Sie sind auf Seite 1von 19

CLINICAL MICROBIOLOGY REVIEWS, Apr. 1993, p. 118-136 Vol. 6, No.

2
0893-8512/93/020118-19$02.00/0
Copyright © 1993, American Society for Microbiology

Pathogenesis and Pathophysiology of Bacterial Meningitis


ALLAN R. TUNKELl* AND W. MICHAEL SCHELD2
Department of Internal Medicine (Infectious Diseases), Medical College of Pennsylvania, Philadelphia,
Pennsylvania 19129,1 and Departments of Internal Medicine (Infectious Diseases) and Neurosurgety,
University of Virginia Health Sciences Center, Charlottesville, Virginia 229082
INTRODUCTION ...................................................... 118
Epidemiology ...................................................... 118
Animal Models ...................................................... 119
MUCOSAL COLONIZATION AND SYSTEMIC INVASION ...................................................... 119
Fimbriae ...................................................... 119
Polysaccharide Capsule ...................................................... 120
Antibodies ...................................................... 120
Other Bacterial Components ...................................................... 121
BACTEREMIA ...................................................... 121
Polysaccharide Capsule ...................................................... 121
Host Defense Mechanisms ...................................................... 121
MENINGEAL INVASION ...................................................... 121
Site of Invasion ...................................................... 121
Fimbriae ...................................................... 122
Monocytes ...................................................... 122
Other Bacterial Components ...................................................... 122
Secondary Bacteremia ...................................................... 122
ALTERATIONS OF THE BBB ...................................................... 122
LPS ...................................................... 123
Cytokines ...................................................... 123
Localization of BBB Injury ...................................................... 123
In Vitro Model of the BBB ...................................................... 124
BACTERIAL SURVIVAL WITHIN THE SUBARACHNOID SPACE .............................................. 124
CSF Complement ...................................................... 124
CSF Antibody ...................................................... 125
CSF Leukocytes ...................................................... 125
INDUCTION OF SUBARACHNOID SPACE INFLAMMATION .................................................... 125
Cell Wall ..................................................... 126
LPS ..................................................... 126
Inflammatory Mediators ..................................................... 126
INCREASED INTRACRANIA L PRESSURE ..................................................... 127
CEREBRAL VASCULITIS ..................................................... 128
ALTERATIONS IN CEREBRAL BLOOD FLOW ..................................................... 128
ADJUNCTIVE THERAPEUTIC STRATEGIES ..................................................... 129
Experimental Studies ...................................................... 129
Clinical Trials ..................................................... 131
REFERENCES ..................................................... 132

INTRODUCTION during 1986 were lower (e.g., 19% for meningitis due to S.
pneumoniae) (178), suggesting that improvements in early
Epidemiology detection and antibiotic treatment may have occurred in the
1980s. Bacterial meningitis also remains a significant prob-
Bacterial meningitis remains a relatively common and lem in other parts of the world. A recent review of all cases
devastating disease with an overall annual attack rate in the of bacterial meningitis admitted to an isolation-fever hospital
United States of -3.0 cases per 100,000 population. Mortal- in Salvador, Brazil, for the decade 1973 through 1982 re-
ity rates associated with the three most common causative vealed an approximate annual incidence of 45.8 cases per
agents of bacterial meningitis, Haemophilus influenzae, 100,000 population and an overall mortality rate of 33% (24).
Neisseria meningitidis, and Streptococcus pneumoniae, The three common meningeal pathogens (H. influenzae, N.
were 6.0, 10.3, and 26.3%, respectively, in the United StatesS.
1981 (136). Casese fatality rates mengtdsadS.ppneumonae) accounted for 72% offal all
es in a
from 1978 through uoie)conedfr7%
from1
subsequent study
h
ofh
of fe Can fanglt
s s and Los
five states Angeles Cunty
County cases and 70% of the deaths. In addition to this unacceptable
mortality, there is a high rate of neurologic sequelae in
children and adults who survive their episodes of bacterial
* Corresponding author. meningitis (17, 33, 107, 109, 156). This stable but unsatisfac-
118
VOL. 6, 1993 PATHOGENESIS OF BACTERIAL MENINGITIS 119

tory situation indicates the ongoing need to study the patho- NASOPHARYNGEAL COLONIZATION
genesis and pathophysiology of bacterial meningitis in an
attempt to improve the response to conventional antimicro- LOCAL INVASION
bial therapy (126, 148, 165, 166).
BACTEREMIA
Animal Models
Z ENDOTHEUAL CELL INJURY
Experimental animal models have been employed exten-
sively during the past 2 decades to increase our understanding INCREASED BBB
of the pathogenesis and pathophysiology of bacterial menin- PERMEABIUTY MENINGEAL INVASION
gitis (163). In the most commonly employed infant rat model, - CEREBRAL
animals developed meningitis following intranasal challenge SUBARACHNOID SPACE INFLAMMATION M VASCULJTIS
with H. influenzae type b (84). This model most closely
simulates the presumed pathogenesis of H. influenzae menin- INCREASED CSF OUTFLOW RESISTANCE
gitis in humans, since there was an initial nasopharyngeal
focus followed by bacteremia and an age-dependent suscep- HYDROCEPHALUS
tibility to meningeal invasion (83). The incidence of bacterial
meningitis, irrespective of rat host age, was directly related to VASOGENIC EDEMA INTERSTITIAL EDEMA CYTCDTOXIC EDEMbJA |
the intensity of bacteremia. Infant rats also developed men-
ingitis after orogastric challenge with Eschenchia coli (82). INCREASED INTRACRANIAL PRESSURE
CEREBRAL
INFARCTION
The infant rat model of H. influenzae meningitis has been I/
used primarily to study the early pathogenic events of bacte- DECREASED CEREBRAL BLOOD FLOW
rial meningitis. These include the determinants of nasopha-
ryngeal colonization and translocation into the bloodstream, FIG. 1. Scheme depicting the pathogenesis and pathophysiology
intravascular survival of the organism, bacteremia, and the of bacterial meningitis.
mechanisms of central nervous system (CNS) invasion. The
animal's small size is a disadvantage of the infant rat model
because only small samples (7 to 25 RI) of cerebrospinal fluid cells in organ culture in vitro with meningococci or H.
(CSF) are obtainable from 5- to 10-day-old rats, precluding influenzae type b, several events are observed (143): (i)
frequent sampling of CSF. Therefore, this model is less association with mucus independent of capsular polysaccha-
suitable for study of the pathophysiologic consequences of ride, fimbriae, or immunoglobulin A subclass 1 (IgAl) pro-
bacterial meningitis, since frequent sampling of CSF is usu- tease production; (ii) cytotoxicity characterized by break-
ally required for studying these events. Infant primates have down of epithelial-cell tight junctions, sloughing of ciliated
also been used in the study of the pathogenesis of bacterial cells, and ciliostasis; (iii) selective attachment to nonciliated
meningitis. In one study (130), bacteremia and meningitis epithelial cells; (iv) multiplication and formation of micro-
developed in 89 and 94% of animals, respectively, following colonies on the epithelial surface; (v) invasion of the epithe-
the atraumatic intranasal inoculation of H. influenzae type b. lium by intracellular or intercellular routes; and (vi) passage
Although this model closely simulates the spectrum of inva- of organisms to the submucosa. These events require viable
sive H. influenzae disease seen in humans, it is employed organisms and are not completed by commensal species. In
infrequently because of expense. addition, meningococci and H. influenzae type b appear to
Experimental models of meningitis in adult rabbits or rats invade nasopharyngeal mucosa by different mechanisms:
rely on the direct intracisternal inoculation of bacteria for meningococci utilize parasite-directed endocytosis, and H.
initiation of infection (30, 110). These animals reliably develop influenzae type b adheres to cells, causing a breakdown in
lethal infections with a predictable time course, although the tight junctions between epithelial cells and leading to inva-
natural bacteremia-meningitis sequence is bypassed, thereby sion by an intercellular mechanism. The bacterial virulence
creating an artificial pathogenesis. However, these models factors and the host defense mechanisms responsible for
have been extremely useful for the study of the pathophysi- these events are discussed in detail below.
ologic consequences of bacterial meningitis after organisms
have reached the subarachnoid space. CSF samples from Fimbriae
these adult animals may be obtained often.
The following sections briefly review the pathogenesis and Many of the major meningeal pathogens possess surface
pathophysiology of bacterial meningitis, as depicted in Fig. characteristics that enhance mucosal colonization. Fim-
1, emphasizing the relationships between specific bacterial briae, or pili, are specific organelles found on many bacteria
virulence factors and host defense mechanisms that are and often mediate adhesion of bacteria to host cells (10). The
responsible for clinical expression of disease (126, 165, 166). fimbriae of N. meningitidis mediate adherence of the organ-
New areas of investigation, as suggested by several of the ism to nasopharyngeal epithelial cells (Fig. 2) (143, 145).
steps depicted in Fig. 1, may lead to improvements in the Meningococci, like gonococci, possess fimbriae that differ in
refractory mortality and unacceptable morbidity in patients their morphologic, antigenic, and binding properties (51).
with bacterial meningitis. Fimbriae appear morphologically as aggregated bundles or
single filaments. The aggregated bundles are found primarily
MUCOSAL COLONIZATION AND SYSTEMIC among disease isolates and exhibit a low degree of adherence
INVASION to human buccal epithelial cells, whereas the single filaments
are found predominantly among colonizing isolates with
Initiation of most cases of bacterial meningitis begins with medium to high adherence characteristics. These fimbriated
host acquisition of a new organism by nasopharyngeal strains account for 80% of primary meningococcal isolates
colonization. Following infection of human nasopharyngeal from nasopharyngeal carriers and from the CSF of patients
120 TUNKEL AND SCHELD CLIN. MICROBIOL. REV.

lated types of H. influenzae (a through f) type b strains


constitute less than 5% of nasopharyngeal isolates, although
more than 95% of meningeal and systemic infections are
caused by type b strains. Experimental studies with infant
rats and laboratory transformants selected by capsular type
have shown that, while all encapsulated strains of H. influ-
enzae have the potential for systemic invasion after intrap-
eritoneal inoculation, type b strains are the most virulent and
are the only capsular types capable of systemic invasion
following intranasal inoculation (85, 120). Indeed, the pres-
ence of serum antibodies to polyribosyl-ribitol phosphate,
the capsular polysaccharide of type b isolates, is protective
against invasive disease (2). Antibodies to type b capsule are
almost uniformly detectable in humans by the age of 4 years,
even in the absence of known exposure to H. influenzae type
b. The presence of antibodies to type b capsule may be
related to the abilities of other encapsulated strains of H.
influenzae to produce some type b capsular material. It has
also been suggested that the source of bacterial capsule may
be secondary to acquisition of new DNA from organisms
colonizing the gastrointestinal tract (81, 138). For example,
E. coli K100 possesses a capsule that is immunologically
related to the type b capsule of H. influenzae and may
stimulate the production of cross-reacting anticapsular anti-
bodies by the host, suggesting that the protection against
as -- mm vw infection with H. influenzae type b is due to priming of serum
anticapsular antibodies.
FIG. 2. Scanning electron micrograph of nasopharyngeal organ Polysaccharide capsule may also be an important viru-
cultures 6 h after infection with encapsulated N. meningitidis 34NP, lence factor for invasive disease caused by S. pneumoniae.
showing attachment of meningococci to nonciliated epithelial cells Of the 84 pneumococcal serotypes known, 18 are responsi-
and formation of a microcolony on the epithelial surface (magnifi- ble for 82% of cases of bacteremic pneumococcal disease (9,
cation, x8,775). Reproduced from reference 143 with permission of
The University of Chicago Press. 39), and there is a close correlation between bacteremic
subtypes and those implicated in meningitis (20, 44, 50).
