You are on page 1of 20

CLINICAL MICROBIOLOGY REVIEWS, Oct. 2008, p. 666–685 Vol. 21, No.

4
0893-8512/08/$08.00⫹0 doi:10.1128/CMR.00012-08
Copyright © 2008, American Society for Microbiology. All Rights Reserved.

Animal Models of Streptococcus pneumoniae Disease


Damiana Chiavolini,§ Gianni Pozzi, and Susanna Ricci*
Laboratory of Molecular Biology and Biotechnology (LA.M.M.B.), Department of Molecular Biology, University of Siena, Siena, Italy

INTRODUCTION .......................................................................................................................................................666
ANIMAL MODELS OF PNEUMOCOCCAL DISEASE .......................................................................................667

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


Considerations of Animal and Pneumococcal Strains ......................................................................................667
ANIMAL MODELS OF PNEUMONIA ...................................................................................................................668
Mouse Models .........................................................................................................................................................668
i.t. models.............................................................................................................................................................669
i.n. models............................................................................................................................................................669
Remarks on murine pneumococcal pneumonia..............................................................................................670
Rat and Rabbit Models..........................................................................................................................................670
ANIMAL MODELS OF SEPSIS ..............................................................................................................................671
Mouse Models .........................................................................................................................................................672
Rat and Rabbit Models..........................................................................................................................................673
ANIMAL MODELS OF MENINGITIS ...................................................................................................................673
Mouse Models .........................................................................................................................................................674
Direct induction of meningitis ..........................................................................................................................674
Hematogenous meningitis..................................................................................................................................675
Nonhematogenous meningitis ...........................................................................................................................675
Rat and Rabbit Models..........................................................................................................................................675
i.cist. models for rats..........................................................................................................................................675
Hematogenous and otologic models for rats...................................................................................................676
i.cist. infection in rabbits...................................................................................................................................677
ANIMAL MODELS OF OTITIS MEDIA................................................................................................................677
Chinchilla, Gerbil, and Guinea Pig Models........................................................................................................678
Rat and Mouse Models ..........................................................................................................................................678
CONCLUSIONS .........................................................................................................................................................679
ACKNOWLEDGMENTS ...........................................................................................................................................679
REFERENCES ............................................................................................................................................................680

INTRODUCTION age die of pneumonia and invasive diseases in developing


countries (177). Mortality rates for pneumonia and menin-
Streptococcus pneumoniae (the pneumococcus) is a major
gitis are especially high for young children, the elderly, and
human pathogen that colonizes the upper respiratory tract
immunocompromised individuals (55), including patients
and causes both life-threatening diseases such as pneumo-
with human immunodeficiency virus (135).
nia, sepsis, and meningitis and milder but common diseases,
like sinusitis and otitis media (91). Colonization of the na- Pneumococcal diseases are treated with ␤-lactams (penicil-
sopharynx by S. pneumoniae begins early on in life and peaks lin G, amoxicillin, cephalosporins), respiratory fluoroquino-
(up to 55%) at the age of 3 years (31). Colonization not only lones, macrolides, and vancomycin (159). However, efforts to
provides the basis for pneumococcal horizontal spread but is treat pneumococcal diseases have been complicated by in-
also necessary for invasive disease (31). The pneumococcus creasing resistance to antibiotics. A recent work reported high
produces a plethora of virulence factors that allow bacteria rates of resistance for some antimicrobials such as penicillin
to spread from the upper to the lower respiratory tract, (34.2%), trimethoprim-sulfamethoxazole (31.9%), and eryth-
leading to pneumonia and invasive disease (126). The latest romycin (29.5%) (74). For the prevention of pneumococcal
report from the Centers for Disease Control and Prevention disease, there is a licensed 23-valent polysaccharide vaccine
in the United States estimated that in 2006 pneumococci which is effective against 23 out of 90 pneumococcal serotypes
caused 41,400 invasive infections and 5,000 deaths, most of but does not confer protection in children younger than 2 years
which were due to bacteremic pneumonia (55). However, of age or in the elderly. A 7-valent conjugate vaccine has also
each year at least 1 million children younger than 5 years of been available since 2000 and is effective in children, although
its cost is high and protection is induced only against serotypes
included in the vaccine formulation (32). Both 9- and 11-valent
* Corresponding author. Mailing address: LA.M.M.B., Università di conjugate vaccines will be licensed in the near future (16).
Siena, Policlinico Le Scotte, V lotto 1° piano, Viale Bracci, 53100 Protein-based or protein subunit vaccines common to different
Siena, Italy. Phone: 39 0577 233100. Fax: 39 0577 233334. E-mail:
capsular serotypes represent attractive alternatives to improve
riccisus@unisi.it.
§ Present address: Evans Biomedical Research Center, 650 Albany protection and to address the current limitations of polysac-
Street, Boston University School of Medicine, Boston, MA 02118. charide vaccines (16).

666
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 667

ANIMAL MODELS OF PNEUMOCOCCAL DISEASE tion, their lower cost makes them attractive alternatives to
inbred strains.
Animal experimentation is an essential tool for the study of Experimental pneumococcal disease has been investigated
infectious diseases. Numerous animal models of diseases by use of different rodents and the rabbit, but undoubtedly the
caused by S. pneumoniae are currently available for clarifying mouse represents the most commonly used animal model. In-
mechanisms of disease pathogenesis, testing novel drugs and bred mouse strains, including BALB/c, C57BL/6, DBA, and
vaccine candidates, and characterizing the role of bacterial and CBA mice, have largely been exploited for the analysis of
host factors. Recent review articles have specifically addressed pneumococcal pneumonia (12, 113, 131, 176, 186, 201, 234),
the value of animal models to test pneumococcal protein vac- sepsis (15, 42, 43, 163, 186), meningitis (86, 95, 136, 173, 230),
cines (225) and the utility of murine models of pneumonia to and otitis media (162, 164). Experimental sepsis has also been

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


assess the efficacy of antimicrobials (175). Some authors have induced in immunodeficient mice, such as the CBA/N strain,
examined mouse genetic susceptibility to pneumococcal dis- which carries the Xid-linked immune deficiency (38, 43).
ease (121), while others have discussed the value of knockout CBA/N mice are unable to produce antibodies against pneu-
mice to investigate the pathophysiology of bacterial meningitis mococcal polysaccharides and phosphocholine and are thereby
(191). In 1999, Giebink effectively reviewed the utility of the highly susceptible to infection by S. pneumoniae (41). How-
chinchilla model for the study of middle ear (ME) pneumo- ever, outbred mice, such as MF1, CD-1 (Swiss), Swiss-Webster,
coccal infection (90), while Sabirov and Metzger have lately and NMRI, have increasingly become more popular. They
evaluated the importance of the mouse to test mucosal vac- have widely been used for analyzing pathogenicity mechanisms
cines against otitis media (211). A recent review on animal of pneumonia (50, 58, 114, 120, 125, 131), sepsis (109, 131,
models of invasive pneumococcal disease focused on the im- 251), and meningitis (59, 95, 160, 268). In addition, outbred
portance of sepsis and pneumonia in experimental animal mice have also been employed to evaluate active and passive
models for studying vaccine efficacy and also provided very protection against pneumonia and sepsis (4, 115, 163) and the
useful considerations on technical aspects (39). Meningitis and efficacy of antibiotic therapy against invasive pneumococcal
otitis media models were mentioned by the authors, although disease (13, 141, 169, 220, 223). The host genetic factors con-
to a lesser extent (39). The present report is a more compre- trolling the response to infection by S. pneumoniae are still
hensive review of animal models that have significantly con- unknown. Mouse susceptibility and resistance to pneumococ-
tributed to our knowledge of all the clinically relevant pneu- cal disease have been investigated in more detail in several
mococcal diseases, highlighting the importance of readouts, studies (72, 92, 130, 215) and discussed in a few reviews (41,
defining the advantages and the drawbacks, and suggesting 121). The rat has also been exploited for studying invasive
potential applications of each one of these systems. Models of pneumococcal disease and otitis media. Outbred strains, in-
colonization are briefly mentioned throughout the text but not cluding Wistar, Sprague-Dawley, and CD (derived from Spra-
discussed in detail. The reader interested in animal models of gue-Dawley) rats, have been extensively utilized as experimen-
pneumococcal colonization should refer to a recently pub- tal models (2, 49, 71, 79, 101, 103, 145, 147, 166, 193, 214, 221,
lished review (157). We fully appreciate the fundamental im- 242, 253). The New Zealand White rabbit is the most com-
portance of nasopharyngeal carriage as a crucial step for inva- monly used strain to induce pneumococcal pneumonia, sepsis,
sive disease, but we have chosen to focus on animal models of and meningitis (26, 57, 69, 98, 106, 119, 196, 246). Finally, the
acute disease, such as pneumonia, sepsis, meningitis, and otitis chinchilla (only outbred strains are available), gerbil, and
media. For easier reading, major experimental animal models guinea pig are commonly used animal models of experimental
are listed in tables according to the pneumococcal disease and otitis media (81, 89, 90, 260).
the animal species. Representative models were chosen based There is large variation in the outcomes of infection by S.
on historical importance, innovative improvement of preexist- pneumoniae in experimental animal models, depending on the
ing models, widespread use within the pneumococcus research pneumococcal strain used. It is well accepted that the virulence
community, and usefulness for future studies. of S. pneumoniae strains is strongly influenced by the capsular
serotype in both humans and mice (38, 129, 215); however, the
Considerations of Animal and Pneumococcal Strains genetic background of the strain also seems to be relevant for
disease development (29, 129, 215). Certain human isolates are
The choice of both animal and bacterial strains should be poorly virulent in vivo, whereas other pneumococcal strains
carefully considered before approaching the study of pneumo- that are highly virulent in mice have less relevance for human
coccal disease in vivo. It is beyond the scope of this review to disease (38, 39). Typically, strains of capsular serotypes 2, 3, 4,
include a detailed discussion on animal or S. pneumoniae 5, and 6 are virulent in mice, whereas strains of types 14, 19,
strains to be used in experimental models of disease; however, and 23 are relatively avirulent (21, 39). The low virulence of
we provide general considerations on the subject. many pneumococcal strains in rodents can be balanced by
Mouse and rat inbred strains afford more-uniform responses either using larger challenge doses (40) or neutropenic hosts
to experimental treatments due to their tightly controlled im- (19, 49, 169, 221, 223).
mune system. For this reason, they have extensively been em- Experimental pneumococcal disease has been induced with
ployed as models for infection, vaccination, and drug efficacy both historical laboratory strains and clinical isolates. The use
studies. Outbred mouse strains are instead generated to main- of classic strains, able to generate consistent features of dis-
tain maximum heterozygosity. Their phenotypic diversity, de- ease, allows comparison of results among research groups. Of
spite being lower than that of humans, may be important to particular relevance is the type 2 D39 reference strain (142),
mimic the natural variation in response to infection. In addi- which several research groups have employed to study the
668 CHIAVOLINI ET AL. CLIN. MICROBIOL. REV.

TABLE 1. Mouse models of pneumonia


Route of Pneumococcal
Mouse strain(s) Aim(s) of study Readouts Utility Reference(s)
infection serotype(s)

i.t. C57BL/6 3 Antibiotic efficacy, CFU, histology, survival, Drug efficacy and 12
model description pharmacokinetics pharmacokinetics
i.t. ddY NSa Gastric juice in Survival, CFU, PMNb, Pathogenesis of 114
pneumonia lung histology pneumonia in the
elderly
i.n. MF1 2 Role of pneumolysin CFU, lung histology Host-parasite interaction, 50
and autolysin, model disease pathogenesis,
description vaccine and drug

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


studies
i.n. Neonatal and 1 Immunization in early Antibody titers, CFU Neonatal and early-life 115
infant NMRI life vaccinology
i.n. BALB/c and OF1 14 (for BALB/c) Therapy in neutropenic CFU, survival, Antibiotic efficacy, 201, 223
and 3, 9 (for mice pharmacokinetics immunotherapy studies
OF1)
i.n. CBA/J 3, 11, 19 Model of penicillin- Survival, CFU, Antibiotic efficacy in 234
resistant pneumonia histology, immunocompetent
pharmacokinetics mice
Aerosol nebulizer CBA/J 28 Model of chronic CFU, serum Disease pathogenesis 113
obstructive immunoglobulin G
pneumonia
Aerosol nebulizer CBA/J 3 Low-dose model, drug Survival, CFU, histology Efficacy of tissue-directed 176
efficacy antimicrobials
a
NS, not specified.
b
PMN, polymorphonuclear cells.

pathogenesis of pneumococcal disease and test vaccine anti- The pathogenesis of pneumococcal pneumonia is a complex
gens over the years (25, 40, 50, 92, 103, 104, 109, 123, 124, 131, interplay between pneumococcal virulence and the host im-
180–182, 186). Serotype 3 strains, in particular A66 (9) and mune response (91). Since the late 1970s, when an important
WU2 (43), are also widely used for inducing sepsis in the model of pneumococcal pneumonia was set up for the rat (14),
mouse and the rat (2, 40, 43, 109, 129, 163) and to study many different animal models of pneumonia have been devel-
invasive pneumococcal disease in the rabbit (246, 247). More oped. Pneumonia due to S. pneumoniae has been studied
recently, laboratory strains with sequenced genomes, such as largely by use of the mouse, probably the most commonly
the type 4 TIGR4 strain (238) and the type 19F G54 strain employed system today for the characterization of this disease,
(75), have also been employed by different research groups (58, due to the ease of manipulation and data reproducibility in
59, 80, 131, 187, 197, 215). The availability of clinical isolates is large numbers of animals.
also important in attempting the reproduction of human dis-
ease in laboratory animals. Several groups have preferred se- Mouse Models
rotype 3 and 6 strains isolated from patients with pneumococ-
cal meningitis and have successfully induced this disease in the Different research groups have reproduced the features of
mouse, the rat, and the rabbit (26, 35, 69, 76, 85, 95, 145, 192, pneumococcal pneumonia in the mouse (Table 1). Mouse
268, 269). Clinical isolates of serotypes 3, 6, 9, and 19 have also models of pneumonia allow the analysis of different parame-
been employed in laboratory animal models to elucidate fea- ters, including animal survival after infection, the presence of
tures of pneumococcal pneumonia and sepsis (12, 14, 40, 49, bacteria in lungs and blood, levels of inflammation, and histol-
196, 223, 234, 251). For experimental otitis media, strains of ogy of lung tissue. Additionally, quantification of antibody ti-
serotypes 3, 4, 6, 7, 9, 11, 14, 19, and 23, which are the ones ters and antimicrobials performed in vaccine and drug phar-
most commonly isolated from children with acute otitis, have macokinetic studies, respectively, are also feasible. Two main
been used (18, 80, 81, 89, 101, 154). routes of infection are currently used to induce pneumonia: the
intratracheal (i.t.) and the intranasal (i.n.). The i.t. model re-
quires a complex and invasive technique for disease induction
ANIMAL MODELS OF PNEUMONIA
but offers the advantage of allowing 99% delivery of the bac-
S. pneumoniae is the leading causative agent of community- terial inoculum to the lungs (208). The i.n. route includes both
acquired pneumonia (159). In 2006, bacteremic pneumonia the standard aspiration method (50) and the aerosol nebulizer
constituted more than 66% of invasive pneumococcal disease system (176). The model of infection through i.n. aspiration is
in the United States (55). Typically, pneumococcal pneumonia the most commonly used, as it is fast and easy to perform
is lobar. However, other radiographic patterns are also com- without invasive surgical procedures and also because it mim-
monly found, including lobular bronchopneumonia and occa- ics the natural route of infection in humans. The i.n. aerosol
sionally interstitial or mixed types of pneumonia (127, 188). model instead requires an exposure chamber with a nebulizer,
The proportion of pneumococcal pneumonia associated with but it is suitable for the simultaneous infection of many mice.
bacteremia is estimated to be approximately 20 to 25% (7). A key aspect of i.n. aspiration models is the need for anesthesia
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 669

to allow infection of the lower respiratory tract (58), in com- (40). PsaA has also been reported to be unable to provide
parison to absence of anesthesia in both colonization (157, significant protection against i.p. challenge (180). In passive-
258) and i.n. aerosol (113, 176) pneumonia models. When immunization studies, capsular polysaccharide antibodies ob-
animal sedation is required, the choice of anesthetic should tained from vaccinated infants were also able to protect mice
also be evaluated for experimental design and result interpre- against lung infection (213). The protective efficacies of novel
tation. It has been reported that pentobarbital and halothane protein targets and live attenuated pneumococci (with dele-
have distinct effects on the pathogenesis of pneumococcal tions in genes encoding the capsule, pneumolysin, and PspA)
pneumonia in mice (207). have also been tested as vaccine candidates in this type of
i.t. models. The i.t. pneumonia model was first described by pneumonia challenge model (100, 205). Of importance for
Azoulay-Dupuis et al. to test the efficacies of different antibi- vaccine studies was also an early-life infection model (115). As

