Stoiber, D.,
33. Cole, J. N., Barnett, T. C., Nizet, V., Walker, M. J. (2011) Molecular Lienenklaus, S., Kovarik, P. (2016) Type I interferon signaling prevents
insight into invasive group A streptococcal disease. Nat. Rev. Microbiol. 9, IL-1b-driven lethal systemic hyperinflammation during invasive
724–736. bacterial infection of soft tissue. Cell Host Microbe 19, 375–387.

www.jleukbio.org Volume 103, February 2018 Journal of Leukocyte Biology 15


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
54. Veckman, V., Miettinen, M., Matikainen, S., Lande, R., Giacomini, E., nonredundant role for recognition of Streptococcus pyogenes. J. Immunol.
Coccia, E. M., Julkunen, I. (2003) Lactobacilli and streptococci induce 195, 1092–1099.
inflammatory chemokine production in human macrophages that 77. Krüger, A., Oldenburg, M., Chebrolu, C., Beisser, D., Kolter, J.,
stimulates Th1 cell chemotaxis. J. Leukoc. Biol. 74, 395–402. Sigmund, A. M., Steinmann, J., Schäfer, S., Hochrein, H., Rahmann, S.,
55. Latvala, S., Mäkelä, S. M., Miettinen, M., Charpentier, E., Julkunen, I. Wagner, H., Henneke, P., Hornung, V., Buer, J., Kirschning, C. J. (2015)
(2014) Dynamin inhibition interferes with inflammasome activation Human TLR8 senses UR/URR motifs in bacterial and mitochondrial
and cytokine gene expression in Streptococcus pyogenes-infected human RNA. EMBO Rep. 16, 1656–1663.
macrophages. Clin. Exp. Immunol. 178, 320–333. 78. Campbell, G. R., Spector, S. A. (2012) Toll-like receptor 8 ligands
56. Dennis, E. A., Norris, P. C. (2015) Eicosanoid storm in infection and activate a vitamin D mediated autophagic response that inhibits human
inflammation. Nat. Rev. Immunol. 15, 511–523. immunodeficiency virus type 1. PLoS Pathog. 8, e1003017.
57. Goldmann, O., Hertzén, E., Hecht, A., Schmidt, H., Lehne, S., Norrby- 79. Nakagawa, I., Amano, A., Mizushima, N., Yamamoto, A., Yamaguchi, H.,
Teglund, A., Medina, E. (2010) Inducible cyclooxygenase released Kamimoto, T., Nara, A., Funao, J., Nakata, M., Tsuda, K., Hamada, S.,
prostaglandin E2 modulates the severity of infection caused by Yoshimori, T. (2004) Autophagy defends cells against invading group A
Streptococcus pyogenes. J. Immunol. 185, 2372–2381. Streptococcus. Science 306, 1037–1040.
58. Serezani, C. H., Ballinger, M. N., Aronoff, D. M., Peters-Golden, M. 80. Latz, E., Xiao, T. S., Stutz, A. (2013) Activation and regulation of the
(2008) Cyclic AMP: master regulator of innate immune cell function. inflammasomes. Nat. Rev. Immunol. 13, 397–411.
Am. J. Respir. Cell Mol. Biol. 39, 127–132. 81. Harder, J., Franchi, L., Muñoz-Planillo, R., Park, J. H., Reimer, T.,
59. Soares, E. M., Mason, K. L., Rogers, L. M., Serezani, C. H., Faccioli, Núñez, G. (2009) Activation of the Nlrp3 inflammasome by Streptococcus
L. H., Aronoff, D. M. (2013) Leukotriene B4 enhances innate immune pyogenes requires streptolysin O and NF-kappa B activation but proceeds
defense against the puerperal sepsis agent Streptococcus pyogenes. J. independently of TLR signaling and P2X7 receptor. J. Immunol. 183,
Immunol. 190, 1614–1622. 5823–5829.
60. Goldmann, O., Sastalla, I., Wos-Oxley, M., Rohde, M., Medina, E. (2009) 82. Hancz, D., Westerlund, E., Bastiat-Sempe, B., Sharma, O., Valfridsson,
Streptococcus pyogenes induces oncosis in macrophages through the C., Meyer, L., Love, J. F., O’Seaghdha, M., Wessels, M. R., Persson, J. J.
activation of an inflammatory programmed cell death pathway. Cell. (2017) Inhibition of inflammasome-dependent interleukin 1b
Microbiol. 11, 138–155. production by Streptococcal NAD(+)-glycohydrolase: evidence for
