You are on page 1of 13

REVIEwS

Streptococcus pneumoniae:
transmission, colonization
and invasion
Jeffrey N. Weiser   1*, Daniela M. Ferreira2 and James C. Paton3
Abstract | Streptococcus pneumoniae has a complex relationship with its obligate human host.
On the one hand, the pneumococci are highly adapted commensals, and their main reservoir
on the mucosal surface of the upper airways of carriers enables transmission. On the other
hand, they can cause severe disease when bacterial and host factors allow them to invade
essentially sterile sites, such as the middle ear spaces, lungs, bloodstream and meninges.
Transmission, colonization and invasion depend on the remarkable ability of S. pneumoniae to
evade or take advantage of the host inflammatory and immune responses. The different stages
of pneumococcal carriage and disease have been investigated in detail in animal models and,
more recently , in experimental human infection. Furthermore, widespread vaccination and the
resulting immune pressure have shed light on pneumococcal population dynamics and
pathogenesis. Here, we review the mechanistic insights provided by these studies on the
multiple and varied interactions of the pneumococcus and its host.

Upper respiratory tract


Streptococcus pneumoniae (also known as pneumococ- of disease is due to the combination of high carriage
(URT). Includes the nasal cavity, cus) is a Gram-​positive, extracellular, opportunistic path- rates, its genetic adaptability and its ability to shift from
paranasal sinuses, mouth, ogen that colonizes the mucosal surfaces of the human a commensal to a pathogenic interaction with its host.
pharynx and larynx and forms upper respiratory tract (URT). Up to 27–65% of children In this Review, we discuss the bacterial, environmental
the major passages above the
and <10% of adults are carriers of S. ­pneumoniae and and host factors that contribute to the different stages of
trachea.
carriage involves a commensal relationship between the pneumococcal disease.
Community-​acquired bacterium and the host1,2. Local spread, aspiration or
pneumonia seeding to the bloodstream results in invasive inflam- Transmission of S. pneumoniae
Infection of the lung acquired
matory diseases3 (Fig. 1). S. pneumoniae is a leading Until recently, all that was known about pneumococcal
outside of hospitals or nursing
facilities.
bacterial cause of a wide range of infections, including contagion was that spread requires close contact with
otitis media, community-​acquired pneumonia, sepsis and a carrier and/or carriers (especially young children),
Natural competence meningitis. As all of these diseases are ‘dead ends’ in the is more frequent during drier, colder months when
The endogenous ability of a life cycle of the organism, the bacterial factors that cause airway secretions are more copious and is more likely
bacterium to alter its genes by
invasive diseases must also be adaptive for colonization to occur in conjunction with viral infections of the
taking up extracellular DNA
from its environment through and/or transmission. URT5–7. This general ignorance about transmission was
transformation. In 2017, the WHO included S. pneumoniae as one a consequence of a lack of tractable animal models and
of 12 priority pathogens. The continued high burden an inability to study human-​to-human transmission in
of disease and rising rates of resistance to penicillin sufficient detail. In 2010, airborne transmission among
and other antibiotics have renewed interest in preven- closely housed ferrets co-​infected with influenza A
tion. The widespread use of pneumococcal conjugate virus (IAV) was described8. Another group modelled
1
New York University, New vaccines (PCVs) has reduced invasive disease of sero- murine transmission from index pups colonized at
York, NY, USA.
types with the capsular polysaccharide (CPS) types 4 days of age to littermate contact pups in the setting
2
Liverpool School of Tropical
that are included in the vaccine4 (Box 1). The remark- of IAV co-​infection9. Similar to human transmission,
Medicine, Liverpool, UK.
able capacity of S. pneumoniae to remodel its genome viral infection, close contact and younger age increased
3
University of Adelaide,
Adelaide, Australia.
through the uptake and incorporation of exogenous transmission. This infant mouse model has now ena-
DNA (natural competence) from other pneumococci or bled the study of the major steps during host-​to-host
*e-​mail: jeffrey.weiser@
nyumc.org closely related oral streptococci has facilitated the spread spread, including exit from a colonized host (shed-
https://doi.org/10.1038/ of antibiotic resistance and evasion of vaccine-​induced ding), survival in the environment and acquisition by
s41579-018-0001-8 immunity. The prominence of S. pneumoniae as a cause a new host.

Nature Reviews | Microbiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

bottleneck during transmission would explain the need


S. pneumoniae for large numbers of shed pneumococci for at least one
Mucus to be successful in reaching a new host. Accordingly,
Colonization increasing total shedding per cage by increasing the pro-
Nasal epithelium portion of colonized index pups per cage to 50% made
transmission to ~30% of contacts possible without the
need for IAV co-​infection14.
During early childhood, rhinorrhoea is pronounced,
and clinical surveys demonstrate a relationship between
Transmission
secretion volume and S. pneumoniae density15. In the
infant mouse model, dampening inflammation by intra-
nasal dexamethasone treatment or the use of Tlr2−/− index
Shedding
mice reduces shedding and transmission16. The single
pneumococcal toxin, pneumolysin (Ply), has strong
Invasion pro-​inflammatory effects, and Ply-​induced inflam-
mation hastens clearance of bacteria from the URT17.
Both a ply-​knockout mutant and a point mutant, in
Aspiration Bacteraemia Local spread which the toxin is unable to oligomerize to form pores
after membrane insertion, reduced URT inflammation,
shedding and the ability to transmit to littermates16.
Additionally, intranasal administration of the purified
toxin, but not the inactive toxoid (PdB), could com-
plement the inflammation, shedding and transmission
defect of the ply mutant. This is the first example of a
pneumococcal factor that is specifically required for
transmission. These findings with Ply also provide a
Pneumonia Otitis media link between pneumococcal virulence and transmis-
Meningitis sion18, suggesting that factors such as Ply that contri­
bute to the disease state by enhancing inflammation
Fig. 1 | The life cycle of Streptococcus pneumoniae and also promote the transmission of S. pneumoniae.
the pathogenesis of pneumococcal disease. As epidemiological studies show that the prevalence
Streptococcus pneumoniae colonizes the mucosa of the
of different serotypes is highly variable, the role of cap-
upper respiratory tract (URT). This carriage is the
prerequisite for both transmission to other individuals and
sule type and amount on shedding and transmission was
invasive disease in the carrier. Carriers can shed S. tested using isogenic serotype-​switch and cps-​promoter
pneumoniae in nasal secretions and thereby transmit the switch mutants19. Some serotype-​switch mutants colo-
bacterium. Dissemination beyond its niche along the nasal nized at wild-​type levels but were shed and transmitted
epithelium, either by aspiration, bacteraemia or local poorly in infant mice. Mutants with lower expression of
spread, can lead to invasive diseases, such as pneumonia, CPS and thinner capsules were also shed and transmit-
meningitis and otitis media. ted poorly. The capsule layer shields underlying surface
adhesins, and mutants with reduced shedding and trans-
Exit from the colonized host. IAV-​induced inflammation mission showed increased binding to URT mucins in
stimulates both the expression of mucin glycoproteins an in vitro assay. Encapsulation, therefore, may facilitate
and the flow of mucus10,11. There are more pneumo- shedding by allowing escape from the mucus that lines
cocci in nasal secretions of pups with IAV co-​infection the airway surface, with a thicker capsule or capsule of
(Fig. 2), and only young mice shed S. pneumoniae at levels certain serotypes being more effective.
permissive for transmission12. Moreover, levels of shed-
ding correlate with the extent of URT inflammation in Survival in the environment. The extent of airborne trans-
response to IAV infection. Toll-​like receptor 2 (TLR2)- mission (as demonstrated by the ferret studies) versus
deficiency, which is associated with an increased viral contact-​dependent transmission (as shown by the infant
load and, subsequently, greater inflammation, results in mouse model) is unclear. A number of recent studies have
higher rates of transmission, and this effect is specific examined factors that affect survival of S. pneumoniae
to the index mice12. Furthermore, the effect of IAV is outside the host. Transmission through secretions
recapitulated by intranasal treatment of the index mice of carriers could involve direct person-​t o-person
with the TLR3 ligand polyIC13. contact or spread involving bacteria on contaminated
PolyIC
The size of population bottlenecks in the infant surfaces. As evidence of the latter, in the mouse model,
Polyinosinic:polycytidylic acid
is an agonist of Toll-​like mouse model during transmission was estimated by the co-​housed dam is not colonized but has large num-
receptor 3 and mimics double-​ using marked isogenic bacterial strains13. In this study, bers of S. pneumoniae on her teats, and in-​cage switch
stranded RNA found in some all constructs colonized, shed and could be acquired experiments can serve as a source of contagion9,16.
viruses. in similar numbers by all pups. By contrast, after the S. pneumoniae can also be easily cultured from common
Dexamethasone
index pup is simultaneously colonized with the marked objects, such as soft toys recently handled by colonized
An anti-​inflammatory mutants, in the majority of transmission events, only children20. Under ambient, nutrient-​sufficient condi-
corticosteroid. one of the mutants was successful. This tight population tions, such as in ex vivo human saliva, pneumococci