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


otics (12). It is based on an inoculation technique previously children younger than 2 years of age are major targets of
described for the study of pneumonia caused by Staphylococcus pneumococcal vaccination, the mouse model of i.n. infection
aureus and Klebsiella pneumoniae (78). Briefly, mice were sus- with aspiration set up for adult animals (50) was adjusted to
pended vertically on a board by supporting the lower incisors neonatal (1-week-old) and infant (3-week-old) mice, which
on a wire loop and retaining the lower incisors with a rubber mimic the immune systems of human neonates and infants,
band. The oropharynges were transilluminated, and the tra- respectively. By using this approach, several aspects of vacci-
cheas were cannulated with needles attached to microliter sy- nology in early life were investigated, including the efficacies of
ringes containing the pneumococcal suspension (12). This polysaccharide conjugate vaccines delivered by the mucosal
model is still used in several studies on drug efficacy, host route (115), the role of T-cell responses to pneumococcal con-
response to infection, and the role of pneumococcal virulence jugates (116), and the importance of maternal antibodies for
factors in the disease (1, 11, 19, 168, 169, 208, 237). Another i.t. protecting offspring against S. pneumoniae (204).
mouse model, based on pneumococcal inoculation via the oro- Other i.n. aspiration models of pneumonia have been devel-
pharynx, was developed by Iwasaki et al. to investigate the oped for mice rendered neutropenic after treatment with cy-
combined effects of bacteria and gastric juice commonly ob- clophosphamide: Ramisse et al. described the role of human
served for elderly patients with aspiration pneumonia (114). immunoglobulins in passive and active immunotherapy against
Finally, studies on neutrophil migration after induction of pneumonia (201), while Soriano et al. focused on the pharma-
pneumonia were carried out by injection of bacteria directly cokinetics of penicillins and cefotaxime in the disease (223).
into the exposed trachea (167, 232). While both systems described above employ immunosup-
All i.t. infection models have the advantage of allowing the pressed animals, Tateda et al. established an i.n. model of
delivery of the entire bacterial inoculum into the lower respi- penicillin-resistant pneumococcal pneumonia in immunocom-
ratory tract and directly causing pneumonia without interme- petent CBA/J mice (234). This experimental system represents
diate pathogenic steps. By using different mouse strains, it is the first example of penicillin-resistant pneumonia in uncom-
possible to induce either acute or subacute forms of disease promised mice, which are generally quite insensitive to infec-
(13). Swiss mice develop acute pneumonia and die rapidly (2 to tion by penicillin-resistant strains (169). The last model was
4 days), while C57BL/6 animals develop a subacute progressive employed both for therapeutic studies (229, 233) and for an-
disease leading to death in 8 to 10 days (13). alyzing the host inflammatory response during pneumonia
i.n. models. In the late 1980s, two studies demonstrated the (228).
importance of pneumolysin (25) and autolysin (24) in pneu- Finally, experimental pneumococcal pneumonia in mice can
mococcal virulence by using in vivo models, including an i.n. also be induced by the i.n. aerosol route using an exposure
pneumonia model based on bacterial instillation into the chamber and a nebulizer. The technique less closely resembles
mouse nostrils. However, the first detailed i.n. aspiration the development of pneumococcal pneumonia in humans,
model was described by Canvin et al. in 1995, who introduced where the disease generally follows aspiration of bacteria from
the practice of infecting animals with mouse-passaged pneu- the upper respiratory tract. The first example of aerosol-in-
mococci (Table 1) (50). Pneumococci recovered from the duced pneumococcal pneumonia originates from an early work
blood of intraperitoneally (i.p.) infected animals were inocu- by Coil et al., who described that splenectomized mice were
lated into the nostrils of anesthetized mice, which rapidly de- more susceptible to type 3 pneumococci delivered by means of
veloped bronchopneumonia with bacteremia (50). This model an aerosolized suspension (61). Another attempt at an i.n.
has been employed largely in studies on host immune response aerosol pneumonia model was made to analyze the develop-
to pneumococcal lung infection (23, 70, 122, 123, 130), mouse ment of pneumonia after airway obstruction in mice persis-
susceptibility and resistance to the disease (92), invasion by S. tently colonized by S. pneumoniae (113). This model intends to
pneumoniae of bronchoepithelial cells in vivo (124), the role of mimic the occurrence of pneumonia in humans with altered
virulence determinants in pneumonia (30, 58, 120, 125, 158), mucociliary clearance mechanisms, chronic obstructive pulmo-
and the efficacy of antibiotics and anti-inflammatory drugs nary disease, or concomitant viral infections (113). However, a
(250, 252). Furthermore, many investigations on protection more exhaustive model was established by Nuermberger et al.
from i.n. challenge after immunization with vaccine candidates (176). They used a low bacterial inoculum to generate a murine
have been performed. Pneumolysin (4), PspA (40, 257), PspC disease with a resemblance to human pneumonia that was
(15), and phosphorylcholine (241) have successfully been em- closer than those seen for traditional mouse i.n. and i.t. mod-
ployed in vaccine studies followed by i.n. challenge. Whereas els, which rely on larger doses to produce fulminant infections
PsaA has been able to confer protection against colonization due to concomitant sepsis (12, 50). This model is especially
(37, 183), it did not elicit protection against focal pneumonia suitable for evaluating the efficacy of antimicrobials at the site
670 CHIAVOLINI ET AL. CLIN. MICROBIOL. REV.

TABLE 2. Rat models of pneumonia


Route of Pneumococcal
Rat strain Aim(s) of study Readouts Utility Reference(s)
infection serotype(s)

i.t. R strain albino 3 Antimicrobial therapy, CFU, histology Pathogenesis of lobar 14


model description pneumonia, drug
efficacy studies
i.t. Sprague-Dawley 3 Model in cirrhotic and Survival, CFU, Pneumonia in 71, 166
chronically hematology and alcoholic and
intoxicated rats chemistry tests cirrhotic patients
Intrabronchial Wistar 3 Effect of pneumolysin Lung histology Pathogenesis of lobar 79

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


on lungs pneumonia,
detailed surgery
information
Intrabronchial Infant CD 6 Model of penicillin- Gross pathology and Drug efficacy studies 221
resistant pneumonia CFU
in neutropenic rats
Intrapulmonary CD 2, 6B Model of penicillin- CFU, lung histology, Antibiotic efficacy 49
resistant pneumonia pharmacokinetics studies
in neutropenic rats
Intrapulmonary Infant Sprague- 1, 3, 4, 5, 6B, 7F, Low-dose pneumonia Survival, CFU, lung Pathogenesis of 214
Dawley 9V, 14, 19F, model histology pneumonia in
23F children, vaccine
studies

of infection rather than in the bloodstream, as the experimen- models of pneumococcal pneumonia have been set up with
tal approach allows the development of an indolent subacute these animals.
pneumonia (175). The main disadvantage of this model is the Table 2 summarizes the principal characteristics of rat mod-
use of neutropenic mice, which might not optimally represent els of pneumococcal pneumonia. Main readouts include ani-
the human host (175, 176). mal survival, observation of gross pathology, lung histology,
Remarks on murine pneumococcal pneumonia. i.t. models and bacterial counts in lungs and blood. The rat has been
generally cause lobar pneumonia (12), while systems based on employed mainly for clarifying features of pneumonia occur-
i.n. aspiration of bacteria induce bronchopneumonia (50). In ring in asplenic, neutropenic, cirrhotic, or alcoholic subjects,
both cases, there are concomitant pneumonia and sepsis: in- thus representing a valuable experimental system for studying
fection readily leads to death of animals, which most likely pneumonia in the immunocompromised host. Alcoholism is a
succumb to septic shock rather than to pulmonary disease. predisposing factor for the development of pneumococcal
Clearly, models of focal pneumonia in the absence of gener- pneumonia (140), and mortality rates from bacteremic pneu-
alized sepsis are more helpful to study the pathogenesis of monia double (up to 40%) in alcoholic patients with hepatic
pulmonary disease, vaccine protection, and antibiotic efficacy insufficiency (8). The models in ethanol-intoxicated (71) and
exclusively in the lungs. In addition, they better reproduce the cirrhotic (166) rats were developed over 15 years ago by using
clinical manifestations in humans, where pneumonia is usually the i.t. route of infection to deliver pneumococci into the left
not associated with bacteremia (7). The use of pneumococcal lobe of the lung following intubation of the main stem bron-
strains of serotypes 14, 19, and 23, which are poorly virulent in chus (14). Infection caused lobar pneumonia and sepsis (14).
mice, is a necessary requisite for causing this type of disease. So The model in ethanol-intoxicated animals was based on con-
far, type 19 strains have been employed to induce progressive tinuous ethanol feeding leading to chronic intoxication (71),
focal pneumonia without sepsis (40, 234). Alternatively, neu- while experimental cirrhosis was induced in rats by adminis-
tropenic mice infected with low doses of virulent pneumococ- tration of a hepatotoxin that caused cirrhosis and ascites (166).
cal strains (e.g., type 3) also develop primary pneumonia with In both cases, rats showed increased susceptibility to pneumo-
delayed dissemination (approximately 4 days after infection) coccal pneumonia. These models have been used by the same
(176). research group in studies on phagocytosis and antibiotic effi-
cacy (84, 184, 199). A sophisticated surgical method based on
Rat and Rabbit Models the delivery of bacteria into the apical lobe bronchi of adult
rats was described in the early 1990s (79). Briefly, rats were
Rat and rabbit models are uncommonly employed compared anesthetized and intubated before the thoracic cage was
to the mouse. Their larger size allows the collection of more- opened to reveal the apical lobe of the lung. The apical lobe
substantial samples, but one drawback is that experimental bronchus was exposed and partially sutured to impair muco-
groups are normally smaller, thus potentially compromising ciliary clearance, and the inoculum was injected through the
statistical significance. Sophisticated surgical procedures are bronchial wall into the lumen toward the lung periphery (79).
required to induce pneumonia via i.t., intrabronchial, or in- Histological examination of rat lungs showed clear consolida-
trapulmonary routes of infection, and to our knowledge, no i.n. tion of the apical lobes (lobar pneumonia) (79). Two other
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 671

TABLE 3. Rabbit models of pneumonia


Route of Pneumococcal
Rabbit strain Aim of study Readouts Utility Reference
infection serotype(s)

i.t. New Zealand 2, 3 Inflammation induced by CFU, protein content, Disease pathogenesis, efficacy 246
White cell wall leukocytes in lungs of antimicrobial and
adjunctive therapy
Intrabronchial New Zealand 9, 19 Model of penicillin- Survival, CT scanb, lung Efficacy and 196
White resistant pneumonia histology, pharmacokinetics of drugs
pharmacokinetics (model of human therapy)
Intrabronchial New Zealand NSa Ventilator-associated CFU, lung histology Pneumonia in ICUc 57

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


White pneumonia
Intrapulmonary New Zealand NS Neutrophil activity in FDGd uptake in lungs Acute and chronic lung 119
White lungs inflammation in
pneumonia
a
NS, not specified.
b
CT scan, computerized tomography scan.
c
ICU, intensive care units.
d
FDG, 18F-labeled fluoro-2-deoxy-D-glucose.

models of lobar pneumonia induced either intrabronchially in approach, analyses of the roles of pneumococcal cell surface
infant rats (221) or intrapulmonarily in adult rats (49) ren- components in the induction of pulmonary inflammation
dered neutropenic were employed to study the efficacy of an- (246), of host oligosaccharides in preventing pneumococcal
tibiotics against penicillin-resistant lobar pneumonia (49, 221, colonization of the nasopharynx and subsequent lung infection
222). As already described for the mouse, the use of immuno- (110), and of the platelet-activating factor in the pathogenesis
compromised animals is necessary to produce pneumonia by of pneumonia (47) were performed. Two further models were
penicillin-resistant strains of S. pneumoniae in rats. The above- developed for immunocompetent rabbits based on intrabron-
described infant rat model is based on a nonsurgical intrabron- chial instillation of bacteria (57, 196). The former reproduces
chial instillation of bacteria resuspended in cooled melted agar ventilator-associated pneumonia (57), while the latter mimics
(as an adjuvant and growth medium) (221). Another group penicillin-resistant pneumonia (196). In the model of antibiot-
developed a refined intrapulmonary pneumonia model for in- ic-resistant pneumonia, pathology—if untreated—was charac-
fant rats based on the nonsurgical inoculation of a low bacte- terized by initial unilobar lesions and subsequent consolidation
rial inoculum (1 to 400 CFU per rat) (214). The right chests of of other lobes and confluent bronchopneumonia with bactere-
pups were prepared with ethanol, and rat-passaged bacteria mia (196). The system mimics lethal pneumococcal pneumonia
were delivered transthoracically into the mid-right lungs. Prior in patients and allows simulation in the rabbit of human anti-
to inoculation, pneumococci were entrapped in cooled agar biotic pharmacokinetics. As this model simulates “human-like
particles to provide both protection from clearance and adju- treated pneumonia,” it constitutes an extremely valuable tool
vant activity. The virulences of 10 different serotypes prevalent for assessing drug efficacy and pharmacokinetics in humans
in children were compared (214). Despite being a model of (67, 195, 196), and it may also be useful for evaluating the
focal pneumonia, it differs from the one described for the selection of resistant pneumococcal strains in vivo (68). As
mouse by Briles et al. (40) in that experimental pneumonia aforementioned, the same research group also developed a
may be accompanied by bacteremia and occasionally by men- novel infection model based on the induction of ventilator-
ingitis, mimicking the disease seen for children of developing associated pneumococcal pneumonia in adult rabbits (57). This
countries. The model was utilized in both neonatal (4-day-old) approach proved to be instrumental to assess antibiotic efficacy
and infant (3-week-old) rats to assess passive protection against pneumonia associated with mechanical ventilation,
against intrapulmonary challenge with S. pneumoniae (155, which is a serious health concern in intensive care units (56).
156, 224). Finally, positron emission tomography was also exploited to
The rabbit has also been employed to study pneumococcal study intrapulmonary experimental pneumonia localized in the
pneumonia, although fewer models are available. Readout pa- upper lung lobes of rabbits (119).
rameters include animal survival, determination of leukocyte
numbers in lungs and blood, histology of the lungs, and assess- ANIMAL MODELS OF SEPSIS
ment of drug concentration in serum (Table 3). All models
described below are based on i.t., intrabronchial, or intrapul- The definition of sepsis and related terms (e.g., bacteremia,
monary routes and are useful systems to study disease patho- septicemia, severe sepsis, septic shock) has been controversial.
genesis and perform drug efficacy studies. However, all of them In 1989, sepsis was defined as the host systemic response to
necessitate surgical expertise for disease induction. An early i.t. infection according to specific measurements of clinical param-
model was established in 1987 and it is based on the instillation eters (e.g., blood leukocyte counts, body temperature, and
of pneumococci in rabbits by means of a catheter inserted into heart and respiratory rates), in contrast to bacteremia, which is
the animal trachea (246), as previously described for experi- defined only according to the presence of bacteria in the blood
mental pneumonia caused by S. aureus (185). By using this (33). S. pneumoniae is an important cause of sepsis in humans,
672 CHIAVOLINI ET AL. CLIN. MICROBIOL. REV.

TABLE 4. Mouse models of sepsis


Route of Pneumococcal
Mouse strain(s) Aim(s) of study Readouts Utility Reference
infection serotype

i.v. CBA/N, DBA/2, 3 Protection by anti-PCa Survival, CFU Bacterial clearance 43


BALB/c antibodies, model mechanisms, passive and
description active protection
i.v. CD-1 3 Host response to sepsis CFU, hematological Pathogenesis of sepsis 251
parameters,
cytokines,
histology

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


i.p. CBA/N, BALB/c 3 Protection by anti-PC Survival, CFU Passive and active protection, 43
antibodies, model drug efficacy and virulence
description studies
a
Anti-PC, anti-phosphocholine.

generally occurring as a form of an advanced stage of pneu- tail vein, researchers have used an infrared lamp to allow
monia (60). Predisposing conditions are splenectomy (219), vasodilatation (109). i.p. inoculation is technically easier but
hepatic cirrhosis, respiratory insufficiency, and cardiovascular presents disadvantages including the risk of causing tissue
disease (140). In children of less than 3 years of age, a common damage or pain to the animal and may not allow the study of
clinical manifestation is occult bacteremia (without a focus) bacterial clearance by the reticuloendothelial system (38).
(153). In discussions of experimental animal models developed Briles et al. observed that certain pneumococcal strains that
to study pneumococcal disease, the terms sepsis and bactere- were lethal following injection of mice by the i.p. route were
mia are commonly found and used interchangeably. Experi- avirulent by the i.v. route, indicating that i.p.-induced sepsis is
mental sepsis by S. pneumoniae can be generated either by much more severe (38, 43). For this reason, the i.p. route is
inoculating bacteria directly into the bloodstream or by injec- widely used to mouse passage pneumococci in order to render
tion into the peritoneal cavity. While sepsis induced by the i.p. them more virulent (50).
route is secondary to peritoneal infection, with the concomi- Studies performed by the group of Briles et al. in the early
tant strong peritoneal inflammation, intravenous (i.v.) inocu- 1980s employed the i.v. mouse sepsis model mainly for vaccine
lation produces a “cleaner” model, where the clearance of studies. These works largely contributed to demonstrate S.
bacteria by the reticuloendothelial system can be easily stud- pneumoniae clearance from the bloodstream promoted by nat-
ied. In addition, i.p. infection represents neither an important urally occurring anti-phosphocholine antibodies (43, 163, 261).
clinical manifestation of invasive pneumococcal disease nor The model was employed and is still used by this group and
the natural route of infection by S. pneumoniae in humans. others, resulting in several investigations on the role of pneu-
mococcal virulence factors during sepsis and in studies aimed
Mouse Models at analyzing the efficacy of vaccines (15, 20, 30, 38, 42, 109, 131,
186, 200, 202, 227, 245). For instance, thanks to the use of the
Besides a few early studies carried out in guinea pigs to
i.v. infection route, it was possible to unambiguously establish
analyze the role of spleen and complement in experimental
the role of PspC as a key virulence factor in pneumococcal
pneumococcal sepsis (44, 45), the mouse is the most exten-
sepsis (109). In a work by Loeffler et al., the therapeutic value
sively used animal model for inducing sepsis by S. pneumoniae.
of a novel antimicrobial, the Cpl-1 phage enzyme, was proven
Most research groups have analyzed the occurrence of murine
sepsis principally by determining the presence of bacteria in by using an i.v. mouse sepsis model (149). One of the most
the blood and by observing animal survival after infection detailed studies of the pathogenesis of sepsis in the mouse was
(Table 4). Sepsis in the mouse can be induced by either the i.p. made by Wang et al., who investigated the systemic host re-
or the i.v. route of infection. Sepsis can also develop secondary sponses to sepsis in normal and immunosuppressed mice in-
to pneumonia induced by i.n./i.t. routes or to meningitis fol- fected by the i.v. route (251). A complete analysis of different
lowing intracranial injection, but the kinetics of bacterial tran- parameters, such as bacterial loads in blood and lungs, lung
sit from lungs or brain into the bloodstream cannot be con- injury, biochemical markers, and hematological and inflamma-
trolled experimentally. Consequently, performing viable tory mediators, was carried out (251).
counts in the blood after i.n./i.t. or intracranial inoculations The i.p. route of infection is one of the earliest techniques
does not constitute a direct measure of bacterial clearance employed to induce experimental sepsis. This method was used
operating in those districts and it is not the finest approach to by Briles et al. in early studies of blood clearance mechanisms
evaluate immune mechanisms and efficacy of drugs or vaccines (43). i.p. challenge models are still widely employed in studies
in experimental pneumonia or meningitis. As above men- on the efficacies of vaccine candidates, including the surface
tioned, the i.v. model is a direct and valuable system for in- proteins PspA, PspC, and PsaA; a toxoid derivative of pneu-
ducing infection in the bloodstream and thus for studying the molysin (PdB); the caseinolytic protease (ClpP); the compo-
mechanisms of bacterial clearance, although the technique can nents of two iron uptake transporters, PiaA and PiuA; and the
be time-consuming and difficult to perform compared to i.p. more recently discovered pilus protein subunits (46, 51, 87,
infection. To facilitate i.v. injection of bacteria into the mouse 118, 180, 181). The i.p. model also proved to be useful to clarify
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 673

TABLE 5. Rat and rabbit models of sepsis


Route of Pneumococcal
Animals Aim(s) of study Readouts Utility Reference
infection serotype

i.v. Sprague-Dawley 25 Model description for Survival, CFU Sepsis in 147


rats splenectomized rats immunocompromised
hosts
i.v. Sprague-Dawley 3 Role of complement in Survival, CFU Sepsis in cirrhotic 2
rats cirrhotic rats patients
i.p. New Zealand White 1 Role of lysosomes in Lysosomal enzymes in plasma Clinical and metabolic 98
rabbits sepsis and organs, CFU, vital parameters during