However, low degrees of adherence to human pharyngeal
with meningitis (32), although all fimbriae were lost on serial
epithelial cells have been observed among pneumococcal
strains isolated from patients with serious infections such as
subculture in the laboratory. Following attachment via a septicemia and meningitis (4), suggesting that adherence
specific cell surface receptor, meningococci are transported may be less important in pneumococcal pathogenicity. A
within a phagocytic vacuole across nonciliated nasopharyn- study of S. pneumoniae adherence to nasopharyngeal cells
geal columnar epithelial cells (74, 144); this series of events demonstrated that these organisms were more often found
appears to be essential for development of invasive menin- on desquamated cells than on cells taken from intact epithe-
gococcal disease. Fimbriae have also been implicated in the lium (71). Nasopharyngeal mucus may provide a protected
adherence of H. influenzae to upper respiratory tract epithe- nidus from which pneumococci can spread (45), although the
lial cells (72, 146), and lack of fimbrial expression impairs the various factors responsible for invasiveness among certain
ability of H. influenzae type b to colonize the nasopharynx pneumococcal serotypes remain unknown.
(177). However, fimbriae have not been found on H. influ-
enzae type b isolated from the CSF or blood of patients with
invasive disease (72, 106), suggesting that although fimbriae Antibodies
play a role in initial adherence within the nasopharynx, their Natural antibodies such as IgA, found predominantly in
presence is not necessary for the organism to cause menin- mucosal secretions, may inhibit the adherence of microor-
gitis. Acquisition and colonization of H. influenzae type b ganisms to mucosal surfaces. Formation of these antibodies
may also be promoted following viral infection by a variety is stimulated by colonization of organisms that share cross-
of respiratory viruses, including influenza A Victoria and reactive antigens with pathogenic strains (138). However,
respiratory syncytial virus (140). The precise role of a the presence of high concentrations of circulating IgA anti-
preceding upper respiratory viral infection in the enhance- bodies to N. meningitidis may paradoxically permit the
ment of nasopharyngeal colonization and the subsequent development or exacerbate the progression of invasive dis-
development of meningitis is controversial, however, and ease by preferentially binding to the organism, thereby
requires further study. blocking the beneficial effects of IgG and IgM antibodies (53,
54, 64). In addition, many pathogenic Neisseria, Haemoph-
Polysaccharide Capsule ilus, and Streptococcus species produce IgAl proteases that
cleave IgA in the hinge region of the immunoglobulin mole-
Bacterial encapsulation may be important for nasopharyn- cule (108); these enzymes may have a pathogenic role by
geal colonization and systemic invasion of meningeal patho- facilitating adherence of bacterial strains to mucosal surfaces
gens. Strains of H. influenzae colonize the nasopharynges of through local destruction of IgA (86). The exact role of IgA
most children by the age of 3 months, although most of these protease production in the pathogenic sequelae of bacterial
strains are unencapsulated (140). Among the six encapsu- meningitis remains unclear, however.
VOL. 6, 1993 PATHOGENESIS OF BACTERIAL MENINGITIS 121

Other Bacterial Components 23). Impairment of the alternative complement pathway, as


in patients with sickle cell disease (103) and in patients who
Other studies have examined various surface components have undergone splenectomy, predisposes to the develop-
(e.g., lipopolysaccharide [LPS] and outer membrane pro- ment of pneumococcal meningitis. The complement cascade
teins) of H. influenzae to determine their roles in the entry is also activated by H. influenzae type b (113). C3-depleted
stages of pathogenesis. Antibodies against these components rats show a greater incidence and magnitude of bacteremia
confer protection against repeated challenge with the organ- after either intravenous or intraperitoneal challenge with H.
ism (3, 55, 56, 65). More recently, hemocin, the bacteriocin influenzae of various serotypes (a, b, c, or d) than do normal
produced by H. influenzae, has been shown to be strongly rats (27, 29, 187). Although the incidence of bacteremia due
associated with type b encapsulated strains and may play a to type b organisms increases from 63 to 95% in comple-
role in host nasopharyngeal colonization and/or systemic ment-depleted rats, the incidence and severity of meningitis
invasion by this organism. After intranasal inoculation of are unaffected.
infant rats with an equal mixture of a non-hemocin-produc- Activation of the complement system is an essential host
ing strain and its hemocin-producing transformant, organ- defense mechanism in protection against invasive disease by
isms capable of hemocin production predominated in iso- N. meningitidis. Patients with deficiencies in the terminal
lates from both nasopharyngeal and blood cultures (70). complement components (C5, C6, C7, C8, and perhaps C9),
However, the precise roles of capsular polysaccharide, LPS, the so-called membrane attack complex, are particularly
hemocin, outer membrane proteins, and other surface com- prone to infection with neisserial species, including N.
ponents in mucosal translocation and bloodstream invasion meningitidis, though usually with a favorable outcome for
of H. influenzae and other major meningeal pathogens re- the patient when appropriate treatment is instituted (122).
main incompletely defined. The reasons for the decreased mortality rate in the comple-
ment-deficient patients are not clear. It has been suggested
BACTEREMIA that the presence of complement-activating products (i.e.,
the membrane attack complex), in concert with other medi-
ators, may contribute to the development of multiorgan
Polysaccharide Capsule failure and death. A qualitative relationship exists among the
Once the mucosal barrier is crossed, bacteria gain access level of circulating meningococcal LPS, a fatal outcome, and
to the bloodstream and must then overcome host defense the degree of complement activation (19), indicating that
mechanisms to survive and invade the CNS. Surface encap- prognosis is worse for patients with intact complement
sulation is the most important virulence factor in this regard. systems.
Bacterial capsule, by effectively inhibiting neutrophil phago-
cytosis and resisting classic complement-mediated bacteri- MENINGEAL INVASION
cidal activity, may enhance bloodstream survival of the
organism and facilitate intravascular replication (166). In- The mechanisms by which bacterial pathogens gain access
deed, the most common meningeal pathogens (H. influen- to the CNS are largely unknown. One factor may relate to
zae, N. meningitidis, S. pneumoniae, E. coli, and Strepto- the concentration of organisms in the blood (84). In the
coccus agalactiae) are all encapsulated. Furthermore, experimental infant rat model, the intranasal inoculation of
certain specific capsular types among the great many re- H. influenzae type b initially produced a low-grade bactere-
quired are associated disproportionately with the develop- mia (about 102 CFU/ml) and no organisms were present in
ment of bacterial meningitis. For example, about 84% of the CSF (141). Culture-positive meningitis was observed
cases of neonatal meningitis due to E. coli are caused by only after an intense bacteremia (>10W CFU/ml) had been
strains bearing the Kl antigen, which is antigenically related present for at least 6 h (100). Meningitis was also induced in
to the capsular material of serogroup B meningococci and an age-dependent manner, with a higher incidence in 5-day-
type III group B streptococci (119). In the absence of old than in 20-day-old rats (83). In the animals that ultimately
Kl-specific host antibody, these organisms are profoundly developed meningitis, sustained bacteremia, as opposed to
resistant to phagocytosis (28). Human monoclonal antibod- transient bacteremia, was documented. However, sustained
ies with specific reactivity for epitopes on the Kl capsule of bacteremia is not the only factor responsible for meningeal
E. coli and/or the group B polysaccharide of N. meningitidis invasion, because many other organisms (e.g., viridans
may be useful for prevention and/or treatment of blood- streptococci) that produce continuous bacteremia during
stream infections caused by these organisms (114); however, infective endocarditis rarely produce bacterial meningitis.
this concept remains conjectural for humans.
Site of Invasion
Host Defense Mechanisms
The exact sites of CNS invasion by meningeal pathogens
Several host defense mechanisms can counteract the are unclear. Early studies with the experimental rat model
antiphagocytic effects of bacterial capsule. For example, the suggested that the route of invasion from the bloodstream to
capsular polysaccharides of S. pneumoniae activate the the CSF was through the dural venous sinus system (141).
alternative complement pathway, resulting in cleavage of C3 However, subsequent experiments with the same animal
and subsequent attachment of C3b to the bacterial surface, model suggested that during the ensuing bacteremia a non-
thereby facilitating opsonization, phagocytosis, and intra- specific, sterile, focal inflammation above the cribriform
vascular clearance of the organism (38). Although antipneu- plate facilitated invasion of the CNS at that site. Further
mococcal cell wall antibody and antipneumococcal capsular studies of infant rats and primates demonstrated that bacte-
antibody promote the efficient deposition of C3b on the ria may enter the CSF via the choroid plexus, which has an
pneumococcal surface, C3b deposited on the surface of exceptionally high rate of blood flow (-200 ml/g/min), per-
pneumococcal capsule is a more efficient opsonin in vitro haps because more bacterial organisms are delivered to this
and in vivo than C3b activated by anti-cell wall antibody (22, site than to other anatomic locations in the CNS per unit of
122 TUNKEL AND SCHELD CLIN. MICROBIOL. REV.

time. Sampling of CSF compartments early during bacterial titis than do strains of subtype 1 (151), perhaps because of
meningitis has demonstrated higher bacterial densities in the the ability of each subtype to release LPS under appropriate
lateral ventricles than in the cisterna magna, lumbar sub- circumstances. However, the precise role of outer mem-
arachnoid space, or supracortical subarachnoid space. Al- brane protein subtypes in meningeal invasion is unclear.
though with time equilibrium is reached in these other
locations, the data suggest initial bacterial entry into CSF in Secondary Bacteremia
the lateral ventricles, presumably through the choroid plexi.
Following bacterial invasion of the subarachnoid space, a
Fimbriae secondary bacteremia may result from the local CNS sup-
purative process, allowing the meningeal pathogen to con-
Recent experimental studies have suggested that receptors tinuously enter and leave the CSF compartment under quite
for some meningeal pathogens are present on cells in the dynamic circumstances. In an experimental canine model of
choroid plexus and/or cerebral capillaries, which may facil- pneumococcal meningitis, the early transport of bacteria
itate movement of these pathogens into the subarachnoid from CSF to blood was presumed to be transendothelial
space. In cryostat sections of infant rat brain cortical slices, through arachnoid villi containing pores large enough to
strains of E. coli possessing S fimbriae are seen to bind to the accommodate bacteria, which would then enter the superior
luminal surfaces of the vascular endothelium and the epithe- sagittal sinus and return to the central venous blood (135).
lium lining the choroid plexus and brain ventricles (101). This transport occurred only following active bacterial mul-
Pretreatment of the brain sections with neuraminidase or the tiplication in CSF and before the height of the febrile
trisaccharide receptor analog of S fimbriae abolished this response or CSF pleocytosis. This phenomenon was also
binding. Subsequent experiments demonstrated that 1 h after observed with H. influenzae type b, inoculation of which
intraperitoneal challenge with the S-fimbriated strain of E. into the cisterna magna of experimental animals produced an
coli, about 50% of organisms in the CSF were S fimbriated almost instantaneous bacteremia (140).
and 50% were nonfimbriated (128), suggesting that phase
variation to the nonfimbriated form may be necessary for ALTERATIONS OF THE BBB
these bacteria to invade the CNS. Nevertheless, the specific
adherence of meningeal pathogens to sites within the CNS is Bacterial meningitis, like many other disease states, in-
one hypothesis raised to explain bacterial neurotropism and creases the permeability of the blood-brain barrier (BBB).
currently is an area of intense investigation. The major sites of the BBB are the arachnoid membrane,
choroid plexus epithelium, and cerebral microvascular en-
Monocytes dothelium. Previous extensive morphologic studies have
demonstrated intact arachnoid membranes in animals with
Additional studies to determine the pathogenic mecha- bacterial meningitis (176). Therefore, the increased BBB
nisms responsible for meningeal invasion utilized histologic permeability seen in this disorder must occur at the level of
and scanning microscopic techniques to examine the the choroid plexus epithelium, the cerebral microvascular
neuraxes of pigs inoculated intravenously with a pathogenic endothelium, or both; the cerebral microvascular endothe-
strain of Streptococcus suis type 2 (180). The animals were lium has been the site of intensive study in recent years as a
sacrificed 17 to 47 h after intravenous challenge, and the only result of techniques for isolation of cerebral microvessels or
pathologic lesions detected were associated with the choroid endothelial cells or both. The features that distinguish cere-
plexus, manifested as disruption of the plexus brush border, bral capillaries from other capillaries throughout the body
decrease in the number of Kolmer cells, and exudation of are (i) adjacent endothelial cells fused together by pentalam-
fibrin and inflammatory cells into the ventricles. Intracellular inar tight junctions (zonulae occludens) that prevent inter-
bacteria were demonstrated in the parenchyma of the cho- cellular transport; (ii) rare or absent pinocytotic vesicles;
roid plexus, in the ventricular monocytes, and within periph- and (iii) abundant mitochondria (18, 49). Therefore, the
eral blood monocytes. Circulating monocytes, which were increased BBB permeability that occurs during bacterial
also found to contain phagocytized bacterium-sized parti- meningitis at the level of the cerebral capillary endothelial
cles, migrated into the CSF via the choroid plexus, suggest- cell may result from separation of intercellular tight junc-
ing that bacteria may gain access to the CSF in association tions, from increased pinocytosis, from both alterations, or
with monocytes migrating along normal pathways (the so- by processes as yet unknown.
called Trojan horse hypothesis for CNS invasion). An adult rat model of bacterial meningitis was used to
investigate the propensity for bacterial meningitis to induce
Other Bacterial Components functional and morphologic alterations of the BBB (110).