www.nature.com/nrmicro
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Box 1 | Streptococcus pneumoniae vaccination following antigen exposure. Furthermore, the effective-
ness of S. pneumoniae agglutination of airway secretions
Streptococcus pneumoniae has a high genetic diversity, and certain lineages are after PCV vaccination correlates with protection during
particularly successful. an important source of strain-​to-strain variation is the structure experimental human colonization28. An additional effect
of the capsular polysaccharide (CPs), which is the major virulence determinant and
of immunity demonstrated in the infant mouse model is
immunodominant surface structure of S. pneumoniae. Currently, 98 immunologically
a decrease in shedding by index pups24. Moreover, immu-
and structurally distinct CPs types are recognized, but only a relatively small subset of
these types is commonly found to cause carriage and disease. therefore, CPs-​based nity in either the index or contact pups alone is sufficient
vaccines target only a limited number of serotypes. when covalently conjugated to an to reduce rates of transmission, indicating that decreased
immunogenic protein carrier, CPS is recognized as a T cell-​dependent antigen, which shedding and protection from acquisition both contrib-
stimulates a more effective humoral immune response (including immunoglobulin class ute. These experiments were carried out with serotype-​
switching, affinity maturation and memory) than polysaccharide-​alone antigens, specific antibody. It is unclear whether immunity to other
particularly in young children. since its introduction in 2000, the pneumococcal S. pneumoniae surface targets can block transmission.
conjugate vaccine (PCv) has been highly effective in preventing invasive pneumococcal In this regard, immunization with Ply shows no effect
diseases. an unexpected benefit of the high levels of serotype-​specific immunoglobulin on shedding and transmission, even though the toxin
G generated by PCv has been reduced rates of carriage in and transmission from
is required for spread between pups16. This result is not
immunized children, which also protects unimmunized populations (herd immunity)145.
unexpected as Ply is not actively secreted, is not present
However, the protection elicited by PCv is incomplete, as current formulations contain
only 10 to 13 of the 97 known CPs types146. a further issue is the rising prevalence of on the cell surface and might be released only when pneu-
non-​vaccine serotypes in carriage and disease (serotype replacement) as a mococci are lysed within the phagosome and therefore
consequence of the immune pressure from widespread use of PCv147,148. Current efforts are not exposed to antibody29. When pneumococci are
to improve prevention through vaccination are directed at increasing the number of killed by lysozyme within the phagosome, the released
serotypes covered by PCv or adding conserved pneumococcal proteins that induce Ply forms pores, which enable bacterial products to access
serotype-​independent immunity. the host cell cytosol and trigger the production of pro-​
inflammatory chemokines and cytokines30–32. In this
can survive for days21. Under nutrient-​poor conditions, manner, S. pneumoniae responds to an influx of profes-
such as in airway surface fluid, bacterial expression of Ply sional phagocytes when it finds itself in a host that is no
increases ex vivo survival16. This effect can be explained longer hospitable. Triggering inflammation and mucus
by toxin-​dependent inflammation and, consequently, secretions drives its transit to a new, more hospitable host.
increased nutrients levels in secretions. Capsule expres-
sion from the cps locus increases survival in nutrient-​ Colonization by S. pneumoniae
poor environmental conditions, perhaps by providing a Nasopharyngeal carriage is the source of S. pneumo-
reserve of glycans22. Furthermore, pneumococci survive niae spread between hosts and the first step towards
desiccation for many days, and biofilm bacteria retain invasive disease. Several bacterial factors are required
viability in vitro better than planktonic bacteria20,23. for S. pneumoniae to colonize and persist on the
mucosal surface at a density and duration that is suf-
Acquisition by the new host. Given the importance of ficient for transmission to occur (Fig. 3). For example,
PCV in reducing transmission from immunized children, S. pneumoniae expresses two enzymes, peptidoglycan-​
the infant mouse model has been used to explore the role N-acetylglucosamine deacetylase (PgdA) and attenuator
of immunity in spread24. Pre-​existing S. pneumoniae col- of drug resistance (Adr), that modify its peptidoglycan
onization of contact pups inhibits the acquisition of a and render it resistant to the lytic effects of lysozyme,
new strain13. This bacterial interference could affect the which is abundant on the mucosal surface of the URT33.
frequency of co-​colonizing strains. Passive immunization The main features that facilitate colonization are adher-
of contact pups with anti-​capsular polysaccharide immu- ence to host cells and tissues, subversion of mucosal
noglobulin G (IgG) is also sufficient to block acquisition, innate and adaptive immunity, and evasion of clearance
although this effect requires high levels of antibody and by mucociliary flow.
can be overcome by a large inoculum25. The protective
activity of specific antibodies during acquisition is inde- Adherence to the nasopharynx. The first defence that
pendent of Fc fragment-​mediated effects but requires their S. pneumoniae encounters in the nasopharynx is mucus
agglutinating function, which could facilitate mechanical entrapment. The glycocalyx overlying the URT epithe-
Fc fragment clearance by the mucociliary flow. However, S. pneumo- lium is composed of gel-​like mucin glycoproteins and
The tail region of an antibody niae evades clearance that is mediated by IgA1, the most contains antimicrobial peptides and immunoglobu-
that interacts with cell surface
receptors and some proteins of
abundant immunoglobulin on mucosal surfaces of the lins34. S. pneumoniae, like other residents of the URT,
the complement system. human URT26. The pneumococcal zinc metalloprotease is found predominantly in and on this mucus layer35.
ZmpA (also known as IgA1 protease), which cleaves the Although the mucus layer keeps the bacteria away
Agglutinating function hinge region of human IgA1, eliminates the agglutinat- from the underlying cell surface, adherence to mucin
The clumping of antigens
ing activity of this immunoglobulin25. Thus, PCV likely is glycans helps the bacteria to remain in the nasophar-
through multivalent binding by
antibodies. effective because it induces IgG, which is not sensitive to ynx and provides a favourable niche and nutrients. On
the protease, at levels high enough to reach the mucosal the other hand, CPSs, which are almost all negatively
Mucociliary flow surface and block pneumococcal acquisition. In a model charged, repel the sialic acid-​rich mucopolysaccha-
A non-​immunological defence of experimental human colonization with S. pneumo- rides in mucus36. By avoiding entrapment in the nasal
mechanism that involves ciliary
action and the flow of mucus; it
niae, levels of CPS-​specific memory B cells correlate with mucus, S. pneumoniae might access and attach to the
clears the respiratory tract of protection from acquisition27. Such memory B cells can surface of epithelial cells. Much of our understanding
pathogens and particles. quickly differentiate into antibody-​secreting plasma cells of S. pneumoniae–host cell interactions comes primarily

Nature Reviews | Microbiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Carriage Pneumolysin Capsule type Anticapsule IgG Anticapsule IgA1 Viral co-infection
and amount

Shedding Shedding Shedding No effect Shedding

Inflammation Inhibition of Agglutination Bacterial protease Increased bacterial load


promotes secretions mucin entrapment blocks release relieves agglutination and mucus production

Neutrophil IgA1
Mucus
S. pneumoniae
IgG

Virus

Nasal epithelium

Fig. 2 | bacterial and host factors affecting pneumococcal shedding from carriers. Streptococcus pneumoniae is found
predominantly in the mucus layer overlying the epithelial surface of the upper respiratory tract. Inflammation (indicated by
the presence of neutrophils), which is induced by the pore-​forming toxin pneumolysin or by co-​infection with influenza virus
or other respiratory viruses, stimulates secretions and increases shedding. By contrast, agglutinating antibodies such as
anti-​capsule immunoglobulin G (IgG) and IgA1 decrease shedding unless they are cleaved by the human IgA1-specific
pneumococcal protease. Capsule type and amount also influence mucus association and numbers of shed bacteria.

from models that use cultured human epithelial cells. Interactions with the nasopharyngeal flora. The
S. pneumoniae uses several surface components for bind- success of S. pneumoniae as a colonizer requires inter-
ing, but their relative importance in natural carriage has actions with the nasopharyngeal microbiota, which
not been established. Examples of these adhesins are likely are extensive and complex. These interactions
surface-​located pneumococcal adherence and virulence can either be cooperative or competitive52. For example,
protein A (PavA), PavB and enolase (Eno), all of which detection of Gram-​negative peptidoglycan through the
bind to the extracellular matrix proteins fibronectin and sensor nucleotide-​binding oligomerization domain-​
plasminogen37–39. Phosphorylcholine (ChoP) moieties on containing protein 1 (NOD1) by neutrophils triggers
cell wall teichoic acid bind to the platelet-​activating factor killing of S. pneumoniae53. During experimental human
receptor (PAFR), and choline-​binding protein A (CbpA; colonization, increased diversity of the microbiota is
also known as PspC) binds the secretory component of associated with increased acquisition of S. pneumoniae
the polymeric immunoglobulin receptor40,41. CbpA also following intranasal challenge54. S. pneumoniae coloni-
binds the host proteins factor H and vitronectin. Other zation was also found to promote microbial heteroge­
major classes of host cell receptors include carcinoembry- neity in these studies. Similarly, during the first 2 years of
onic antigen-​related cell adhesion molecule 1 (CEACAM) life, S. pneumoniae colonization was associated with less
and intercellular adhesion molecule 1 (ICAM1)42. S. stable microbiome profiles55. Co-​colonizing pneumo-
pneumoniae increases expression of many of its epi- cocci compete with one another through a diverse array
thelial surface receptors and thereby increases its of bacteriocins (pneumocins) and related peptides with
adherence in response to inflammatory stimuli43. The antimicrobial activity56–58. Lysis of susceptible strains
surface-​exposed lipoproteins foldase protein PrsA (also not only allows for predation but also provides a source
known as PpmA)44 and peptidyl-​prolyl isomerase SlrA45 of DNA for the adaptation of the predator.
also contribute to adherence to epithelial cells. CbpL facili­ In general, inflammatory conditions in the URT
tates migration of S. pneumoniae from the nasopharynx favour the presence of S. pneumoniae. A common and
to the lungs and blood46. S. pneumoniae encodes at least important example is infection with URT viruses. Nasal
ten extracellular glycosidases, some of which have been inflammation in response to infection with respira-
shown to enhance adherence by modifying host glyco- tory viruses, such as IAV, modulates the expression of
conjugates to reveal glycan receptors47. In addition, two of pro-​inflammatory chemokines, upregulates epithelial
these surface glycosidases, Neuraminidase A (NanA) and receptors used for S. pneumoniae adherence, com-
the β-​galactosidase BgaA, have lectin domains and seem promises the integrity of the epithelium and provides
Lectin domains
to function as adhesins independently of their enzymatic a more nutrient-​rich inflammatory milieu. Together,
The carbohydrate-​binding activities48,49. N-​acetylglucosamine-β-(1,3)-galactose these effects of viral co-​infection increase the suscep-
domains on proteins. inhibits pneumococcal adherence to epithelial cells, and tibility to acquisition and the density of colonizing
S. pneumoniae is one of many pathogens that bind to S. pneumoniae59–61. Recent data from murine models
Bacteriocins N-​acetylglucosamine-β-(1,4)-galactose50,51. These adhe- and clinical studies have shown that the live attenuated
The proteinaceous or peptidic
toxins produced by bacteria to
sive interactions with the epithelial surface may be influenza vaccine also increases the number of coloniz-
inhibit the growth of similar or needed for colonization but also comprise the initial step ing S. pneumoniae62–64. A higher pneumococcal density
closely related bacteria. in the invasion process (see below). in the nasopharynx is likely to facilitate transmission

www.nature.com/nrmicro
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Interaction with Apolactoferrin influence on carriage duration66. This pan-​genome-wide