61. Gryllos, I., Grifantini, R., Colaprico, A., Cary, M. E., Hakansson, A., extracellular activity. MBio 8, e00756-17.
Carey, D. W., Suarez-Chavez, M., Kalish, L. A., Mitchell, P. D., White, 83. Lin, A. E., Beasley, F. C., Keller, N., Hollands, A., Urbano, R., Troemel,
G. L., Wessels, M. R. (2008) PerR confers phagocytic killing resistance E. R., Hoffman, H. M., Nizet, V. (2015) A group A Streptococcus ADP-
and allows pharyngeal colonization by group A Streptococcus. PLoS Pathog. ribosyltransferase toxin stimulates a protective interleukin 1b-
4, e1000145. dependent macrophage immune response. MBio 6, e00133.
62. Franklin, L., Nobbs, A. H., Bricio-Moreno, L., Wright, C. J., Maddocks, 84. LaRock, C. N., Todd, J., LaRock, D. L., Olson, J., O’Donoghue, A. J.,
S. E., Sahota, J. S., Ralph, J., O’Connor, M., Jenkinson, H. F., Kadioglu, Robertson, A. A., Cooper, M. A., Hoffman, H. M., Nizet, V. (2016) IL-
A. (2013) The AgI/II family adhesin AspA is required for respiratory 1beta is an innate immune sensor of microbial proteolysis. Sci. Immunol.
infection by Streptococcus pyogenes. PLoS One 8, e62433. 1, eaah3539.
63. Barnett, T. C., Liebl, D., Seymour, L. M., Gillen, C. M., Lim, J. Y., Larock, 85. Medina, E., Anders, D., Chhatwal, G. S. (2002) Induction of NF-kappaB
C. N., Davies, M. R., Schulz, B. L., Nizet, V., Teasdale, R. D., Walker, nuclear translocation in human respiratory epithelial cells by group A
M. J. (2013) The globally disseminated M1T1 clone of group A streptococci. Microb. Pathog. 33, 307–313.
Streptococcus evades autophagy for intracellular replication. Cell Host 86. Chandrasekaran, S., Caparon, M. G. (2016) The NADase-negative
Microbe 14, 675–682. variant of the Streptococcus pyogenes toxin NAD+ glycohydrolase
64. Thulin, P., Johansson, L., Low, D. E., Gan, B. S., Kotb, M., McGeer, A., induces JNK1-mediated programmed cellular necrosis. MBio 7,
Norrby-Teglund, A. (2006) Viable group A streptococci in macrophages e02215–e15.
during acute soft tissue infection. PLoS Med. 3, e53. 87. Okahashi, N., Sakurai, A., Nakagawa, I., Fujiwara, T., Kawabata, S.,
65. Loof, T. G., Rohde, M., Chhatwal, G. S., Jung, S., Medina, E. (2007) The Amano, A., Hamada, S. (2003) Infection by Streptococcus pyogenes induces
contribution of dendritic cells to host defenses against Streptococcus the receptor activator of NF-kappaB ligand expression in mouse
pyogenes. J. Infect. Dis. 196, 1794–1803. osteoblastic cells. Infect. Immun. 71, 948–955.
66. Veckman, V., Miettinen, M., Pirhonen, J., Sirén, J., Matikainen, S., 88. Chung, W. O., Dale, B. A. (2004) Innate immune response of oral and
Julkunen, I. (2004) Streptococcus pyogenes and Lactobacillus rhamnosus foreskin keratinocytes: utilization of different signaling pathways by
differentially induce maturation and production of Th1-type cytokines various bacterial species. Infect. Immun. 72, 352–358.
and chemokines in human monocyte-derived dendritic cells. J. Leukoc. 89. Klenk, M., Koczan, D., Guthke, R., Nakata, M., Thiesen, H. J.,
Biol. 75, 764–771. Podbielski, A., Kreikemeyer, B. (2005) Global epithelial cell
67. Ye, J., Ortaldo, J. R., Conlon, K., Winkler-Pickett, R., Young, H. A. transcriptional responses reveal Streptococcus pyogenes Fas regulator
(1995) Cellular and molecular mechanisms of IFN-gamma production activity association with bacterial aggressiveness. Cell. Microbiol. 7,
induced by IL-2 and IL-12 in a human NK cell line. J. Leukoc. Biol. 58, 1237–1250.
225–233. 90. Hai, T., Curran, T. (1991) Cross-family dimerization of transcription