Following the intracisternal inoculation of either E. coli, S.
Other bacterial virulence factors have been studied to pneumoniae, or H. influenzae into rats, a uniform host
determine their possible roles in meningeal invasion. The response to all three encapsulated pathogens was observed
liberation of LPS from N. meningitidis may contribute to the at the level of the cerebral capillary endothelial cell, charac-
pathogenicity of this organism in invasive infections (1). terized morphologically by an early and sustained increase in
Meningococci vary in their ability to liberate endotoxin, with pinocytotic vesicle formation and a progressive increase in
increased amounts liberated from patients with invasive separation of intercellular tight junctions from 4 to 18 h
disease. For example, serogroup B meningococcal strains postinoculation (Table 1). These morphologic changes cor-
released slightly more free endotoxin when isolated from related with the functional penetration of albumin across the
blood or CSF than when isolated from the nasopharynges of BBB, with the highest values of albumin entry occurring 18
presumably healthy persons. Outer membrane proteins may h after intracisternal inoculation, when both morphologic
also be important. One report has suggested that H. influen- changes were evident. Following intracisternal inoculation
zae strains with outer membrane protein subtype lc cause of an unencapsulated strain of H. influenzae (Rd strain),
more episodes of meningitis and fewer episodes of epiglot- there was an increase in pinocytotic vesicle formation, but
VOL. 6, 1993 PATHOGENESIS OF BACT7ERIAL MENINGITIS 123

TABLE 1. Correlation of morphologic and functional 108


v-
alterations of the BBB in an experimental rat
model of H. influenzae-caused meningitisa 50000 -
8 Bi I}
Time after
treatment Inoculum (n)
% Penetration
of "-albumin
CBBB mor-
at 40000 -
6 X t
E
(h)
~~~~~~~~(meanSE)
± phooy
LL W 30000 -
m co
en cn
C. +1
*4 0
4 Saline (3) 0.26 ± 0.08 C 20000 -

H. influenzae Rd (3) 4.12 + 1.25c t PV co


H. influenzae type b (4) 3.80 + 0.75c t PV 10000 -
*2 co
44
18 Saline (4) 1.25 ± 0.27
H. influenzae Rd (4) 4.64 ± 0.80c t PV 0
H. influenzae type b (4) 8.10 ± 1.20cd T PV + t SJ 1

a
Reproduced from reference 110 with permission of the American Society
for Clinical Investigation. TIME (H)
b PV, pinocytotic vesicles; SJ, separated junctions; T, increase.
c P < 0.05 compared with control. FIG. 3. Kinetics of changes in concentration of leukocytes
d p < 0.05 compared with H.
influenzae type b at 4 h and H. influenzae Rd (WBC) in CSF and in BBB permeability (BBBP) after inoculation
at 18 h. with LPS in an experimental rat model. Reproduced from reference
183 with permission of the American Society for Clinical Investiga-
tion. SE, standard error of the mean.
separation of intercellular tight junctions did not occur. This
discrepancy between encapsulated and unencapsulated of LPS) and neutrophil acyloxyacyl hydrolase (which re-
strains of H. influenzae likely occurred secondary to the moves nonhydroxylated fatty acids from the lipid A region of
removal of unencapsulated organisms from the CSF by host the LPS molecule) inhibited the effect of LPS on BBB
defense mechanisms, whereas deficient opsonic mechanisms permeability, strongly implicating the lipid A region of LPS
in the CSF (see below) permitted sustained concentrations of in the observed effects in the CNS. Monoclonal antibodies
the encapsulated strain. Therefore, encapsulation of H. directed against the oligosaccharide portion of LPS did not
influenzae was not essential for BBB injury but facilitated decrease permeability. No change in BBB permeability was
the progression of such injury by avoidance of host defense observed following intracisternal inoculation of LPS into
mechanisms. leukopenic rats. Similar results in rats were obtained follow-
The effect of the host leukocyte response on altered BBB ing intracistemal inoculation of H. influenzae type b outer
permeability was subsequently examined in the experimen- membrane vesicles (182), which may represent a relevant,
tal rat model by first rendering the animals leukopenic 4 days nonreplicating vehicle for the delivery of the toxic moieties
following intraperitoneal injection of cyclophosphamide of LPS to host cells.
(68). Functional increases in BBB permeability, assessed by
penetration of radioactive albumin from blood to CSF, were Cytokines
observed in both normal and leukopenic rats at 18 h follow-
ing inoculation of either encapsulated or unencapsulated Because the increased BBB permeability induced by LPS
strains of H. influenzae, but permeability was greater after in the experimental rat model was not maximal until 4 h
challenge with the encapsulated strain. Significant increases following intracisternal inoculation, it was suggested that a
in BBB permeability occurred in the near absence of leuko- common host mediator(s) was responsible. Therefore, it was
cytes in CSF late in the disease process, although the next determined whether specific inflammatory cytokines,
presence of leukocytes augmented changes in permeability. which mediate many of the deleterious effects of LPS, also
At 18 h following inoculation, alterations of BBB permeabil- increased permeability (112). Intracisternal inoculation of
ity correlated with concentrations of bacteria in the CSF. human recombinant interleukin-lp (IL-1ip) into rats led to a
peak increase in BBB permeability about 3 h after inocula-
LPS tion (Fig. 4), which is earlier than the peak response ob-
served with LPS (at 4 h). This effect was significantly
Since bacterial capsule was not essential for BBB injury in attenuated by preincubation of the cytokine with a mono-
the experimental rat model of bacterial meningitis and since clonal antibody to IL-1l3 and was totally abolished in leuko-
it was recognized that pneumococcal capsule did not induce penic animals. Preincubation of IL-1lB with polymyxin B did
inflammation within the subarachnoid space (see below), not alter IL-1,B activity. No permeability changes were
BBB permeability was examined following intracisternal observed following intracisternal inoculation of human re-
inoculation of purified H. influenzae type b LPS. After combinant tumor necrosis factor alpha (TNF-a) into rats,
intracisternal inoculation of LPS into rats, the following although rabbit TNF-ot clearly elicits subarachnoid space
results were observed (183): (i) dose-dependent increases in inflammation following intracistemal inoculation of the ho-
BBB permeability from 2 pg to 20 ng, with attenuation in mologous species. All available evidence suggests that both
peak response after challenge with 500 ng or 1 ,ug; (ii) cytokines are important and that they act synergistically,
time-dependent increases in BBB permeability, with maxi- since inoculation with submaximal doses of IL-lp plus
mal alteration at 4 h and complete reversal at 18 h (Fig. 3); TNF-a at concentrations that produced no changes individ-
(iii) greater increases in permeability after challenge with ually enhanced BBB permeability.
LPS than after challenge with the live parent strain despite
identical LPS concentrations; and (iv) close correlation
between BBB permeability and CSF pleocytosis 4 h after Localization of BBB Injury
intracisternal inoculation. Preincubation of LPS with poly- The precise localization of BBB injury in bacterial menin-
myxin B (a cationic antibiotic that binds to the lipid A region gitis was subsequently examined by in situ tracer perfusion
124 TUNKEL AND SCHELD CLIN. MICROBIOL. REV.

5 mechanisms responsible for this increased ability to perme-


0
Co ate monolayers in the in vitro model, the effects of LPS on
x IL-1I the formation of various second messenger systems in the
0
4 O TNFa cerebral microvascular endothelial cells in response to LPS
'D
0

* Control
stimulation were examined (161). Production of both cyclic
E AMP and cyclic GMP was increased in response to LPS, but
3 production of cyclic AMP only was evident prior to the
immLu increased permeation of the cell monolayer. This observa-
0.
tion suggests that the increased BBB permeability within the
2 cerebral microvascular endothelium during bacterial menin-
m')0
gitis occurs via a cyclic AMP-dependent process.
w
cn In contrast, other investigators, utilizing primary cultures
+1
C
1 of bovine brain microvascular endothelial cells (102), found
c
that H. influenzae type b or purified type b LPS caused
E
I.
marked cytotoxicity of these cells in culture, an effect that
0
0 2 4 6 8 10
could be completely blocked by polymyxin B. The presence
of serum was essential for the LPS-induced cytotoxic effect,
Time Post Inoculation (hr) and a monoclonal antibody against CD14, a receptor in-
FIG. 4. Kinetics of changes in CSF traversal of systemically
volved in mediating the actions of LPS in monocytes,
administered 1251-BSA in an experimental rat model after intracis- completely blocked the cytotoxic effect. Further studies are
ternal inoculation with recombinant IL-1i, recombinant TNF-a, necessary, however, to precisely define the cellular mecha-
and controls. *, P < 0.05. BBBP, BBB permeability; SE, standard nisms of altered BBB permeability during bacterial menin-
error of the mean. Reproduced from reference 112 with permission gitis.
of the American Society for Clinical Investigation.
BACTERIAL SURVIVAL WITHIN THE
SUBARACHNOID SPACE
and immunolabeling procedures to identify the topography
and microvascular exit pathways of bovine serum albumin CSF Complement
(BSA) (111). Two tracers were used: colloidal gold, which is
useful in ultrastructural definition of albumin exit pathways, Once meningeal pathogens penetrate the subarachnoid
and monomeric BSA, which has a faster rate of transcytosis space, host defense mechanisms are inadequate to control
than colloidal gold. After intracisternal challenge of rats with the infection (131). Complement components in CSF are
E. coli (O111:B4) LPS, an inducible increase in immunode- usually absent or present in only minimal concentrations (25,
tectable monomeric BSA binding to the luminal membranes 115, 139, 157). Meningeal inflammation leads to increased,
of all microvascular segments in the pia-arachnoid and but low, concentrations of complement in CSF. The impor-
superficial brain cortex was observed by transmission elec- tance of this relative complement deficiency in normal and
tron microscopy. Uptake of colloidal gold and monomeric infected CSF may be critical, since specific antibody and/or
BSA by plasmalemmal vesicles was similar, but there was no complement is essential for opsonization of encapsulated
detectable transcytosis to the abluminal side of the endothe- meningeal pathogens, efficient phagocytosis, and removal of
lium. Exit of both perfused colloidal gold-BSA and immuno- pathogens by the spleen (21). Opsonic and bactericidal
detectable BSA was through open intercellular junctions of activities are absent or barely detectable in patients with
venules in the pia-arachnoid, specifically and topographi- various forms of meningitis; similar observations have been
cally localizing the BBB injury in bacterial meningitis to the made in experimental animal models of bacterial meningitis.
meningeal venules. A serum-sensitive E. coli strain was not opsonized in vitro
when the strain was incubated with CSF from rabbits
In Vitro Model of the BBB challenged intracisternally with E. coli Kl, although CSF
from animals with Staphylococcus aureus-caused meningitis
Further studies were performed to delineate the precise was opsonically active in vitro against E. coli (13). The
effect of H. influenzae type b LPS on the cerebral microvas- absence of opsonization after E. coli challenge may have
culature. Purified preparations of cerebral microvascular been due to the absorption of a specific opsonin by E. coli
endothelial cells (identified by positive immunofluorescent early in the course of infection. Alternatively, more serum
staining for factor VIII-related antigen and by the ability to protein, and therefore opsonins, may have penetrated into
take up acetylated low-density lipoproteins) from rat cere- CSF in staphylococcal rather than E. coli meningitis. Al-
bral cortices were grown on a semipermeable support to though there are low functional and bactericidal activities in
create an in vitro model of the BBB (162, 164). Isolated purulent CSF, the presence of some measurable opsonic
cerebral microvascular endothelium has been shown to activity may correlate with a favorable outcome. For exam-
retain many of the characteristics of an intact BBB, includ- ple, outcome was better in patients whose concentrated CSF
ing the ability to form intercellular tight junctions in vitro. samples demonstrated opsonic activity (15 of 27 patients
Treatment of intact cerebral microvascular endothelial cell [186]), suggesting that complement-mediated opsonic activ-
monolayers with LPS (0.1 ,ug/ml) in serum-free media led to ity can appear in the CSF during bacterial meningitis,
a statistically significant increase in the percentage of radio- particularly in patients who recover completely.
active albumin able to permeate the monolayer (2.2 to 5.6%; Several explanations of the low concentrations of comple-
P < 0.001) in the absence of host inflammatory cells. This ment in CSF during meningitis have been advanced: (i)
increase in permeability was not associated with cytotoxicity insufficient traversal across the BBB; (ii) variable subarach-
as assessed by release of lactate dehydrogenase into the noid space inflammation; (iii) enhanced clearance or removal
media. To determine the putative intracellular regulatory from the subarachnoid space; (iv) low production rates in the
VOL. 6, 1993 PATHOGENESIS OF BACTERIAL MENINGITIS 125

CNS; and (v) degradation at the site of infection (131). This generated CSF pleocytosis during bacterial meningitis.
last possibility has been investigated in patients and in However, despite the entry of leukocytes, host defense
animal models of meningitis. Leukocyte proteases have been mechanisms in CSF remain suboptimal because of the rela-
shown to degrade functional complement components (e.g., tive lack of functional opsonic and bactericidal activity.