complement Impaired neutrophil association study also identified prophage sequences as
system recruitment
Metal-binding Lactoferrin having the greatest negative impact on carriage duration,
Ply PAF (ChoP proteins independent of serotype.
moieties)
One important characteristic that enables S. pneu-
PepO
Eno moniae to successfully thrive in this competitive
CbpA CbpE PsaA
PspA PiaA PiuA niche is its ability to evade and sometimes hijack
Pht Ply host responses during colonization. In mouse mod-
Capsule els, acquisition of the organism leads to a mild acute
Cell wall inflammatory response in the URT that is ineffective
Cell membrane Interaction with at completely clearing the organism67. By contrast, pre-​
epithelial receptors existing inflammation is the most closely associated
StrH BgaA ZmpA susceptibility factor in the human challenge model68.
Pilus NanA IgA1
Cilia Many of the factors contributing to the eventual clear-
ChoP
Eno PavA CbpA ance of S. pneumoniae have been delineated. Studies in
First contact Mucus PIGR Factor H Ply mice suggest that clearance requires TLR2-dependent
with the degradation responses that result in the recruitment of additional
RrgA epithelium Mucus macrophages from the monocyte pool into the nasal
PAFR
Glycosaminoglycans Reduction of lumen. Positive feedback and additional recruitment
cilia beating of macrophages are required for the gradual elimina-
tion of colonization69. The cellular immune responses
to S. pneumoniae are greatly accelerated by cytosolic
sensing of the pathogen, which requires the pore-​
Respiratory epithelium forming function of Ply29,30. Accordingly, ply-​deficient
mutants, or mutants unable to form pores, show pro-
Fig. 3 | Molecular mechanisms of pneumococcal colonization of host surfaces. Key longed colonization and diminished production of
functions that enable Streptococcus pneumoniae colonization are: establishing the first key inflammatory mediators needed for clearance,
contact with the epithelium and epithelial receptors, interaction with the complement including interleukin-1β (IL-1β), CXC-​m otif and
system, mucus degradation, metal binding, impairment of neutrophil activity and the CC-​m otif chemokines, and type 1 interferons 29,30,32.
pro-​inflammatory effects of the toxin pneumolysin (Ply). The pneumococcal enzymes These macrophage-​d ependent responses are dys-
Neuraminidase A (NanA), β-​galactosidase (BgaA) and β-​N-acetylglucosaminidase (StrH) functional in both infant and aged mice, which might
degrade mucus and thereby inhibit mucociliary clearance. Furthermore, the LytA (autolysin)- explain the higher incidence of infection among the
facilitated release of Ply damages the epithelium and reduces ciliary beating. Negatively very young and old 70,71. The importance of cellular
charged capsular polysaccharide (CPS) inhibits bacterial mucus entrapment. CPS and clearance mechanisms is likely to be a consequence
several pneumococcal proteins, including pneumococcal surface protein A (PspA),
of the inability of specific antibodies that are induced
choline-​binding protein A (CbpA), enolase (Eno) and pneumococcal histidine triad
protein (Pht), directly and indirectly block complement deposition. PspA also binds to during carriage to clear the organism once it is
lactoferrin to acquire iron and blocks the antimicrobial effect of apolactoferrin. established on the mucosal surface72,73.
Endopeptidase (PepO), which is released from the pneumococcal surface, binds to C1q
and thereby depletes complement components. Pneumococcal CbpE impairs neutrophil Immunizing effect of colonization. Colonization
recruitment by degrading platelet-​activating factor (PAF), a host-​derived inflammatory increases anti-​capsular (serotype-​specific) and anti-​
phospholipid. CbpA interacts with factor H interactions to facilitate adherence and protein (non-​s erotype-specific) antibody levels74–77.
subsequent internalization of S. pneumoniae via cell glycosaminoglycans. CbpA also Experimental data from murine models show that col-
binds to polymeric immunoglobulin receptor (PIGR) to promote adherence. The zinc onization is an immunizing event and protects against
metalloprotease ZmpA (also known as immunoglobulin A1 protease) subverts mucosal subsequent colonization and disease78,79. Experimental
humoral immunity by cleaving IgA1. Phosphorylcholine (ChoP) on teichoic acid mimics
human carriage studies have confirmed that colonization
host PAF and allows binding to its receptor. Piliated strains express an ancillary pilus
subunit tip adhesin called RrgA . Other S. pneumoniae adhesins include enolase (Eno) and increases nasal, lung and serum antibody levels74,80,81.
adherence and virulence protein A (PavA). PAFR , platelet-​activating factor receptor. Moreover, these studies corroborated observations in
murine models, demonstrating the protective effect of
colonization against reacquisition of the same strain
and microaspiration to the lungs, thereby increasing the up to 1 year following the first colonization episode75.
likelihood of progression to disease65. Serotype-​specific or strain-​specific immunity seems to
be required for this protection, as challenge of volunteers
Bacterial and host factors involved in clearance. following a known natural carriage episode with a strain
Individual carriage episodes typically last for weeks to of a different serotype did not result in increased protec-
months. Through the use of a model for calculating tion27. These infection studies also showed that coloni-
the duration of carriage episodes from a longitudinal zation increases levels of S. pneumoniae-​specific CD4+
carriage study and combining these results with whole-​ T memory cells in the blood and lungs in humans80. In
genome sequence data, it was recently estimated that mice, anti-​pneumococcal CD4+ T cells are sufficient, and
Type 1 interferons S. pneumoniae genomic variation accounts for 63% of the T helper 17 (TH17) cell response is necessary for effi-
A group of signalling proteins
expressed and released by
the variation in carriage duration, whereas measured cient clearance82,83. The importance of TH17 immunity in
host cells to regulate immune host traits (such as age and previous carriage) accounted natural colonization has yet to be confirmed, although
responses to pathogens. for less than 5%. Serotype was found to have a major a low ratio of TH17 to T regulatory (Treg) cells correlates

Nature Reviews | Microbiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

with colonization in children and increases with age as the metal binding site in manganese ABC transporter
colonization frequency decreases84. substrate-​binding lipoprotein (PsaA)95, but unlike Mn,
which is passed from PsaA to the PsaBC transporter for
Invasive pneumococcal disease uptake, Zn binds irreversibly to PsaA and thereby blocks
From an evolutionary perspective, stable nasopharyn- the transport pathway, starving the bacterium of Mn96.
geal colonization ought to be the principal modus Thus, host Zn release contributes to nutritional immu-
operandi of S. pneumoniae, as this enables ready trans- nity and may explain why dietary Zn deficiency increases
mission to new hosts. As noted above, induction of pro-​ rates of pneumococcal disease97,98. Pneumococcal surface
inflammatory chemokines and cytokines, upregulation protein A (PspA) also interacts with host lactoferrin, an
of target receptors and damage to the respiratory epithe- Fe-​sequestering glycoprotein, and this protects the bac-
lium caused by viral infection of the URT increases bac- terium from killing by apolactoferrin (the Fe-​free form
terial loads in the nasopharynx. This facilitates bacterial of lactoferrin)99. Recent work has shown that the varia-
transmission but also increases the likelihood of pene- ble capacity of different S. pneumoniae strains to bind
tration of host tissues and progression to localized or lactoferrin depends on PspA and differences in CPS100.
invasive disease. Progression to invasive disease is more Optimal utilization of carbon sources available in
likely in young children, elderly people and patients distinct host niches is also critical for pathogenesis.
with specific lifestyle traits and comorbidities. There S. pneumoniae is totally dependent on carbohydrates
are also marked differences in the capacity of specific as a carbon source, and its genome encodes roughly
S. pneumoniae strains to cause invasive disease, which 30 carbohydrate-​specific phosphotransferase systems
is unsurprising given the vast genetic and phenotypic (PTSs) and ABC transporters capable of importing
heterogeneity of this bacterium. S. pneumoniae factors a wide range of sugars101. Many of these have previ-
and pathways that contribute to tissue adherence and ously been shown to contribute to growth and survival
invasion are outlined in Fig. 4. in vivo102. Although glucose is available in the blood,
free sugars may be in low abundance at sites such as the
Niche adaptation. Translocation from the nasophar- mucosa of the upper and lower respiratory tracts. In
ynx to deeper tissues exposes S. pneumoniae to dis- these niches, pneumococci scavenge sugars by sequential
tinct microenvironmental niches, requiring extensive cleavage of host cell surface N-​linked glycoconjugates,
adjustments to gene expression patterns. The impor- which is mediated by surface-​associated exoglycosidases
tance of such adaptations for pathogenesis was ini- such as NanA, BgaA and the β-​N-acetylglucosaminidase
tially suggested by genome-​wide screens, for example, StrH. The released sugars (sialic acid, galactose and N-​
signature-​tagged mutagenesis , which showed that in acetylglucosamine) may then be taken up by the relevant
addition to known virulence genes, a large number of ABC and PTS transporters and metabolized. At the same
metabolic and transporter genes were required either time, mannose residues are unmasked on the core gly-
for colonization or for local or invasive infections but can structure, which may function as surface receptors
not necessarily for growth of S. pneumoniae in vitro85–87. for pneumococcal adherence103. S. pneumoniae also has
Subsequent studies using genomic microarray analysis a surface-​associated endoglycosidase, EndoD104, which
identified substantial differences in expression patterns of releases the residual mannose3-N-​acetylglucosamine2
these non-​traditional virulence genes between pneumo- (Man3GlcNAc2) structure from host glycoconjugates.
cocci growing in distinct host niches (nasopharynx, lungs Terminal mannose can also be released from high man-
and blood) and compared with cells grown in vitro88,89. nose N-​glycans by an α-(1,2)-mannosidase, SpGH92,
Acquisition of metal ions, particularly transition met- and taken up by the mannose PTS. Meanwhile, resid-
als such as iron (Fe), manganese (Mn) and zinc (Zn), ual mannose5-N-​acetylglucosamine2 (Man5GlcNAc2)
is crucial for growth and survival of S. pneumoniae in is also released from these host structures by EndoD
multiple host niches, where availability of free ions may and taken up along with Man3GlcNAc2 by an ABC
be restricted. These metals are essential cofactors for transporter, with further deconstruction occurring in
many metabolic and other enzymes, and in the case of the pneumococcal cytoplasm104. The various released
Mn, they also mediate resistance to oxidative stress90. sugars can have substantial intracellular effects by regu-
Unsurprisingly, genes encoding the metal-​binding lating carbohydrate metabolism through the catabolite
components of ATP-​binding cassette (ABC) transport- repressor CcpA102. Sialic acid released by NanA has been
ers responsible for uptake of Fe (piuA, piaA and pitA), shown to act as a signal, increasing bacterial loads in the
Signature-​tagged
Mn (psaA) and Zn (adcA and adcAII) are preferentially nasopharynx of mice colonized with S. pneumoniae and
mutagenesis expressed in the host environment, and the respective facilitating invasion of nasal tissue and progression to
A genetic technique using DNA S. pneumoniae knockout mutants are heavily atten- pneumonia and meningitis105,106. Such signalling involves
signature tags (molecular uated in vivo in models of both carriage and invasive the two-​component response regulator transcriptional reg-
barcodes) to identify mutants
disease91–93. Indeed, the absolute requirement for psaA ulatory protein CiaR and requires sialic acid uptake by
in mixed populations.
in vivo makes it a valid target for novel antimicrobials94. the transporter SatABC, and this results in increased
Two-​component response Certain metals may also be deleterious in excess, and, pneumococcal resistance to antimicrobial reactive oxy-
regulator hence, intracellular concentrations must be strictly reg- gen species107. NanA can also trigger TGFβ signalling
The transcription factor ulated by coordination of uptake and efflux systems90. pathways, leading to endothelial cell invasion108.
component of a stimulus-​
response mechanism for
In addition, excess Zn released into the extracellular The role of biofilms in the ability of S. ­pneumoniae
bacteria to sense and respond compartment by leukocytes poses a particular problem to persist at various sites of infection is not well under-
to environmental changes. for invading pneumococci. Zn can compete with Mn for stood, and their contribution to invasive disease remains

www.nature.com/nrmicro
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

a Respiratory tract
Evasion of clearance Adherence Endocytosis Paracellular invasion
S. pneumoniae
CPS Eno
Hyl
NanA ZmpA or GAPDH CbpE
BgaA StrH Ply Plasmin
Mucus Pilus PsrP ChoP CbpA
RrgA PavA PAFR PIGR H2O2 Ply
Cilia