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


parameters sepsis
Intracardiac New Zealand White 3 Role of spleen in CFU, splenic mass, and blood Sepsis in 106
rabbits clearance, model flow splenectomized
description patients
i.v. Chinchilla rabbits NSa Pharmacokinetics of CFU, drug concentrations in Drug efficacy studies 82
cephalosporins plasma
a
NS, not specified.

the role of pneumococcal virulence factors in systemic infec- similar experimental strategy was applied to investigate pneu-
tions (182, 262) and to evaluate efficacy of antibiotics (53, 141). mococcal sepsis in cirrhotic rats (2). Patients with alcohol-
The i.p. sepsis model was also employed to assess passive induced hepatic cirrhosis are predisposed to pneumococcal
protection conferred by a mouse hyperimmune serum (263) infection, partially due to decreased liver production of com-
and by human anticapsular (117) and anti-pneumolysin anti- plement factors. In this model, cirrhotic rats are also hypo-
bodies (171). Passive immunization with anticapsular antibod- complementemic compared to healthy animals (2, 166). By
ies in infant (13- to 15-day-old) mice demonstrated that anti- using this approach, a detrimental role of pneumolysin was
capsular opsonophagocytic titers are better predictors of observed in both depleting complement components and re-
protection than are enzyme-linked immunosorbent assay im- ducing serum opsonic activity (3), resulting in enhanced pneu-
munoglobulin G titers (117). Very few studies focused on the mococcal virulence in the cirrhotic host (2). To our knowledge,
host response to pneumococcal sepsis by describing the func- there are no reports of i.p. models of sepsis in the rat.
tion of tumor necrosis factor alpha in controlling systemic One of the first models of pneumococcal sepsis in the rabbit
infection (22, 178) and increasing the permeability of the was described in 1970 by Guckian et al., and it represents a fine
blood-brain barrier (BBB) (244). example of analysis of clinical parameters (e.g., body temper-
ature, cardiac output, arterial blood pressure) detected during
Rat and Rabbit Models the course of this disease (98). Infection was established by the
i.p. route, and the model was employed to analyze the roles of
Experimental models of sepsis in the rat and the rabbit have lysosomes and cathepsin inhibitor in plasma (98), metabolic
been used mainly from the beginning of the 1970s throughout alterations in the course of disease (96), and the effects of
the 1980s, but presently they have mostly been replaced by the splenectomy and drug therapy in the disease (97). An intra-
mouse due to the latter’s greater ease in manipulation and cardiac model of experimental sepsis in New Zealand White
smaller size. Similarly to that induced in the mouse model, rabbits was utilized to investigate the key role played by the
sepsis induced in the rat and rabbit models is studied by ob- spleen during the disease (106). This represents another ex-
serving animal survival over time and by determining the pres- ample of refined surgery and measurements of clinical param-
ence and number of bacteria in the blood (Table 5). As in the eters in the rabbit in the course of infection. Effects of
case of rat models of pneumococcal pneumonia (Table 2), rat hemisplenectomy, total splenectomy, spleen transplant, and
models of sepsis are particularly valuable for studying the dis- spleen repair on the clearance of S. pneumoniae during exper-
ease in immunocompromised animals. The first model of sep- imental sepsis were evaluated (62, 106). Finally, an i.v. model
sis in the rat was set up in 1972 by Leung et al., who infected of sepsis was established in chinchilla rabbits to study the
splenectomized rats by injecting pneumococci into the tail pharmacokinetics of cephalosporins (82).
veins (147). Despite being an early work, the authors made
significant observations, including the importance of (i) the use
ANIMAL MODELS OF MENINGITIS
of i.v. rather than i.p. challenge to show a difference between
normal and immunocompromised animals in susceptibility to S. pneumoniae is one of the major infectious agents respon-
infection and (ii) vaccination to protect splenectomized rats sible for acute bacterial meningitis, which can be fatal in 5% to
from fatal sepsis (147). The model was also employed to eval- 40% of patients (137, 146). Up to 30% of survivors suffer from
uate the efficacies of penicillin and steroid drugs (99), of hu- neurological sequelae (e.g., learning, hearing, and memory im-
man gamma globulins (179), and of a type 6 capsular polysac- pairment, seizures, and motor deficits) due to brain damage
charide vaccine (111) to treat and/or prevent pneumococcal (146). In patients with meningitis, brain injury can manifest
postsplenectomy sepsis. Starting from a previously described with several patterns, including vasculitis, cortical necrosis, and
model of pneumococcal pneumonia in cirrhotic rats (166), a neuronal apoptosis in the hippocampus (137, 146). Develop-
674 CHIAVOLINI ET AL. CLIN. MICROBIOL. REV.

TABLE 6. Mouse models of meningitis


Pneumococcal
Route of infection Mouse strain(s) Aim of study Readouts Utility Reference
serotype(s)

Intracerebral (ventricular) CD-1 NSa Drug efficacy CFU in brain and Therapeutic studies 220
organs
Intracerebral (right C57BL/6 3 Model description Immunohistochemistry, Meningitis pathophysiology, 86
forebrain) CFU, clinical scores, drug and virulence
behavioral tests studies
i.cist. (cisterna magna) C57BL/6 3 Role of eNOSb Survival, clinical scores, Meningitis pathophysiology, 136
cytokines, and hearing loss, therapeutic

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


histology studies
Intracerebral MF1 2, 3, 4 Model description Histology, survival, Virulence studies, host- 59
(subarachnoidal) CFU parasite interaction
i.cist. (cisterna magna) Infant BALB/c 3 Model of brain Survival, CFU in CSF, Meningitis in children, 95
C57BL/6, damage histology, and brain damage
CD-1 apoptosis
i.p. Infant C57/129 6 Role of ICAM-1 Survival, CFU in Hematogenous meningitis, 230
organs, cytokines drug studies
i.n. Swiss 6 Model description Brain histology, CFU Pathogenesis via natural 268
in CSF, cytokines infection, drug studies
i.n. Swiss 2 Model description CFU in brain and Disease pathogenesis via 160
other organs natural infection
a
NS, not specified.
b
eNOS, endothelial nitric oxide synthase.

ment of meningitis generally initiates from the colonization of analyzing different parameters, including animal survival; as-
the nasopharynx by S. pneumoniae, which reaches the lungs sessment of clinical scores; viable bacterial counts in the brain,
and then invades the bloodstream with subsequent crossing of cerebrospinal fluid (CSF), and other organs; histological anal-
the BBB (146). Meningitis can also be caused either by con- ysis of the brain tissue; and determination of leukocyte and
tiguous spread of pneumococci infecting the sinuses or the ME cytokine levels in CSF and serum (Table 6). One disadvantage
or by accidental traumatic inoculation of bacteria into the presented by the mouse model is the difficulty of collecting
central nervous system (CNS) (137). Experimental animal CSF samples (only up to 10 ␮l can be obtained) due to the
models are essential tools to study the pathogenesis of menin- small size of the animal (52).
gitis and the efficacies of different drugs against the disease. If Direct induction of meningitis. The meningitis model of
the mouse is a key laboratory animal for the induction of Gerber et al., described in 2001 and based on a previously
experimental pneumonia and sepsis, rabbits and rats represent established Listeria monocytogenes meningoencephalitis model
the counterparts for the study of experimental meningitis. (218), is an attractive system for analyzing pneumococcal men-
ingitis following the infection of inbred mice into the right
Mouse Models frontal lobe of the brain (86). Meningitis was studied by per-
forming bacterial counts for different organs, histology of the
As previously mentioned, the mouse has become an exper-
brain, clinical score calculations, and behavioral tests. This
imental system for studying pneumococcal meningitis only re-
model has been employed to study the efficacy of rifampin
cently, while most studies between the mid-1970s and the 1990s
against meningitis (173) and has also been used for analyzing
were carried out in the rat and the rabbit. Two major types of
S. pneumoniae murine meningitis models exist: (i) direct infec- host and pneumococcal factors involved in disease pathogen-
tion by the intracerebral or the intracisternal (i.cist.) route and esis (35, 76, 198, 255). In another work by the same research
(ii) infection induced by the i.p. or i.n. route. Direct bacterial group, mice surviving pneumococcal meningitis presented with
inoculation into the CNS mimics the contiguous spread of clear deficits in motor skills, spatial memory, and learning
bacteria from the nasopharynx or the ME or the inoculation of when subjected to behavioral tests (e.g., Morris water maze
pneumococci into the brain due to trauma. This system allows test, tightrope test) (254). Therefore, this mouse model also
the study of host-pathogen interactions once infection is estab- enables the assessment of postinfectious sequelae (254). The
lished in the meninges but does not enable the analysis of the induction of experimental meningitis in the mouse was also
different pathogenic steps occurring from colonization to dis- accomplished by inoculating pneumococci directly into the cis-
ease in the CNS (137). In contrast, meningitis induced via i.n. ternae magnae (i.cist.) of anesthetized C57BL/6 mice (136). By
or i.p. routes is useful for the analysis of pathogenesis accord- use of this model, Koedel et al. described the role of nitric
ing to the natural way of infection but has the drawback that oxide synthase in the disease, and the same system has been
approximately 50% of infected animals will die due to sepsis used to study innate immune responses to infection, including
without ever developing meningitis (243, 268). Characteriza- cytokine expression patterns and the role of caspase-1, MyD88,
tion of meningitis in the mouse model has been performed by and complement factors (133, 134, 136, 138, 209). As all mice
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 675

infected by the i.cist. route presented with hearing loss, this animals developed the disease (268). The same research group
system allowed the establishment of the first experimental ex- largely used the above-described model to study the role of
ample of pneumococcal meningitis-associated hearing loss in interleukin-1 (IL-1), IL-10, and IL-18 in the disease (264, 266,
the mouse (133). The two models described above finely re- 267). Recently, the model was also employed to assess the
semble pneumococcal meningitis in humans and offer the op- therapeutic effects of the inhibitor of complement factor C1,
portunity to study the pathophysiologic changes during men- which was effective at promoting bacterial clearance and re-
ingitis, the effects of drug administration, and postinfection ducing both mouse illness and inflammation in the CNS (265).
neurological sequelae. Finally, the model described by Marra and Brigham is also
Most mouse models of meningitis employ inbred mice, but based on i.n. infection of Swiss mice with pneumococci (with-
studies employing outbred strains also exist. Shapiro et al. out hyaluronidase) (160). The course of meningitis and bacte-

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


induced meningitis by the intracerebral-ventricular route in rial counts in brains, lungs, and blood following infection with
outbred CD-1 mice and studied the therapeutic efficacy of different challenge doses were evaluated in that study (160).
clinafloxacin (220). Another model of meningitis in outbred Nonhematogenous meningitis. A study conducted by van
MF1 mice was developed by our research group by inoculating Ginkel et al. reported a novel in vivo route for pneumococci to
S. pneumoniae strains of different capsular serotypes via the enter the CNS along olfactory nerves: nasal colonization by S.
intracranial-subarachnoidal route (59). The establishment of pneumoniae resulted in bacterial entry into the brain via ret-
meningitis was confirmed by histological analysis of the brain, rograde axonal transport without causing bacteremia (249). To
and animal survival and bacterial counts in different organs our knowledge, this is the first report describing a nonhemat-
were also evaluated. Because levels of disease development ogenous meningitis caused via i.n. infection of mice.
and outcomes were comparable among strains of serotypes 2,
3, and 4 (59), this model may be instrumental for screening the Rat and Rabbit Models
virulences of different serotypes and possibly for assessing at-
tenuation of isogenic mutants. The model is based on a mod- Rat and rabbit models are still currently employed by several
ified version of a method describing the induction of crypto- research groups to study pneumococcal meningitis. They rely
coccal meningoencephalitis in the mouse (28). Recently, a on the induction of disease via the i.cist. route, with the excep-
novel model of brain damage in pneumococcal meningitis has tion of two models of either hematogenous or otologic men-
been described for infant (11-day-old) mice of both inbred ingitis developed in the rat (206, 253). As already mentioned
(C57BL/6, BALB/c) and outbred (CD-1) strains: all mice in- for the pneumonia models in rats and rabbits, more-sophisti-
fected via the i.cist. route rapidly developed the disease and cated surgical procedures are here employed to cause the dis-
some animals also exhibited brain damage (95). Among the ease; however, animal size has the advantage of allowing large
mouse strains tested, BALB/c mice showed the largest suscep- samples of and multiple sampling for CSF and blood compared
tibility to infection, while C57BL/6 mice were the most resis- to what is possible with the mouse model. Except for an oto-
tant and the ones with major neuronal injury both in the cortex logic model (253), none of the methods used to inoculate these
and in the hippocampus. The model was established from animals mimic the natural route of infection occurring in hu-
previous studies of brain injury to infant rats (143) and is mans, but they enable the study of disease once infection is
promising for the study of neuronal damage in children (95). established in the meninges (Table 7). The diseases induced in
Hematogenous meningitis. Tan et al. developed one of the the two animal species differ in some aspects, including brain
first i.p. meningitis model for inbred infant mice in order to damage. Apoptosis in the dentate gyrus and cortical necrosis
study the role of intercellular adhesion molecule 1 (ICAM-1) are the predominant features of damage in the rabbit (269)
in this disease (230). Similarly, two additional murine models and in the adult rat (103), respectively. Conversely, the infant
of meningitis via the i.p. route were described later on by rat is unique, as it reproducibly manifests both hippocampal
Iizawa et al., who analyzed the efficacy of cefozopran against apoptosis and cortical necrosis (143), similar to what is ob-
the disease (112), and by Tsao et al., who focused on elucidat- served for patient brain damage (137, 146).
ing the role of tumor necrosis factor alpha in regulating the i.cist. models for rats. Both infant and adult rats have been
opening of the BBB during experimental meningitis (243). In employed extensively in different studies on pneumococcal
the latter model, antibiotics were given soon after infection to meningitis and represent valuable tools for studying disease
avoid mouse death due to septic shock before the development characteristics, host immune response to infection, and efficacy
of meningitis. of antimicrobials and anti-inflammatory drugs (Table 7). In-
The model of meningitis induced by the i.n. route mimics the fant (6- to 12-day-old) rats are generally more susceptible than
natural way of infection. The work of Kostyukova et al. repre- adults to pneumococcal infection, and despite their smaller
sents one of the first attempts to analyze the roles of different size, repeated collection of CSF and blood is still feasible. As
pneumococcal virulence factors (e.g., hyaluronidase, capsule, aforementioned, another advantage of infant compared to
and pneumolysin) in the pathogenic steps leading to meningitis adult rats is the possibility of inducing brain damage both in
following i.n. infection (139). However, the first detailed model the hippocampus (apoptosis) and in the cortex (necrosis)
of hematogenous meningitis should be accredited to a work (143). Thus, the infant rat represents a robust and reliable
conducted by Zwijnenburg et al., who infected Swiss mice via laboratory animal to study brain damage (27, 143, 145).
the i.n. route using a mixture of bacteria and purified pneu- In the early 1990s, the i.cist. infant rat meningitis model was
mococcal hyaluronidase to favor systemic invasion from the used by different groups, mainly for therapeutic studies (242,
nasopharyngeal mucosa (268). As aforementioned, the disad- 259). In 1990, Tsai et al. established an important i.cist. model
vantage of this system relies in the fact that only 50% of in infant Sprague-Dawley rats for the study of cefepime phar-
676 CHIAVOLINI ET AL. CLIN. MICROBIOL. REV.

TABLE 7. Rat and rabbit models of meningitis


Route of Pneumococcal
Animal Aim(s) of study Readouts Utility Reference(s)
infection serotype(s)

i.cist. Infant Sprague- 3 Drug efficacy, MMPa in Drug pharmacokinetics, Therapeutic studies, host 145, 242
Dawley rat brain damage histology, CFU, immune response,
MMP in CSF brain damage
i.cist. Wistar rat 3, 6 Microvascular changes ICPb, rCBFc, brain Meningitis 193
edema pathophysiology
i.cist. Wistar rat 2 Damage to brain Ciliary beat frequency, Virulence studies, host 103
ependyma CFU, CSF response to infection,

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


parameters, SEMd of brain damage
brain
i.p. Infant Sprague- 3 Pathogenesis of inner CFU in blood and CSF, Meningitis secondary to 206
Dawley rat ear invasion histology of ME and ear infection, drug
brain efficacy studies
Otologic, i.p. Wistar rat 2 Cochlear implantation in Gram staining, Post-cochlear 253
meningitis histology, CFU implantation
counts meningitis, therapeutic
studies
i.cist. New Zealand White 3 Model description, drug ␤-Lactam Disease pathophysiology, 69
rabbit study concentrations in host response, drug
CSF and virulence studies
i.cist. New Zealand White 3 Hearing assessment Hearing loss, histology Hearing loss due to 26
rabbit of inner ear and meningitis, drug
brain efficacy
a
MMP, metal metalloproteinases.
b
ICP, intracranial pressure.
c
rCBF, regional cerebral blood flow.
d
SEM, scanning electron microscopy.

macokinetics (242). However, the inoculation technique was parameters reached stable baseline levels, meningitis was in-
fully described later in studies conducted by Leib et al. (145) duced by injecting live pneumococci in a volume of 75 ␮l (192,
and is based on a previously described model for meningitis 193). The method was instrumental to assess the detrimental
caused by group B streptococci (132). Briefly, pups were in- effect of the oxidant peroxynitrite in the disease (128) and the
fected i.cist. with 10 ␮l of bacterial suspension by use of a function of transforming growth factor ␤2 in suppressing
32-gauge needle and then monitored for seizures due to injec- cerebrovascular alterations and brain edema in early experi-
tion (132, 145). Development of meningitis was proven by mental meningitis (192). Another adult rat i.cist. model was
culturing bacteria present in the CSF (132, 145). The menin- developed by Hirst et al., who investigated the effects of pneu-
gitis model in the infant rat has largely been used by Leib and mococcal infection on cerebral ciliated ependyma during ex-
coworkers in studies on the involvement of matrix metallopro- perimental meningitis. An accurate description of the animal
teinases (143, 145, 165) and endothelin (194) in neuronal surgical preparation is given, based on the implantation of a
damage. By means of this model, the efficacies of several ther- 19-gauge cannula into the cisterna magna, which allowed both
apeutic approaches against meningitis and/or postinfectious pneumococcal infection and CSF sampling during animal fol-
sequelae, including antioxidants (6), adjunctive therapy drugs low-up. A detailed report on treatment and analysis of CSF,
(27, 143, 144, 194), and antimicrobials such as daptomycin (94) blood, and brain tissue is also provided (103). This technique
and phage lysins (93), have been evaluated. It is interesting was successfully employed to show that S. pneumoniae caused
that in this experimental system, the use of dexamethasone as loss of and damage to ependymal cells and ciliary functions,
an adjuvant therapy increased hippocampal cell injury and which may contribute to several neuropathological aspects of
reduced the learning capacities of rats (144). More recently, meningitis (103). Recently, the same research group also
the same model was also employed to analyze the key role of showed that both autolysin and pneumolysin play key roles in
choline in the pathogenesis of meningitis and brain damage the pathogenesis of pneumococcal meningitis (104).
(83). Hematogenous and otologic models for rats. Hematogenous
The adult rat model of pneumococcal meningitis was origi- and otologic models for rats were established to study menin-
nally described in detail in a work carried out to study the gitis secondary to sepsis or otitis media. In the model of Ro-
microvascular alterations during the establishment of menin- driguez et al., infant Sprague-Dawley rats were inoculated by
gitis (193). Wistar rats were anesthetized, tracheotomized, and the i.p. route, and some of the animals infected developed
artificially ventilated before the insertion of catheters into the meningitis (about 50%) and cochlear inflammation (20 to
cisternae magnae. A craniotomy was then performed in the 80%). This model may be useful to study the pathogenesis of
right parietal bone to place a probe for measuring regional inner ear invasion by S. pneumoniae during hematogenously
cerebral blood flow and intracranial pressure, and once these acquired experimental meningitis (206). Recently, another
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 677