C3b) in CSF from patients with meningococcal meningitis, Therefore, phagocytosis is inefficient at this protected site,
with the formation of nonopsonic products (e.g., C3d) (179). leading to huge concentrations of bacteria in the CSF during
In the experimental rabbit model of pneumococcal meningi- meningitis (36, 37).
tis, the intracisternal inoculation of phenylmethylsulfonyl There is controversy over the precise role of neutrophils in
fluoride, a nonspecific protease inhibitor, led to a decline in host defense within the CNS during bacterial meningitis. In
pneumococcal concentrations in CSF compared with those experimental animal models, low concentrations of leuko-
of saline-inoculated controls (131). Therefore, during bacte- cytes in CSF have generally been associated with increased
rial meningitis, complement components crossing the BBB mortality rates (46, 134). Although a study of dogs with
may be degraded by leukocyte proteases, resulting in ineffi- pneumococcal meningitis revealed that leukopenic animals
cient opsonic activity at the site of infection similar to had a higher survival rate than animals with normal periph-
mechanisms postulated to occur in the pleural space during eral leukocyte counts (62 versus 47 h) (104), the small
the development of an empyema. number of animals studied precluded statistical analysis. In
an experimental rabbit model of pneumococcal meningitis,
CSF Antibody the parameters of bacterial growth rate; final concentrations
of bacteria in CSF; and concentrations of protein, glucose,
Immunoglobulin concentrations are also low in normal and lactate in CSF were no different in animals rendered
CSF, with an average blood/CSF ratio of IgG of about 800:1. leukopenic by the prior intravenous inoculation of nitrogen
Immunoglobulin concentrations in CSF increase during bac- mustard than in nonleukopenic animals. However, the re-
terial meningitis but remain low compared with simultaneous sultant bacteremia was about 100-fold greater in leukopenic
concentrations in serum, and IgG does not appear in the CSF animals (34), suggesting that neutrophils either prevented
until late in the course of disease (142, 179). An experimental traversal of pneumococci from CSF to blood or enhanced
rabbit model of pneumococcal meningitis has been utilized neutrophil-mediated bacterial elimination from the blood-
to study the importance of antibodies in host defense against stream at extraneural sites.
bacterial meningitis. Intracisternal inoculation of type-spe- The precise pathway by which neutrophils enter the
cific antibodies against S. pneumoniae decreased pneumo- subarachnoid space remains unknown. Adherence of neu-
coccal concentrations in CSF, but at a much slower rate than trophils to vascular endothelial cells is a likely prerequisite
did treatment with appropriate bactericidal antibiotics (131). for traversal into the CSF. Pretreatment of endothelial cells
The systemic administration of type-specific monoclonal with cytokines induces formation of specific adhesion mol-
antibodies has been examined in the experimental rabbit ecules such as endothelial leukocyte adhesion molecule 1
model. Intravenous injection of a bactericidal monoclonal (16). Neutrophil adherence to vascular endothelial cells is
antibody against the polyribosyl-ribitol phosphate of H. enhanced by pretreatment of the endothelial cells with LPS
influenzae type b produced high concentrations of antibody (137) or with inflammatory cytokines such as IL-1 and TNF
in serum, but there was poor BBB penetration (c5.5%), (15, 137, 165). Similar mechanisms may also enhance neu-
even in the presence of meningeal inflammation (47). These trophil binding to cerebral vascular endothelium (11). A
results suggested that systemic administration of type-spe- recent study of an experimental rabbit model has demon-
cific antibodies alone is suboptimal. The pioneering work of strated that the intravenous inoculation of a monoclonal
Flexner (42), who demonstrated that systemic and intrathe- antibody (IB4) against the CD18 family of receptors on
cal administration of antimeningococcal antiserum raised in leukocytes blocked the accumulation of leukocytes in the
horses was effective in reducing the mortality rate in menin- subarachnoid space despite intracisternal challenge with H.
gococcal meningitis from approximately 80 to 30%, suggests influenzae type b, N. meningitidis, pneumococcal cell wall,
that combined sites of administration may be useful. It or LPS (170). The monoclonal antibody also attenuated a
remains to be determined, however, whether the intrathecal parameter of BBB permeability (i.e., increased protein con-
administration of antibodies may be useful in the adjunctive centrations in the CSF). Furthermore, development of cere-
treatment of bacterial meningitis. bral edema and death were prevented in monoclonal anti-
body-treated animals challenged with lethal doses of S.
CSF Leukocytes pneumoniae. Penetration of antibiotics into CSF, bacterial
concentrations in CSF, and bactericidal response to ampi-
One of the hallmarks of bacterial meningitis is the devel- cillin therapy were not affected by monoclonal antibody
opment of a neutrophilic pleocytosis within the CSF, al- administration, although the animals exhibited a delay in the
though the precise mechanisms of leukocyte traversal across onset of bacteremia and there was an attenuated CSF
the BBB remain to be defined (52, 184). In an experimental inflammatory response after ampicillin-induced bacterial
animal model of pneumococcal meningitis, the complement killing. These results suggest that systemic inoculation of
component C5a has been suggested as one chemotactic monoclonal antibodies directed at leukocyte-endothelium
substance in CSF (35, 96, 158), with chemotactic activity interactions can block leukocyte-mediated damage within
appearing 2 to 4 h before neutrophil influx into the CSF. The the CNS during bacterial meningitis.
precise role of C5a as a chemotactic substance has recently
been examined in the experimental rabbit model, in which INDUCTION OF SUBARACHNOID SPACE
the intracistemal inoculation of C5a caused a rapid early INFLAMMATION
influx of leukocytes into CSF 1 h after inoculation (60). This
response was attenuated by coadministration of CSa with The ability of meningeal pathogens to induce a marked
prostaglandin E2 (PGE2) in a dose-dependent manner, sug- subarachnoid space inflammatory response contributes to
gesting a direct anti-inflammatory action of PGE2 on CSa- many of the pathophysiologic consequences of bacterial
126 TUNKEL AND SCHELD CLIN. MICROBIOL. REV.

meningitis (e.g., cerebral edema and increased intracranial antibody directed against epitopes on the oligosaccharide
pressure). For example, this inflammatory response may be portion of the LPS molecule did not reduce the inflammatory
a crucial factor in the development of sensorineural deafness potential of the LPS. Similar results were observed in an
in patients with bacterial meningitis. Utilizing an infant rat experimental rat model following the intracisternal inocula-
model of pneumococcal meningitis, multiple intraperitoneal tion of purified LPS, with the maximal amount of inflamma-
inocula (1 x 108 to 10 x 108 CFU every 24 h for 3 days) of tion observed 8 h after inoculation of a 20-ng dose (Fig. 3)
S. pneumoniae in 5-day-old rats led to bacteremia and (183). In addition, the intracisternal inoculation of outer
meningitis in 50 and 46% of animals, respectively (121). membrane vesicles from H. influenzae type b induced men-
Hematoxylin-eosin-stained sections of brain tissue from rats ingeal inflammation in both rabbits and rats in a dose- and
with positive CSF cultures revealed inflammation in the time-dependent manner (92, 182); this response was blocked
meninges and scala tympani but not in the scala media. In by polymyxin B but not by two monoclonal antibodies
addition, perilymphatic inflammation occurred significantly directed against surface epitopes of the oligosaccharide side
(P < 0.001) more than did endolymphatic inflammation, chains of LPS within outer membrane vesicles. This obser-
suggesting that bacteria reached the cochlea via the cochlear vation supports the concept that the delivery of LPS via
duct, thereby providing a connection between perilymphatic outer membrane vesicles induces subarachnoid space in-
fluid and CSF. Recent studies in other animal model systems flammation.
have focused on the bacterial virulence factors that are
responsible for this inflammatory response, providing many
new, exciting areas of investigation. Inflammatory Mediators

Cell Wall Recently, evidence that supports the hypothesis that


pneumococcal cell wall or H. influenzae type b LPS induces
Despite the fact that bacterial capsule is largely responsi- subarachnoid space inflammation through the local CNS
ble for intravascular and subarachnoid space survival of release of inflammatory mediators such as IL-1, TNF (14,
meningeal pathogens, capsular polysaccharides are remark- 80, 159), and/or prostaglandins (77, 95) has been accumulat-
ably noninflammatory when injected in purified form into the ing. Intact pneumococci, pneumococcal cell wall, or lipotei-
subarachnoid space. In contrast, cell walls of gram-positive choic acid induced IL-1 production by human peripheral
bacteria are potent inducers of inflammation, and S. pneu- monocytes in vitro (118). IL-1 production by monocytes was
moniae cell walls activate the alternative complement path- dramatically reduced by chemical alteration of phosphoryl-
way (181). Various surface components of S. pneumoniae choline, the major determinant of teichoic acid, or by
have been examined to determine their roles in subarachnoid pretreatment with a phosphorylcholine antibody. TNF pro-
space inflammation. A CSF inflammatory response was duction was not induced in monocytes incubated with pneu-
invoked in an experimental rabbit model by intracisternal mococcal cell wall in vitro.
inoculation of live encapsulated and unencapsulated S. In an experimental rat model, the intracisternal inocula-
pneumoniae cells, heat-killed unencapsulated pneumococci, tion of H. influenzae type b LPS led to elevated concentra-
and pneumococcal cell walls (169); no inflammation was tions of IL-1 and TNF in CSF within 30 to 120 min (166).
induced by intracisternal inoculation of either heat-killed Elevated concentrations of TNF in CSF have also been
encapsulated strains or isolated capsular polysaccharide. observed in the experimental rabbit model following intra-
Pneumococcal cell walls inoculated intracisternally were the cisternal inoculation of LPS (90). TNF activity in CSF was
most potent inducers of CSF inflammation among these first detected 45 min after intracisternal challenge with LPS,
substances (168). In addition, independent intracisternal with peak activity at 2 h and persistence for about 5 h.
injection of the major components of the pneumococcal cell Following intracisternal challenge with live H. influenzae
wall, teichoic acid and peptidoglycan, also induced CSF type b, peak TNF activity in CSF was comparable to that
inflammation. Teichoic acid had the highest specific activity after LPS challenge, although activity persisted for -14 h.
of the cell wall fractions tested, and activity was markedly TNF activity and meningeal inflammation (i.e., CSF pleocy-
reduced if either component (teichoic acid or peptidoglycan) tosis) in CSF were significantly reduced by therapy with
was extensively degraded. These findings suggest that pneu- dexamethasone or by the intracisternal administration of
mococcal cell wall lytic products released during antibiotic- goat anti-human TNF-ao antibodies. Interestingly, simulta-
induced autolysis during treatment of bacterial meningitis neous analysis of serum samples revealed no detectable
contribute to the host inflammatory response in the sub- TNF activity, indicating that TNF was principally produced
arachnoid space. within the CNS. This local production of TNF-ot has also
been observed in patients with bacterial meningitis (79). In
LPS addition, the finding of increased CSF concentrations of
TNF-a may be specific for bacterial meningitis, as was
Intracisternal inoculation of purified H. influenzae type b suggested in a recent study. TNF-ot concentrations, mea-
LPS (in contrast to purified capsular polysaccharide) also sured in mice or humans with either bacterial or viral
induces subarachnoid space inflammation. In an experimen- meningitis, were elevated in CSF only during bacterial
tal rabbit model, the intracisternal inoculation of purified meningitis (67); however, only a few patients were analyzed.