Endocytosis
Respiratory
epithelium

Exocytosis

Interstitium or

Capillary endothelium ChoP CbpA


PAFR PIGR

Blood CPS
Haematogenous spread
S. pneumoniae
PspA

Eno
Hyl
b Brain GAPDH CbpE
Plasmin
Blood Ply
CbpA CbpA ChoP RrgA RrgA H2O2
PIGR LR PAFR PIGR PECAM1
Blood–brain
barrier Tight junction

Cerebrospinal fluid

Fig. 4 | Stages in pneumococcal adherence and invasion. a | Several steps are required for invasion of the respiratory
tract. Streptococcus pneumoniae evades entrapment in mucus and mucociliary clearance by negatively charged capsular
polysaccharide (CPS) and proteolytic degradation of secretory immunoglobulin A1 (IgA1) by the zinc metalloprotease
ZmpA (also known as IgA1 protease). Neuraminidase A (NanA), β-​galactosidase (BgaA) and β-​N-acetylglucosaminidase
(StrH) deglycosylate mucus and unmask glycan targets for adhesins on the epithelium. Finally , pneumolysin (Ply) inhibits
ciliary beating. Adherence to the apical surface of epithelial cells is mediated by diverse surface structures, including
phosphorylcholine (ChoP), choline-​binding protein A (CbpA), the ancillary pilus subunit RrgA at the tip of pili, adherence
and virulence protein A (PavA) and large surface-​exposed glycoprotein (PsrP). S. pneumoniae binds through ChoP to
platelet-​activating factor receptor (PAFR) and through CbpA to polymeric immunoglobulin receptor (PIGR), and by
subverting the respective host receptor recycling pathways, it induces its own endocytosis, which is followed by release of
pneumococci at the basolateral surface. Alternatively , Ply and hydrogen peroxide (H2O2) directly damage the epithelium,
and hyaluronate lyase (Hyl) and plasmin, which is bound to the pneumococcal surface through enolase (Eno), glyceralde-
hyde-3-phosphate dehydrogenase (GAPDH) or CbpE, degrade the extracellular matrix. This breaks down the epithelial
barrier and provides a pathway for paracellular invasion. ChoP–PAFR and CbpA–PIGR interactions also enable
pneumococci to traverse the endothelium and enter the bloodstream. Upregulation of PAFR by inflammatory cytokines
amplifies ChoP–PAFR-​mediated invasion. CPS and other virulence factors, including pneumococcal surface protein A
(PspA), CbpA and Ply, facilitate evasion of opsonophagocytosis. b | To penetrate the blood–brain barrier, S. pneumoniae
uses ChoP–PAFR , CbpA–PIGR and CbpA–laminin receptor (LR) binding. Strains that express pili also use RrgA to bind to
PIGR and platelet endothelial cell adhesion molecule 1 (PECAM1). Similar to invasion of the respiratory tract, Ply , H2O2
generated by α-​glycerophosphate oxidase (GlpO) and activated plasmin bound to the pneumococcal surface proteins
Eno, GAPDH and CbpE can compromise the blood–brain barrier.

controversial. Most studies of pneumococcal biofilms and in the middle ear cleft of chinchillas co-​infected
have been carried out in vitro, and in vivo data are limited. with S. pneumoniae and Haemophilus influenzae110. In
Pneumococcal biofilm structures have been detected biopsy samples from volunteers colonized in experi­
in biopsy samples from patients with otitis media109 mental human studies, S. pneumoniae was found in

Nature Reviews | Microbiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

microcolonies68, although it has not been determined route involves interaction between the pneumococcal
whether these have characteristics of biofilms, such as an surface protein CbpA and polymeric immunoglobulin
extracellular matrix. The production of an extracellular receptor (PIGR) on human respiratory epithelial cells.
matrix has a major impact on the ability of S. pneumoniae Subversion of the PIGR recycling pathway enables inter-
that has been grown in a biofilm in vitro to subse- nalization and transmigration of S. pneumoniae across
quently translocate from the nasopharynx to the lungs polarized epithelial cell monolayers41. Interestingly,
in a murine infection model111. A recent report has also another region of CbpA has been shown to bind to the
suggested that NanA-​mediated cleavage of sialic acid laminin receptor on brain microvascular endothelium,
promotes biofilm formation in vivo and increases carbon and this facilitates penetration of the BBB during devel-
availability during colonization112. Murine experiments opment of pneumococcal meningitis118. CbpA, as well as
suggested that the large surface-​exposed glycoprotein laminin receptor and PAFR, are also necessary for inva-
PsrP is particularly important for bacterial attachment sion of cardiomyocytes and formation of cardiac microle-
to lung cells and biofilm formation by intraspecies inter- sions, which can occur as a complication of invasive
action113. PsrP seems to be required for bacterial persis- pneumococcal disease119. Recently, the ancillary pilus sub-
tence in the lower airway but not for nasal colonization unit RrgA, the tip adhesin of the pneumococcal pilus 1,
or survival in the bloodstream during sepsis114. has also been shown to interact with PIGR and platelet
Quorum sensing (QS) and phase variation also have an endothelial cell adhesion molecule 1 (PECAM1) on brain
important role in modulating pneumococcal niche adap- microvascular endothelium, and antibody blockade or
tations. It has been known for many years that S. pneumo- deletion of these two receptors reduced brain invasion
niae colonies can switch between transparent and opaque in a mouse meningitis model120. Currently, the relative
phenotypes in a process known as phase variation. These importance of these uptake mechanisms and the extent
variants differ in levels of expression of key virulence of cooperation between them are uncertain. Furthermore,
proteins, such as PspA and CbpA, as well as CPS and cell many S. pneumoniae strains are not piliated and thus
wall teichoic acid. The transparent phenotype is favoured cannot use RrgA-​dependent pathways. It should also be
in the nasopharyngeal niche, and the opaque phenotype emphasized that bacteraemia is not an essential prerequi-
is favoured in the blood115. A more recent study has site for meningitis, as localized infections such as sinusitis
shown that the underlying mechanism involves a type I or mastoiditis can also lead to meningitis. When mod-
restriction-​modification system, SpnIII, within a genetic elled in mice, meningitis may also develop as a conse-
locus containing inverted repeats that enable sponta- quence of the interaction of pneumococci colonizing the
neous rearrangement of alternative specificity domain nasopharynx with gangliosides on the surface of olfactory
genes. This generates six different SpnIII target specif- neurons, triggering cell invasion and direct entry of pneu-
icities, each with distinct genome-​wide DNA methyl- mococci into the central nervous system by retrograde
ation patterns, gene expression profiles and virulence axonal transport121. Such non-​haematogenous spread is
phenotypes116. Moreover, pneumococci were shown to stimulated by exogenous sialic acid106.
readily switch between SpnIII alleles during progression Regardless of the mechanism or site of invasion, the
of disease in a murine model116. Differentially expressed pneumococcal capsule impedes adherence to and inva-
genes included the CPS biosynthesis locus cps, various sion of host cells because it may sterically hinder inter-
sugar transporters, the Mn transporter psaBCA and luxS. actions between cell wall ChoP or surface proteins and
The luxS gene is of particular interest, as it is involved their cognate host receptors122. However, pneumococci
in the synthesis of the ubiquitous QS molecule autoin- markedly reduce capsule thickness when in close contact
ducer 2 (AI-2), which is an important regulator of bio- with epithelial cells and during the invasion process123.
film formation and virulence in pneumococci117. Recent This process of capsule shedding has recently been shown
studies show that AI-2 accumulating in the extracellular to depend on the major pneumococcal autolysin LytA
compartment is sensed by the fructose PTS transporter and is triggered by exposure to cationic antimicrobial
subunit IIC FruA, leading to upregulation of the galac- peptides that are released by the host cells124.
tose ABC transporter and the Leloir pathway105. Galactose
Quorum sensing is an important carbon source for S. pneumoniae in the Several pneumococcal virulence factors that directly
(QS). A system of stimuli and respiratory tract, and AI-2-mediated QS seems to be damage host tissues or induce host inflammatory
responses that is correlated to essential for its uptake and metabolism. Upregulation of responses also facilitate tissue invasion. One of the
microbial population density. the Leloir pathway increases the availability of activated most notable examples is Ply, which, in addition to
Restriction-​modification
sugar precursors, leading to increased production of CPS wide-​ranging pro-​inflammatory effects, directly lyses or
system and a hypervirulent phenotype105. induces apoptosis of diverse cell types, including lung
A bacterial defence system in epithelium and endothelial cells at the BBB125. Ply also
which restriction Penetration of tissues. Invasive pneumococcal disease inhibits mucociliary clearance in human lungs, separates
endonucleases cleave and
requires breaching of epithelial and/or endothelial barri- tight junctions between cells (which enables tissue pene-
inactivate specific target
sequences in foreign DNA (for ers and penetration of tissues, ultimately providing access tration), and exposes new sites for pneumococcal attach-
example, from phages); to the bloodstream. In the case of meningitis, this involves ment126. The pneumococcal pyruvate oxidase SpxB and
cleavage sites in host DNA are breaching the blood–brain barrier (BBB). Invasion α-​glycerophosphate oxidase GlpO produce hydrogen per-
protected by methylation. involves interaction between ChoP moieties and PAFR oxide, which also contributes to tissue damage in the lung
Leloir pathway
on the surface of cytokine-​activated respiratory epithelial and at the BBB127. Surface-​exposed hydrolytic enzymes,
The predominant route of and vascular endothelial cells, followed by hijacking of the including neuraminidases, hyaluronate lyase128 and metal-
cellular galactose metabolism. PAFR recycling pathway to gain entry40. An alternative loproteinases129, can also directly damage host tissues. Two

www.nature.com/nrmicro
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Table 1 | Major pneumococcal virulence factors