TABLE 8. Animal models of otitis media


Route of Pneumococcal
Animal(s) Aim(s) of study Readouts Utility Reference
infection serotype(s)

Intratympanic Chinchilla 23 Model description, Exudation in ME, Disease pathogenesis, 89


antibody antibody titers, host response, vaccine
response CFU and drug studies
Intrabullar Mongolian gerbil, 3 Model description Otoscopy, IEa Antibiotic efficacy and 81
chinchilla histology, CFU kinetics, vaccine and
virulence studies
Intratympanic Guinea pig 23F Model description CFU in ME fluid, Drug efficacy studies, 239

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


ME histology disease pathogenesis
Intrabullar Sprague-Dawley 3, 6A Model description Survival, CFU in ME Evaluation of drugs and 101
rat and blood vaccines
i.n. Rivm:WU (CBP) 14 Model description, ME histology, TEMb, Immunization and 248
rat vaccine study antibody titers correlates of
protection in a
natural-route model
Intrabullar BALB/c, C57BL/6 3, 6, 9, 19 Model description CFU in blood and Host inflammatory 164
mice ear, survival, response, drug and
otomicroscopy vaccine studies
i.n. BALB/c mice 19F Prevention of Bacterial Otitis media in children, 162
otitis media by multiplication in vaccine studies
phage lysin vivo
a
IE, inner ear.
b
TEM, transmission electron microscopy.

model was set up in adult rats inoculated via both otologic standing of several aspects of pneumococcal meningitis, in-
(inner ear and ME) and i.p. routes of infection to establish cluding bacterial replication in the CSF, host defenses and
whether cochlear implantation increases the risk of pneumo- inflammation, BBB alteration, and also the systemic effects of
coccal meningitis (253). The use of three infection routes cov- meningitis on animal respiration and circulation (236). Numer-
ers the potential routes used by S. pneumoniae to reach the ous studies have been undertaken to evaluate antibiotic effi-
CNS: direct spread from the inner ear, contiguous or hema- cacy (34, 64, 73, 85, 172, 189, 203, 226, 231) and the role of host
togenous spread from the ME, and invasion of the CNS from and bacterial factors in disease pathogenesis (10, 36, 247).
the bloodstream. Meningitis was induced in all animals with Interestingly, the effectiveness of a bactericidal but nonbacte-
cochlear implants, regardless of the infection route (253). riolytic antibiotic, such as daptomycin, in clearing pneumococ-
i.cist. infection in rabbits. Rabbit models of pneumococcal cal infection from the CSF and in reducing inflammation was
meningitis have contributed substantially to our present knowl- proven by means of both this rabbit model (226) and a previ-
edge of the disease and have significantly helped the evaluation ously described rat model (94). Also, of no less importance is
of antimicrobial drug efficacy. The i.cist. rabbit model is an the fact that this rabbit model is a useful tool to analyze
advantageous system that allows accurate analysis of disease damage in the hippocampus due to meningitis (269). Similarly,
features, as well as multiple sampling and large samples of CSF Lindquist et al. described the establishment of a rabbit model
and blood (235). Due to the large quantity of CSF available based on the use of a conventional head holder and on the
from this animal, many types of analysis can be performed, percutaneous puncture by hand of the cisternae magnae of
providing information on bacterial and leukocyte counts and rabbits (148). Finally, Bhatt et al. developed a model of me-
concentrations of glucose, proteins, and other metabolites (Ta- ningogenic labyrinthitis via i.cist. infection of New Zealand
ble 7). A limitation of the model is that experimental groups rabbits with the aim of assessing hearing loss secondary to the
are generally small due to animal size, representing an issue for induction of meningitis. Data obtained from this model indi-
the performance of dependable statistical analysis. Another cate that the severity of hearing loss strongly correlates with
obvious drawback relates to the unnatural method of infection, the duration of infection (26). The model largely mimics hear-
based on the instillation of relatively large numbers of bacteria ing loss due to meningitis in humans and may be instrumental
into the cisterna magna. for testing the efficacies of therapeutic drugs.
The adult rabbit model, originally developed by Dacey and
Sande in 1974, is one of the first important models of pneu- ANIMAL MODELS OF OTITIS MEDIA
mococcal meningitis based on an extremely accurate technique
for inducing the disease. Briefly, animals were secured in a Otitis media is one of the most common diseases in child-
frame through a dental helmet attached to the skull: their hood. It manifests in different forms depending on the dura-
cisternae magnae were then punctured with needles mounted tion and type of exudate (e.g., purulent, mucoid, and serous)
in a geared electrode introducer, and S. pneumoniae was inoc- and it may lead to sequelae, including hearing loss, learning
ulated (69). The use of this model greatly helped the under- disabilities, and speech impediments (65). S. pneumoniae is the
678 CHIAVOLINI ET AL. CLIN. MICROBIOL. REV.

main cause of ME disease in children; other common causes for the study of pathological aspects of the disease as well as
include Haemophilus influenzae and Moraxella catarrhalis. The the efficacies of drugs and vaccines (90).
pathogenesis of the disease depends on different factors in- The acute otitis media model in the gerbil is also a well-
cluding concomitant bacterial and/or viral (e.g., influenza) in- established system for studying disease pathogenesis and com-
fection of the upper respiratory tract, impaired immunity, age, paring the efficacies of different drugs. The anatomies and
genetic predisposition, and dysfunction of the Eustachian tube histologies of Eustachian tubes and MEs in gerbils and chin-
(65). Different models of pneumococcal otitis media have been chillas are similar, with both allowing easy inoculation through
developed using the chinchilla, gerbil, guinea pig, rat, and, the tympanic membrane or the bulla. The model described by
more recently, the mouse in order to understand different Fulghum et al. in a comparative study between gerbils and
aspects of the disease and test novel antimicrobials and poten- chinchillas (81) was developed following the method originally

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


tial vaccine candidates. Description of disease by use of these used by Giebink et al. (89). Advantages of using the gerbil as
animal models has been done by observing different parame- a model of ME infection include the availability of inbred
ters, including the presence of bacteria, polymorphonuclear strains, the reasonable cost, and the susceptibility of this ani-
cells, and inflammatory molecules in ME fluid as well as his- mal to common pathogens of otitis media, including S. pneu-
tological analysis of Eustachian tube, the ME, and the inner moniae, H. influenzae, and M. catarrhalis (81). Infection was
ear (Table 8). carried out by instilling bacteria directly into the ME by trans-
bullar challenge (81). This system was employed in investiga-
tions on the efficacy and pharmacokinetics of ␤-lactams against
Chinchilla, Gerbil, and Guinea Pig Models
otitis media caused by penicillin- and cephalosporin-resistant
Historically, the chinchilla has been the preferred model for S. pneumoniae (17, 18, 54, 190). Intrabullar challenge of gerbils
mimicking pneumococcal otitis media and studying its charac- with virulent pneumococci was also carried out to investigate
teristics (Table 8). Giebink very elegantly reviewed the impor- the pathogenesis of pneumococcal meningitis secondary to oti-
tance of this animal model, emphasizing how infection can be tis media (160, 170).
induced by small bacterial inocula administered either directly Finally, it is also worth mentioning the guinea pig as a model
into the ME or i.n. (90). One major advantage of this experi- of pneumococcal otitis media (239). Although less used than
mental system is its feasibility for the study of disease patho- the chinchilla and the gerbil, it represents a reliable and rela-
genesis, the immunogenicity of vaccine antigens, and the effi- tively inexpensive otosurgical animal for which inbred strains
cacy of antimicrobials at the mucosal site without causing and experimental reagents are also available (211). Investiga-
disseminated disease (90). In addition, chinchillas have large tions on the cytotoxic effects triggered by live pneumococci in
bullae, which are easily accessible for both inoculation and the cochlea (63) and also therapeutic (105) and vaccine (260)
repetitive sampling of ME fluid (211). Two disadvantages re- studies were performed.
late to the unavailability of inbred chinchilla strains and the
high costs of the animal compared to those of other rodents. Rat and Mouse Models
The chinchilla model was first described in 1976 by Giebink
and colleagues. Acute otitis media was induced by inoculation In recent years, interest in both rat and mouse models of
of pneumococci directly into the ME cavity through the dorsal otitis media has increased, since these animals—especially the
bulla (89). Pathogenic aspects of otitis media, as well as the mouse—are less expensive and more characterized in terms of
immune response evoked after infection, were analyzed (89). immunological and genetic information than are chinchillas,
Both this group and other research teams have employed this gerbils, and guinea pigs.
model in several investigations, including characterization of Otitis media has been induced in the rat by two different
the inflammatory response to infection (174, 216), analysis of routes of infection: direct inoculation into the ME and i.n.
the role of pneumococcal virulence factors in disease (217, infection. In a model described in 1988, S. pneumoniae was
240) and of the efficacies and kinetics of antimicrobials against directly instilled into the tympanic cavities of rats to investigate
purulent and serous otitis media (48, 107, 108), and vaccine bacterial replication in the ME and blood (101). Inoculation of
studies (90, 150). As seen for other pneumococcal disease rat ME with pneumococci caused otitis media with either pu-
agents, the virulence of S. pneumoniae in experimental otitis rulent or serous effusion, closely resembling the disease in the
media induced in the chinchilla varies largely by serotype (90). human host (101). This system has been used to evaluate the
A comparative study was recently conducted to analyze the efficacy of penicillin for the treatment and prevention of per-
virulences of different pneumococcal strains in chinchillas in- manent mucosal changes due to pneumococcal acute otitis (5,
oculated via the transbullar route. Fourteen strains of sero- 102) and to assess protection against experimental otitis media
types 3, 4, 6A, 9V, 11, 14, 19F, and 23F were tested in the conferred either by anticapsular antibodies (256) or by active
model, and data showed how even within a single serotype, immunizations with PspA (256) or whole killed bacteria (66).
strains differ in their capabilities to cause local and systemic Moreover, the virulences of pneumococcal strains with differ-
disease (80). Besides the classic intratympanic route, Giebink ent opacity phenotypes (151) and various levels of susceptibil-
et al. also established a chinchilla model of otitis media based ity to penicillin (154) were also evaluated by intratympanic
on simultaneous i.n. infection of S. pneumoniae and influenza challenge of rats. van der Ven et al. described a novel rat
A virus: approximately 70% of animals developed the disease, model based on the i.n. instillation of bacteria, thus mimicking
while only 20% of chinchillas presented with otitis after inoc- the natural route of infection of otitis media in humans (248).
ulation with bacteria only (88). This last model more closely Histamine was inoculated into Wistar rats [strain Rivm:WU
resembles the clinical manifestations in humans and is valuable (CBP)] by the intratympanic route in order to impair ciliary
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 679

activity and contribute to Eustachian tube dysfunction, and imentation remains an unquestionably valuable tool to clarify
bacteria were then delivered i.n. with an infusion catheter. the pathogenic mechanisms of disease. This review provides a
Histamine pretreatment facilitated the ascent of bacteria from perspective on the development and applications of different
the nasopharynx to the ME, allowing assessment of mucosal animal models used to mimic and study the major diseases
immunity directly in the Eustachian tube and ME. The model caused by S. pneumoniae. We have highlighted the progress
was utilized in a vaccination study to analyze protection against made over the years, indicating how certain historical models
otitis media following immunization with a tetanus toxoid- represented milestones for developing novel and more-effec-
conjugated type 14 capsular polysaccharide (248). The impor- tive methods, while others are still in use largely to investigate
tance of this model was also revealed in the work of Eriksson pneumococcal diseases. Some of the techniques for inducing
and Hellström, who provided clear evidence of acute otitis disease were described to point out how thoroughly these an-

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


media development in the rat after multiple i.n. inoculations of imal models were set up. Characteristics of pneumococcal
pneumococci (77). pneumonia, sepsis, and meningitis have successfully been re-
Less than 10 years ago, only a few mouse models of pneu- produced in the mouse, the rat, and the rabbit. In the case of
mococcal otitis media were available (210). Clearly, the small otitis media, the chinchilla, the gerbil, and the rat have been
size of the animal together with the poor accessibility of the the preferred animal models throughout the years, although
ME for inoculation and sampling makes the mouse a tricky recently researchers have also reproduced the disease in the
choice to study otitis media. Two different routes of infection mouse. All these models were and continue to be helpful tools
are used in the mouse model: either direct injection (intrab- for elucidating aspects of disease pathogenesis, characterizing
ullar or intratympanic) into the ME or i.n. inoculation. Direct innate and adaptive immunities to S. pneumoniae, and testing
microbial injection into the ME warrants reproducibility in the efficacies of antibiotics and other therapies as well as po-
disease initiation and development and allows the use of small tential vaccine candidates. Various infection routes and meth-
bacterial inocula, whereas the i.n. route mimics the natural ods have been employed to induce disease, and we have dis-
mode of ME infection, but induction of experimental otitis is cussed the importance of different readouts and emphasized
more sporadic and requires larger doses of bacteria. In 2003, the main advantages and limitations of the models. The utility
Melhus and Ryan established a model of otitis media caused by of each animal model is also highlighted to facilitate research-
S. pneumoniae and H. influenzae in BALB/c and C57BL/6 mice ers in the choice of the appropriate system. The mouse is
inoculated via the intrabullar route (164). The course of dis- comparatively inexpensive and easy to handle and allows the
ease was dependent upon both bacterial and mouse strains, screening of drugs and vaccines with high statistical power. The
with BALB/c being the most susceptible mouse strain. The commercial availability of plentiful murine experimental re-
authors also found that highly virulent pneumococcal serotypes agents and strains, including transgenic and knockout strains,
(types 3 and 6) consistently induced lethal sepsis with or with- makes the mouse an attractive model to address the interplay
out ME infection, while less virulent strains (types 9 and 19) between bacteria and host factors. Furthermore, the comple-
produced otitis (164, 210). Direct inoculation of bacteria into tion of the mouse genome sequence and the availability of
the ME can also be instrumental in assessing inflammation in technologies to introduce targeted mutations into the genome
the ME following the injection of heat-killed pneumococci provide a powerful and extensive toolkit for studying mouse
(152, 161). As acute bacterial otitis media is an inflammatory susceptibility to pneumococcal disease. In contrast, larger an-
response to replicating microorganisms, the use of heat-killed imals present the advantage of obtaining larger samples in
bacteria does not mimic the natural disease but still allows addition to an increased ease in performing inoculation and
measurements of inflammatory parameters in the ME, such as surgical procedures, but they are more expensive, and the
the amount and type of effusions, the thickness of the tympanic results obtained may lack statistical significance due to the use
membrane, and the recruitment of polymorphonuclear cells of smaller animal groups.
(152). The mouse intratympanic/intrabullar model of pneumo- The development of pneumococcal disease depends upon
coccal otitis media found applications in both vaccine (211, both bacterial (e.g., capsular serotype and other virulence de-
212) and therapy (161) research fields. Finally, a recent work terminants) and host (e.g., genetic background, immune re-
elegantly described the induction and prevention, by means of sponse, age, sex) factors. Thus, in addition to the selection of
phage lysins, of otitis media in BALB/c mice stably colonized the appropriate in vivo model, the use of several animal and
by bioluminescent S. pneumoniae (162). In the model, recur- pneumococcal strains is highly recommended to test a research
rent otitis media was induced by infecting the colonized mice hypothesis. We also wish to emphasize how in vivo experimen-
via the i.n. route with influenza virus (to favor bacterial spread tation should preferably be combined with in vitro assays (e.g.,
from the nasopharynx to the ME) and was efficiently treated cell culture assays) to understand and reproduce essential
with the Cpl-1 lysin (162). This novel model of naturally oc- characteristics of the pathogenesis of infectious diseases and to
curring otitis media mimics infection in children and may elucidate key features of host-pathogen interaction. Finally,
prove useful in studies on the prevention and treatment of this worth considering is the fact that certain host-microbe inter-
important disease. actions are species specific, thus suggesting additional caution
to scientists who wish to mimic, in an animal model, pathoge-
nicity mechanisms which occur only in the human host.
CONCLUSIONS
Pneumococcal disease in humans is varied and multifaceted, ACKNOWLEDGMENTS
and no experimental animal model is able to completely mimic We thank Tiziana Braccini, Riccardo Parigi, and Velia Braione for
human disease. However, despite its limitations, animal exper- their excellent expertise with animal experiments offered along the
680 CHIAVOLINI ET AL. CLIN. MICROBIOL. REV.

years. We also acknowledge Marina Piccinin for secretarial work. We factor alpha in the host response of mice to bacteria caused by pneumoly-
are grateful to Aras Kadioglu (University of Leicester, United King- sin-deficient Streptococcus pneumoniae. Infect. Immun. 66:839–842.
dom) for advice and critical reading of the manuscript. 23. Bergeron, Y., N. Ouellet, A. M. Deslauriers, M. Simard, M. Olivier, and
M. G. Bergeron. 1998. Cytokine kinetics and other host factors in response
to pneumococcal pulmonary infection in mice. Infect. Immun. 66:912–922.
REFERENCES 24. Berry, A. M., R. A. Lock, D. Hansman, and J. C. Paton. 1989. Contribution
1. Abgueguen, P., E. Azoulay-Dupuis, V. Noel, P. Moine, V. Rieux, B. Fantin, of autolysin to virulence of Streptococcus pneumoniae. Infect. Immun. 57:
and J. P. Bédos. 2007. Amoxicillin is effective against penicillin-resistant 2324–2330.
Streptococcus pneumoniae strains in a mouse pneumonia model simulating 25. Berry, A. M., J. Yother, D. E. Briles, D. Hansman, and J. C. Paton. 1989.
human pharmacokinetics. Antimicrob. Agents Chemother. 51:208–214. Reduced virulence of a defined pneumolysin-negative mutant of Strepto-
2. Alcantara, R. B., L. C. Preheim, and M. J. Gentry. 1999. Role of pneumo- coccus pneumoniae. Infect. Immun. 57:2037–2042.
lysin’s complement-activating activity during pneumococcal bacteremia in 26. Bhatt, S., C. Halpin, W. Hsu, B. A. Thedinger, R. A. Levine, E. Tuomanen,
cirrhotic rats. Infect. Immun. 67:2862–2866. and J. B. J. Nadol. 1991. Hearing loss and pneumococcal meningitis: an
3. Alcantara, R. B., L. C. Preheim, and M. J. Gentry-Nielsen. 2001. Pneumo- animal model. Laryngoscope 101:1285–1292.