LPS (2 fg to 200 ng) produced a dose-dependent increase in These results were confirmed in a subsequent prospective
concentrations of leukocytes in CSF, whereas no inflamma- study of patients with infectious meningitis. The presence of
tion was induced by intracisternal inoculation of purified H. TNF-a in CSF was indicative of a bacterial etiology, al-
influenzae type b capsular polysaccharide (149). This inflam- though the absence of TNF-ao did not exclude the diagnosis
matory response was blocked by polymyxin B and neutro- of bacterial meningitis (93). In addition, elevated concentra-
phil acyloxyacyl hydrolase, a fact that supports the impor- tions of PGE2, prostacyclin, IL-11, and TNF in CSF were
tance of the lipid A region of the LPS in the induction of CSF found in the majority of infants and children with bacterial
inflammation. Pretreatment of the LPS with a monoclonal meningitis (91). Systemic dexamethasone administration sig-
VOL. 6, 1993 PATHOGENESIS OF BACTERIAL MENINGITIS 127

TABLE 2. Concentrations of endotoxin in CSF and bacterial titers before and after antibiotic therapy in eight patientsa

Time TotaTiter
CFU/ml)
(l (log10 of endotoxin/mi (mean
Loglo ngBound + SE)
CFU/ml) ~Total Free (%
Before therapy 6.69 + 0.71 2.06 + 0.27 2.04 ± 0.28 0.75 + 0.21 (8.5 ± 4.2)
After therapy 5.24 + 0.85 1.53 + 0.29 1.06 ± 0.33 1.29 + 0.23 (63.5 + 11.5)
P value b <0.005 <0.02 <0.01 <0.01 (<0.005)
a Data from reference 5 with permission of The University of Chicago Press.
b
Significance of paired t test (two tailed).

nificantly reduced concentrations of PGE2 and IL-lp in CSF neutrophilic infiltration into CSF (174), have also been
and improved several indices of meningeal inflammation. observed in patients with bacterial meningitis (58, 123, 175).
Inoculation of purified rabbit TNF-a or human recombi- These experimental and clinical studies strongly suggest that
nant IL-1B into the CSF of rabbits also produced significant release of inflammatory mediators into the CSF during
CSF inflammation (117). Simultaneous administration of bacterial meningitis is responsible for induction of a marked
lower doses of each cytokine resulted in an apparent syner- subarachnoid space inflammatory response and may possi-
gistic inflammatory response that was manifested by a more bly correlate with morbidity and mortality from this disor-
rapid and significantly increased influx of leukocytes into der.
CSF than occurred with administration of each cytokine The source of these inflammatory cytokines within the
alone. In contrast, in an experimental rabbit model of CSF of patients with bacterial meningitis is unclear. LPS
pneumococcal meningitis, CSF leukocytosis, BBB perme- stimulation of astrocytes and microglia in vitro leads to
ability, and brain edema were induced by intracisternal release of various cytokines (43), and vascular endothelial
inoculation of human recombinant TNF-a, macrophage in- cells in culture produce IL-1 in response to stimulation with
flammatory proteins 1 and 2, and IL-la but not by intracis- either LPS or TNF (69, 78, 94). One study utilizing purified
ternal inoculation with IL-lp (127). Antibodies homologous preparations of cerebral microvascular endothelial cells from
to each mediator inhibited leukocytosis and brain edema. In rats has demonstrated that these cells release IL-6 in vitro in
rabbits treated with a monoclonal antibody to CD18 to response to LPS stimulation (160). Further studies, how-
render neutrophil-endothelial cell interactions dysfunctional, ever, are warranted to address these issues as they pertain to
each cytokine lost the ability to cause leukocytosis and brain the patient with bacterial meningitis.
edema. Therefore, these cytokines have multiple complex
and interrelated activities in the CNS that contribute to INCREASED INTRACRANIAL PRESSURE
tissue damage during pneumococcal meningitis.
These findings have implications with regard to outcomes The major element that contributes to an increase in
in patients with bacterial meningitis. Outcome from menin- intracranial pressure during bacterial meningitis is the devel-
gitis due to gram-negative bacilli has been correlated with opment of cerebral edema, which may be vasogenic, cyto-
persistence of organisms and higher concentrations of endo- toxic, and/or interstitial in origin and may result in life-
toxin in CSF (as detected by the Limulus lysate assay) (73). threatening cerebral herniation and other complications (26,
A study of children with H. influenzae meningitis docu- 40, 41, 57, 98). Vasogenic cerebral edema is principally a
mented that treatment with ceftriaxone induced a marked consequence of increased BBB permeability (see above).
increase in concentrations of free LPS in CSF within 2 to 6 Cytotoxic cerebral edema results from swelling of the cellu-
h (Table 2) that correlated with the Herson-Todd severity lar elements of the brain, most likely due, in bacterial
score and the number of febrile hospital days (5). These data meningitis, to release of toxic factors from neutrophils or
suggest that early release of "free" LPS (i.e., LPS not bacteria or both. Secretion of antidiuretic hormone also
present in outer membranes of viable organisms) after anti- contributes to the pathogenesis of cytotoxic edema, with
microbial therapy enhances the host subarachnoid space resultant hypotonicity of extracellular fluid and increased
inflammatory response. The degree of elevated concentra- permeability of the brain to water (62). Interstitial edema
tions of IL-1lB in CSF also correlated significantly with reflects obstruction of flow in normal CSF pathways (e.g.,
outcome from neonatal gram-negative bacillary meningitis in from the subarachnoid space to blood), as in hydrocephalus.
children (88). In another study of infants and children In an experimental rabbit model of pneumococcal or E.
predominantly with H. influenzae type b meningitis, patients coli-caused meningitis, the CSF outflow resistance, defined
with IL-113 concentrations in CSF of .500 pg/ml were more (and quantified) as factors that inhibit the flow of CSF from
likely to develop neurologic sequelae (87). Although ele- the subarachnoid space to the major dural sinuses, was
vated concentrations of TNF in CSF were observed in 50 to markedly elevated (133); these alterations remained for as
75% of patients with bacterial meningitis, there was no long as 2 weeks despite rapid CSF sterilization with penicil-
correlation between concentrations of TNF in CSF and lin therapy. An increase in the outflow resistance to CSF
outcome. The roles of other inflammatory cytokines in the movement may cause interstitial brain edema and/or the
induction of subarachnoid space inflammation are less clear. resultant hydrocephalus during bacterial meningitis.
Elevated concentrations of platelet-activating factor in CSF Subsequent studies have examined these concepts in more
have been demonstrated in children with H. influenzae detail by measuring the water content of the brain (indicative
meningitis (6) and correlated with bacterial density and with of cerebral edema if elevated), concentrations of lactate in
LPS and TNF-a concentrations in CSF. These increased CSF, and CSF pressure in animals with pneumococcal
concentrations of TNF-a and platelet-activating factor were meningitis (154). All three parameters were elevated in
associated with severity of disease. Increased concentra- infected animals. Treatment with ampicillin sterilized the
tions of IL-6, occurring after release of TNF-ot and before CSF rapidly and normalized the water content of the brain
128 TUNKEL AND SCHELD CLIN. MICROBIOL. REV.

and intracranial pressure within 24 h, but the concentration quently lead to vasodilatation or organic stenosis or both
of lactate in the CSF remained elevated. The bacterial cell later in the course of disease (185). Phlebitis of the major
components responsible for the production of brain edema cortical draining vessels or dural sinuses or both may result
were studied in an experimental model of E. coli-caused in thrombosis with secondary brain infarction, focal neuro-
meningitis (155). Treatment with cefotaxime but not chlor- logic deficits, and prominent seizure activity.
amphenicol induced a marked rise in concentrations of
endotoxin in the CSF that were associated with an increase ALTERATIONS IN CEREBRAL BLOOD FLOW
in the water content of the brain. These effects were neu-
tralized by either polymyxin B or a monoclonal antibody In combination with increased intracranial pressure, cere-
against lipid A, indicating that increased concentrations of bral vasculitis may result in altered cerebral blood flow in
endotoxin in CSF may be associated with brain edema. The patients with bacterial meningitis. In the infant rhesus mon-
putative role of the leukocyte in these processes was re- key model of H. influenzae meningitis, cerebral blood flow
cently examined in an experimental meningitis model (152). (measured by an autoradiographic technique utilizing
In sterile meningitis induced by the intracisternal inoculation [14C]antipyrine) was lower in certain areas of the cortex
of N-formylmethionylleucylphenylalanine (fMLP), a chemo- (postcentral, temporal, and occipital areas) than in the
tactic peptide, both high (10-' M) and low (10-5 M) doses hypothalamus and midbrain while the brain stem was hyper-
induced a CSF pleocytosis, although only high doses pro- perfused, suggesting that one of the initial physiologic
duced an increase in the water content of the brain; intra- changes in H. influenzae meningitis is cerebral cortical
cranial pressure and the concentrations of lactate and pro- hypoperfusion with resultant relative cerebral anoxia (141).
tein in the CSF were unaltered. No increase in the water A recent report has demonstrated that cerebrovascular au-
content of the brain was observed in neutropenic animals. toregulation is lost in experimental bacterial meningitis
When high doses of N-formylmethionylleucylphenylalanine (171). Cerebral blood flow was increased when systemic
were injected during the course of pneumococcal meningitis blood pressure was raised and decreased when blood pres-
in rabbits, the results were similar, suggesting that neutro- sure was lowered, indicating that flow was pressure passive.
phils contributed to cerebral edema if adequately stimulated. Similar changes were observed in intracranial pressure;
The parameters of increased intracranial pressure and in- increased blood flow led to increased intracranial pressure.
creased concentrations of lactate and protein in the CSF Furthermore, in an experimental rat model of meningitis
seemed unrelated to the presence of neutrophils. This area (105), an increase in cerebral blood flow was observed within
remains controversial, however, because neutrophils are the first few hours of intracisternal inoculation of either live
required for the increased BBB permeability seen in re- pneumococci or pneumococcal cell wall fragments. These
sponse to intracisternal injection of bacterial products and results suggested that maintenance of adequate intravascular
inflammatory mediators (112, 183). Additional studies are volume status and minimization of stimuli that increase
needed to more precisely define the role of the neutrophil systemic blood pressure may be important in the treatment
within the CNS in the pathophysiology of bacterial menin- of bacterial meningitis and of potentially practical clinical
gitis. relevance. Measurement of cerebral blood flow (by the
Variability among bacterial strains may also be an impor- xenon-133 intra-arterial injection method) in an earlier study
tant determinant in the production of brain edema. A recent of patients with bacterial meningitis revealed a 30 to 40%
study found that intracisternal injection of three different reduction in average total blood flow in five patients with
pneumococcal isolates resulted in pronounced differences in pneumococcal meningitis (mean age, 54 years) but not in five
the pathophysiologic profiles 24 h after challenge (153). patients with meningococcal meningitis (mean age, 20 years)
Following intracisternal inoculation of pneumococcal cell (165). An inverse relationship between cerebral blood flow
wall fragments, the chemical composition of the fragments, velocity and intracranial pressure has been seen in infants
specifically the degree of teichoication, was found to influ- with bacterial meningitis (76). Among eight patients, these
ence the induction of brain edema during bacterial meningi- alterations were detected only in the four older infants (ages,
tis. Spontaneous release of cell wall is reduced for certain 3 to 10 months) and not in the four neonates (ages, 5 to 30
pneumococcal strains and may thus limit their inflammatory days), in whom no changes in cerebral blood flow velocity
potential. Further work is needed, however, to determine were observed.