68. Schroder, K., Sweet, M. J., Hume, D. A. (2006) Signal integration factors Fos/Jun and ATF/CREB alters DNA binding specificity. Proc.
between IFNgamma and TLR signalling pathways in macrophages. Natl. Acad. Sci. USA 88, 3720–3724.
Immunobiology 211, 511–524. 91. Wang, B., Ruiz, N., Pentland, A., Caparon, M. (1997) Keratinocyte
69. Goldmann, O., Chhatwal, G. S., Medina, E. (2005) Contribution of proinflammatory responses to adherent and nonadherent group A
natural killer cells to the pathogenesis of septic shock induced by streptococci. Infect. Immun. 65, 2119–2126.
Streptococcus pyogenes in mice. J. Infect. Dis. 191, 1280–1286. 92. Kalinski, P. (2012) Regulation of immune responses by prostaglandin
70. Loof, T. G., Goldmann, O., Gessner, A., Herwald, H., Medina, E. (2010) E2. J. Immunol. 188, 21–28.
Aberrant inflammatory response to Streptococcus pyogenes in mice lacking 93. Agard, M., Asakrah, S., Morici, L. A. (2013) PGE(2) suppression of
myeloid differentiation factor 88. Am. J. Pathol. 176, 754–763. innate immunity during mucosal bacterial infection. Front. Cell. Infect.
71. Fieber, C., Kovarik, P. (2014) Responses of innate immune cells to Microbiol. 3, 45.
group A Streptococcus. Front. Cell. Infect. Microbiol. 4, 140. 94. Shea, P. R., Virtaneva, K., Kupko III, J. J., Porcella, S. F., Barry, W. T.,
72. Tsatsaronis, J. A., Walker, M. J., Sanderson-Smith, M. L. (2014) Host Wright, F. A., Kobayashi, S. D., Carmody, A., Ireland, R. M., Sturdevant,
responses to group a streptococcus: cell death and inflammation. PLoS D. E., Ricklefs, S. M., Babar, I., Johnson, C. A., Graham, M. R., Gardner,
Pathog. 10, e1004266. D. J., Bailey, J. R., Parnell, M. J., Deleo, F. R., Musser, J. M. (2010)
73. Loof, T. G., Goldmann, O., Medina, E. (2008) Immune recognition Interactome analysis of longitudinal pharyngeal infection of
of Streptococcus pyogenes by dendritic cells. Infect. Immun. 76, cynomolgus macaques by group A Streptococcus. Proc. Natl. Acad. Sci. USA
2785–2792. 107, 4693–4698.
74. Gratz, N., Siller, M., Schaljo, B., Pirzada, Z. A., Gattermeier, I., Vojtek, I., 95. Vega, L. A., Malke, H., McIver, K. S. (2016) Virulence-related
Kirschning, C. J., Wagner, H., Akira, S., Charpentier, E., Kovarik, P. transcriptional regulators of Streptococcus pyogenes. In Streptococcus
(2008) Group A streptococcus activates type I interferon production and pyogenes: Basic Biology to Clinical Manifestations. (J. J., Ferretti, D. L.,
MyD88-dependent signaling without involvement of TLR2, TLR4, and Stevens, V. A., Fischetti, eds.), University of Oklahoma Health Sciences
TLR9. J. Biol. Chem. 283, 19879–19887. Center, Oklahoma City, OK.
75. Oldenburg, M., Krüger, A., Ferstl, R., Kaufmann, A., Nees, G., Sigmund, 96. Egesten, A., Eliasson, M., Johansson, H. M., Olin, A. I., Morgelin, M.,
A., Bathke, B., Lauterbach, H., Suter, M., Dreher, S., Koedel, U., Akira, Mueller, A., Pease, J. E., Frick, I. M., Bjorck, L. (2007) The CXC
S., Kawai, T., Buer, J., Wagner, H., Bauer, S., Hochrein, H., Kirschning, chemokine MIG/CXCL9 is important in innate immunity against
C. J. (2012) TLR13 recognizes bacterial 23S rRNA devoid of Streptococcus pyogenes. J. Infect. Dis. 195, 684–693.
erythromycin resistance-forming modification. Science 337, 1111–1115. 97. Linge, H. M., Sastalla, I., Nitsche-Schmitz, D. P., Egesten, A., Frick,
76. Eigenbrod, T., Pelka, K., Latz, E., Kreikemeyer, B., Dalpke, A. H. (2015) I. M. (2007) Protein FOG is a moderate inducer of MIG/CXCL9,
TLR8 senses bacterial RNA in human monocytes and plays a and group G streptococci are more tolerant than group A