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


lysin-induced complement depletion during experimental pneumococcal 27. Bifrare, Y.-D., J. Kummer, P. Joss, M. G. Tauber, and S. L. Leib. 2005.
bacteremia. Infect. Immun. 69:3569–3575. Brain-derived neurotrophic factor protects against multiple forms of brain
4. Alexander, J. E., R. A. Lock, C. A. M. Peeters, J. T. Poolman, P. W. Andrew, injury in bacterial meningitis. J. Infect. Dis. 191:40–45.
T. J. Mitchell, D. Hansman, and J. C. Paton. 1994. Immunization of mice 28. Blasi, E., R. Barluzzi, F. Bistoni, P. Mosci, and F. Bistoni. 1992. Experi-
with pneumolysin toxoid confers a significant degree of protection against mental model of intracerebral infection with Cryptococcus neoformans:
at least nine serotypes of Streptococcus pneumoniae. Infect. Immun. 62: roles of phagocytes and opsonization. Infect. Immun. 60:3682–3688.
5683–5688. 29. Blue, C. E., and T. J. Mitchell. 2003. Contribution of a response regulator
5. Andersen, H., J. Thomsen, and P. Caye-Thomasen. 2005. Penicillin reduces to the virulence of Streptococcus pneumoniae is strain dependent. Infect.
eustachian tube gland tissue changes in acute otitis media. Otolaryngol. Immun. 71:4405–4413.
Head Neck Surg. 133:206–210. 30. Blue, C. E., G. K. Paterson, A. Kerr, M. Berge, J. P. Claverys, and T. J.
6. Auer, M., L.-A. Pfister, D. Leppert, M. G. Tauber, and S. L. Leib. 2000. Mitchell. 2003. ZmpB, a novel virulence factor of Streptococcus pneumoniae
Effects of clinically used antioxidants in experimental pneumococcal men- that induces tumor necrosis factor alpha production in the respiratory tract.
ingitis. J. Infect. Dis. 182:347–350. Infect. Immun. 71:4925–4935.
7. Austrian, R. 1986. Pneumococcal pneumonia. Diagnostic, epidemiologic, 31. Bogaert, D., R. de Groot, and P. W. M. Hermans. 2004. Streptococcus
therapeutic and prophylactic considerations. Chest 90:738–743. pneumoniae colonization: the key to pneumococcal disease. Lancet 4:144–
8. Austrian, R., and J. Gold. 1964. Pneumococcal bacteremia with especial 154.
reference to bacteremic pneumococcal pneumonia. Ann. Intern. Med. 60: 32. Bogaert, D., P. M. Hermans, P. V. Adrian, H. C. Rumke, and R. de Groot.
759–776. 2004. Pneumococcal vaccines: an update on current strategies. Vaccine
9. Avery, O. T., C. M. MacLeod, and M. McCarty. 1944. Studies on the 22:2209–2220.
chemical nature of the substance inducing transformation of pneumococcal 33. Bone, R. C., C. J. J. Fisher, T. P. Clemmer, G. J. Slotman, C. A. Metz, R. A.
types. Induction of tranformation by a desoxyribonucleic acid fraction iso- Balk, et al. 1989. Sepsis syndrome: a valid clinical entity. Crit. Care Med.
lated from Pneumococcus type III. J. Exp. Med. 79:137–158. 17:389–393.
10. Azeh, I., M. Mader, A. Smirnov, W. Beuche, R. Nau, and F. Weber. 1998. 34. Böttcher, T., H. Ren, M. Goiny, J. Gerber, J. Lykkesfeldt, U. Kuhnt, M.
Experimental pneumococcal meningitis in rabbits: the increase of matrix Lotz, S. Bunkowski, C. Werner, I. Schau, A. Spreer, S. Christen, and R.
metalloproteinase-9 in cerebrospinal fluid correlates with leucocyte inva- Nau. 2004. Clindamycin is neuroprotective in experimental Streptococcus
sion. Neurosci. Lett. 256:127–130. pneumoniae meningitis compared with ceftriaxone. J. Neurochem. 91:1450–
11. Azoulay-Dupuis, E., J. P. Bédos, J. Mohler, P. Moine, C. Cherbuliez, G. 1460.
Peytavin, B. Fantin, and T. Köhler. 2005. Activity of gemifloxacin against 35. Böttcher, T., A. Spreer, I. Azeh, R. Nau, and J. Gerber. 2003. Matrix
quinolone-resistant Streptococcus pneumoniae strains in vitro and in a metalloproteinase-9 deficiency impairs host defense mechanisms against
mouse pneumonia model. Antimicrob. Agents Chemother. 49:1046–1054. Streptococcus pneumoniae in a mouse model of bacterial meningitis. Neu-
12. Azoulay-Dupuis, E., J. P. Bédos, E. Vallee, D. J. Hardy, R. N. Swanson, and rosci. Lett. 338:201–204.
J. J. Pocidalo. 1991. Antipneumococcal activity of ciprofloxacin, oxofloxacin 36. Braun, J. S., J. E. Sublett, D. Freyer, T. J. Mitchell, J. L. Cleveland, E. I.
and temafloxacin in an experimental mouse pneumonia model at various Tuomanen, and J. R. Weber. 2002. Pneumococcal pneumolysin and H2O2
stages of the disease. J. Infect. Dis. 163:319–324. mediate brain cell apoptosis during meningitis. J. Clin. Investig. 109:19–27.
13. Azoulay-Dupuis, E., J. P. Bédos, E. Vallee, and J. J. Pocidalo. 1991. Com- 37. Briles, D. E., E. Ades, J. C. Paton, J. S. Sampson, G. M. Carlone, R. C.
parative activity of fluorinated quinolones in acute and subacute Strepto- Huebner, A. Virolainen, E. Swiatlo, and S. K. Hollingshead. 2000. Intra-
coccus pneumoniae pneumonia models: efficacy of temafloxacin. J. Antimi- nasal immunization of mice with a mixture of the pneumococcal proteins
crob. Chemother. 28:45–53. PsaA and PspA is highly protective against nasopharyngeal carriage of
14. Bakker-Woudenberg, I. A. J. M., J. Y. T. de Jong-Hoenderop, and M. F. Streptococcus pneumoniae. Infect. Immun. 68:796–800.
Michel. 1979. Efficacy of antimicrobial chemotherapy in experimental rat 38. Briles, D. E., M. J. Crain, B. M. Gray, C. Forman, and J. Yother. 1992.
pneumonia: effects of impaired phagocytosis. Infect. Immun. 25:366–375. Strong association between capsular type and virulence for mice among
15. Balachandran, P., A. Brooks-Walter, A. Virolainen-Julkunen, S. K. Holl- human isolates of Streptococcus pneumoniae. Infect. Immun. 60:111–116.
ingshead, and D. E. Briles. 2002. Role of pneumococcal surface protein C 39. Briles, D. E., S. K. Hollingshead, and I. Jónsdóttir. 2008. Animal models of
in nasopharyngeal carriage and pneumonia and its ability to elicit protec- invasive pneumococcal disease, p. 47–58. In G. R. Siber, K. P. Klugman,
tion against carriage of Streptococcus pneumoniae. Infect. Immun. 70:2526– and P. H. Mäkelä (ed.), Pneumococcal vaccines: the impact of conjugate
2534. vaccine. ASM Press, Washington, DC.
16. Barocchi, M. A., S. Censini, and R. Rappuoli. 2007. Vaccines in the era of 40. Briles, D. E., S. K. Hollingshead, J. C. Paton, E. W. Ades, L. Novak, F. W.
genomics: the pneumococcal challenge. Vaccine 25:2963–2973. Van Ginkel, and W. H. Benjamin. 2003. Immunizations with pneumococcal
17. Barry, B., M. Muffat-Joly, J. Bauchet, F. Faurisson, P. Gehanno, J. J. surface protein A and pneumolysin are protective against pneumonia in a
Pocidalo, and C. Carbon. 1996. Efficacy of single-dose ceftriaxone in ex- murine model of pulmonary infection with Streptococcus pneumoniae. J. In-
perimental otitis media induced by penicillin- and cephalosporin-resistant fect. Dis. 188:339–348.
Streptococcus pneumoniae. Antimicrob. Agents Chemother. 40:1977–1982. 41. Briles, D. E., J. Horowitz, L. S. McDaniel, W. H. Benjamin, J. L. Claflin,
18. Barry, B., M. Muffat-Joly, P. Gehanno, and J. J. Pocidalo. 1993. Effect of C. L. Brooker, G. Scott, and C. Forman. 1986. Genetic control of the
increased dosages of amoxicillin in treatment of experimental middle ear susceptibility to pneumococcal infection. Curr. Top. Microbiol. Immunol.
otitis due to penicillin-resistant Streptococcus pneumoniae. Antimicrob. 124:103–120.
Agents Chemother. 37:1599–1603. 42. Briles, D. E., J. D. King, M. A. Gray, L. S. McDaniel, E. Swiatlo, and K. A.
19. Bédos, J. P., V. Rieux, J. Bauchet, M. Muffat-Joly, C. Carbon, and E. Benton. 1996. PspA, a protection eliciting pneumococcal protein: immuno-
Azoulay-Dupuis. 1998. Efficacy of trovafloxacin against penicillin-suscepti- genicity of isolated native PspA in mice. Vaccine 14:858–867.
ble and multiresistant strains of Streptococcus pneumoniae in a mouse 43. Briles, D. E., M. H. Nahm, K. Schroer, J. Davie, P. Baker, J. Kearney, and
pneumonia model. Antimicrob. Agents Chemother. 42:862–867. R. Barletta. 1981. Antiphosphocholine antibodies found in normal mouse
20. Benton, K. A., M. P. Everson, and D. E. Briles. 1995. A pneumolysin- serum are protective against intravenous infection with type 3 Streptococcus
negative mutant of Streptococcus pneumoniae causes chronic bacteremia pneumoniae. J. Exp. Med. 153:694–705.
rather than acute sepsis in mice. Infect. Immun. 63:448–455. 44. Brown, E. J., S. W. Hosea, and M. M. Frank. 1981. The role of the spleen
21. Benton, K. A., J. C. Paton, and D. E. Briles. 1997. Differences in virulence in experimental pneumococcal bacteremia. J. Clin. Investig. 67:975–982.
for mice among Streptococcus pneumoniae strains of capsular types 2, 3, 4, 45. Brown, E. J., S. W. Hosea, and M. M. Frank. 1983. The role of antibody and
5, and 6 are not attributable to differences in pneumolysin production. complement in the reticuloendothelial clearance of pneumococci from the
Infect. Immun. 65:1237–1244. bloodstream. Rev. Infect. Dis. 5:(Suppl.)S797–S805.
22. Benton, K. A., J. L. VanCott, and D. E. Briles. 1998. Role of tumor necrosis 46. Brown, J. S., D. A. Ogunniyi, M. C. Woodrow, D. W. Holden, and J. C.
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 681

Paton. 2001. Immunization with components of two iron uptake ABC fluid concentrations of penicillin and cephalosporin derivatives. Antimi-
transporters protects mice against systemic Streptococcus pneumoniae in- crob. Agents Chemother. 6:437–441.
fection. Infect. Immun. 69:6702–6706. 70. Dallaire, F., N. Ouellet, Y. Bergeron, V. Turmel, M. C. Gauthier, M.
47. Cabellos, C., D. E. MacIntyre, M. Forrest, M. Burroughs, S. Prasad, and E. Simard, and M. G. Bergeron. 2001. Microbiological and inflammatory fac-
Tuomanen. 1992. Differing roles for platelet-activating factor during in- tors associated with development of pneumococcal pneumonia. J. Infect.
flammation of lung and subarachnoid space. The special case of Strepto- Dis. 184:292–300.
coccus pneumoniae. J. Clin. Investig. 90:612–618. 71. Davis, C. C., M. A. Mellencamp, and L. C. Preheim. 1991. A model of
48. Canafax, D. M., N. Nonomura, G. R. Erdmann, C. T. Le, S. K. Juhn, and pneumococcal pneumonia in chronically intoxicated rats. J. Infect. Dis.
G. S. Giebink. 1989. Experimental animal models for studying antimicrobial 163:799–805.
pharmacokinetics in otitis media. Pharm. Res. 6:279–285. 72. Denny, P., E. Hopes, N. Gingles, K. W. Broman, W. McPheat, J. Morten, J.
49. Candiani, G., M. Abbondi, M. Borgonovi, and R. Williams. 1997. Experi- Alexander, P. W. Andrew, and S. D. Brown. 2003. A major locus conferring
mental lobar pneumonia due to penicillin-susceptible and penicillin-resis- susceptibility to infection by Streptococcus pneumoniae in mice. Mamm.
tant Streptococcus pneumoniae in immunocompetent and neutropenic rats: Genome 14:448–453.
efficacy of penicillin and teicoplanin treatment. J. Antimicrob. Chemother.