whether these differences affect the clinical expression of A subsequent study that measured total and regional
disease. cerebral blood flow by stable xenon computed tomography
in 20 children seriously ill with bacterial meningitis revealed
CEREBRAL VASCULITIS a global decrease in cerebral blood flow and even more
regional variability (7). Autoregulation of cerebral blood flow
Bacterial meningitis exerts profound effects on blood was preserved in the patients studied, although hyperventi-
vessels coursing through the subarachnoid space (116). The lation reduced cerebral blood flow below the ischemic
resulting vasculitis leads to narrowing and/or thrombosis of threshold, raising important concerns about the routine use
cerebral blood vessels and the propensity for ischemia of hyperventilation in the management of increased intracra-
and/or infarction of underlying brain. Arteriography in chil- nial pressure in patients with bacterial meningitis. Some
dren with bacterial meningitis uniformly demonstrates leak- authors have suggested that in infants and children with
age of contrast material or other vascular abnormalities in bacterial meningitis and initially normal computed tomogra-
the subarachnoid space, although these changes reverted to phy or magnetic resonance imaging scans, the chance that
normal following successful antibiotic therapy. Severe neu- cerebral blood flow would be reduced to ischemic levels is
rologic complications (e.g., hemiparesis and quadriparesis) unlikely, and in this situation hyperventilation may safely
with permanent sequelae may result from involvement of the reduce elevated intracranial pressure for the first 24 to 48 h
large arteries at the base of the brain (59). Vasospasm may before its effect diminishes (8). However, in children with
also occur secondary to release of humoral factors elabo- cerebral edema revealed by neuroimaging studies, cerebral
rated within the CSF or blood vessel wall and may subse- blood flow is more likely to be normal or reduced, so that
VOL. 6, 1993 PATHOGENESIS OF BACTERIAL MENINGITIS 129

TABLE 3. Effect of cyclooxygenase and lipoxygenase inhibitors on CSF leukocytosis in rabbits after intracisternal
injection of 30 p.g of pneumococcal cell walls'

Compound (mg/k
anDoseand Times (h)C CSF leukocytosisd after:
route" ~~~~~~~
~~5
h 7h ~ 24 h

Control 677 34 1,545 ±90 870 ± 92


Methylprednisolone 30, i.m. -1 50 ± 20 198 ± 30e 54 ± 20
Diclofenac sodium 5, i.v. -1, +2 110 ± 40 433 ± 210e 1,183 ± 150
Indomethacin 5, p.o. -1, +2, +5 155 ± 60 300 ± 50 320 ± 50
Nordihydroguaiaretic acid 5, p.o. -1, +2, +5 1,093 - 500 870 ± 240
Oxindanac 5, p.o. -1, +2, +5 118 ± 57 59 ± 14e 500 ± 325
a Reproduced from reference 167 with permission of The University of Chicago Press.
bi.m., intramuscular; i.v., intravenous; p.o., oral.
c Time of administration in reference to time in hours of cell wall challenge dose.
d Mean number of leukocytes per microliter of CSF + standard deviation. Value at 0
h, 28 + 14 cells per p.I.
e p < 0.01 compared with control at 7 h.

hyperventilation may decrease the intracranial pressure at sponse 24 h after challenge with cell wall. An inhibitor of the
the expense of a significant reduction of cerebral blood flow, lipoxygenase pathway, nordihydroguaiaretic acid, was inef-
possibly approaching ischemic thresholds. These patients fective in preventing cell wall-induced inflammation. Similar
may benefit from early use of diuretics (e.g., furosemide) or results were observed with these adjunctive agents after
osmotically dehydrating agents such as mannitol, providing intracisternal challenge with live pneumococci. There was a
that intravascular volume is protected. Corticosteroids may correlation between the concentrations of the arachidonic
also be useful in this situation. However, it must be stressed acid metabolite PGE2 and of leukocytes in the CSF after
that controlled clinical trials examining these issues have yet intracisternal challenge with either live pneumococci or
to be performed. Although the definitive changes in cerebral pneumococcal cell walls, and inhibition of the cyclooxyge-
blood flow during bacterial meningitis are controversial and nase pathway reduced CSF inflammation and the concentra-
may vary with the stage of disease, these blood flow alter- tion of PGE2 in the CSF. Administration of the nonsteroidal
ations may lead to regional hypoxia, increased concentra- anti-inflammatory agent indomethacin also led to a decrease
tions of lactate in the brain secondary to utilization of in both the water content of the brain and concentrations of
glucose by anaerobic glycolysis, and CSF acidosis, which PGE2 during experimental rabbit pneumococcal meningitis,
may be a precursor to encephalopathy. This phenomenon although there was no reduction in intracranial pressure
has recently been examined in an experimental rabbit model (172). In addition, an anti-inflammatory agent (dexametha-
of pneumococcal meningitis (173) in which animals given a sone or oxindanac) lessened the massive influx of serum
lower fluid regimen (50 ml/kg per 24 h) of normal saline had albumin and other proteins of high and low molecular masses
lower mean arterial blood pressure, lower cerebral blood into the CSF during the early phases of experimental pneu-
flow, and higher concentrations of lactate in the CSF com- mococcal meningitis (61). Ampicillin given alone or in com-
pared with animals that received a higher fluid regimen (150 bination with indomethacin was ineffective in preventing this
ml/kg per 24 h). These results suggest that intravascular influx, and the abnormal protein profile in the CSF persisted
volume status may be a critical factor in determination of for up to 30 days after the initiation of the experimental
cerebral blood flow and, therefore, the degree of cerebral infection.
ischemia in meningitis. Several corticosteroid agents have been evaluated in ex-
perimental animal models of bacterial meningitis. Early
ADJUNCTIVE THERAPEUTIC STRATEGIES studies revealed a significant reduction in the mass of
leukocytes within the meninges of rabbits with pneumococ-
Experimental Studies cal meningitis following methylprednisolone administration
compared with concentrations in infected controls (97);
Because of the information supporting subarachnoid space chemotactic activity, chemotactic response, and phagocyto-
inflammation as a major factor contributing to morbidity and sis in CSF were, however, not altered by methylpred-
mortality from bacterial meningitis, several studies in exper- nisolone treatment, although there was an attenuation of
imental animal models have examined whether attenuation rabbit neutrophil adherence to a nylon fiber column. CSF
of this inflammatory response might be beneficial. As stated outflow resistance (defined as factors that inhibit the flow of
above, the generation of pneumococcal cell wall components CSF from the subarachnoid space to the major dural sinuses
after treatment with bacteriolytic antibiotics in the experi- and quantified following infusion of mock CSF into the
mental rabbit model may contribute to an increased inflam- subarachnoid space) was also reduced by methylpred-
matory response within the subarachnoid space (168, 169). nisolone therapy and to a greater extent than in untreated or
This CSF inflammatory response was reduced by agents penicillin-treated rabbits with pneumococcal meningitis
known to exert their effects by inhibition of the cyclooxyge- (133). The reduction in resistance was apparent within 4 h of
nase pathway of arachidonic acid metabolism (Table 3) the second injection of methylprednisolone (at a dose of 30
(167). Treatment with methylprednisolone or oxindanac in mg/kg of body weight intramuscularly 16 and 20 h following
addition to antimicrobial agents was particularly effective in intracistemal inoculation); a rebound increase in CSF out-
decreasing pneumococcal cell wall-induced inflammation, flow resistance was not seen after corticosteroid therapy was
whereas another inhibitor, diclofenac sodium, was effective discontinued.
only when administered 5 and 7 h after inoculation of The effects of methylprednisolone or dexamethasone on
bacteria into CSF, but did not produce an inhibitory re- water content of the brain, CSF pressure, and lactate con-
130 TUNKEL AND SCHELD CLIN. MICROBIOL. REV.

TABLE 4. Influence of methylprednisolone and dexamethasone on manifestations of a 24-h infection with pneumococci in rabbitsa

Treatment Bacterial titer Leukocyte count Concn of lactate Change in CSF Water content (g
(log10o CFU/ml) (103/mm3) in CSF (mg/dl) pressure (mm Hg) [dry wt]) of brain
No infection ND <0.01 14.0 ± 2.6 +0.8 ± 1.4 396 ± 14
24-h infection
No treatment 7.0 ± 1.2 2.5 ± 6.6 75.3 ± 25.6 +7.5 ± 6.5 410 ± llb
Methylprednisolone 7.6 ± 0.7c 3.9 ± 4.9 64.3 ± 33.1 +7.8 ± 5.4 395 ± 9
Dexamethasone 6.7 ± 0.6 2.1 ± 2.2 43.8 ± 12.3 +1.8 ± 2.7d 399 ± 12
a Reproduced from reference 154 with permission of The University of Chicago Press. All values are expressed as means + standard deviations. ND, not
determined.
b p < 0.02 in comparison with other groups.
c P < 0.05 in comparison with dexamethasone-treated group.
d p < 0.02 in comparison with other groups infected for 24 h.

centrations in the CSF of rabbits with pneumococcal men- meningitis, which is due in part to the development of
ingitis have also been examined (Table 4) (154). Administra- labyrinthritis following spread of infection to the inner ear.
tion of both agents completely reversed the development of The infant rat model has been utilized to determine the
brain edema at the times studied, but only dexamethasone influence of corticosteroid administration on the inflamma-
reduced the increases in CSF pressure and lactate; methyl- tory reaction in the cochleas of infected animals (63). Infant
prednisolone, but not dexamethasone, was associated with rats were inoculated intraperitoneally with H. influenzae
an increase in concentrations of bacteria in CSF. However, type b, and 24 h later they were treated with ampicillin or
neither agent was superior to therapy with ampicillin alone in ampicillin plus dexamethasone. At 48 h, concentrations of
reducing cerebral edema or intracranial pressure. No com- leukocytes in CSF were significantly lower in the dexameth-
parison between ampicillin alone and ampicillin plus corti- asone group than in the ampicillin-alone group, although no
costeroids, a comparison that would be relevant to the histologic differences in the degree of cochlear inflammation
potential clinical efficacy of adjunctive corticosteroids in were noted between groups. The extent of cochlear inflam-
humans, was made. In another study, treatment with ceftri- mation was minimal only in the ampicillin group, however.
axone was compared with treatment with ceftriaxone plus Pentoxifylline, a phosphodiesterase inhibitor that de-
dexamethasone in an experimental rabbit model of H. influ- creases endotoxin-induced TNF-a production and attenu-
enzae meningitis (150). Combination therapy consistently ates the inflammatory action of IL-1 and TNF on leukocyte
reduced the water content of the brain, CSF pressure, and function (147), has also been examined in the experimental
the lactate concentration in the CSF to a greater degree than rabbit model of H. influenzae type b meningitis (125).
ceftriaxone alone, although the differences were not statis- Administration of pentoxifylline 20 min before intracisternal
tically significant. By 29 h after inoculation (a well-estab- challenge with H. influenzae type b LPS significantly re-
lished disease in this model), the values were comparable duced concentrations of leukocytes, protein, and lactate in
whether the animals received antibiotic alone, dexametha- CSF. Peak concentrations of TNF in CSF were reduced by
sone alone, or the combination. The authors suggested that more than one-third in pentoxifylline-treated animals, al-
dexamethasone might be more beneficial if administered though this reduction was not statistically significant and
early during antibiotic therapy after (or even before) antibi- was unlikely to be solely responsible for the marked modu-
otic-induced lysis and release of microbial products. In a lation of meningeal inflammation. Dexamethasone was su-
subsequent analysis utilizing the rabbit model of H. influen- perior to pentoxifylline in modulation of these inflammatory
zae type b meningitis (89), a significant increase in concen- changes in CSF, and no appreciable synergism was observed
trations of endotoxin in CSF was documented 2 h following when dexamethasone and pentoxifylline were used together.