16 Journal of Leukocyte Biology Volume 103, February 2018 www.jleukbio.org


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
Soderholm et al. Group A streptococcal pharyngitis and host immune responses

streptococci to this chemokine’s antibacterial effect. Microbiology 117. Fernie-King, B. A., Seilly, D. J., Lachmann, P. J. (2004) The
153, 3800–3808. interaction of streptococcal inhibitor of complement (SIC) and its
98. Verma, V., Kumar, P., Dhanda, R. S., Yadav, M. (2016) Kinetics of proteolytic fragments with the human beta defensins. Immunology
cytokine profile in response to Mycobacterium bovis BCG and Streptococcus 111, 444–452.
pyogenes activated cells. Data Brief 7, 445–448. 118. Peschel, A., Sahl, H. G. (2006) The co-evolution of host cationic
99. Sumby, P., Zhang, S., Whitney, A. R., Falugi, F., Grandi, G., Graviss, antimicrobial peptides and microbial resistance. Nat. Rev. Microbiol. 4,
E. A., Deleo, F. R., Musser, J. M. (2008) A chemokine-degrading 529–536.
extracellular protease made by group A Streptococcus alters pathogenesis 119. Frick, I. M., Akesson, P., Rasmussen, M., Schmidtchen, A., Björck, L.
by enhancing evasion of the innate immune response. Infect. Immun. 76, (2003) SIC, a secreted protein of Streptococcus pyogenes that inactivates
978–985. antibacterial peptides. J. Biol. Chem. 278, 16561–16566.
100. Egesten, A., Olin, A. I., Linge, H. M., Yadav, M., Mörgelin, M., Karlsson, 120. Fernie-King, B. A., Seilly, D. J., Davies, A., Lachmann, P. J. (2002)
A., Collin, M. (2009) SpeB of Streptococcus pyogenes differentially Streptococcal inhibitor of complement inhibits two additional
modulates antibacterial and receptor activating properties of human components of the mucosal innate immune system: secretory
chemokines. PLoS One 4, e4769. leukocyte proteinase inhibitor and lysozyme. Infect. Immun. 70,
101. Shelburne III, S. A., Granville, C., Tokuyama, M., Sitkiewicz, I., Patel, P., 4908–4916.
Musser, J. M. (2005) Growth characteristics of and virulence factor 121. Lukomski, S., Hoe, N. P., Abdi, I., Rurangirwa, J., Kordari, P., Liu, M.,
production by group A Streptococcus during cultivation in human saliva. Dou, S. J., Adams, G. G., Musser, J. M. (2000) Nonpolar inactivation of
Infect. Immun. 73, 4723–4731. the hypervariable streptococcal inhibitor of complement gene (sic) in
102. Svensson, M. D., Scaramuzzino, D. A., Sjöbring, U., Olsén, A., Frank, C., serotype M1 Streptococcus pyogenes significantly decreases mouse mucosal
Bessen, D. E. (2000) Role for a secreted cysteine proteinase in the colonization. Infect. Immun. 68, 535–542.
establishment of host tissue tropism by group A streptococci. Mol. 122. Hollands, A., Gonzalez, D., Leire, E., Donald, C., Gallo, R. L.,
Microbiol. 38, 242–253. Sanderson-Smith, M., Dorrestein, P. C., Nizet, V. (2012) A bacterial
103. Courtney, H. S., Ofek, I., Hasty, D. L. (1997) M protein mediated pathogen co-opts host plasmin to resist killing by cathelicidin
adhesion of M type 24 Streptococcus pyogenes stimulates release of antimicrobial peptides. J. Biol. Chem. 287, 40891–40897.
interleukin-6 by HEp-2 tissue culture cells. FEMS Microbiol. Lett. 151, 123. Love, J. F., Tran-Winkler, H. J., Wessels, M. R. (2012) Vitamin D and the
65–70. human antimicrobial peptide LL-37 enhance group a streptococcus
104. Miettinen, M., Veckman, V., Latvala, S., Sareneva, T., Matikainen, S., resistance to killing by human cells. MBio 3, e00394-12.
Julkunen, I. (2008) Live Lactobacillus rhamnosus and Streptococcus 124. Lauth, X., von Köckritz-Blickwede, M., McNamara, C. W., Myskowski, S.,
pyogenes differentially regulate Toll-like receptor (TLR) gene Zinkernagel, A. S., Beall, B., Ghosh, P., Gallo, R. L., Nizet, V. (2009) M1
expression in human primary macrophages. J. Leukoc. Biol. 84, protein allows Group A streptococcal survival in phagocyte extracellular
1092–1100. traps through cathelicidin inhibition. J. Innate Immun. 1, 202–214.
105. Leão, S. C., Leal, I. O., Rocha, H. M., Rodrigues, T. M. (2015) 125. LaRock, C. N., Döhrmann, S., Todd, J., Corriden, R., Olson, J.,
Evaluation of cytokines produced by b-hemolytic streptococcus in acute Johannssen, T., Lepenies, B., Gallo, R. L., Ghosh, P., Nizet, V. (2015)
pharyngotonsillitis. Bras. J. Otorhinolaryngol. 81, 402–407. Group A Streptococcal M1 protein sequesters cathelicidin to evade
106. Kapur, V., Majesky, M. W., Li, L. L., Black, R. A., Musser, J. M. (1993) innate immune killing. Cell Host Microbe 18, 471–477.
Cleavage of interleukin 1 beta (IL-1 beta) precursor to produce active 126. Fernie-King, B. A., Seilly, D. J., Willers, C., Würzner, R., Davies, A.,
IL-1 beta by a conserved extracellular cysteine protease from Lachmann, P. J. (2001) Streptococcal inhibitor of complement (SIC)
Streptococcus pyogenes. Proc. Natl. Acad. Sci. USA 90, 7676–7680. inhibits the membrane attack complex by preventing uptake of C567
107. Sakurai, A., Okahashi, N., Nakagawa, I., Kawabata, S., Amano, A., onto cell membranes. Immunology 103, 390–398.
Ooshima, T., Hamada, S. (2003) Streptococcus pyogenes infection induces 127. Serruto, D., Rappuoli, R., Scarselli, M., Gros, P., van Strijp, J. A. (2010)
septic arthritis with increased production of the receptor activator of Molecular mechanisms of complement evasion: learning from