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


73. Destache, C. J., C. B. Pakiz, C. Larsen, H. Owens, and A. K. Dash. 2001.
39:199–207. Cerebrospinal fluid penetration and pharmacokinetics of levofloxacin in an
50. Canvin, J. R., A. P. Marvin, M. Sivakumaran, J. C. Paton, G. J. Boulnois, experimental rabbit meningitis model. J. Antimicrob. Chemother. 47:611–
P. W. Andrew, and T. J. Mitchell. 1995. The role of pneumolysin and 615.
autolysin in the pathology of pneumonia and septicemia in mice infected 74. Doern, G. V., S. S. Richter, A. Miller, N. Miller, C. Rice, K. Heilmann, and
with a type 2 pneumococcus. J. Infect. Dis. 172:119–123. S. Beekmann. 2005. Antimicrobial resistance among Streptococcus pneu-
51. Cao, J., D. Chen, W. Xu, T. Chen, S. Xu, J. Luo, Q. Zhao, B. Liu, D. Wang, moniae in the United States: have we begun to turn the corner on resistance
X. Zhang, Y. Shan, and Y. Yin. 2007. Enhanced protection against pneu- to certain antimicrobial classes? Clin. Infect. Dis. 41:139–148.
mococcal infection elicited by immunization with the combination of PspA, 75. Dopazo, J., A. Mendoza, J. Herrero, F. Caldara, Y. Humbert, L. Friedli, M.
PspC, and ClpP. Vaccine 25:4996–5005. Guerrier, E. Grand-Schenk, C. Gandin, M. de Francesco, A. Polissi, G.
52. Carp, R. I., A. I. Davidson, and P. A. Merz. 1971. A method for obtaining Buell, G. Feger, E. Garcia, M. Peitsch, and J. F. Garcia-Bustos. 2001.
cerebrospinal fluid from mice. Res. Vet. Sci. 12:499. Annotated draft genomic sequence from Streptococcus pneumoniae type
53. Casal, J., L. Aguilar, I. Jado, J. Yuste, M. J. Gimenez, J. Prieto, and A. 19F clinical isolate. Microb. Drug Resist. 7:99–125.
Fenoll. 2002. Effects of specific antibodies against Streptococcus pneu- 76. Echchannaoui, H., K. Frei, C. Schnell, S. L. Leib, W. Zimmerli, and R.
moniae on pharmacodynamic parameters of ␤-lactams in a mouse sepsis Landmann. 2002. Toll-like receptor 2-deficient mice are highly susceptible
model. Antimicrob. Agents Chemother. 46:1340–1344. to Streptococcus pneumoniae meningitis because of reduced bacterial clear-
54. Cenjor, C., C. Ponte, A. Parra, E. Nieto, G. Garcia-Calvo, M. J. Gimenez, ing and enhanced inflammation. J. Infect. Dis. 186:798–806.
L. Aguilar, and F. Soriano. 1998. In vivo efficacies of amoxicillin and 77. Eriksson, P. O., and S. Hellström. 2003. Acute otitis media develops in the
cefuroxime against penicillin-resistant Streptococcus pneumoniae in a gerbil rat after intranasal challenge of Streptococcus pneumoniae. Laryngoscope
model of acute otitis media. Antimicrob. Agents Chemother. 42:1361–1364. 113:2047–2051.
55. Centers for Disease Control and Prevention. 2008. Active bacterial core
78. Esposito, A. L., and J. E. Pennington. 1983. Effects of aging on antibacterial
surveillance (ABCs) report, Emerging Infections Program Network, Strep-
mechanisms in experimental pneumonia. Am. Rev. Respir. Dis. 128:662–
tococcus pneumoniae 2006. http://www.cdc.gov/abcs.
667.
56. Charles, P. E., M. Etienne, D. Croisier, L. Piroth, C. Lequeu, J. Pugin, H.
79. Feldman, C., N. C. Munro, P. K. Jeffery, T. J. Mitchell, P. W. Andrew, G. J.
Portier, and P. Chavanet. 2005. The impact of mechanical ventilation on
Boulnois, D. Guerreiro, J. A. L. Rohde, H. C. Todd, P. J. Cole, and R.
the moxifloxacin treatment of experimental pneumonia caused by Strepto-
Wilson. 1991. Pneumolysin induces the salient histologic features of pneu-
coccus pneumoniae. Crit. Care Med. 33:1029–1035.
mococcal infection in the rat lung in vivo. Am. J. Respir. Cell Mol. Biol.
57. Charles, P. E., L. Piroth, N. Desbiolles, C. Lequeu, L. Martin, H. Portier,
5:416–423.
and P. Chavanet. 2002. New model of ventilator-associated pneumonia in
80. Forbes, M. L., E. Horsey, N. L. Hiller, F. J. Buchinsky, J. D. Hayes, J. M.
immunocompetent rabbits. Crit. Care Med. 30:2278–2283.
Compliment, T. Hillman, S. Ezzo, K. Shen, R. Keefe, K. Barbadora, J. C.
58. Chiavolini, D., G. Memmi, T. Maggi, F. Iannelli, G. Pozzi, and M. R.
Post, F. Z. Hu, and G. D. Ehrlich. 2008. Strain-specific virulence pheno-
Oggioni. 2003. The three extra-cellular zinc metalloproteinases of Strepto-
types of Streptococcus pneumoniae assessed using the Chinchilla laniger
coccus pneumoniae have a different impact on virulence in mice. BMC
model of otitis media. PLoS ONE 3:e1969.
Microbiol. 3:14.
59. Chiavolini, D., S. Tripodi, R. Parigi, M. R. Oggioni, E. Blasi, M. Cintorino, 81. Fulghum, R. S., J. E. Brinn, A. M. Smith, H. J. Daniel III, and P. J.
G. Pozzi, and S. Ricci. 2004. Method for inducing experimental pneumo- Loesche. 1982. Experimental otitis media in gerbils and chinchillas with
coccal meningitis in outbred mice. BMC Microbiol. 22:36. Streptococcus pneumoniae, Haemophilus influenzae, and other aerobic and
60. Chiou, C., and V. L. Yu. 2006. Severe pneumococcal pneumonia: new anaerobic bacteria. Infect. Immun. 36:802–810.
strategies for management. Curr. Opin. Crit. Care 12:470–476. 82. Ganzinger, U., and A. Haslberger. 1985. Pharmacokinetics of cephalospo-
61. Coil, J. A., J. D. Dickerman, and E. Boulton. 1978. Increased susceptibility rins in normal and septicemic rabbits. Antimicrob. Agents Chemother.
of splenectomized mice to infection after exposure to an aerosolized sus- 28:473–477.
pension of type III Streptococcus pneumoniae. Infect. Immun. 21:412–416. 83. Gehre, F., S. L. Leib, D. Grandgirard, J. Kummer, A. Buhlmann, F. Simon,
62. Coln, D., J. Horton, M. Ogden, and S. Williams. 1983. Clearance of pneu- R. Gaumann, A. S. Kharat, M. G. Tauber, and A. Tomasz. 2008. Essential
mococcal organisms after repair of injured spleens. J. Pediatr. Surg. 18: role of choline for pneumococcal virulence in an experimental model
280–281. of meningitis. J. Intern. Med. doi: 10.1111/j.1365-2796.2008.01930.x.
63. Cook, R. D., D. S. Postma, G. M. Brinson, J. Prazma, and H. C. Pillsbury. 84. Gentry, M. J., M. U. Snitily, and L. C. Preheim. 1995. Phagocytosis of
1999. Cytotoxic changes in hair cells secondary to pneumococcal middle-ear Streptococcus pneumoniae measured in vitro and in vivo in a rat model of
infection. J. Otolaryngol. 28:325–331. carbon tetrachloride-induced liver cirrhosis. J. Infect. Dis. 171:350–355.
64. Cottagnoud, P., C. M. Gerber, F. Acosta, M. Cottagnoud, K. Neftel, and 85. Gerber, J., K. Pohl, V. Sander, S. Bunkowski, and R. Nau. 2003. Rifampin
M. G. Tauber. 2000. Linezolid against penicillin-sensitive and-resistant followed by ceftriaxone for experimental meningitis decreases lipoteichoic
pneumococci in the rabbit meningitis model. J. Antimicrob. Chemother. acid concentrations in cerebrospinal fluid and reduces neuronal damage in
46:981–985. comparison to ceftriaxone alone. Antimicrob. Agents Chemother. 47:1313–
65. Cripps, A. W., and J. Kyd. 2003. Bacterial otitis media: current vaccine 1317.
development strategies. Immunol. Cell Biol. 81:46–51. 86. Gerber, J., G. Raivich, A. Wellmer, C. Noeske, T. Kunst, A. Werner, W.
66. Cripps, A. W., and J. M. Kyd. 2007. Comparison of mucosal and parenteral Bruck, and R. Nau. 2001. A mouse model of Streptococcus pneumoniae
immunisation in two animal models of pneumococcal infection: otitis media meningitis mimicking several features of human disease. Acta Neuropathol.
and acute pneumonia. Vaccine 25:2471–2477. 101:499–508.
67. Croisier, D., P. Chavanet, C. Lequeu, A. Ahanou, A. Nierlich, C. Neuwirth, 87. Gianfaldoni, C., S. Censini, M. Hilleringmann, M. Moschioni, C. Facciotti,
L. Piroth, M. Duong, M. Buisson, and H. Portier. 2002. Efficacy and phar- W. Pansegrau, V. Masignani, A. Covacci, R. Rappuoli, M. A. Barocchi, and
macodynamics of simulated human-like treatment with levafloxacin on ex- P. Ruggiero. 2007. Streptococcus pneumoniae pilus subunits protect mice
perimental pneumonia induced with penicillin-resistant pneumococci with against lethal challenge. Infect. Immun. 75:1059–1062.
various susceptibilities to fluoroquinolones. J. Antimicrob. Chemother. 50: 88. Giebink, G. S., I. K. Berzins, S. C. Marker, and G. Schiffmann. 1980.
349–360. Experimental otitis media after nasal inoculation of Streptococcus pneu-
68. Croisier, D., M. Etienne, E. Bergoin, P. E. Charles, C. Lequeu, L. Piroth, H. moniae and influenza A virus in chinchillas. Infect. Immun. 30:445–450.
Portier, and P. Chavanet. 2004. Mutant selection window in levofloxacin 89. Giebink, G. S., E. E. Payne, E. L. Mills, S. K. Juhn, and P. G. Quie. 1976.
and moxifloxacin treatments of experimental pneumococcal pneumonia in Experimental otitis media due to Streptococcus pneumoniae: immunopatho-
a rabbit model of human therapy. Antimicrob. Agents Chemother. 48: genic response in the chinchilla. J. Infect. Dis. 134:595–604.
1699–1707. 90. Giebink, G. S. 1999. Otitis media: the chinchilla model. Microb. Drug
69. Dacey, R. G., and M. A. Sande. 1974. Effect of probenecid on cerebrospinal Resist. 5:57–72.
682 CHIAVOLINI ET AL. CLIN. MICROBIOL. REV.

91. Gillespie, S. H. 1989. Aspects of pneumococcal infection including bacterial 116. Jakobsen, H., S. Hannesdóttir, S. P. Bjarnarson, D. Schulz, E. Trannoy,
virulence, host response and vaccination. J. Med. Microbiol. 28:237–248. C. A. Siegrist, and I. Jónsdóttir. 2006. Early life T cell responses to pneu-
92. Gingles, N. A., J. E. Alexander, A. Kadioglu, P. W. Andrew, A. Kerr, T. J. mococcal conjugates increase with age and determine the polysaccharide-
Mitchell, E. Hopes, P. Denny, S. Brown, H. B. Jones, S. Little, G. C. Booth, specific antibody response and protective efficacy. Eur. J. Immunol. 36:287–
and W. L. McPheat. 2001. Role of genetic resistance in invasive pneumo- 295.
coccal infection: identification and study of susceptibility and resistance in 117. Johnson, S. E., L. Rubin, S. Romero-Steiner, J. K. Dykes, L. B. Pais, A.
inbred mouse strains. Infect. Immun. 69:426–434. Rivzi, E. Ades, and G. M. Carlone. 1999. Correlation of opsonophagocytosis
93. Grandgirard, D., J. M. Loeffler, V. A. Fischetti, and S. L. Leib. 2008. Phage and passive protection assay using human anticapsular antibodies in an
lytic enzyme Cpl-1 for antibacterial therapy in experimental pneumococcal infant mouse model of bacteremia for Streptococcus pneumoniae. J. Infect.
meningitis. J. Infect. Dis. 197:1519–1522. Dis. 180:133–140.
94. Grandgirard, D., C. Schurch, P. Cottagnoud, and S. L. Leib. 2007. Prevention 118. Jomaa, H., J. Yuste, J. C. Paton, C. Jones, G. Dougan, and J. S. Brown.
of brain injury by the nonbacteriolytic antibiotic daptomycin in experimental 2005. Antibodies to the iron uptake ABC transporter lipoproteins PiaA and
pneumococcal meningitis. Antimicrob. Agents Chemother. 51:2173–2178. PiuA promote opsonophagocytosis of Streptococcus pneumoniae. Infect.
95. Grandgirard, D., O. Steiner, M. G. Tauber, and S. L. Leib. 2007. An infant Immun. 73:6852–6859.

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


mouse model of brain damage in pneumococcal meningitis. Acta Neuro- 119. Jones, H. A., R. J. Clark, C. G. Rhodes, J. B. Schofield, T. Krausz, and C.
pathol. 114:609–617. Haslett. 1994. In vivo measurement of neutrophil activity in experimental
96. Guckian, J. C. 1973. Role of metabolism in pathogenesis of bacteremia due lung inflammation. Am. J. Respir. Crit. Care Med. 149:1635–1639.
to Diplococcus pneumoniae in rabbits. J. Infect. Dis. 127:1–8. 120. Jounblat, R., A. Kadioglu, T. J. Mitchell, and P. W. Andrew. 2003. Pneu-
97. Guckian, J. C. 1976. Coagulopathy in experimental sepsis with Streptococ- mococcal behaviour and host responses during bronchopneumonia are
cus pneumoniae in rabbits: effect of drug therapy and splenectomy. J. Infect. affected differently by the cytolytic and complement-activating activities of
Dis. 134:150–157. pneumolysin. Infect. Immun. 71:1813–1819.
98. Guckian, J. C., B. F. Morrey, and H. B. Kirby. 1970. Role of lysosomes and 121. Kadioglu, A., and P. W. Andrew. 2005. Susceptibility and resistance to
cathepsin inhibitor in plasma during pneumococcal infection. J. Infect. Dis. pneumococcal disease in mice. Brief. Funct. Genomics Proteomics 4:241–
122:290–302. 247.
99. Gullstrand, P., A. Alwmark, and C. Schalen. 1982. Effect of steroids on the 122. Kadioglu, A., W. Coward, M. J. Colston, E. Hewitt, and P. W. Andrew. 2004.
outcome of penicillin treatment in pneumococcal sepsis in splenectomized CD4-T-lymphocyte interactions with pneumolysin and pneumococci sug-
rats. Surgery 91:222–225. gest a crucial protective role in the host response to pneumococcal infec-
100. Hamel, J., N. Charland, I. Pineau, C. Ouellet, S. Rioux, D. Martin, and tion. Infect. Immun. 72:2689–2697.
B. R. Brodeur. 2004. Prevention of pneumococcal disease in mice immu- 123. Kadioglu, A., N. A. Gingles, K. Grattan, A. Kerr, T. J. Mitchell, and P. W.
nized with conserved surface-accessible proteins. Infect. Immun. 72:2659– Andrew. 2000. Host cellular immune response to pneumococcal lung infec-
2670. tion in mice. Infect. Immun. 68:492–501.
101. Hermansson, A., P. Emgard, K. Prellner, and S. Hellström. 1988. A rat 124. Kadioglu, A., J. A. Sharpe, I. Lazou, C. Svanborg, C. Ockleford, T. J.
model for pneumococcal otitis media. Am. J. Otolaryngol. 9:97–101. Mitchell, and P. W. Andrew. 2001. Use of green fluorescent protein in
102. Hermansson, A., S. Hellström, and K. Prellner. 1991. Mucosal changes visualisation of pneumococcal invasion of broncho-epithelial cells in vivo.
induced by experimental pneumococcal otitis media are prevented by pen- FEMS Microbiol. Lett. 194:105–110.
icillin V. Otolaryngol. Head Neck Surg. 105:578–585. 125. Kadioglu, A., S. Taylor, F. Iannelli, G. Pozzi, T. J. Mitchell, and P. W.
103. Hirst, R. A., B. Gosai, A. Rutman, P. W. Andrew, and C. O’Callaghan. 2003. Andrew. 2002. Upper and lower respiratory tract infection by Streptococcus
Streptococcus pneumoniae damages the ciliated ependyma of the brain pneumoniae is affected by deficiency of pneumolysin and by differences in
during meningitis. Infect. Immun. 71:6095–6100. serotype. Infect. Immun. 70:2886–2890.
104. Hirst, R. A., B. Gosai, A. Rutman, C. J. Guerin, P. Nicotera, P. W. Andrew, 126. Kadioglu, A., J. N. Weiser, J. C. Paton, and P. W. Andrew. 2008. The role
and C. O’Callaghan. 2008. Streptococcus pneumoniae deficient in pneumo- of Streptococcus pneumoniae virulence factors in host respiratory coloniza-
lysin or autolysin has reduced virulence in meningitis. J. Infect. Dis. 197: tion and disease. Nat. Rev. Immunol. 6:288–301.
745–751.
127. Kantor, H. G. 1981. The many radiologic facies of pneumococcal pneumo-
105. Hori, R., H. Araki, M. Yonezawa, S. Minami, and Y. Watanabe. 2000.
nia. Am. J. Roentgenol. 137:1213–1220.
Therapeutic effects of parenteral beta-lactam antibiotics on experimental
128. Kastenbauer, S., U. Koedel, and H.-W. Pfister. 1999. Role of peroxynitrite
otitis media caused by penicillin-resistant Streptococcus pneumoniae in guinea-
as a mediator of pathophysiological alterations in experimental pneumo-
pigs. J. Antimicrob. Chemother. 45:311–314.
coccal meningitis. J. Infect. Dis. 180:1164–1170.
106. Horton, J., M. E. Ogden, S. Williams, and D. Coln. 1982. The importance
129. Kelly, T., J. P. Dillard, and J. Yother. 1994. Effect of genetic switching of
of splenic blood flow in clearing pneumococcal organisms. Ann. Surg.
capsular type on virulence of Streptococcus pneumoniae. Infect. Immun.
195:172–176.
62:1813–1819.
107. Hotomi, M., M. Suzumoto, K. Itahashi, J. Nagura, T. Fukushima, J. Shi-
mada, D. S. Billal, K. Yamauchi, K. Fujihara, and N. Yamanaka. 2007. 130. Kerr, A., J. J. Irvine, J. J. Search, N. A. Gingles, A. Kadioglu, P. W. Andrew,
Efficacy of a novel oral carbapenem, tebipenem pivoxil (TBM-PI), against W. L. McPheat, G. C. Booth, and T. J. Mitchell. 2002. Role of inflammatory
experimental otitis media caused by penicillin resistant Streptococcus pneu- mediators in resistance and susceptibility to pneumococcal infection. Infect.
moniae in chinchilla. Vaccine 25:2478–2484. Immun. 70:1547–1557.
108. Huang, Y., P. Ji, A. Inano, Z. Yang, G. S. Giebink, and R. J. Sawchuk. 2001. 131. Kerr, A. R., G. K. Paterson, J. McCluskey, F. Iannelli, M. R. Oggioni, G.
Microdialysis studies of the middle ear distribution kinetics of amoxicillin in Pozzi, and T. J. Mitchell. 2006. The contribution of PspC to pneumococcal
the awake chinchilla. J. Pharm. Sci. 90:2088–2098. virulence varies between strains and is accomplished by both complement
109. Iannelli, F., D. Chiavolini, S. Ricci, M. R. Oggioni, and G. Pozzi. 2004. evasion and complement-independent machanisms. Infect. Immun. 74:5319–
Pneumococcal surface protein C contributes to sepsis caused by Streptococ- 5324.
cus pneumoniae in mice. Infect. Immun. 72:3077–3080. 132. Kim, Y. S., R. A. Sheldon, B. R. Elliott, Q. Liu, D. M. Ferriero, and M. G.
110. Idanpaan-Heikkila, I., P. M. Simon, D. Zopf, T. Vullo, P. Cahill, K. Sokol, Tauber. 1995. Brain injury in experimental neonatal meningitis due to
and E. Tuomanen. 1997. Oligosaccharides interfere with the establishment group B streptococci. J. Neuropathol. Exp. Neurol. 54:531–539.
and progression of experimental pneumococcal pneumonia. J. Infect. Dis. 133. Klein, M., C. Schmidt, S. Kastenbauer, R. Paul, C. J. Kirschning, H.
176:704–712. Wagner, B. Popp, H.-W. Pfister, and U. Koedel. 2007. MyD88-dependent
111. Iinuma, H., and K. Okinaga. 1989. Prevention of pneumococcal bacteremia immune response contributes to hearing loss in experimental pneumococ-
by immunization with type 6 pneumococcal capsular polysaccharide vaccine cal meningitis. J. Infect. Dis. 195:1189–1193.
in splenectomized rats. J. Infect. Dis. 160:66–75. 134. Klein, M., J. R. Paul, B. Angele, B. Popp, H.-W. Pfister, and U. Koedel.
112. Iizawa, Y., K. Hiroe, M. Nakao, and K. Okonogi. 1998. Therapeutic efficacy 2006. Protein expression pattern in experimental pneumococcal meningitis.
of cefozopran in a murine model of haematogenous pneumococcal menin- Microbes Infect. 8:974–983.
gitis. Chemotherapy 44:265–271. 135. Klugman, K. P., S. Madhi, and C. Feldman. 2007. HIV and pneumocococ-
113. Iizawa, Y., K. Kitamoto, K. Hiroe, and M. Nakao. 1996. Streptococcus cal disease. Curr. Opin. Infect. Dis. 20:11–15.
pneumoniae in the nasal cavity of mice causes lower respiratory tract infec- 136. Koedel, U., R. Paul, F. Winkler, S. Kastenbauer, P. L. Huang, and H.-W.
tion after airway obstruction. J. Med. Microbiol. 44:490–495. Pfister. 2001. Lack of endothelial nitric oxide synthase aggravates murine
114. Iwasaki, K., T. Ohrui, Q. Wang, K. Sekizawa, T. Akaike, H. Maeda, and H. pneumococcal meningitis. J. Neuropathol. Exp. Neurol. 60:1041–1050.
Sasaki. 1999. Combined effects of both bacteria and gastric juice on pneu- 137. Koedel, U., W. M. Scheld, and H.-W. Pfister. 2002. Pathogenesis and patho-
monia in mice. Respir. Physiol. 116:201–209. physiology of pneumococcal meningitis. Lancet Infect. Dis. 2:721–736.
115. Jakobsen, H., S. Bjarnarson, G. Del Giudice, M. Moreau, C. A. Siegrist, 138. Koedel, U., F. Winkler, B. Angele, A. Fontana, R. A. Flavell, and H. W.
and I. Jónsdóttir. 2002. Intranasal immunization with pneumococcal con- Pfister. 2002. Role of caspase-1 in experimental pneumococcal meningitis:
jugate vaccines with LT-K63, a nontoxic mutant of heat-labile enterotoxin, evidence from pharmacologic caspase inhibition and caspase-1-deficient
as adjuvant rapidly induces protective immunity against lethal pneumococ- mice. Ann. Neurol. 51:319–329.
cal infections in neonatal mice. Infect. Immun. 70:1443–1452. 139. Kostyukova, N. N., M. O. Volkova, V. V. Ivanova, and A. S. Kvetnaya. 1995.
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 683