Virulence factor Description Function in pathogenesis
CPS • Major surface antigen • Prevents entrapment by mucus during colonization
• 98 structurally distinct serotypes • Inhibits opsonophagocytosis by preventing the interaction of iC3b and
the Fc fragment of IgG bound to deeper bacterial surface structures with
receptors on phagocytic cells
ChoP on teichoic acid PAFR ligand Binds PAFR on surface of epithelial and endothelial cells, facilitating
adherence and invasion
Lipopeptides, lipoteichoic Pathogen-​associated molecular patterns Promote inflammation
acid and peptidoglycan
fragments
Ply • Pore-​forming toxin • Cytotoxic and pro-​apoptotic for a wide variety of host cells
• TLR4 ligand • Activates classical complement pathway and depletes serum opsonic
activity
• Highly pro-​inflammatory at sub-​lytic levels
• Activates TLR4, NLRP3 inflammasome and p38–MAPK pathways
PspA CBP • Limits C3 deposition on pneumococcal surface
• Protects against bactericidal effects of free lactoferrin
CbpA (also known as PspC) CBP • Binds C3 and factor H and limits C3b deposition on pneumococcal surface
• Binds PIGR and laminin receptor through separate domains
• Facilitates adherence and invasion of respiratory epithelium and
blood–brain barrier
LytA • CBP • Digests cell wall
• Autolysin • Releases Ply and pro-​inflammatory cell wall fragments
• Mediates capsule shedding during cellular invasion
CbpD • CBP • Mediates fratricide and release of extracellular DNA
• Murein hydrolase • Promotes biofilm formation
CbpE (also known as Pce) • CBP • Decreases neutrophil activity by inactivation of host PAF
• Phosphorylcholine esterase • Binds plasminogen
CbpG CBP serine protease • Cell-​attached form promotes adherence
• Extracellular form degrades fibronectin
• Important for mucosal and invasive disease
CbpL CBP • Binds collagen, elastin and C-​reactive protein
• Promotes dissemination from nasopharynx to lungs and blood by
inhibiting phagocytosis
NanA • Neuraminidase A • Cleaves terminal sialic acid from host mucin and cell surface
• LPXTG glycoconjugates
• Unmasks receptors for adhesins
• Important role in otitis media
• Triggers TGF-​β signalling to facilitate endothelial invasion
BgaA • β-​Galactosidase Sequentially cleaves sugars from host glycoconjugates
• LPXTG
StrH • β-​N-acetylglucosaminidase Sequentially cleaves sugars from host glycoconjugates
• LPXTG
EndoD • Endo-​N-acetylglucosaminidase Sequentially cleaves sugars from host glycoconjugates
• LPXTG
Hyl • Hyaluronate lyase • Degrades extracellular matrix
• LPXTG • Facilitates tissue penetration
PrtA • Cell wall-​associated serine protease • Cleaves lactoferrin
• LPXTG • Possible adhesin
ZmpA (also known as IgA1 • Zinc metalloprotease Cleaves human IgA1
protease) • LPXTG
ZmpB • Zinc metalloprotease Possible adhesin
• LPXTG
ZmpC • Zinc metalloprotease Cleaves human matrix metalloproteinase 9
• LPXTG
PepO Endopeptidase • Binds fibronectin and plasminogen
• Facilitates adherence and invasion
• Binds C1q to inhibit classical complement pathway
PsrP • Very large O-​glycosylated serine-​rich • Adhesin
repeat protein • Binds to lung cells via keratin 10
• LPXTG • Mediates bacterial aggregation and biofilm formation in lung tissue

Nature Reviews | Microbiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Table 1 (cont.) | Major pneumococcal virulence factors


Virulence factor Description Function in pathogenesis
RrgA , RrgB and RrgC • LPXTG proteins • Adhesins
• Structural components of pilus 1 • Binds to a range of glycans
• Encoded by rlrA pathogenicity islet • Facilitate colonization and biofilm formation
• RrgA is tip adhesin • RrgA also binds PIGR and PECAM1 on endothelium of the blood–brain
barrier, which promotes brain invasion
PsaA • Lipoprotein • Mn uptake in host environment
• Solute-​binding component of • Essential for pneumococcal resistance to oxidative stress in vivo
Mn-​specific
• ABC transporter
AdcA and AdcAII • Lipoproteins Zn acquisition in vivo
• Solute-​binding components of a single
Zn-​specific ABC transporter
PiuA , PiaA and PitA • Lipoprotein Fe acquisition in vivo
• Solute-​binding components of
iron-​specific ABC transporters
SlrA and PpmA • Lipoproteins Contribute to nasopharyngeal colonization
• Peptidyl-​prolyl isomerases
PhtA , PhtB, PhtD and PhtE Family of surface proteins with unusual • May reduce C3 deposition on pneumococcal surface by binding factor H
His-​triad motifs • Putative adhesins
• Facilitate Zn acquisition together with AdcAII
PavA and PavB • Fibronectin-​binding proteins • Adhere to host surfaces
• NCSP • Important during sepsis and meningitis
Eno • Enolase • Binds and activates plasminogen
• NCSP • Facilitates tissue invasion
GAPDH • Glyceraldehyde-3-phosphate • Binds and activates plasminogen
dehydrogenase • Facilitates tissue invasion
• NCSP
SpxB Pyruvate oxidase Generates H2O2
GlpO α-​Glycerophosphate oxidase Generates H2O2
SodA Mn-​dependent superoxide dismutase Resistance to oxidative stress
Etrx1 and Etrx2 Surface-​exposed thioredoxin-​family Resistance to oxidative stress
lipoproteins
SpMsrAB2 Methionine sulfoxide reductase Redox partner of Etrx1 and Etrx2
ABC, ATP-​binding cassette; CBP, choline-​binding protein; ChoP, phosphorylcholine; CPS, capsular polysaccharide; Etrx, surface-​exposed thioredoxin family
lipoprotein; Fe, iron; H2O2, hydrogen peroxide; iC3b, inactivated C3b; IgG, immunoglobulin G; LPXTG, sortase-​anchored surface protein; MAPK , mitogen-​activated
protein kinase; Mn, manganese; NCSP, non-​classical surface protein lacking secretion signals or anchorage motifs; NLRP3, NACHT, LRR and PYD domains-​containing
protein 3; PAF, platelet-​activating factor ; PAFR , PAF receptor ; Pav, adherence and virulence protein; PECAM1, platelet endothelial cell adhesion molecule 1; PIGR ,
polymeric immunoglobulin receptor ; Ply , pneumolysin; PpmA , foldase protein PrsA ; PspA , pneumococcal surface protein A; Zn, zinc.

glycolytic enzymes, Eno and glyceraldehyde-3-phosphate potent activator of neutrophils133. Many of the virulence
dehydrogenase, are also surface-​exposed and function determinants of S. pneumoniae target components of the
as plasminogen-​binding proteins along with CbpE (also complement system to minimize opsonophagocytosis
known as Pce). They sequester and activate host plasmino- and clearance of invading pneumococci (reviewed in
gen at the pneumococcal surface and facilitate adherence ref.134). The CPS is undoubtedly the most important
to and penetration of the extracellular matrix130,131. An defence against the host immune system. For exam-
overview of pneumococcal surface proteins and other fac- ple, although non-​e ncapsulated pneumococci can
tors contributing to adherence and invasion is provided colonize the URT and cause superficial eye infections,
in Table 1. they rarely cause invasive infection. CPS covers deeper
bacterial surface structures and thereby inhibits bind-
Evasion and subversion of host defences. S. pneumo- ing of immunoglobulins, complement components
niae expresses a plethora of factors that mediate immune and C-​reactive protein. It reduces opsonization with
evasion and subversion (Table  1). As an extracellu- C3b and inactivated C3b (iC3b) and physically impairs
lar pathogen, S. pneumoniae must evade neutrophil-​ interactions between C3b, iC3b and Fc regions of immu-
mediated killing to survive the acute inflammation that noglobulins with their receptors on phagocytic cells135.
accompanies tissue invasion. Neutrophils can readily kill Capsular serotypes differ in the effectiveness with which
phagocytized pneumococci by releasing serine proteases they inhibit opsonophagocytosis and the level of inhi-
from neutrophil granules132. One mechanism to evade bition correlates with their ability to cause invasive dis-
neutrophil recruitment involves CbpE, which functions ease. Studies of capsule-​switch mutants have shown an
as a ChoP esterase. CbpE cleaves ChoP moieties on inverse relationship between the amount of C3b and
host-​derived platelet-​activating factor (PAF), which is a iC3b deposition and binding to factor H, which inhibits

www.nature.com/nrmicro
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

Opsonophagocytosis
the alternative complement pathway136. Increased levels Short-​chain forms have minimal surface area and
A process by which a of factor H in nasal lavages of asymptomatic individ- are less likely to trigger complement activation and
microorganism is labelled uals infected with a URT virus predispose the host to therefore are more likely to evade opsonophagocytic
(opsonized) by host immune acquisition of S. pneumoniae137. Factor H mainly binds clearance during invasive disease144.
factors to facilitate uptake by
CbpA on the pneumococcal surface. CbpA can also bind
phagocytic cells.
directly to C3 and, in some strains, the classical com- Conclusions and outlook
plement pathway inhibitor C4b-​binding protein (C4BP) S. pneumoniae has proved to be a truly resilient foe. It
in an interaction that is inhibited by CbpA binding to has overcome selective pressure from multiple classes of
vitronectin134,138,139. Thus, CPS and CbpA on the pneu- antibiotics and now seems to be adapting to the immune
mococcal surface are both important for resistance to pressure of widespread immunization. These develop-
opsonophagocytosis. ments demonstrate that we cannot be complacent, and
PspA also interferes with complement deposition by further insights are needed to combat pneumococcal dis-
binding factor B and blocking formation of or accel- ease. This Review has highlighted the current state of our
erating the dissociation of the alternative pathway understanding of the three key stages in the pathogene-
C3 ­convertase140. Furthermore, Ply released from the sis of S. pneumoniae — transmission, colonization and
bacterium activates the classical complement pathway invasion. In particular, our understanding has profited
through a domain with structural similarity to the from progress in defining the molecular events involved
Fc component of IgG, thereby depleting serum opsonic in invasion and new models of transmission in infant
activity125,141. The combined functions of PspA and mice and of carriage in humans. Further progress will
Ply are essential for S. pneumoniae to successfully likely come from a broader perspective that takes into
cause septicaemia142. Other pneumococcal proteins account pneumococcal ecology. In this regard, there are
that interfere with opsonophagocytosis include the now more than 8,000 publicly available whole-​genome
exoglycosidases NanA, BgaA and StrH, presuma- sequences of S. pneumoniae, which are providing a more
bly by deglycosylating human glycoproteins that are comprehensive view of the species and the remarkable
important for complement deposition 143. Eno and extent of its diversity. Additional insight will come from
glyceraldehyde-3-phosphate dehydrogenase (GAPDH) studies of the interactions of S. pneumoniae with other
bind and activate plasminogen, and this can also con- members of the microbiota and a better understanding
tribute to immune evasion through the degradation of of its niche in the human URT.
complement pathway components134. S. pneumoniae
Published online xx xx xxxx
grows in both short-​c hain and long-​c hain forms.