the NF-kappaB ligand. Infect. Immun. 71, 6019–6026. staphylococci and meningococci. Nat. Rev. Microbiol. 8, 393–399.
108. Lappalainen, U., Whitsett, J. A., Wert, S. E., Tichelaar, J. W., Bry, K. 128. Thern, A., Stenberg, L., Dahlbäck, B., Lindahl, G. (1995) Ig-binding
(2005) Interleukin-1beta causes pulmonary inflammation, emphysema, surface proteins of Streptococcus pyogenes also bind human C4b-binding
and airway remodeling in the adult murine lung. Am. J. Respir. Cell Mol. protein (C4BP), a regulatory component of the complement system. J.
Biol. 32, 311–318. Immunol. 154, 375–386.
109. Kazmi, S. U., Kansal, R., Aziz, R. K., Hooshdaran, M., Norrby-Teglund, 129. Horstmann, R. D., Sievertsen, H. J., Knobloch, J., Fischetti, V. A. (1988)
A., Low, D. E., Halim, A. B., Kotb, M. (2001) Reciprocal, temporal Antiphagocytic activity of streptococcal M protein: selective binding of
expression of SpeA and SpeB by invasive M1T1 group a streptococcal complement control protein factor H. Proc. Natl. Acad. Sci. USA 85,
isolates in vivo. Infect. Immun. 69, 4988–4995. 1657–1661.
110. Olsen, R. J., Raghuram, A., Cantu, C., Hartman, M. H., Jimenez, F. E., 130. Johnsson, E., Berggård, K., Kotarsky, H., Hellwage, J., Zipfel, P. F.,
Lee, S., Ngo, A., Rice, K. A., Saddington, D., Spillman, H., Valson, C., Sjöbring, U., Lindahl, G. (1998) Role of the hypervariable region in
Flores, A. R., Beres, S. B., Long, S. W., Nasser, W., Musser, J. M. (2015) streptococcal M proteins: binding of a human complement inhibitor. J.
The majority of 9,729 group A streptococcus strains causing disease Immunol. 161, 4894–4901.
secrete SpeB cysteine protease: pathogenesis implications. Infect. Immun. 131. Carlsson, F., Sandin, C., Lindahl, G. (2005) Human fibrinogen bound to
83, 4750–4758. Streptococcus pyogenes M protein inhibits complement deposition via the
111. Yang, D., Chertov, O., Oppenheim, J. J. (2001) The role of classical pathway. Mol. Microbiol. 56, 28–39.
mammalian antimicrobial peptides and proteins in awakening of 132. Ly, D., Taylor, J. M., Tsatsaronis, J. A., Monteleone, M. M., Skora, A. S.,
innate host defenses and adaptive immunity. Cell. Mol. Life Sci. 58, Donald, C. A., Maddocks, T., Nizet, V., West, N. P., Ranson, M., Walker,
978–989. M. J., McArthur, J. D., Sanderson-Smith, M. L. (2014) Plasmin(ogen)
112. Frick, I. M., Nordin, S. L., Baumgarten, M., Mörgelin, M., Sørensen, acquisition by group A Streptococcus protects against C3b-mediated
O. E., Olin, A. I., Egesten, A. (2011) Constitutive and inflammation- neutrophil killing. J. Innate Immun. 6, 240–250.
dependent antimicrobial peptides produced by epithelium are 133. Von Bernuth, H., Picard, C., Puel, A., Casanova, J. L. (2012)
differentially processed and inactivated by the commensal Finegoldia Experimental and natural infections in MyD88- and IRAK-4-deficient
magna and the pathogen Streptococcus pyogenes. J. Immunol. 187, mice and humans. Eur. J. Immunol. 42, 3126–3135.
4300–4309. 134. Picard, C., Casanova, J. L., Puel, A. (2011) Infectious diseases in patients
113. Dorschner, R. A., Pestonjamasp, V. K., Tamakuwala, S., Ohtake, T., with IRAK-4, MyD88, NEMO, or IkBa deficiency. Clin. Microbiol. Rev. 24,
Rudisill, J., Nizet, V., Agerberth, B., Gudmundsson, G. H., Gallo, R. L. 490–497.
(2001) Cutaneous injury induces the release of cathelicidin anti- 135. Mortensen, R., Nissen, T. N., Blauenfeldt, T., Christensen, J. P.,
microbial peptides active against group A Streptococcus. J. Invest. Dermatol. Andersen, P., Dietrich, J. (2015) Adaptive immunity against Streptococcus
117, 91–97. pyogenes in adults involves increased IFN-g and IgG3 responses
114. Yu, J., Mookherjee, N., Wee, K., Bowdish, D. M., Pistolic, J., Li, Y., compared with children. J. Immunol. 195, 1657–1664.
Rehaume, L., Hancock, R. E. (2007) Host defense peptide LL-37, in 136. Agren, K., Brauner, A., Andersson, J. (1998) Haemophilus influenzae and
synergy with inflammatory mediator IL-1beta, augments immune Streptococcus pyogenes group A challenge induce a Th1 type of cytokine
responses by multiple pathways. J. Immunol. 179, 7684–7691. response in cells obtained from tonsillar hypertrophy and recurrent
115. Quinn, G. A., Cole, A. M. (2007) Suppression of innate immunity by a tonsillitis. ORL J. Otorhinolaryngol. Relat. Spec. 60, 35–41.
nasal carriage strain of Staphylococcus aureus increases its colonization on 137. Hyland, K. A., Brennan, R., Olmsted, S. B., Rojas, E., Murphy, E., Wang,
nasal epithelium. Immunology 122, 80–89. B., Cleary, P. P. (2009) The early interferon response of nasal-associated
116. Bell, S., Howard, A., Wilson, J. A., Abbot, E. L., Smith, W. D., Townes, lymphoid tissue to Streptococcus pyogenes infection. FEMS Immunol. Med.
C. L., Hirst, B. H., Hall, J. (2012) Streptococcus pyogenes infection of tonsil Microbiol. 55, 422–431.
explants is associated with a human b-defensin 1 response from control 138. Raeder, R. H., Barker-Merrill, L., Lester, T., Boyle, M. D., Metzger, D. W.
but not recurrent acute tonsillitis patients. Mol. Oral Microbiol. 27, (2000) A pivotal role for interferon-gamma in protection against group
160–171. A streptococcal skin infection. J. Infect. Dis. 181, 639–645.