ceftriaxone administration; this increase was followed by a Recent studies have examined the effects of a monoclonal
rise in TNF concentrations in CSF (Fig. 5). Simultaneous antibody (IB4) directed against the CD18 family of receptors
administration of dexamethasone and ceftriaxone did not on leukocytes on reduction of subarachnoid space inflam-
affect the appearance of endotoxin in CSF, but there was a mation. In one study, intravenous inoculation of IB4 blocked
marked attenuation in concentrations of TNF in CSF mea- the accumulation of leukocytes in the subarachnoid space
sured 8 h later. Adjunctive dexamethasone therapy also despite intracisternal challenge with H. influenzae type b, N.
resulted in a significant decrease in the resultant CSF leuko- meningitidis, pneumococcal cell wall, or LPS (170). In
cytosis and a trend towards earlier improvement in concen- addition, the parameters of BBB permeability, development
trations of glucose, lactate, and protein in CSF. of cerebral edema, and death were prevented in the animals
Therefore, adjunctive dexamethasone therapy in concert challenged with lethal doses of S. pneumoniae that also
with antimicrobial agents appeared to improve a number of received the monoclonal antibody. In a second study, IB4
parameters of subarachnoid space inflammation in experi- and dexamethasone were administered together in a rabbit
mental animal models of bacterial meningitis. These param- model of H. influenzae type b meningitis. The result was a
eters improved without any apparent decrease in the rate of marked attenuation of all indices of meningeal inflammation
bacterial killing within the CSF in vivo, although other and a reduction in water accumulation in the brain compared
experimental studies have shown that administration of with the results obtained when each agent was given alone
methylprednisolone decreased the entry of ampicillin and and when infected animals were left untreated (124). Despite
gentamicin into CSF (132). this profound amelioration of meningeal inflammation, clear-
The subarachnoid space inflammatory response may con- ance rates of bacteria from CSF to blood and from vascular
tribute to the pathogenesis of hearing loss in bacterial compartments were unaffected. These results indicate that
VOL. 6, 1993 PATHOGENESIS OF BACTERIAL MENINGITIS 131

25 pathogenesis and pathophysiology of bacterial meningitis


had been elucidated, and the studies also suffered from
several flaws (e.g., inadequate corticosteroid dose and/or
20 control group). Therefore, we concentrate on evaluation of
E several recent and relevant clinical studies published in the
C
literature.
15 One study was a double-blind, placebo-controlled trial of
adjunctive dexamethasone therapy in infants and children
1o
with bacterial meningitis (66). The patients received an
UA.
z
antibiotic (cefuroxime or ceftriaxone) with either dexameth-
asone or placebo. The patients who received an antibiotic
plus dexamethasone became afebrile sooner, had more rapid
normalization of CSF parameters (concentrations of glu-
cose, protein, and lactate), and were significantly less likely
to acquire moderate to severe bilateral sensorineural hearing
loss (15.5 versus 3.3%). In addition, concentrations of IL-1,B
but not TNF-cx in CSF were significantly lower 18 to 36 h
later in patients given adjunctive dexamethasone. However,
these findings were significant only in patients with menin-
gitis caused by H. influenzae type b, and the benefits in
terms of morbidity (sensorineural hearing loss) were statis-
tically significant only in patients receiving cefuroxime and
E not in those receiving ceftriaxone. The latter point is impor-
E tant, because cefuroxime has recently been shown to be
-)
inferior to ceftriaxone in a randomized prospective study of
the therapy of childhood bacterial meningitis (129). In addi-
co tion, four patients who received adjunctive dexamethasone
developed gastrointestinal hemorrhage, and two of these
patients required blood transfusions.
A second study, from Egypt, of children and adults with
bacterial meningitis demonstrated a significant reduction in
mortality rate and overall neurologic sequelae in patients
with pneumococcal meningitis who received adjunctive
dexamethasone therapy concomitant with antibiotics (ampi-
cillin plus chloramphenicol) (48). However, no significant
Time (hours) differences between groups in time to afebrility or improve-
FIG. 5. (Upper) Comparative changes in concentrations of ment in CSF parameters were observed; furthermore, the
TNF-at in CSF in rabbits that received ceftriaxone (CTX) alone at 6 antibiotics were given intramuscularly, there was no docu-
h (*, K; n = 5), CTX plus dexamethasone (DXM) simultaneously at mentation of possible adverse effects, and an extraordinarily
6 h (0, 0; n = 6), CTX at 6 h and DXM at 7 h (A, A; n 4) or no
=
high percentage of patients presented in a comatose state. In
treatment (controls) (U, El; n = 5). (Lower) Counts of leukocytes fact, most patients (370 of 429) had received inadequate
(WBC) in CSF for the same four groups of rabbits as in the upper therapy for the 3 to 5 days prior to hospitalization. There
panel. Meningitis was induced at time zero. Reproduced from were no differences between the two groups in mortality rate
reference 89 with permission of The University of Chicago Press.
or rate of hearing impairment for patients with meningococ-
cal or H. influenzae meningitis, but children with H. influ-
enzae meningitis were too young to be tested audiometri-
dual therapy with agents directed against the CSF produc- cally.
tion of cytokines and recruitment of leukocytes into the In a third recently published trial centered in Costa Rica
subarachnoid space may be associated with improved out- (99), infants and children with bacterial meningitis were
come in bacterial meningitis. randomized in a placebo-controlled, double-blind fashion to
receive cefotaxime with or without adjunctive dexametha-
Clinical Trials sone therapy; in this study, the dexamethasone was admin-
istered 15 to 20 min before the first dose of cefotaxime in an
Taken together, the experimental studies described above effort to attenuate the CSF inflammatory response associ-
indicate that adjunctive dexamethasone therapy diminishes ated with administration of bacteriolytic antibiotics. Twelve
the CSF inflammatory response and the subsequent patho- hours after treatment was begun, meningeal inflammation
physiologic consequences of this inflammation, specifically, and concentrations of TNF-a and platelet-activating factor
elevated water content in the brain and elevated CSF pres- in CSF had decreased more rapidly in dexamethasone-
sure. On the basis of these observations, several trials were treated patients. In addition, by 24 h, the clinical conditions
undertaken to examine the effects of adjunctive corticoster- and mean prognostic scores were significantly better among
oids on outcomes in patients with bacterial meningitis. Early patients receiving adjunctive dexamethasone therapy. When
clinical trials performed in the 1960s failed to show any the patients were monitored for a mean of 15 months, those
benefit for adjunctive corticosteroids (either methylpred- who had received adjunctive dexamethasone had a signifi-
nisolone or dexamethasone) (12, 31). However, these trials cantly decreased incidence of one or more neurologic se-
were performed before much of the recent information on quelae, although reduction of audiologic impairment was
132 TUNKEL AND SCHELD CLIN. MICROBIOL. REV.

only a trend. Overall mortality was not reduced in the 15. Bevilacqua, M. P., J. S. Pober, M. E. Wheeler, R. S. Cotran,
dexamethasone group. and M. A. Gimbrone, Jr. 1985. Interleukin 1 acts on cultured
Despite the studies described above, controversy regard- human vascular endothelium to increase the adhesion of poly-
ing the routine use of adjunctive dexamethasone therapy in morphonuclear leukocytes, monocytes, and related leukocyte
all patients with bacterial meningitis remains (75, 165). The cell lines. J. Clin. Invest. 76:2003-2011.
16. Bevilacqua, M. P., S. Stengelin, M. A. Gimbrone, Jr., and B.
data support the use of adjunctive dexamethasone in infants Seed. 1989. Endothelial leukocyte adhesion molecule 1: an
and children with bacterial meningitis caused by H. influen- inducible receptor for neutrophils related to complement reg-
zae type b. Routine use of dexamethasone therapy for ulatory proteins and lectins. Science 243:1160-1165.
adults, however, cannot be presently recommended, pend- 17. Bohr, V., 0. Paulson, and N. Rasmussen. 1984. Pneumococcal
ing results of ongoing studies. If dexamethasone is used, the meningitis: late neurologic sequelae and features of prognostic
timing of administration is crucial. Administration before or impact. Arch. Neurol. 41:1045-1049.
with antibiotics is optimal for attenuating the subarachnoid 18. Bradbury, M. W. 1984. The structure and function of the
space inflammatory response. Patients receiving such ther- blood-brain barrier. Fed. Proc. 43:186-190.
apy also need careful monitoring (in addition to the usual 19. Brandtzaeg, P., T. E. Mollnes, and P. Kierulf. 1989. Comple-
ment activation and endotoxin levels in systemic meningococ-
routine for critically ill patients) for the possibility of gas- cal disease. J. Infect. Dis. 160:58-65.
trointestinal hemorrhage. Future studies of the pathogenesis 20. Broome, C. V., R. R. Facklam, J. R. Allen, D. W. Fraser, and
and pathophysiology of bacterial meningitis should lead to R. Austrian. 1980. Epidemiology of pneumococcal serotypes in
the development of other adjunctive treatment strategies the United States. J. Infect. Dis. 141:119-123.
that may improve the outcome of this disorder. 21. Brown, E. J., S. W. Hosea, and M. M. Frank. 1981. The role of
the spleen in experimental pneumococcal bacteremia. J. Clin.
REFERENCES Invest. 67:975-982.
1. Andersen, B. M., and 0. Solberg. 1984. Endotoxin liberation 22. Brown, E. J., S. W. Hosea, C. H. Hammer, C. G. Burch, and
and invasivity of Neisseria meningitidis. Scand. J. Infect. Dis. M. M. Frank. 1982. A quantitative analysis of the interaction of
16:247-254. antipneumococcal antibody and complement in experimental
2. Anderson, P., R. B. Johnston, Jr., and D. H. Smith. 1972. pneumococcal bacteremia. J. Clin. Invest. 69:85-98.
Human serum activities against Haemophilus influenzae type 23. Brown, E. J., K. A. Joiner, R. M. Cole, and M. Berger. 1983.
b. J. Clin. Invest. 51:31-38. Localization of complement component 3 on Streptococcus
3. Anderson, P. A., A. Flesher, S. Shaw, A. L. Harding, and D. H. pneumoniae: anti-capsular antibody causes complement depo-
Smith. 1980. Phenotypic and genetic variation in the suscepti- sition on the pneumococcal capsule. Infect. Immun. 39:403-
bility of Haemophilus influenzae type b to antibodies to 409.
somatic antigens. J. Clin. Invest. 65:885-891. 24. Bryan, J. P., H. R. de Silva, A. Tavares, H. Roche, and W. M.
4. Andersson, B., B. Eriksson, E. Falsen, A. Fogh, L. A. Hanson, Scheld. 1990. Etiology and mortality of bacterial meningitis in
0. Nylen, H. Peterson, and C. Svanborg-Eden. 1981. Adhesion Northeastern Brazil. Rev. Infect. Dis. 12:128-135.
of Streptococcus pneumoniae to human pharyngeal epithelial 25. Buchanen, N., and G. S. McNab. 1972. Cerebrospinal fluid
cells in vitro: differences in adhesive capacity among strains complement and immunoglobulins in meningitis and encepha-
isolated from subjects with otitis media, septicemia, or menin- litis. S. Afr. Med. J. 46:1376-1382.
gitis or from healthy carriers. Infect. Immun. 32:311-317. 26. Chan, P. H., and R. A. Fishman. 1979. Brain edema: induction
5. Arditi, M., L. Ables, and R. Yogev. 1989. Cerebrospinal fluid in cortical slices by polyunsaturated fatty acids. Science 201:
endotoxin levels in children with H. influenzae meningitis 358-360.
before and after administration of intravenous ceftriaxone. J. 27. Corral, C. J., J. A. Winkelstein, and E. R. Moxon. 1982.
Infect. Dis. 160:1005-1011. Participation of complement in host defense against encapsu-
6. Arditi, M., K. R. Manogue, M. Caplan, and R. Yogev. 1990. lated Haemophilus influenzae types a, c, and d. Infect. Immun.