1. Abdullahi, O. et al. The prevalence and risk factors for 12. Richard, A. L., Siegel, S. J., Erikson, J. & Weiser, J. N. 21. Verhagen, L. M. et al. Genome-​wide identification of
pneumococcal colonization of the nasopharynx among TLR2 signaling decreases transmission of genes essential for the survival of Streptococcus
children in Kilifi District, Kenya. PLoS ONE 7, e30787 Streptococcus pneumoniae by limiting bacterial pneumoniae in human saliva. PLoS. ONE. 9, e89541
(2012). shedding in an infant mouse Influenza A co-​infection (2014).
2. Yahiaoui, R. Y. et al. Prevalence and antibiotic model. PLoS Pathog. 10, e1004339 22. Hamaguchi, S., Zafar, M. A., Cammer, M. &
resistance of commensal Streptococcus pneumoniae (2014). Weiser, J. N. Capsule prolongs survival of
in nine European countries. Future Microbiol. 11, 13. Kono, M. et al. Single cell bottlenecks in the Streptococcus pneumoniae during starvation. Infect.
737–744 (2016). pathogenesis of Streptococcus pneumoniae. PLoS Immun. https://doi.org/10.1128/IAI.00802-17
3. Bogaert, D., De Groot, R. & Hermans, P. W. Pathog. 12, e1005887 (2016). (2018).
Streptococcus pneumoniae colonisation: the key 14. Zafar, M. A., Kono, M., Wang, Y., Zangari, T. & 23. Walsh, R. L. & Camilli, A. Streptococcus pneumoniae
to pneumococcal disease. Lancet Infect. Dis. 4, Weiser, J. N. Infant mouse model for the study of is desiccation tolerant and infectious upon
144–154 (2004). shedding and transmission during Streptococcus rehydration. mBio 2, e00092–11 (2011).
4. Whitney, C. G. et al. Decline in invasive pneumococcal pneumoniae monoinfection. Infect. Immun. 84, 24. Zangari, T., Wang, Y. & Weiser, J. N. Streptococcus
disease after the introduction of protein-​ 2714–2722 (2016). pneumoniae transmission is blocked by type-​specific
polysaccharide conjugate vaccine. N. Engl. J. Med. 15. Rodrigues, F. et al. Relationships between rhinitis immunity in an infant mouse model. mBio 8,
348, 1737–1746 (2003). symptoms, respiratory viral infections and e00188–17 (2017).
5. Musher, D. How contagious are common respiratory nasopharyngeal colonization with Streptococcus 25. Roche, A. M., Richard, A. L., Rahkola, J. T.,
tract infections? N. Engl. J. Med. 348, 1256–1266 pneumoniae. Haemophilus influenza and Janoff, E. N. & Weiser, J. N. Antibody blocks
(2003). Staphylococcus aureus in children attending daycare. acquisition of bacterial colonization through
6. Numminen, E. et al. Climate induces seasonality in Pediatr. Infect. Dis. J. 32, 227–232 (2013). agglutination. Mucosal Immunol. 8, 176–185
pneumococcal transmission. Sci. Rep. 5, 11344 (2015). 16. Zafar, M. A., Wang, Y., Hamaguchi, S. & Weiser, J. N. (2015).
7. Gwaltney, J. J., Sande, M., Austrian, R. & Hendley, J. Host-​to-host transmission of Streptococcus 26. Janoff, E. N. et al. Pneumococcal IgA1 protease
Spread of Streptococcus pneumoniae in families. II. pneumoniae is driven by its inflammatory toxin, subverts specific protection by human IgA1. Mucosal
Relation of transfer of S. pneumoniae to incidence of pneumolysin. Cell Host Microbe 21, 73–83 (2017). Immunol. 7, 249–256 (2014).
colds and serum antibody. J. Infect. Dis. 132, 62–68 This study provides evidence that the toxin 27. Pennington, S. H. et al. Polysaccharide-​specific
(1975). Ply promotes mucosal inflammation, which memory b cells predict protection against
8. McCullers, J. et al. Influenza enhances susceptibility to facilitates pneumococcal transmission in experimental human pneumococcal carriage. Am. J.
natural acquisition of and disease due to infant mice. Respir. Crit. Care Med. 194, 1523–1531 (2016).
Streptococcus pneumoniae in ferrets. J. Infect. Dis. 17. Matthias, K. A., Roche, A. M., Standish, A. J., 28. Mitsi, E. et al. Agglutination by anti-​capsular
202, 1287–1295 (2010). Shchepetov, M. & Weiser, J. N. Neutrophil-​toxin polysaccharide antibody is associated with protection
9. Diavatopoulos, D. A. et al. Influenza A virus facilitates interactions promote antigen delivery and mucosal against experimental human pneumococcal carriage.
Streptococcus pneumoniae and disease. FASEB. J. 24, clearance of Streptococcus pneumoniae. J. Immunol. Mucosal Immunol. 10, 385–394 (2017).
1789–1798 (2010). 180, 6246–6254 (2008). This study shows that the agglutinating activity of
This study demonstrates the role of influenza virus 18. Lipsitch, M. & Moxon, E. R. Virulence and anticapsular antibody mediates protection from
in pneumococcal transmission in an infant mouse transmissibility of pathogens: what is the relationship? experimental pneumococcal carriage in humans.
model. Trends Microbiol. 5, 31–37 (1997). 29. Lemon, J. K. & Weiser, J. N. Degradation products of
10. Barbier, D. et al. Influenza A induces the major 19. Zafar, M. A., Hamaguchi, S., Zangari, T., Cammer, M. the extracellular pathogen Streptococcus pneumoniae
secreted airway mucin MUC5AC in a protease-​EGFR- & Weiser, J. N. Capsule type and amount affect access the cytosol via its pore-​forming toxin. mBio 6,
extracellular regulated kinase-​Sp1-dependent pathway. shedding and transmission of Streptococcus e02110–e02114 (2015).
Am. J. Respir. Cell. Mol. Biol. 47, 149–157 (2012). pneumoniae. mBio 8, e00989–17 (2017). 30. Davis, K., Nakamura, S. & Weiser, J. Nod2 sensing of
11. Siegel, S., Roche, A. & Weiser, J. Influenza promotes 20. Marks, L. R., Reddinger, R. M. & Hakansson, A. P. lysozyme-​digested peptidoglycan promotes
pneumococcal growth during coinfection by providing Biofilm formation enhances fomite survival of macrophage recruitment and clearance of S.
host sialylated substrates as a nutrient source. Streptococcus pneumoniae and Streptococcus pneumoniae colonization in mice. J. Clin. Invest. 121,
Cell Host Microbe 16, 55–67 (2014). pyogenes. Infect. Immun. 82, 1141–1146 (2014). 3666–3676 (2011).