www.jleukbio.org Volume 103, February 2018 Journal of Leukocyte Biology 17


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
139. Dong, C. (2008) IL-23/IL-17 biology and therapeutic considerations. J. probiotic Streptococcus salivarius K12 in preventing recurrent pharyngitis
Immunotoxicol. 5, 43–46. and/or tonsillitis caused by Streptococcus pyogenes and recurrent acute
140. Korn, T., Bettelli, E., Oukka, M., Kuchroo, V. K. (2009) IL-17 and Th17 otitis media. Int. J. Gen. Med. 5, 991–997.
cells. Annu. Rev. Immunol. 27, 485–517. 162. Di Pierro, F., Colombo, M., Zanvit, A., Rottoli, A. S. (2016) Positive
141. Linehan, J. L., Dileepan, T., Kashem, S. W., Kaplan, D. H., Cleary, P., clinical outcomes derived from using Streptococcus salivarius K12 to
Jenkins, M. K. (2015) Generation of Th17 cells in response to intranasal prevent streptococcal pharyngotonsillitis in children: a pilot
infection requires TGF-b1 from dendritic cells and IL-6 from CD301b+ investigation. Drug Healthc. Patient Saf. 8, 77–81.
dendritic cells. Proc. Natl. Acad. Sci. USA 112, 12782–12787. 163. Di Pierro, F., Colombo, M., Giuliani, M. G., Danza, M. L., Basile, I.,
142. Dileepan, T., Linehan, J. L., Moon, J. J., Pepper, M., Jenkins, M. K., Bollani, T., Conti, A. M., Zanvit, A., Rottoli, A. S. (2016) Effect of
Cleary, P. P. (2011) Robust antigen specific th17 T cell response to administration of Streptococcus salivarius K12 on the occurrence of
group A Streptococcus is dependent on IL-6 and intranasal route of streptococcal pharyngo-tonsillitis, scarlet fever and acute otitis
infection. PLoS Pathog. 7, e1002252. media in 3 years old children. Eur. Rev. Med. Pharmacol. Sci. 20,
143. Rizzo, A., Losacco, A., Carratelli, C. R., Domenico, M. D., Bevilacqua, N. 4601–4606.
(2013) Lactobacillus plantarum reduces Streptococcus pyogenes virulence by 164. Di Pierro, F., Adami, T., Rapacioli, G., Giardini, N., Streitberger, C.
modulating the IL-17, IL-23 and Toll-like receptor 2/4 expressions in (2013) Clinical evaluation of the oral probiotic Streptococcus
human epithelial cells. Int. Immunopharmacol. 17, 453–461. salivarius K12 in the prevention of recurrent pharyngitis and/or
144. Carey, A. J., Weinberg, J. B., Dawid, S. R., Venturini, C., Lam, A. K., tonsillitis caused by Streptococcus pyogenes in adults. Expert Opin. Biol.
Nizet, V., Caparon, M. G., Walker, M. J., Watson, M. E., Ulett, G. C. Ther. 13, 339–343.
(2016) Interleukin-17A contributes to the control of Streptococcus 165. Wessels, M. R. (2016). Pharyngitis and scarlet fever. In Streptococcus
pyogenes colonization and inflammation of the female genital tract. Sci. pyogenes: Basic Biology to Clinical Manifestations. (J. J., Ferretti, D. L.,
Rep. 6, 26836. Stevens, V. A., Fischetti, eds.), University of Oklahoma Health Sciences
145. Chong, D. L., Ingram, R. J., Lowther, D. E., Muir, R., Sriskandan, S., Center, Oklahoma City, OK.
Altmann, D. M. (2012) The nature of innate and adaptive interleukin- 166. Marijon, E., Mirabel, M., Celermajer, D. S., Jouven, X. (2012)
17A responses in sham or bacterial inoculation. Immunology 136, Rheumatic heart disease. Lancet 379, 953–964.
325–333. 167. Seckeler, M. D., Hoke, T. R. (2011) The worldwide epidemiology of
146. Joosten, L. A., Abdollahi-Roodsaz, S., Heuvelmans-Jacobs, M., Helsen, acute rheumatic fever and rheumatic heart disease. Clin. Epidemiol. 3,
M. M., van den Bersselaar, L. A., Oppers-Walgreen, B., Koenders, M. I., 67–84.
van den Berg, W. B. (2008) T Cell dependence of chronic destructive 168. Carapetis, J. R., Steer, A. C., Mulholland, E. K., Weber, M. (2005) The
murine arthritis induced by repeated local activation of Toll-like global burden of group A streptococcal diseases. Lancet Infect. Dis. 5,
receptor-driven pathways: crucial role of both interleukin-1beta and 685–694.
interleukin-17. Arthritis Rheum. 58, 98–108. 169. Carapetis, J. R., McDonald, M., Wilson, N. J. (2005) Acute rheumatic
147. Olofsson, K., Hellström, S., Hammarström, M. L. (1998) The surface fever. Lancet 366, 155–168.
epithelium of recurrent infected palatine tonsils is rich in gammadelta 170. Birdi, N., Hosking, M., Clulow, M. K., Duffy, C. M., Allen, U., Petty, R. E.
T cells. Clin. Exp. Immunol. 111, 36–47. (2001) Acute rheumatic fever and poststreptococcal reactive arthritis:
148. Martin, B., Hirota, K., Cua, D. J., Stockinger, B., Veldhoen, M. (2009) diagnostic and treatment practices of pediatric subspecialists in Canada.
Interleukin-17-producing gammadelta T cells selectively expand in J. Rheumatol. 28, 1681–1688.
response to pathogen products and environmental signals. Immunity 31, 171. Gerber, M. A., Baltimore, R. S., Eaton, C. B., Gewitz, M., Rowley, A. H.,
321–330. Shulman, S. T., Taubert, K. A. (2009) Prevention of rheumatic fever and
149. Costalonga, M., Cleary, P. P., Fischer, L. A., Zhao, Z. (2009) Intranasal diagnosis and treatment of acute Streptococcal pharyngitis: a scientific
bacteria induce Th1 but not Treg or Th2. Mucosal Immunol. 2, 85–95. statement from the American Heart Association Rheumatic Fever,
150. Taylor, A. L., Cross, E. L., Llewelyn, M. J. (2012) Induction of contact- Endocarditis, and Kawasaki Disease Committee of the Council on