A study of pathogenic factors of Streptococcus pneumoniae strains causing Fischetti. 2007. Novel strategy to prevent otitis media caused by colonizing
meningitis. FEMS Immunol. Med. Microbiol. 10:133–138. Streptococcus pneumoniae. PloS Pathog. 3:e28.
140. Kuikka, A., J. Syrjanen, O. V. Renkonen, and V. V. Valtonen. 1992. Pneu- 163. McDaniel, L. S., W. H. Benjamin, C. Forman, and D. E. Briles. 1984. Blood
mococcal bacteraemia during a recent decade. J. Infect. 24:157–168. clearance by anti-phosphocholine antibodies as a mechanism of protection
141. Lacy, M. K., D. P. Nicolau, M. A. Banevicius, C. H. Nightingale, and R. in experimental pneumococcal bacteremia. J. Immunol. 133:3308–3312.
Quintiliani. 1999. Protective effect of trovafloxacin, ciprofloxacin and am- 164. Melhus, Å., and A. F. Ryan. 2003. A mouse model for acute otitis media.
picillin against Streptococcus pneumoniae in a murine sepsis model. J. An- APMIS 111:989–994.
timicrob. Chemother. 44:477–481. 165. Meli, D. N., S. Christen, and S. L. Leib. 2003. Matrix metalloproteinase-9
142. Lanie, J. A., W. L. Ng, K. M. Kazmierczak, T. M. Andrzejewski, T. M. in pneumococcal meningitis: activation via an oxidative pathway. J. Infect.
Davidsen, K. J. Wayne, H. Tettelin, J. I. Glass, and M. E. Winkler. 2007. Dis. 187:1411–1415.
Genome sequence of Avery’s virulent serotype 2 strain D39 of Streptococ- 166. Mellencamp, M. A., and L. C. Preheim. 1991. Pneumococcal pneumonia in
cus pneumoniae and comparison with that of unencapsulated laboratory a rat model of cirrhosis: effects of cirrhosis on pulmonary defense mecha-
strain R6. J. Bacteriol. 189:38–51. nisms against Streptococcus pneumoniae. J. Infect. Dis. 163:102–108.
143. Leib, S. L., J. M. Clements, R. L. P. Lindberg, C. Heimgartner, J. M. 167. Mizgerd, J. P., B. B. Meek, G. J. Kutkoski, D. C. Bullard, A. L. Beaudet,

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


Loeffler, L.-A. Pfister, M. G. Tauber, and D. Leppert. 2001. Inhibition of and C. M. Doerschuk. 1996. Selectins and neutrophil traffic: margination
matrix metalloproteinases and tumour necrosis factor ␣ converting enzyme and Streptococcus pneumoniae-induced emigration in murine lungs. J. Exp.
as adjuvant therapy in pneumococcal meningitis. Brain 124:1734–1742. Med. 184:639–645.
144. Leib, S. L., C. Heimgartner, Y.-D. Bifrare, J. M. Loeffler, and M. G. Tauber. 168. Möhler, J., E. Azoulay-Dupuis, C. Amory-Rivier, J. X. Mazoit, J. P. Bedos,
2003. Dexamethasone aggravates hippocampal apoptosis and learning de- V. Rieux, and P. Moine. 2003. Streptococcus pneumoniae strain-dependent
ficiency in pneumococcal meningitis in infant rats. Pediat. Res. 54:353–357. lung inflammation responses in a murine model of pneumococcal pneumo-
145. Leib, S. L., D. Leppert, J. Clements, and M. G. Tauber. 2000. Matrix nia. Int. Care Med. 29:808–816.
metalloproteinases contribute to brain damage in experimental pneumo- 169. Moine, P., E. Vallee, E. Azoulay-Dupuis, P. Bourget, J. P. Bèdos, J.
coccal meningitis. Infect. Immun. 68:615–620. Bauchet, and J. J. Pocidalo. 1994. In vivo efficacy of a broad-spectrum
146. Leib, S. L., and M. G. Tauber. 1999. Pathogenesis of bacterial meningitis. cephalosporin, ceftriaxone, against penicillin-susceptible and -resistant
Infect. Dis. Clin. North Am. 13:527–548. strains of Streptococcus pneumoniae in a mouse pneumonia model. Anti-
147. Leung, L. S. E., G. J. Szal, and R. H. Drachman. 1972. Increased suscep- microb. Agents Chemother. 38:1953–1958.
tibility of splenectomized rats to infection with Diplococcus pneumoniae. 170. Muffat-Joly, M., B. Barry, D. Henin, M. Fay, P. Gehanno, and J. J. Po-
J. Infect. Dis. 126:507–513. cidalo. 1994. Otogenic meningoencephalitis induced by Streptococcus pneu-
148. Lindquist, L., P. Lundbergh, K. G. Hedström, L. O. Hansson, and E. moniae in gerbils. Arch. Otolaryngol. Head Neck Surg. 120:925–930.
Hultman. 1987. Experimental bacterial meningitis in the rabbit: cerebro- 171. Musher, D. M., H. Phan, and R. E. Baughn. 2001. Protection against
spinal fluid changes and its relation to leukocyte response. Scand. J. Infect. bacteremic pneumococcal infection by antibody to pneumolysin. J. Infect.
Dis. 19:263–270. Dis. 183:827–830.
149. Loeffler, J. M., S. Djurkovic, and V. A. Fischetti. 2003. Phage lytic enzyme 172. Nau, R., K. Kaye, M. Sachdeva, E. R. Sande, and M. G. Tauber. 1994.
Cpl-1 as a novel antimicrobial for pneumococcal bacteremia. Infect. Im- Rifampin for therapy of experimental pneumococcal meningitis in rabbits.
mun. 71:6199–6204. Antimicrob. Agents Chemother. 38:1186–1189.
150. Long, J. P., H. H. Tong, and T. F. DeMaria. 2004. Immunization with native 173. Nau, R., A. Wellmer, A. Soto, K. Koch, O. Schneider, H. Schmidt, J. Gerber,
or recombinant Streptococcus pneumoniae neuroaminidase affords protec- U. Michel, and W. Bruck. 1999. Rifampin reduces early mortality in exper-
tion in the chinchilla otitis media model. Infect. Immun. 72:4309–4313. imental Streptococcus pneumoniae meningitis. J. Infect. Dis. 179:1557–1560.
151. Long, J. P., H. H. Tong, P. A. Shannon, and T. F. DeMaria. 2003. Differ- 174. Nonomura, N., G. S. Giebink, S. K. Juhn, T. Harada, and D. Aeppli. 1991.
ential expression of cytokine genes and inducible nitric oxide synthase Pathophysiology of Streptococcus pneumoniae otitis media: kinetics of the
induced by opacity phenotype variants of Streptococcus pneumoniae during middle ear biochemical and cytologic host responses. Ann. Otol. Rhinol.
acute otitis media in the rat. Infect. Immun. 71:5531–5540. Laryngol. 100:236–243.
152. MacArthur, C. J., S. H. Hefeneider, J. B. Kempton, S. K. Parrish, S. L. 175. Nuermberger, E. 2005. Murine models of pneumococcal pneumonia and
McCoy, and D. R. Trune. 2006. Evaluation of the mouse model for acute their applicability to the study of tissue-directed antimicrobials. Pharmaco-
otitis media. Hear. Res. 219:12–23. therapy 25:134S–139S.
153. Madhi, S. A., and S. I. Pelton. 2008. Epidemiology, diagnosis, and treat- 176. Nuermberger, E., K. Helke, and W. R. Bishai. 2005. Low-dose aerosol
ment of serious pneumococcal infections in children, p. 95–116. In G. R. model of pneumococcal pneumonia in the mouse: utility for evaluation of
Siber, K. P. Klugman, and P. H. Mäkelä (ed.), Pneumococcal vaccines: the antimicrobial efficacy. Int. J. Antimicrob. Agents 26:497–503.
impact of conjugate vaccine. ASM Press, Washington, DC. 177. Obaro, S., and R. Adegbola. 2002. The pneumococcus: carriage, disease and
154. Magnusdóttir, A. B., A. Hermanssön, and Å. Melhus. 2000. Experimental conjugate vaccines. J. Med. Microbiol. 51:98–104.
study of the virulence of Streptococcus pneumoniae with reduced suscepti- 178. O’Brien, D. P., D. E. Briles, A. J. Szalai, A. H. Tu, I. Sanz, and M. H. Nahm.
bility to penicillin. Int. J. Ped. Otorinolaryngol. 55:1–9. 1999. Tumor necrosis factor alpha receptor I is important for survival from
155. Malley, R., M. Lipsitch, A. Stack, R. Saladino, G. Fleisher, S. Pelton, C. Streptococcus pneumoniae infections. Infect. Immun. 67:595–601.
Thomson, D. E. Briles, and P. Anderson. 2001. Intranasal immunization 179. Offenbartl, K., P. Christensen, P. Gullstrand, K. Prellner, and R. A. Seger.
with killed unencapsulated whole cells prevents colonization and invasive 1986. Treatment of pneumococcal postsplenectomy sepsis in the rat with
disease by capsulated pneumococci. Infect. Immun. 69:4870–4873. human gamma-globulin. J. Surg. Res. 40:198–201.
156. Malley, R., A. M. Stack, R. N. Husson, C. Thompson, G. R. Fleisher, and 180. Ogunniyi, D. A., R. L. Folland, D. E. Briles, S. K. Hollingshead, and J. C.
R. Saladino. 2004. Development of a model of focal pneumococcal pneu- Paton. 2000. Immunization of mice with combinations of pneumococcal
monia in young rats. J. Immune Based Ther. Vaccines 2:2. virulence proteins elicites enhanced protection against challenge with Strep-
157. Malley, R., and J. N. Weiser. 2008. Animal models of pneumococcal colo- tococcus pneumoniae. Infect. Immun. 68:3028–3033.
nization, p. 59–66. In G. R. Siber, K. P. Klugman, and P. H. Mäkelä (ed.), 181. Ogunniyi, D. A., M. Grabowicz, D. E. Briles, J. Cook, and J. C. Paton. 2007.
Pneumococcal vaccines: the impact of coniugate vaccine. ASM Press, Development of a vaccine against invasive pneumococcal disease based on
Washington, DC. combinations of virulence proteins of Streptococcus pneumoniae. Infect.
158. Manco, S., F. Hernon, H. Yesilkaya, J. C. Paton, P. W. Andrew, and A. Immun. 75:350–357.
Kadioglu. 2006. Pneumococcal neuraminidases A and B both have essential 182. Ogunniyi, D. A., K. S. Le Messurier, R. M. Graham, J. M. Watt, D. E.
roles during infection of the respiratory tract and sepsis. Infect. Immun. Briles, U. H. Stroeher, and J. C. Paton. 2007. Contributions of pneumoly-
74:4014–4020. sin, pneumococcal surface protein A (PspA), and PspC to pathogenicity of
159. Mandell, L. A., R. G. Wunderink, A. Anzueto, J. G. Bartlett, G. D. Camp- Streptococcus pneumoniae D39 in a mouse model. Infect. Immun. 75:1843–
bell, N. C. Dean, S. F. Dowell, T. M. File, D. M. Musher, M. S. Niederman, 1851.
A. Torres, C. G. Whitney, Infectious Diseases Society of America, and 183. Oliveira, M. L., A. P. Areas, I. B. Campos, V. Monedero, G. Perez-Martinez,
American Thoracic Society. 2007. Infectious Diseases Society of America/ E. N. Miyaji, L. C. Leite, K. A. Aires, and P. Lee Ho. 2006. Induction of
American Thoracic Society consensus guidelines on the management of systemic and mucosal immune response and decrease in Streptococcus
community-acquired pneumonia in adults. Clin. Infect. Dis. 44:S27–S72. pneumoniae colonization by nasal inoculation of mice with recombinant
160. Marra, A., and D. Brigham. 2001. Streptococcus pneumoniae causes exper- lactic acid bacteria expressing pneumococcal surface antigen A. Microbes
imental meningitis following intranasal and otitis media infections via a Infect. 8:1016–1024.
nonhematogenous route. Infect. Immun. 69:7318–7325. 184. Olsen, K. M., M. J. Gentry-Nielsen, M. Yue, M. U. Snitily, and L. C.
161. McCoy, S. L., S. E. Kurtz, C. J. MacArthur, D. R. Trune, and S. H. Preheim. 2006. Effect of ethanol on fluoroquinolone efficacy in a rat model
Hefeneider. 2005. Identification of a peptide derived from vaccinia virus of pneumococcal pneumonia. Antimicrob. Agents Chemother. 50:210–219.
A52R protein that inhibits cytokine secretion in response to TLR-depen- 185. Onofrio, J. M., G. B. Toews, M. F. Lipscomb, and A. K. Pierce. 1983.
dent signaling and reduces in vivo bacterial-induced inflammation. J. Im- Granulocyte-alveolar-macrophage interaction in the pulmonary clearance
munol. 174:3006–3014. of Staphylococcus aureus. Am. Rev. Respir. Dis. 127:335–341.
162. McCullers, J. A., A. Karlström, A. R. Iverson, J. M. Loeffler, and V. A. 186. Orihuela, C. J., G. Gao, K. P. Francis, J. Yu, and E. I. Tuomanen. 2004.
684 CHIAVOLINI ET AL. CLIN. MICROBIOL. REV.

Tissue-specific contributions of pneumococcal virulence factors to patho- Alexander, P. W. Andrew, T. J. Mitchell, and E. N. Janoff. 1996. Distinct
genesis. J. Infect. Dis. 190:1661–1669. roles for pneumolysin’s cytotoxic and complement activities in the patho-
187. Orihuela, C. J., G. Gao, M. McGhee, J. Yu, K. P. Francis, and E. Tuo- genesis of pneumococcal pneumonia. Am. J. Respir. Crit. Care Med. 153:
manen. 2003. Organ-specific models of Streptococcus pneumoniae disease. 1339–1346.
Scand. J. Infect. Dis. 35:647–652. 209. Rupprecht, T., B. Angele, M. Klein, J. Heesemann, H.-W. Pfister, M. Botto,
188. Ort, S., J. L. Ryan, G. Barden, and N. D’Esopo. 1983. Pneumococcal and U. Koedel. 2007. Complement C1q and C3 are critical for the innate
pneumonia in hospitalized patients. Clinical and radiological presentations. immune response to Streptococcus pneumoniae in the central nervous sys-
JAMA 249:214–218. tem. J. Immunol. 178:1861–1869.
189. Østergaard, C., T. Klitmøller Sørensen, J. Dahl Knudsen, and N. Frimodt- 210. Ryan, A. F., J. Ebmeyer, M. Furukawa, K. Pak, Å. Melhus, S. I. Wasserman,
Møller. 1998. Evaluation of moxifloxacin, a new 8-methoxyquinolone, for and W.-H. Chung. 2006. Mouse models of induced otitis media. Brain Res.
treatment of meningitis caused by a penicillin-resistant pneumococcus in 1091:3–8.
rabbits. Antimicrob. Agents Chemother. 42:1706–1712. 211. Sabirov, A., and D. W. Metzger. 2008. Mouse models for the study of
190. Parra, A., C. Ponte, C. Cenjor, G. Garcia-Calvo, M. J. Gimenez, L. Aguilar, mucosal vaccination against otitis media. Vaccine 26:1501–1524.
and F. Soriano. 2002. Optimal dose of amoxicillin in treatment of otitis 212. Sabirov, A., and D. W. Metzger. 2006. Intranasal vaccination of neonatal