Nature Reviews | Microbiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

31. Karmakar, M. et al. Neutrophil IL-1beta processing opsonophagocytic killing. PLoS Pathog. 3, with the cell wall polysaccharide. Infect. Immun. 74,
induced by pneumolysin is mediated by the NLRP3/ 1073–1081 (2007). 2187–2195 (2006).
ASC inflammasome and caspase-1 activation and is This study elucidates how H. influenzae signalling 73. van Rossum, A., Lysenko, E. & Weiser, J. Host and
dependent on K+ efflux. J. Immunol. 194, via NOD1 enhances neutrophil killing of bacterial factors contributing to the clearance of
1763–1775 (2015). S. pneumoniae, leading to bacterial clearance. colonization by Streptococcus pneumoniae in a murine
32. Parker, D. et al. Streptococcus pneumoniae DNA 54. Cremers, A. J. et al. The adult nasopharyngeal model. Infect. Immun. 73, 7718–7726 (2005).
initiates type I interferon signaling in the respiratory microbiome as a determinant of pneumococcal 74. McCool, T. L., Cate, T. R., Moy, G. & Weiser, J. N. The
tract. mBio 2, e00016–11 (2011). acquisition. Microbiome 2, 44 (2014). immune response to pneumococcal proteins during
33. Davis, K., Akinbi, H., Standish, A. & Weiser, J. 55. Biesbroek, G. et al. Early respiratory microbiota experimental human carriage. J. Exp. Med. 195,
Resistance to mucosal lysozyme compensates for the composition determines bacterial succession patterns 359–365 (2002).
fitness deficit of peptidoglycan modifications by and respiratory health in children. Am. J. Respir. Crit. 75. Ferreira, D. M. et al. Controlled human infection and
Streptococcus pneumoniae. PLoS Pathog. 4, Care Med. 190, 1283–1292 (2014). rechallenge with Streptococcus pneumoniae reveals
e1000241 (2008). 56. Miller, E. L., Abrudan, M. I., Roberts, I. S. & the protective efficacy of carriage in healthy adults.
34. Rose, M. C. & Voynow, J. A. Respiratory tract mucin Rozen, D. E. Diverse ecological strategies are encoded Am. J. Respir. Crit. Care. Med. 187, 855–864 (2013).
genes and mucin glycoproteins in health and disease. by Streptococcus pneumoniae bacteriocin-​like This study uses a human infection model to
Physiol. Rev. 86, 245–278 (2006). peptides. Genome Biol. Evol. 8, 1072–1090 demonstrate that immunity induced by a previous
35. Feldman, C. et al. The interaction of Streptococcus (2016). colonization episode protects against reacquisition.
pneumoniae with intact human respiratory mucosa 57. Dawid, S., Roche, A. & Weiser, J. The blp bacteriocins 76. Holmlund, E. et al. Antibodies to pneumococcal
in vitro. Eur. Respir. J. 5, 576–583 (1992). of Streptococcus pneumoniae mediate intraspecies proteins PhtD, CbpA, and LytC in Filipino pregnant
36. Nelson, A. L. et al. Capsule enhances pneumococcal competition both in vitro and in vivo. Infect. Immun. women and their infants in relation to pneumococcal
colonization by limiting mucus-​mediated clearance. 75, 443–451 (2007). carriage. Clin. Vaccine Immunol. 16, 916–923 (2009).
Infect. Immun. 75, 83–90 (2007). 58. Bogaardt, C., van Tonder, A. J. & Brueggemann, A. B. 77. Jackson, L. A. et al. Effectiveness of pneumococcal
37. Holmes, A. R. et al. The pavA gene of Streptococcus Genomic analyses of pneumococci reveal a wide polysaccharide vaccine in older adults. N. Engl.
pneumoniae encodes a fibronectin-​binding protein diversity of bacteriocins — including pneumocyclicin, a J. Med. 348, 1747–1755 (2003).
that is essential for virulence. Mol. Microbiol. 41, novel circular bacteriocin. BMC Genom. 16, 554 78. Richards, L., Ferreira, D. M., Miyaji, E. N.,
1395–1408 (2001). (2015). Andrew, P. W. & Kadioglu, A. The immunising effect of
38. Bergmann, S., Rohde, M., Chhatwal, G. S. & 59. Nakamura, S., Davis, K. & Weiser, J. Synergistic pneumococcal nasopharyngeal colonisation;
Hammerschmidt, S. α-​Enolase of Streptococcus stimulation of type I interferons during influenza virus protection against future colonisation and fatal
pneumoniae is a plasmin(ogen)-binding protein coinfection promotes Streptococcus pneumoniae invasive disease. Immunobiology 215, 251–263
displayed on the bacterial cell surface. Mol. Microbiol. colonization in mice. J. Clin. Invest. 121, 3657–3665 (2010).
40, 1273–1287 (2001). (2011). 79. Cohen, J. M., Wilson, R., Shah, P., Baxendale, H. E. &
39. Jensch, I. et al. PavB is a surface-​exposed adhesin of This study demonstrates a mechanism by which Brown, J. S. Lack of cross-​protection against invasive
Streptococcus pneumoniae contributing to concurrent influenza virus infection leads to pneumonia caused by heterologous strains following
nasopharyngeal colonization and airways infections. increased pneumococcal carriage. murine Streptococcus pneumoniae nasopharyngeal
Mol. Microbiol. 77, 22–43 (2010). 60. McCullers, J. A. & Rehg, J. E. Lethal synergism colonisation despite whole cell ELISAs showing
40. Cundell, D. R., Gerard, N. P., Gerard, C., Idanpaan-​ between influenza virus and Streptococcus significant cross-​reactive IgG. Vaccine 31,
Heikkila, I. & Tuomanen, E. I. Streptococcus pneumoniae: characterization of a mouse model and 2328–2332 (2013).
pneumoniae anchor to activated human cells by the the role of platelet-​activating factor receptor. J. Infect. 80. Wright, A. K. et al. Experimental human pneumococcal
receptor for platelet-​activating factor. Nature 377, Dis. 186, 341–350 (2002). carriage augments IL-17A-​dependent T-​cell defence of
435–438 (1995). 61. Avadhanula, V. et al. Respiratory viruses augment the the lung. PLoS Pathog. 9, e1003274 (2013).
41. Zhang, J. R. et al. The polymeric immunoglobulin adhesion of bacterial pathogens to respiratory 81. Wright, A. K. et al. Human nasal challenge with
receptor translocates pneumococci across human epithelium in a viral species- and cell type-​dependent Streptococcus pneumoniae is immunising in the
nasopharyngeal epithelial cells. Cell 102, 827–837 manner. J. Virol. 80, 1629–1636 (2006). absence of carriage. PLoS. Pathog. 8, e1002622
(2000). 62. Mina, M. J., McCullers, J. A. & Klugman, K. P. Live (2012).
42. Hauck, C. R. Cell adhesion receptors - signaling attenuated influenza vaccine enhances colonization of 82. Malley, R. et al. CD4+ T cells mediate antibody-​
capacity and exploitation by bacterial pathogens. Streptococcus pneumoniae and Staphylococcus independent acquired immunity to pneumococcal
Med. Microbiol. Immunol. 191, 55–62 (2002). aureus in mice. mBio 5, e01040–13 (2014). colonization. Proc. Natl Acad. Sci. USA. 102,
43. Kc, R., Shukla, S. D., Walters, E. H. & O’Toole, R. F. 63. Mina, M. J. Generalized herd effects and vaccine 4848–4853 (2005).
Temporal upregulation of host surface receptors evaluation: impact of live influenza vaccine on 83. Trzcinski, K. et al. Protection against nasopharyngeal
provides a window of opportunity for bacterial off-​target bacterial colonisation. J. Infect. 74, (Suppl. 1), colonization by Streptococcus pneumoniae is
adhesion and disease. Microbiology 163, 421–430 S101–s107 (2017). mediated by antigen-​specific CD4+ T cells. Infect.
(2017). 64. Thors, V. et al. The effects of live attenuated influenza Immun. 76, 2678–2684 (2008).
44. Cron, L. E. et al. Surface-​associated lipoprotein vaccine on nasopharyngeal bacteria in healthy 2 to 4 84. Mubarak, A. et al. A dynamic relationship between
PpmA of Streptococcus pneumoniae is involved in year olds. A randomized controlled trial. Am. J. Respir. mucosal T helper type 17 and regulatory T-​cell
colonization in a strain-​specific manner. Microbiology Crit. Care Med. 193, 1401–1409 (2016). populations in nasopharynx evolves with age and
155, 2401–2410 (2009). 65. McCullers, J. The co-​pathogenesis of influenza viruses associates with the clearance of pneumococcal
45. Hermans, P. W. et al. The streptococcal lipoprotein with bacteria in the lung. Nat. Rev. Microbiol. 12, carriage in humans. Clin. Microbiol. Infect. 22, 736.
rotamase A (SlrA) is a functional peptidyl-​prolyl 252–262 (2014). e1–736.e7 (2016).
isomerase involved in pneumococcal colonization. Provides a good overview of how influenza virus 85. Polissi, A. et al. Large-​scale identification of virulence
J. Biol. Chem. 281, 968–976 (2006). co-​infection leads to bacterial superinfection in the genes from Streptococcus pneumoniae. Infect. Immun.
46. Gutierrez-​Fernandez, J. et al. Modular architecture lungs. 66, 5620–5629 (1998).
and unique teichoic acid recognition features of 66. Lees, J. A. et al. Genome-​wide identification of lineage 86. Lau, G. W. et al. A functional genomic analysis of type
choline-​binding protein L (CbpL) contributing to and locus specific variation associated with 3 Streptococcus pneumoniae virulence. Mol.
pneumococcal pathogenesis. Sci. Rep. 6, 38094 pneumococcal carriage duration. eLife 6, e26255 Microbiol. 40, 555–571 (2001).
(2016). (2017). 87. Hava, D. L. & Camilli, A. Large-​scale identification of
47. King, S. J. Pneumococcal modification of host sugars: 67. Kadioglu, A., Weiser, J., Paton, J. & Andrew, P. The serotype 4 Streptococcus pneumoniae virulence
a major contributor to colonization of the human role of Streptococcus pneumoniae virulence factors in factors. Mol. Microbiol. 45, 1389–1406 (2002).
airway? Mol. Oral Microbiol. 25, 15–24 (2010). host respiratory colonization and disease. Nat. Rev. 88. Orihuela, C. J. et al. Microarray analysis of
48. Uchiyama, S. et al. The surface-​anchored NanA Microbiol. 6, 288–301 (2008). pneumococcal gene expression during invasive
protein promotes pneumococcal brain endothelial cell 68. Jochems, S. P. et al. Novel analysis of immune cells disease. Infect. Immun. 72, 5582–5596 (2004).
invasion. J. Exp. Med. 206, 1845–1852 from nasal microbiopsy demonstrates reliable, 89. Ogunniyi, A. D. et al. Identification of genes that
(2009). reproducible data for immune populations, and contribute to the pathogenesis of invasive
49. Limoli, D. H., Sladek, J. A., Fuller, L. A., Singh, A. K. & superior cytokine detection compared to nasal wash. pneumococcal disease by in vitro transcriptomic
King, S. J. BgaA acts as an adhesin to mediate PLoS ONE 12, e0169805 (2017). analysis. Infect. Immun. 80, 3268–3278
attachment of some pneumococcal strains to human 69. Zhang, Z., Clarke, T. & Weiser, J. Cellular effectors (2012).
epithelial cells. Microbiology 157, 2369–2381 mediating Th17-dependent clearance of 90. Honsa, E. S., Johnson, M. D. & Rosch, J. W. The roles
(2011). pneumococcal colonization in mice. J. Clin. Invest. of transition metals in the physiology and
50. Andersson, B. et al. Identification of an active 119, 1899–1909 (2009). pathogenesis of Streptococcus pneumoniae. Front.
dissaccharide unit of a glycoconjugate receptor for This study shows that IL-17A by CD4+ T cells is Cell. Infect. Microbiol. 3, 92 (2013).
pneumococci attaching to human pharyngeal epithelial required for the recruitment of monocytes and 91. Brown, J. S., Gilliland, S. M. & Holden, D. W. A.
cells. J. Exp. Med. 158, 559–570 (1983). macrophages and effective pneumococcal Streptococcus pneumoniae pathogenicity island
51. Krivan, H. C., Roberts, D. D. & Ginsberg, V. Many clearance in unimmunized mice. encoding an ABC transporter involved in iron
pulmonary pathogenic bacteria bind specifically to the 70. Siegel, S., Tamashiro & Weiser, J. Clearance of uptake and virulence. Mol. Microbiol. 40,
carbohydrate sequence GalNAcb1-4 Gal found in pneumococcal colonization in infants is delayed 572–585 (2001).
some glycolipids. Proc. Natl. Acad. Sci. USA 85, through altered macrophage trafficking. PLoS Pathog. 92. McAllister, L. J. et al. Molecular analysis of the psa
6157–6161 (1988). 11, e1005004 (2015). permease complex of Streptococcus pneumoniae.
52. Shak, J. R., Vidal, J. E. & Klugman, K. P. Influence of 71. Puchta, A. et al. TNF drives monocyte dysfunction with Mol. Microbiol. 53, 889–901 (2004).
bacterial interactions on pneumococcal colonization of age and results in impaired anti-​pneumococcal 93. Plumptre, C. D. et al. AdcA and AdcAII employ
the nasopharynx. Trends. Microbiol. 21, 129–135 immunity. PLoS. Pathog 12, e1005368 (2016). distinct zinc acquisition mechanisms and contribute
(2013). 72. Malley, R. et al. Antibody-​independent, interleukin- additively to zinc homeostasis in. Streptococcus
53. Lysenko, E. S. et al. Nod1-signaling overcomes 17A-​mediated, cross-​serotype immunity to pneumoniae. Mol. Microbiol. 91, 834–851
resistance of Streptococcus pneumoniae to pneumococci in mice immunized intranasally (2014).

www.nature.com/nrmicro
© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.
Reviews