dependent CD8(+) regulatory T cells through stimulation with Cardiovascular Disease in the Young, the Interdisciplinary Council on
staphylococcal and streptococcal superantigens. Immunology 135, Functional Genomics and Translational Biology, and the
158–167. Interdisciplinary Council on Quality of Care and Outcomes Research:
151. Price, J. D., Schaumburg, J., Sandin, C., Atkinson, J. P., Lindahl, G., endorsed by the American Academy of Pediatrics. Circulation 119,
Kemper, C. (2005) Induction of a regulatory phenotype in human CD4+ 1541–1551.
T cells by streptococcal M protein. J. Immunol. 175, 677–684. 172. Chalker, V., Jironkin, A., Coelho, J., Al-Shahib, A., Platt, S., Kapatai, G.,
152. Shaler, C. R., Choi, J., Rudak, P. T., Memarnejadian, A., Szabo, P. A., Daniel, R., Dhami, C., Laranjeira, M., Chambers, T., Guy, R., Lamagni,
Tun-Abraham, M. E., Rossjohn, J., Corbett, A. J., McCluskey, J., T., Harrison, T., Chand, M., Johnson, A. P., Underwood, A.; Scarlet
McCormick, J. K., Lantz, O., Hernandez-Alejandro, R., Haeryfar, Fever Incident Management Team. (2017) Genome analysis following a
S. M. M. (2017) MAIT cells launch a rapid, robust and distinct national increase in scarlet fever in England 2014. BMC Genomics 18,
hyperinflammatory response to bacterial superantigens and quickly 224.
acquire an anergic phenotype that impedes their cognate 173. Davies, M. R., Holden, M. T., Coupland, P., Chen, J. H., Venturini, C.,
antimicrobial function: defining a novel mechanism of superantigen- Barnett, T. C., Zakour, N. L., Tse, H., Dougan, G., Yuen, K. Y., Walker,
induced immunopathology and immunosuppression. PLoS Biol. 15, M. J. (2015) Emergence of scarlet fever Streptococcus pyogenes emm12
e2001930. clones in Hong Kong is associated with toxin acquisition and multidrug
153. Crowe, C. C., Sanders, W. E., Jr., Longley, S. (1973) Bacterial resistance. Nat. Genet. 47, 84–87.
interference. II. Role of the normal throat flora in prevention of 174. Tse, H., Bao, J. Y., Davies, M. R., Maamary, P., Tsoi, H. W., Tong, A. H.,
colonization by group A Streptococcus. J. Infect. Dis. 128, 527–532. Ho, T. C., Lin, C. H., Gillen, C. M., Barnett, T. C., Chen, J. H., Lee, M.,
154. Sanders, E. (1969) Bacterial interference. I. Its occurrence among Yam, W. C., Wong, C. K., Ong, C. L., Chan, Y. W., Wu, C. W., Ng, T.,
the respiratory tract flora and characterization of inhibition of Lim, W. W., Tsang, T. H., Tse, C. W., Dougan, G., Walker, M. J., Lok, S.,
group A streptococci by viridans streptococci. J. Infect. Dis. 120, Yuen, K. Y. (2012) Molecular characterization of the 2011 Hong Kong
698–707. scarlet fever outbreak. J. Infect. Dis. 206, 341–351.
155. Belkaid, Y., Hand, T. W. (2014) Role of the microbiota in immunity and 175. Ben Zakour, N. L., Davies, M. R., You, Y., Chen, J. H., Forde, B. M.,
inflammation. Cell 157, 121–141. Stanton-Cook, M., Yang, R., Cui, Y., Barnett, T. C., Venturini, C., Ong,
156. Niers, L. E., Timmerman, H. M., Rijkers, G. T., van Bleek, G. M., van C. L., Tse, H., Dougan, G., Zhang, J., Yuen, K. Y., Beatson, S. A., Walker,
Uden, N. O., Knol, E. F., Kapsenberg, M. L., Kimpen, J. L., Hoekstra, M. J. (2015) Transfer of scarlet fever-associated elements into the group
M. O. (2005) Identification of strong interleukin-10 inducing lactic acid A Streptococcus M1T1 clone. Sci. Rep. 5, 15877.
bacteria which down-regulate T helper type 2 cytokines. Clin. Exp. Allergy 176. Johnson, L. P., L’Italien, J. J., Schlievert, P. M. (1986) Streptococcal
35, 1481–1489. pyrogenic exotoxin type A (scarlet fever toxin) is related to
157. Christensen, H. R., Frøkiaer, H., Pestka, J. J. (2002) Lactobacilli Staphylococcus aureus enterotoxin B. Mol. Gen. Genet. 203,
differentially modulate expression of cytokines and maturation surface 354–356.
markers in murine dendritic cells. J. Immunol. 168, 171–178. 177. Viciani, E., Montagnani, F., Tavarini, S., Tordini, G., Maccari, S.,
158. Saroj, S. D., Maudsdotter, L., Tavares, R., Jonsson, A. B. (2016) Morandi, M., Faenzi, E., Biagini, C., Romano, A., Salerni, L., Finco, O.,
Lactobacilli interfere with Streptococcus pyogenes hemolytic activity and Lazzi, S., Ruggiero, P., De Luca, A., Barocchi, M. A., Manetti, A. G.
adherence to host epithelial cells. Front. Microbiol. 7, 1176. (2016) Paediatric obstructive sleep apnoea syndrome (OSAS) is
159. Capaldo, C. T., Nusrat, A. (2009) Cytokine regulation of tight junctions. associated with tonsil colonisation by Streptococcus pyogenes. Sci. Rep. 6,
Biochim. Biophys. Acta 1788, 864–871. 20609.
160. Burton, J. P., Wescombe, P. A., Moore, C. J., Chilcott, C. N., Tagg, J. R. 178. Viciani, E., Montagnani, F., Tordini, G., Romano, A., Salerni, L., De
(2006) Safety assessment of the oral cavity probiotic Streptococcus Luca, A., Ruggiero, P., Manetti, A. G. (2017) Prevalence of M75
salivarius K12. Appl. Environ. Microbiol. 72, 3050–3053. Streptococcus pyogenes strains harboring slaA gene in patients affected by
161. Di Pierro, F., Donato, G., Fomia, F., Adami, T., Careddu, D., Cassandro, pediatric obstructive sleep apnea syndrome in central Italy. Front.
C., Albera, R. (2012) Preliminary pediatric clinical evaluation of the oral Microbiol. 8, 294.