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


media caused by a penicillin-resistant pneumococcus stran in the gerbil mice with polysaccharide conjugate vaccine for protection against pneumo-
model. Antimicrob. Agents Chemother. 46:859–862. coccal otitis media. Vaccine 24:5584–5592.
191. Paul, R., U. Koedel, and H.-W. Pfister. 2005. Development of adjunctive 213. Saeland, E., H. Jakobsen, G. Ingolfsdóttir, S. T. Sigurdardóttir, and I.
therapies for bacterial meningitis and lessons from knockout mice. Neuro- Jónsdóttir. 2001. Serum samples from infants vaccinated with a pneumo-
crit. Care 2:313–324. coccal conjugate vaccine, PncT, protect mice against invasive infection
192. Pfister, H.-W., K. Frei, B. Ottnad, U. Koedel, A. Tomasz, and A. Fontana. caused by Streptococcus pneumoniae serotypes 6A and 6B. J. Infect. Dis.
1992. Transforming growth factor beta 2 inhibits cerebrovascular changes 183:253–260.
and brain edema formation in the tumor necrosis factor alpha-independent 214. Saladino, R. A., A. M. Stack, G. R. Fleisher, C. M. Thompson, D. E. Briles,
early phase of experimental pneumococcal meningitis. J. Exp. Med. 176: L. Kobzik, and G. R. Siber. 1997. Development of a model of low-inoculum
265–268. Streptococcus pneumoniae intrapulmonary infection in infant rats. Infect.
193. Pfister, H.-W., U. Koedel, R. L. Haberl, U. Dirnagl, W. Feiden, G. Ruck- Immun. 65:4701–4704.
deschel, and K. M. Einhaupl. 1990. Microvascular changes during the early 215. Sandgren, A., B. Albiger, C. J. Orihuela, E. Tuomanen, S. Normark, and B.
phase of experimental bacterial meningitis. J. Cereb. Blood Flow Metab. Henriques-Normark. 2005. Virulence in mice of pneumococcal clonal types
10:914–922. with known invasive disease potential in humans. J. Infect. Dis. 192:791–
194. Pfister, L. A., J. H. Tureen, S. Shaw, S. Christen, D. M. Ferriero, M. G. 800.
Tauber, and S. L. Leib. 2000. Endothelin inhibition improves cerebral 216. Sato, K., C. L. Liebeler, M. K. Quartey, C. T. Le, and G. S. Giebink. 1999.
blood flow and is neuroprotective in pneumococcal meningitis. Ann. Neu- Middle ear fluid cytokine and inflammatory cell kinetics in the chinchilla
rol. 47:329–335. otitis media model. Infect. Immun. 67:1943–1946.
195. Piroth, L., N. Desbiolles, V. Mateo-Ponce, L. Martin, C. Lequeu, P. E. 217. Sato, K., M. K. Quartey, C. L. Liebeler, C. T. Le, and G. S. Giebink. 1996.
Charles, H. Portier, and P. Chavanet. 2001. HMR 3647 human-like treat- Roles of autolysin and pneumolysin in middle ear inflammation caused by
ment of experimental pneumonia due to penicillin-resistant and erythro- a type 3 Streptococcus pneumoniae strain in the chinchilla otitis media
mycin-resistant Streptococcus pneumoniae. J. Antimicrob. Chemother. 47: model. Infect. Immun. 64:1140–1145.
33–42. 218. Schlüter, D., S. Chahoud, H. Lassmann, A. Schumann, H. Hof, and M.
196. Piroth, L., L. Martin, A. Coulon, C. Lequeu, M. Duong, M. Buisson, H. Deckert-Schlüter. 1996. Intracerebral targets and immunomodulation in
Portier, and P. Chavanet. 1999. Development of a new experimental model murine Listeria monocytogenes meningoencephalitis. J. Neuropathol. Exp.
of penicillin-resistant Streptococcus pneumoniae pneumonia and amoxicillin Neurol. 55:14–24.
treatment by reproducing human pharmacokinetics. Antimicrob. Agents 219. Schutze, G. E., E. O. J. Mason, W. J. Barson, K. S. Kim, E. R. Wald, L. B.
Chemother. 43:2484–2492. Givner, T. Q. Tan, J. S. Bradley, R. Yogev, and S. L. Kaplan. 2002. Invasive
197. Polissi, A., A. Pontiggia, G. Feger, M. Altieri, H. Mottl, L. Ferrari, and D. pneumococcal infections in children with asplenia. Pediatr. Infect. Dis. J.
Simon. 1998. Large-scale identification of virulence genes from Streptococ- 21:278–282.
cus pneumoniae. Infect. Immun. 66:5620–5629. 220. Shapiro, M. A., K. D. Donovan, and J. W. Gage. 2000. Comparative efficacy
198. Pracht, D., C. Elm, G. Gerber, S. Bergmann, M. Rohde, M. Seiler, K. S. of clinafloxacin in a pneumococcal meningitis mouse model. J. Antimicrob.
Kim, H. F. Jenkinson, R. Nau, and S. Hammerschmidt. 2005. PavA of Chemother. 45:489–492.
Streptococcus pneumoniae modulates adherence, invasion, and meningeal 221. Smith, G. M., and K. H. Abbott. 1994. Development of experimental re-
inflammation. Infect. Immun. 73:2680–2689. spiratory infections in neutropenic rats with either penicillin-resistant Strep-
199. Preheim, L. C., K. M. Olsen, M. Yue, M. U. Snitily, and M. J. Gentry- tococcus pneumoniae or ␤-lactamase-producing Haemophilus influenzae.
Nielsen. 2005. Effect of cirrhosis on antibiotic efficacy in a rat model of Antimicrob. Agents Chemother. 38:608–610.
pneumococcal pneumonia. Diagn. Microbiol. Infect. Dis. 51:103–111. 222. Smith, G. M., B. Slocombe, K. H. Abbott, and L. W. Mizen. 1998. Activity
200. Quin, L. R., Q. C. Moore III, and L. S. McDaniel. 2007. Pneumolysin, PspA, of amoxicillin-clavulanate against penicillin-resistant Streptococcus pneu-
and PspC contribute to pneumococcal evasion of early innate immune moniae in an experimental respiratory infection model in rats. Antimicrob.
responses during bacteremia in mice. Infect. Immun. 75:2067–2070. Agents Chemother. 42:813–817.
201. Ramisse, F., P. Binder, M. Szatanik, and J. M. Alonso. 1996. Passive and 223. Soriano, F., C. Ponte, E. Nieto, and A. Parra. 1996. Correlation of in vitro
active immunotherapy for experimental pneumococcal pneumonia by poly- activity and pharmacokinetics parameters with in vivo effect of amoxycillin,
valent human immunoglobulin or F(ab⬘)2 fragments administered intrana- co-amoxiclav and cefotaxime in a murine model of pneumococcal pneumo-
sally. J. Infect. Dis. 173:1123–1128. nia. J. Antimicrob. Chemother. 38:227–236.
202. Ren, B., A. Szalai, S. K. Hollingshead, and D. E. Briles. 2003. Both family 224. Stack, A. M., R. Malley, C. M. Thompson, L. Kobzik, G. R. Siber, and R. A.
1 and family 2 PspA proteins can inhibit complement deposition and confer Saladino. 1998. Minimum protective serum concentrations of pneumococ-
virulence to a capsular serotype 3 strain of Streptococcus pneumoniae. In- cal anti-capsular antibodies in infant rats. J. Infect. Dis. 177:986–990.
fect. Immun. 71:75–85. 225. Steinhoff, M. C., and The Pneumococcal Vaccine Animal Model Consensus
203. Ribes, S., F. Taberner, A. Domenech, C. Cabellos, F. Tubau, J. Linares, Group. 2007. Animal models for protein pneumococcal vaccine evaluation:
P. F. Viladrich, and F. Gudiol. 2006. Evaluation of fosfomycin alone and in a summary. Vaccine 25:2465–2470.
combination with ceftriaxone or vancomycin in an experimental model of 226. Stucki, A., M. Cottagnoud, V. Winkelmann, T. Schaffner, and P. Cottag-
meningitis caused by two strains of cefalosphorin-resistant Streptococcus noud. 2007. Daptomycin produces an enhanced bactericidal activity com-
pneumoniae. Antimicrob. Agents Chemother. 57:931–936. pared to ceftriaxone, measured by [3H]choline release in the cerebrospinal
204. Richter, M. Y., H. Jakobsen, A. Birgisdóttir, J. F. Haeuw, U. F. Power, G. fluid, in experimental meningitis due to a penicillin-resistant pneumococcal
Del Giudice, A. Bartoloni, and I. Jónsdóttir. 2004. Immunization of female strain without lysing the cell wall. Antimicrob. Agents Chemother. 51:2249–
mice with glycoconjugates protects their offspring against encapsulated 2252.
bacteria. Infect. Immun. 72:187–195. 227. Swiatlo, E., J. King, G. S. Nabors, B. Mathews, and D. E. Briles. 2003.
205. Roche, A. M., S. J. King, and J. N. Weiser. 2007. Live attenuated Strepto- Pneumococcal surface protein A is expressed in vivo, and antibodies to
coccus pneumoniae strains induce serotype-independent mucosal and sys- PspA are effective for therapy in a murine model of pneumococcal sepsis.
temic protection in mice. Infect. Immun. 75:2469–2475. Infect. Immun. 71:7149–7153.
206. Rodriguez, A. F., S. L. Kaplan, E. P. Hawkins, and E. O. J. Mason. 1991. 228. Takashima, K., K. Tateda, T. Matsumoto, Y. Iizawa, M. Nakao, and K.
Hematogenous pneumococcal meningitis in the infant rat: description of a Yamaguchi. 1997. Role of tumor necrosis factor alpha in pathogenesis of
model. J. Infect. Dis. 164:1207–1209. pneumococcal pneumonia in mice. Infect. Immun. 65:257–260.
207. Rubins, J. B., and D. Charboneau. 2000. Effect of anesthetics on patho- 229. Tamura, S., S. Miyazaki, K. Tateda, A. Ohno, Y. Ishii, T. Matsumoto, N.
genesis of experimentally induced murine pneumococcal pneumonia. Furuya, and K. Yamaguchi. 1998. In vivo antibacterial activities of sanfetri-
Comp. Med. 50:292–295. nem cilexetil, a new oral tricyclic antibiotic. Antimicrob. Agents Che-
208. Rubins, J. B., D. Charboneau, C. Fasching, A. M. Berry, J. C. Paton, J. E. mother. 42:1858–1861.
VOL. 21, 2008 MODELS OF PNEUMOCOCCAL DISEASE 685

230. Tan, T. Q., C. W. Smith, E. P. Hawkins, E. O. J. Mason, and S. L. Kaplan. kinetics in interleukin-10-treated murine pneumococcal pneumonia. J. An-
1995. Hematogenous bacterial meningitis in an intercellular adhesion mol- timicrob. Chemother. 55:721–726.
ecule-1-deficient infant mouse model. J. Infect. Dis. 171:342–349. 251. Wang, E., N. Ouellet, M. Simard, I. Fillion, Y. Bergeron, D. Beauchamp,
231. Tarasi, A., L. L. Dever, and A. Tomasz. 1997. Activity of quinupristin/ and M. G. Bergeron. 2001. Pulmonary and systemic host response to Strep-
dalfopristin against Streptococcus pneumoniae in vitro and in vivo in the tococcus pneumoniae and Klebsiella pneumoniae bacteremia in normal and
rabbit model of experimental meningitis. J. Antimicrob. Chemother. 39: immunosuppressed mice. Infect. Immun. 69:5294–5304.
121–127. 252. Wang, E., M. Simard, N. Ouellet, Y. Bergeron, D. Beauchamp, and M. G.
232. Tasaka, S., S. E. Richer, J. P. Mizgerd, and C. M. Doerschuk. 2002. Very Bergeron. 2000. Modulation of cytokines and chemokines, limited pulmo-
late antigen-4 in CD18-independent neutrophil emigration during acute nary vascular bed permeability, and prevention of septicemia and death
bacterial pneumonia in mice. Am. J. Respir. Crit. Care Med. 166:53–60. with ceftriaxone and interleukin-10 in pneumococcal pneumonia. J. Infect.
233. Tateda, K., T. Matsumoto, S. Miyazaki, and K. Yamaguchi. 1999. Efficacy Dis. 182:1255–1259.
of ␤-lactam antibiotics combined with gentamicin against penicillin-resis- 253. Wei, B. P., R. K. Shepherd, R. M. Robins-Browne, G. M. Clark, and S. J.
tant pneumococcal pneumonia in CBA/J mice. J. Antimicrob. Chemother. O’Leary. 2006. Pneumococcal meningitis: development of a new animal
43:367–371. model. Otol. Neurotol. 27:844–854.

Downloaded from http://cmr.asm.org/ on October 22, 2020 at NUS CENTRAL LIB


234. Tateda, K., K. Takashima, H. Miyazaki, T. Matsumoto, T. Hatori, and K. 254. Wellmer, A., C. Noeske, J. Gerber, U. Menzel, and R. Nau. 2000. Spatial
Yamaguchi. 1996. Noncompromised penicillin-resistant pneumococcal memory and learning deficits after experimental pneumococcal meningitis
pneumonia CBA/J mouse model and comparative efficacies of antibiotics in in mice. Neurosci. Lett. 296:137–140.
this model. Antimicrob. Agents Chemother. 40:1520–1525. 255. Wellmer, A., G. Zysk, J. Gerber, T. Kunst, M. von Mering, S. Bunkowski,
235. Tauber, M. G., and M. A. Sande. 1984. Pathogenesis of bacterial meningitis: H. Eiffert, and R. Nau. 2002. Decreased virulence of a pneumolysin-defi-
contributions by experimental models in rabbits. Infection 12:S3–S10. cient strain of Streptococcus pneumoniae in murine meningitis. Infect. Im-
236. Tauber, M. G., and A. Zwahlen. 1994. Animal models for meningitis. Meth- mun. 70:6504–6508.
ods Enzymol. 235:93–106. 256. White, P., A. Hermansson, C. Svanborg, D. E. Briles, and K. Prellner. 1999.
237. Tessier, P. R., M. K. Kim, W. Zhou, D. Xuan, C. Li, M. Ye, C. H. Night- Effects of active immunization with a pneumococcal surface protein (PspA)
ingale, and D. P. Nicolau. 2002. Pharmacodynamic assessment of clarithro- and of locally applied antibodies in experimental otitis media. J. Otorhino-
mycin in a murine model of pneumococcal pneumonia. Antimicrob. Agents laryngol. Relat. Spec. 61:206–211.
Chemother. 46:1425–1434. 257. Wu, H. Y., M. H. Nahm, Y. Guo, M. W. Russell, and D. E. Briles. 1997.
238. Tettelin, H., K. E. Nelson, I. T. Paulsen, J. A. Eisen, T. D. Read, S. Peterson, Intranasal immunization of mice with PspA (pneumococcal surface protein
J. Heidelberg, R. T. DeBoy, D. H. Haft, R. J. Dodson, A. S. Durkin, M. A) can prevent intranasal carriage, pulmunary infection, and sepsis with
Gwinn, J. F. Kolonay, W. C. Nelson, J. D. Peterson, L. A. Umajama, O. Streptococcus pneumoniae. J. Infect. Dis. 175:839–846.
White, S. L. Salzberg, M. R. Lewis, D. Radune, E. Holtzapple, H. Khouri, 258. Wu, H., A. Virolainen, B. Mathews, J. King, M. W. Russel, and D. E. Briles.
A. M. Wolf, T. R. Utterback, C. L. Hansen, L. A. McDonald, T. V. Feld- 1997. Establishment of a Streptococcus pneumoniae nasopharyngeal coloni-
blyum, S. Angiuoli, T. Dickinson, E. K. Hickey, I. E. Holt, B. J. Loftus, F. zation model in adult mice. Microb. Pathog. 23:127–137.
Yang, H. O. Smith, J. C. Venter, B. A. Dougherty, D. A. Morrison, S. K.
259. Yamaguchi, T., O. Kasuga, S. Okuno, and I. Maezawa. 1989. An experi-
Hollingshead, and C. M. Fraser. 2001. Complete genome sequence of a
mental meningitis with Streptococcus pneumoniae in rats: the therapeutic
virulent isolate of Streptococcus pneumoniae. Science 293:498–506.
effect of aspoxicillin and its penetration into CSF. Drugs Exp. Clin. Res.
239. Thore, M., L. G. Burman, and S. E. Holm. 1982. Streptococcus pneumoniae
15:297–301.
and three species of anaerobic bacteria in experimental otitis media in
260. Yoshimura, H., N. Watanabe, J. Bundo, M. Shinoda, and G. Mogi. 1991.
guinea pigs. J. Infect. Dis. 145:822–828.
Oral vaccine therapy for pneumococcal otitis media in an animal model.
240. Tong, H. H., L. E. Blue, M. A. James, and T. F. DeMaria. 2000. Evaluation
Arch. Otolaryngol. Head Neck Surg. 117:889–894.
of virulence of a Streptococcus pneumoniae neuraminidase-deficient mutant
in nasopharyngeal colonization and development of otitis media in the 261. Yother, J., C. Forman, B. M. Gray, and D. E. Briles. 1982. Protection of
chinchilla model. Infect. Immun. 68:921–924. mice from infection with Streptococcus pneumoniae by anti-phosphocholine
241. Trolle, S., E. Chachaty, N. Kassis-Chikhani, C. Wang, E. Fattal, P. Cou- antibody. Infect. Immun. 36:184–188.
vreur, B. Diamond, J. M. Alonso, and A. Andremont. 2000. Intranasal 262. Yuste, J., M. Botto, J. C. Paton, D. W. Holden, and J. S. Brown. 2005. Additive
immunization with protein-linked phosphorylcholine protects mice against inhibition of complement deposition by pneumolysin and PspA facilitates
a lethal intranasal challenge with Streptococcus pneumoniae. Vaccine 18: Streptococcus pneumoniae septicemia. J. Immunol. 175:1813–1819.
2991–2998. 263. Yuste, J., I. Jado, M. J. Gimenez, L. Aguilar, F. Molero, A. Fenoll, and J.
242. Tsai, Y. H., M. Bies, F. Leitner, and R. E. Kessler. 1990. Therapeutic Casal. 2002. Modification of bacteraemia by specific antibodies in relation
studies of cefepime (BMY 28142) in murine meningitis and pharmacoki- with mortality in a pneumococcal mouse sepsis model. Clin. Exp. Immunol.
netics in neonatal rats. Antimicrob. Agents Chemother. 34:733–738. 128:411–415.
243. Tsao, N., W. W. Chang, C. C. Liu, and H. Y. Lei. 2002. Development of 264. Zwijnenburg, P. J. G., T. van der Poll, S. Florquin, S. Akira, K. Takeda, J. J.
hematogenous pneumococcal meningitis in adult mice: the role of TNF-␣. Roord, and A. M. van Furth. 2003. Interleukin-18 gene-deficient mice show
FEMS Immunol. Med. Microbiol. 32:133–140. enhanced defense and reduced inflammation during pneumococcal menin-
244. Tsao, N., H. P. Hsu, C. M. Wu, C. C. Liu, and H. Y. Lei. 2001. Tumour gitis. J. Neuroimmunol. 138:31–37.
necrosis factor-␣ causes an increase in blood-brain barrier permeability 265. Zwijnenburg, P. J. G., T. van der Poll, S. Florquin, M. M. J. Polfliet, T. K.
during sepsis. J. Med. Microbiol. 50:812–821. van den Berg, C. D. Dijkstra, J. J. Roord, C. E. Hack, and A. M. van Furth.
245. Tu, A. H., R. L. Fulgham, M. A. McCrory, D. E. Briles, and A. J. Szalai. 2007. C1 inhibitor treatment improves host defense in pneumococcal men-
1999. Pneumococcal surface protein A inhibits complement activation by ingitis in rats and mice. J. Infect. Dis. 196:115–123.
Streptococcus pneumoniae. Infect. Immun. 67:4720–4724. 266. Zwijnenburg, P. J. G., T. van der Poll, S. Florquin, J. J. Roord, and A. M.
246. Tuomanen, E., R. Rich, and O. Zak. 1987. Induction of pulmonary inflam- van Furth. 2003. IL-1 receptor type 1 gene-deficient mice demonstrate an
mation by components of the pneumococcal cell surface. Am. Rev. Respir. impaired host defense against pneumococcal meningitis. J. Immunol. 170:
Dis. 135:869–874. 4724–4730.
247. Tuomanen, E., A. Tomasz, B. Hengstler, and O. Zak. 1985. The relative role 267. Zwijnenburg, P. J. G., T. van der Poll, S. Florquin, J. J. Roord, and A. M.
of bacterial cell wall and capsule in the induction of inflammation in pneu- van Furth. 2003. Interleukin-10 negatively regulates local cytokine and
mococcal meningitis. J. Infect. Dis. 151:535–540. chemokine production but does not influence antibacterial host defense
248. van der Ven, L. T., G. P. van den Dobbelsteen, B. Nagarajah, H. van Dijken, during murine pneumococcal meningitis. Infect. Immun. 71:2276–2279.
P. M. Dortant, J. G. Vos, and P. J. M. Roholl. 1999. A new rat model of 268. Zwijnenburg, P. J. G., T. van der Poll, S. Florquin, S. J. H. van Deventer,
otitis media caused by Streptococcus pneumoniae: conditions and applica- J. J. Roord, and A. M. van Furth. 2001. Experimental pneumococcal men-
tion in immunization protocols. Infect. Immun. 67:6098–6103. ingitis in mice: a model of intranasal infection. J. Infect. Dis. 183:1143–
249. van Ginkel, F. W., J. R. McGhee, J. M. Watt, A. Campos-Torres, L. A. 1146.
Parish, and D. E. Briles. 2003. Pneumococcal carriage results in ganglio- 269. Zysk, G., W. Bruck, J. Gerber, Y. Bruck, H. W. Prange, and R. Nau. 1996.
side-mediated olfactory tissue infection. Proc. Natl. Acad. Sci. USA 100: Anti-inflammatory treatment influences neuronal apoptotic cell death in
14363–14367. the dentate gyrus in experimental pneumococcal meningitis. J. Neuro-
250. Wang, E., Y. Bergeron, and M. G. Bergeron. 2005. Ceftriaxone pharmaco- pathol. Exp. Neurol. 55:722–728.

You might also like