94. Bajaj, M. et al. Discovery of novel pneumococcal 115. Kim, J. O. & Weiser, J. N. Association of intrastrain 135. Hyams, C., Camberlein, E., Cohen, J. M., Bax, K. &
surface antigen A (PsaA) inhibitors using a fragment-​ phase variation in quantity of capsular polysaccharide Brown, J. S. The Streptococcus pneumoniae capsule
based drug design approach. ACS Chem. Biol. 10, and teichoic acid with the virulence of Streptococcus inhibits complement activity and neutrophil
1511–1520 (2015). pneumoniae. J. Infect. Dis. 177, 368–377 (1998). phagocytosis by multiple mechanisms. Infect. Immun.
95. McDevitt, C. A. et al. A molecular mechanism for 116. Manso, A. S. et al. A random six-​phase switch 78, 704–715 (2010).
bacterial susceptibility to zinc. PLoS Pathog. 7, regulates pneumococcal virulence via global 136. Hyams, C. et al. Streptococcus pneumoniae capsular
e1002357 (2011). epigenetic changes. Nat. Commun. 5, 5055 (2014). serotype invasiveness correlates with the degree of
96. Counago, R. M. et al. Imperfect coordination This study elucidates the mechanism underlying the factor H binding and opsonization with C3b/iC3b.
chemistry facilitates metal ion release in the Psa phenomenon of colony opacity phase variation in Infect. Immun. 81, 354–363 (2013).
permease. Nat. Chem. Biol. 10, 35–41 (2014). S. pneumoniae. 137. Hammerschmidt, S., Talay, S. R., Brandtzaeg, P. &
97. Kumar, S., Awasthi, S., Jain, A. & Srivastava, R. C. 117. Trappetti, C., Potter, A. J., Paton, A. W., Oggioni, M. R. Chhatwal, G. S. SpsA, a novel pneumococcal surface
Blood zinc levels in children hospitalized with severe & Paton, J. C. LuxS mediates iron-​dependent biofilm protein with specific binding to secretory
pneumonia: a case control study. Indian Pediatr. 41, formation, competence, and fratricide in immunoglobulin A and secretory component.
486–491 (2004). Streptococcus pneumoniae. Infect. Immun. 79, Mol. Microbiol. 25, 1113–1124 (1997).
98. Coles, C. L. et al. Zinc modifies the association 4550–4558 (2011). 138. Dieudonne-​Vatran, A. et al. Clinical isolates of
between nasopharyngeal Streptococcus pneumoniae 118. Orihuela, C. J. et al. Laminin receptor initiates Streptococcus pneumoniae bind the complement
carriage and risk of acute lower respiratory infection bacterial contact with the blood brain barrier in inhibitor C4b-​binding protein in a PspC allele-​dependent
among young children in rural Nepal. J. Nutr. 138, experimental meningitis models. J. Clin. Invest. 119, fashion. J. Immunol. 182, 7865–7877 (2009).
2462–2467 (2008). 1638–1646 (2009). 139. Kohler, S. et al. Binding of vitronectin and Factor H to
99. Hakansson, A. et al. Characterization of binding of 119. Brown, A. O. et al. Streptococcus pneumoniae Hic contributes to immune evasion of Streptococcus
human lactoferrin to pneumococcal surface protein A. translocates into the myocardium and forms unique pneumoniae serotype 3. Thromb. Haemostasis 113,
Infect. Immun. 69, 3372–3381 (2001). microlesions that disrupt cardiac function. PLoS 125–142 (2015).
100. Mirza, S. et al. The effects of differences in pspA Pathog. 10, e1004383 (2014). 140. Tu, A. H., Fulgham, R. L., McCrory, M. A., Briles, D. E.
alleles and capsular types on the resistance of 120. Iovino, F. et al. pIgR and PECAM-1 bind to & Szalai, A. J. Pneumococcal surface protein A inhibits
Streptococcus pneumoniae to killing by apolactoferrin. pneumococcal adhesins RrgA and PspC mediating complement activation by Streptococcus pneumoniae.
Microb. Pathog. 99, 209–219 (2016). bacterial brain invasion. J. Exp. Med. 214, Infect. Immun. 67, 4720–4724 (1999).
101. Bidossi, A. et al. A functional genomics approach to 1619–1630 (2017). 141. Paton, J. C., Rowan-​Kelly, B. & Ferrante, A. Activation
establish the complement of carbohydrate 121. van Ginkel, F. W. et al. Pneumococcal carriage results of human complement by the pneumococcal toxin
transporters in Streptococcus pneumoniae. PLoS ONE in ganglioside-​mediated olfactory tissue infection. pneumolysin. Infect. Immun. 43, 1085–1087 (1984).
7, e33320 (2012). Proc. Natl. Acad. Sci. USA 100, 14363–14367 142. Yuste, J., Botto, M., Paton, J. C., Holden, D. W. &
102. Buckwalter, C. M. & King, S. J. Pneumococcal (2003). Brown, J. S. Additive inhibition of complement
carbohydrate transport: food for thought. Trends 122. Talbot, U. M., Paton, A. W. & Paton, J. C. Uptake of deposition by pneumolysin and PspA facilitates
Microbiol. 20, 517–522 (2012). Streptococcus pneumoniae by respiratory epithelial Streptococcus pneumoniae septicemia. J. Immunol.
103. King, S. J., Hippe, K. R. & Weiser, J. N. Deglycosylation cells. Infect. Immun. 64, 3772–3777 (1996). 175, 1813–1819 (2005).
of human glycoconjugates by the sequential activities 123. Hammerschmidt, S. et al. Illustration of pneumococcal 143. Dalia, A., Standish, A. & Weiser, J. Three surface
of exoglycosidases expressed by Streptococcus polysaccharide capsule during adherence and invasion exoglycosidases from Streptococcus pneumoniae,
pneumoniae. Mol. Microbiol. 59, 961–974 (2006). of epithelial cells. Infect. Immun. 73, 4653–4667 NanA, BgaA, and StrH, promote resistance to
104. Robb, M. et al. Molecular characterization of N-​glycan (2005). opsonophagocytic killing by human neutrophils. Infect.
degradation and transport in Streptococcus 124. Kietzman, C. C., Gao, G., Mann, B., Myers, L. & Immun. 78, 2108–2116 (2010).
pneumoniae and its contribution to virulence. PLoS Tuomanen, E. I. Dynamic capsule restructuring by the 144. Dalia, A. & Weiser, J. Minimization of bacterial size
Pathog.13, e1006090 (2017). main pneumococcal autolysin LytA in response to the allows for complement evasion and is overcome by the
105. Trappetti, C. et al. Autoinducer 2 signaling via the epithelium. Nat. Commun. 7, 10859 (2016). agglutinating effect of antibody. Cell Host Microbe 10,
phosphotransferase frua drives galactose utilization by 125. Mitchell, T. J. & Dalziel, C. E. The biology of 486–496 (2011).
Streptococcus pneumoniae, resulting in pneumolysin. Subcell. Biochem. 80, 145–160 (2014). 145. O’Brien, K. L. et al. Effect of pneumococcal conjugate
hypervirulence. mBio 8, e02269–16 (2017). 126. Rayner, C. F. et al. Interaction of pneumolysin-​sufficient vaccine on nasopharyngeal colonization among
This study was the first to identify an AI-2 receptor and -deficient isogenic variants of Streptococcus immunized and unimmunized children in a community-​
in Gram-​positive bacteria and describe a pneumoniae with human respiratory mucosa. Infect. randomized trial. J. Infect. Dis. 196, 1211–1220 (2007).
mechanism whereby quorum sensing of AI-2 Immun. 63, 442–447 (1995). 146. Geno, K. A. et al. Pneumococcal capsules and their
promotes invasive disease. 127. Mahdi, L. K., Wang, H., Van der Hoek, M. B., types: past, present, and future. Clin. Microbiol. Rev.
106. Hatcher, B. L., Hale, J. Y. & Briles, D. E. Free sialic acid Paton, J. C. & Ogunniyi, A. D. Identification of a 28, 871–899 (2015).
acts as a signal that promotes Streptococcus novel pneumococcal vaccine antigen preferentially 147. Klugman, K. The significance of serotype replacement
pneumoniae invasion of nasal tissue and expressed during meningitis in mice. J. Clin. Invest. for pneumococcal disease and antibiotic resistance.
nonhematogenous invasion of the central nervous 122, 2208–2220 (2012). Adv. Exp. Med. Biol. 634, 121–128 (2009).
system. Infect. Immun. 84, 2607–2615 (2016). 128. Berry, A. M. & Paton, J. C. Additive attenuation of 148. von Gottberg, A. et al. Effects of vaccination on
107. Hentrich, K. et al. Streptococcus pneumoniae senses a virulence of Streptococcus pneumoniae by mutation of invasive pneumococcal disease in South Africa.
human-​like sialic acid profile via the response regulator the genes encoding pneumolysin and other putative N. Engl. J. Med. 371, 1889–1899 (2014).
ciaR. Cell Host Microbe 20, 307–317 (2016). pneumococcal virulence proteins. Infect. Immun. 68,
108. Gratz, N. et al. Pneumococcal neuraminidase activates 133–140 (2000). Acknowledgements
TGF-​beta signalling. Microbiology 163, 1198–1207 129. Chiavolini, D. et al. The three extra-​cellular zinc The authors thank J. Pagano for editorial assistance. J.N.W.
(2017). metalloproteinases of Streptococcus pneumoniae have is funded by grants from the United States Public Health
109. Hall-​Stoodley, L. et al. Direct detection of bacterial a different impact on virulence in mice. BMC Service (AI038446 and AI105168). Research in J.C.P.’s lab-
biofilms on the middle-​ear mucosa of children with Microbiol. 3, 14 (2003). oratory is supported by program grant 1071659 from the
chronic otitis media. JAMA 296, 202–211 (2006). 130. Attali, C., Durmort, C., Vernet, T. & Di Guilmi, A. M. National Health and Medical Research Council of Australia
110. Weimer, K. E. et al. Coinfection with Haemophilus The interaction of Streptococcus pneumoniae with (NHMRC); J.C.P. is an NHMRC Senior Principal Research
influenzae promotes pneumococcal biofilm formation plasmin mediates transmigration across endothelial Fellow. D.M.F. is supported by the Medical Research Council
during experimental otitis media and impedes the and epithelial monolayers by intercellular junction (grant MR/M011569/1) and the Bill and Melinda Gates
progression of pneumococcal disease. J. Infect. Dis. cleavage. Infect. Immun. 76, 5350–5356 (2008). Foundation (grant OPP1117728).
202, 1068–1075 (2010). 131. Bergmann, S., Rohde, M., Preissner, K. T. &
111. Trappetti, C., Ogunniyi, A. D., Oggioni, M. R. & Hammerschmidt, S. The nine residue plasminogen-​ Author contributions
Paton, J. C. Extracellular matrix formation enhances binding motif of the pneumococcal enolase is the All authors researched data for the article, substantially con-
the ability of Streptococcus pneumoniae to cause major cofactor of plasmin-​mediated degradation of tributed to discussion of content, wrote the article and
invasive disease. PLoS ONE 6, e19844 (2011). extracellular matrix, dissolution of fibrin and reviewed and edited the manuscript before submission.
112. Blanchette, K. A. et al. Neuraminidase A-​exposed transmigration. Thromb. Haemostasis 94, 304–311
galactose promotes Streptococcus pneumoniae (2005). Competing interests
biofilm formation during colonization. Infect. Immun. 132. Standish, A. & Weiser, J. Human neutrophils kill The authors declare no competing interests.
84, 2922–2932 (2016). Streptococcus pneumoniae via serine proteases.
113. Sanchez, C. J. et al. The pneumococcal serine-​rich J. Immunol. 183, 2602–2609 (2009). Publisher’s note
repeat protein is an intra-​species bacterial adhesin 133. Hergott, C. B. et al. Bacterial exploitation of Springer Nature remains neutral with regard to jurisdictional
that promotes bacterial aggregation in vivo and in phosphorylcholine mimicry suppresses inflammation claims in published maps and institutional affiliations.
biofilms. PLoS. Pathog 6, e1001044 (2010). to promote airway infection. J. Clin. Invest. 125,
114. Rose, L. et al. Antibodies against PsrP, a novel 3878–3890 (2015). Reviewer information
Streptococcus pneumoniae adhesin, block adhesion 134. Andre, G. O. et al. Role of Streptococcus pneumoniae Nature Reviews Microbiology thanks Sven Hammerschmidt
and protect mice against pneumococcal challenge. proteins in evasion of complement-​mediated and the other anonymous reviewer(s) for their contribution to
J. Infect. Dis. 198, 375–383 (2008). immunity. Front. Microbiol. 8, 224 (2017). the peer review of this work.

Nature Reviews | Microbiology


© 2018 Macmillan Publishers Limited, part of Springer Nature. All rights reserved.

You might also like