18 Journal of Leukocyte Biology Volume 103, February 2018 www.jleukbio.org


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
Soderholm et al. Group A streptococcal pharyngitis and host immune responses

179. Nagiec, M. J., Lei, B., Parker, S. K., Vasil, M. L., Matsumoto, M., Ireland, 186. Thorleifsdottir, R. H., Sigurdardottir, S. L., Sigurgeirsson, B., Olafsson,
R. M., Beres, S. B., Hoe, N. P., Musser, J. M. (2004) Analysis of a novel J. H., Sigurdsson, M. I., Petersen, H., Arnadottir, S., Gudjonsson, J. E.,
prophage-encoded group A Streptococcus extracellular phospholipase Johnston, A., Valdimarsson, H. (2012) Improvement of psoriasis after
A(2). J. Biol. Chem. 279, 45909–45918. tonsillectomy is associated with a decrease in the frequency of
180. Nickoloff, B. J. (1991) The cytokine network in psoriasis. Arch. Dermatol. circulating T cells that recognize streptococcal determinants and
127, 871–884. homologous skin determinants. J. Immunol. 188, 5160–5165.
181. Prinz, J. C. (2001) Psoriasis vulgaris–a sterile antibacterial skin reaction 187. Weisenseel, P., Prinz, J. C. (2005) Incidental detection of S. pyogenes-
mediated by cross-reactive T cells? An immunological view of the DNA in psoriatic skin by PCR. Arch. Dermatol. Res. 296, 573–576.
pathophysiology of psoriasis. Clin. Exp. Dermatol. 26, 326–332. 188. Nakagawa, I., Nakata, M., Kawabata, S., Hamada, S. (2004)
182. Roberson, E. D., Bowcock, A. M. (2010) Psoriasis genetics: breaking the Transcriptome analysis and gene expression profiles of early apoptosis-
barrier. Trends Genet. 26, 415–423. related genes in Streptococcus pyogenes-infected epithelial cells. Cell.
183. Sigmundsdóttir, H., Gudjónsson, J. E., Jónsdóttir, I., Lúdvı́ksson, B. R., Microbiol. 6, 939–952.
Valdimarsson, H. (2001) The frequency of CLA+ CD8+ T cells in the 189. Dileepan, T., Smith, E. D., Knowland, D., Hsu, M., Platt, M., Bittner-
blood of psoriasis patients correlates closely with the severity of their Eddy, P., Cohen, B., Southern, P., Latimer, E., Harley, E., Agalliu, D.,
disease. Clin. Exp. Immunol. 126, 365–369. Cleary, P. P. (2016) Group A Streptococcus intranasal infection promotes
184. Ruiz-Romeu, E., Ferran, M., Sagrista, M., Gomez, J., Gimenez-Arnau, A., CNS infiltration by streptococcal-specific Th17 cells. J. Clin. Invest. 126,
Herszenyi, K., Hollo, P., Celada, A., Pujol, R., Santamaria-Babi, L. F. 303–317.
(2016) Streptococcus pyogenes-induced cutaneous lymphocyte antigen- 190. Kerakawauchi, H., Kurono, Y., Mogi, G. (1997) Immune responses
positive T cell-dependent epidermal cell activation triggers TH17 against Streptococcus pyogenes in human palatine tonsils. Laryngoscope 107,
responses in patients with guttate psoriasis. J. Allergy Clin. Immunol. 138, 634–639.
491–499.e6.
185. Sigurdardottir, S. L., Thorleifsdottir, R. H., Valdimarsson, H., Johnston,
A. (2013) The role of the palatine tonsils in the pathogenesis and KEY WORDS:
treatment of psoriasis. Br. J. Dermatol. 168, 237–242. host immune response Streptococcus Streptococcus pyogenes
• •

www.jleukbio.org Volume 103, February 2018 Journal of Leukocyte Biology 19


Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017
Group A streptococcal pharyngitis: immune responses involved
in bacterial clearance and GAS-associated immunopathologies
Amelia T. Soderholm, Timothy C. Barnett, Matthew J. Sweet, et al.

J Leukoc Biol published online September 26, 2017


Access the most recent version at doi:10.1189/jlb.4MR0617-227RR

Subscriptions Information about subscribing to Journal of Leukocyte Biology is online at


http://www.jleukbio.org/site/misc/Librarians_Resource.xhtml

Permissions Submit copyright permission requests at:


http://www.jleukbio.org/site/misc/Librarians_Resource.xhtml

Email Alerts Receive free email alerts when new an article cites this article - sign up at
http://www.jleukbio.org/cgi/alerts

© Society for Leukocyte Biology

Downloaded from www.jleukbio.org to IP 128.122.230.132. Journal of Leukocyte Biology Vol., No. , pp:, September, 2017

You might also like