Cerebrospinal fluid cachectin/tumor necrosis factor-a and 35:759-763.
platelet-activating factor concentrations and severity of bacte- 28. Cross, A. S., P. Gemski, J. C. Sadoff, F. Orskov, and I. Orskov.
rial meningitis in children. J. Infect. Dis. 162:139-147. 1984. The importance of the Kl capsule in invasive infections
7. Ashwal, S., W. Stringer, L. Tomasi, S. Schneider, J. Thompson, caused by Eschenichia coli. J. Infect. Dis. 149:184-193.
and R. Perkin. 1990. Cerebral blood flow and carbon dioxide 29. Crosson, F. A., Jr., J. A. Winkelstein, and E. R. Moxon. 1976.
reactivity in children with bacterial meningitis. J. Pediatr. Participation of complement in the nonimmune host defense
117:523-530. against experimental Haemophilus influenzae bacteremia and
8. Ashwal, S., L. Tomasi, S. Schneider, R. Perkin, and J. Thomp- meningitis. Infect. Immun. 14:882-887.
son. 1992. Bacterial meningitis in children: pathophysiology 30. Dacey, R. G., Jr., and M. A. Sande. 1974. Effect of probenecid
and treatment. Neurology 42:739-748. on cerebrospinal fluid concentrations of penicillin and cepha-
9. Austrian, R., and J. Gold. 1964. Pneumococcal bacteremia losporin derivatives. Antimicrob. Agents Chemother. 6:437-
with special reference to bacteremic pneumococcal pneumo- 444.
nia. Ann. Intern. Med. 60:759-776. 31. deLemos, R. A., and R. J. Haggerty. 1969. Corticosteroids as
10. Beachey, E. H. 1981. Bacterial adherence: adhesin-receptor an adjunctive to treatment in bacterial meningitis. A controlled
interactions mediating the attachment of bacteria to mucosal clinical trial. Pediatrics 44:30-34.
surfaces. J. Infect. Dis. 143:325-345. 32. Devoe, I. W., and J. E. Gilchrist. 1975. Pili on meningococci
11. Beilke, M. A. 1989. Vascular endothelium in immunology and from primary cultures of nasopharyngeal carriers and cerebro-
infectious disease. Rev. Infect. Dis. 11:273-283. spinal fluid of patients with acute disease. J. Exp. Med.
12. Belsey, M. A., C. W. Hoffpauir, and M. H. D. Smith. 1969. 141:297-305.
Dexamethasone in the treatment of acute bacterial meningitis: 33. Dodge, P. R., H. Davis, R. Feigin, S. J. Holmes, S. L. Kaplan,
the effect of study design on the interpretation of results. D. P. Jubelirer, B. W. Stechenberg, and S. K. Hirsh. 1984.
Pediatrics 44:503-513. Prospective evaluation of hearing impairment as a sequela of
13. Bernhardt, L. L., M. S. Simberkoff, and J. J. Rahal, Jr. 1981. acute bacterial meningitis. N. Engl. J. Med. 311:869-874.
Deficient cerebrospinal fluid opsonization in experimental Esch- 34. Ernst, J. D., J. M. Decazes, and M. A. Sande. 1983. Experi-
erichia coli meningitis. Infect. Immun. 32:411-413. mental pneumococcal meningitis: role of leukocytes in patho-
14. Beutler, B., N. Krochin, I. W. Milsark, C. Leudke, and A. genesis. Infect. Immun. 41:275-279.
Cerami. 1986. Control of cachectin (tumor necrosis factor) 35. Ernst, J. D., K. T. Hartiala, I. M. Goldstein, and M. A. Sande.
synthesis: mechanisms of endotoxin resistance. Science 232: 1984. Complement (C5)-derived chemotactic activity accounts
977-980. for accumulation of polymorphonuclear leukocytes in cerebro-
VOL. 6, 1993 PATHOGENESIS OF BACTERIAL MENINGITIS 133

spinal fluid of rabbits with pneumococcal meningitis. Infect. and J. Van Snick. 1988. Elevated levels of the 26K human
Immun. 46:81-86. hybridoma growth factor (interleukin 6) in cerebrospinal fluid
36. Feldman, W. E. 1976. Concentrations of bacteria in cerebro- of patients with acute infection of the central nervous system.
spinal fluid of patients with bacterial meningitis. J. Pediatr. Clin. Exp. Immunol. 71:320-323.
88:549-552. 59. Igarashi, M., R. C. Gilmartin, B. Gerald, F. Wilburn, and J. T.
37. Feldman, W. E. 1977. Relation of concentrations of bacteria Jabbour. 1984. Cerebral arteritis and bacterial meningitis.
and bacterial antigens in cerebrospinal fluid to prognosis in Arch. Neurol. 41:531-535.
patients with bacterial meningitis. N. Engl. J. Med. 296:433- 60. Kadurugamuwa, J. L., B. Hengstler, M. A. Bray, and 0. Zak.
435. 1989. Inhibition of complement-factor-5a-induced inflamma-
38. Fine, D. P. 1975. Pneumococcal type-associated variability in tory reactions by prostaglandin E2 in experimental meningitis.
alternate complement pathway activation. Infect. Immun. 12: J. Infect. Dis. 160:715-719.
772-778. 61. Kadurugamuwa, J. L., B. Hengstler, and 0. Zak. 1989. Cere-
39. Finland, M. 1973. Excursions into epidemiology: selected brospinal fluid protein profile in experimental pneumococcal
studies during the past four decades at Boston City Hospital. J. meningitis and its alteration by ampicillin and anti-inflamma-
Infect. Dis. 128:76-124. tory agents. J. Infect. Dis. 159:26-34.
40. Fishman, R. A. 1975. Brain edema. N. Engl. J. Med. 293:706- 62. Kaplan, S. L., and R. D. Feigin. 1978. The syndrome of
711. inappropriate secretion of antidiuretic hormone in children
41. Fishman, R. A., K. Sligar, and R. B. Hake. 1977. Effects of with bacterial meningitis. J. Pediatr. 92:758-761.
leukocytes on brain metabolism in granulocytic brain edema. 63. Kaplan, S. L., E. P. Hawkins, M. W. Kline, G. S. Patrick, and
Ann. Neurol. 2:89-94. E. 0. Mason, Jr. 1989. Invasion of the inner ear by Haemo-
42. Flexner, S. 1913. The results of serum treatment in 1300 cases philus influenzae type b in experimental meningitis. J. Infect.
of epidemic meningitis. J. Exp. Med. 17:553-576. Dis. 159:923-930.
43. Fontana, A., F. Kristensen, R. Dubs, D. Gemsa, and E. Weber. 64. Kayhty, H., H. Jousimies-Somer, H. Peltola, and P. H. Makela.
1982. Production of prostaglandin E and an interleukin-1 like 1981. Antibody response to capsular polysaccharides of groups
factor by cultured astrocytes and C6 glioma cells. J. Immunol. A and C Neisseria meningitidis and Haemophilus influenzae
129:2413-2419. type b during bacteremic disease. J. Infect. Dis. 143:32-41.
44. Fraser, D. W., C. C. Geil, and R. A. Feldman. 1974. Bacterial 65. Lam, J. S., D. M. Granoff, J. R. Gilsdorf, and J. W. Costerton.
meningitis in Bernalillo County, New Mexico: a comparison 1980. Immunogenicity of outer membrane derivatives of Hae-
with three other American populations. Am. J. Epidemiol. mophilus influenzae type b. Curr. Microbiol. 3:359-364.
100:29-34. 66. Lebel, M. H., B. J. Frei, G. A. Syrogiannopoulos, D. F.
45. Freter, R. 1980. Prospects for preventing the association of Chrane, M. J. Hoyt, S. M. Stewart, B. D. Kennard, K. D.
harmful bacteria with host mucosal surfaces, p. 441-459. In Olsen, and G. H. McCracken, Jr. 1988. Dexamethasone ther-
E. H. Beachey (ed.), Bacterial adherence, vol. 6. Chapman apy for bacterial meningitis. Results of two double-blind,
and Hall, New York. placebo-controlled trials. N. Engl. J. Med. 319:964-971.
46. Giampaolo, C., W. M. Scheld, J. Boyd, J. Savory, M. Sande, 67. Leist, T. P., K. Frei, S. Kam-Hansen, R. M. Zinkernagel, and
and M. Wills. 1981. Leukocyte and bacterial interrelationships A. Fontana. 1988. Tumor necrosis factor ao in cerebrospinal
in experimental meningitis. Ann. Neurol. 9:328-333. fluid during bacterial, but not viral, meningitis. Evaluation in
47. Gigliotti, F., D. Lee, R. A. Insel, and W. M. Scheld. 1987. IgG murine model infections and in patients. J. Exp. Med. 167:
penetration into the cerebrospinal fluid in a rabbit model of 1743-1748.
meningitis. J. Infect. Dis. 156:394-398. 68. Lesse, A. J., E. R. Moxon, A. Zwahlen, and W. M. Scheld.
48. Girgis, N. I., Z. Farid, I. A. Mikhail, I. Farrag, Y. Sultan, and 1988. Role of cerebrospinal fluid pleocytosis and Haemophilus
M. E. Kilpatrick 1989. Dexamethasone treatment for bacterial influenzae type b capsule on blood brain barrier permeability
meningitis in children and adults. Pediatr. Infect. Dis. J. during experimental meningitis in the rat. J. Clin. Invest.
8:848-851. 82:102-109.
49. Goldstein, G. W., and A. L. Betz. 1986. The blood-brain 69. Libby, P., J. M. Ordovas, K. R. Auger, A. H. Robbins, L. K.
barrier. Sci. Am. 255:74-83. Birinyi, and C. A. Dinarello. 1986. Endotoxin and tumor
50. Gray, B. M., G. M. Converse III, and H. C. Dillon, Jr. 1979. necrosis factor induce interleukin-1 gene expression in adult
Serotypes of Streptococcus pneumoniae causing invasive dis- human vascular endothelial cells. Am. J. Pathol. 124:179-185.
ease. J. Infect. Dis. 140:979-983. 70. LiPuma, J. J., H. Richman, and T. L. Stull. 1990. Haemocin,
51. Greenblatt, J. J., K. Floyd, M. W. Philipps, and C. E. Frasch. the bacteriocin produced by Haemophilus influenzae: species
1988. Morphologic differences in Neisseria meningitidis pili. distribution and role in colonization. Infect. Immun. 58:1600-
Infect. Immun. 56:2356-2362. 1605.
52. Greenwood, B. M. 1978. Chemotactic activity of cerebrospinal 71. Lundberg, C., J. Lonnroth, and C. E. Nord. 1982. Adherence
fluid in pyogenic meningitis. J. Clin. Pathol. 31:213-216. in the colonization of Streptococcus pneumoniae in the naso-
53. Griffis, J. M. 1975. Bactericidal activity of IgA of lytic antibody pharynx of children. Infection 10:63-69.
in human convalescent sera. J. Immunol. 114:1779-1784. 72. Mason, E. O., S. L. Kaplan, B. L. Wiedermann, E. P. Norrod,
54. Griffis, J. M., and M. A. Bertram. 1977. Immunoepidemiology and W. A. Stenback. 1985. Frequency and properties of natu-
of meningococcal disease in military recruits. II. Blocking of rally occurring adherent piliated strains of Haemophilus influ-
serum bactericidal activity by circulating IgA early in the enzae type b. Infect. Immun. 49:98-103.
course of invasive disease. J. Infect. Dis. 136:733-739. 73. McCracken, G. H., Jr., M. M. Mustafa, 0. Ramilo, K. D.
55. Gulig, P. A., G. H. McCracken, Jr., C. F. Frisch, K. H. Olsen, and R. C. Risser. 1989. Cerebrospinal fluid interleukin
Johnston, and E. J. Hansen. 1982. Antibody response of infants 1-beta and tumor necrosis factor concentrations and outcome
to cell surface-exposed outer membrane proteins of Hae- from neonatal gram-negative enteric bacillary meningitis. Pe-
mophilus influenzae type b after systemic Haemophilus dis- diatr. Infect. Dis. J. 8:155-159.
ease. Infect. Immun. 37:82-88. 74. McGee, Z. A., D. S. Stephens, L. H. Hoffman, W. F. Schlech
56. Hansen, E. J., C. F. Frisch, R. L. McDade, Jr., and K. H. III, and R. G. Horn. 1983. Mechanisms of mucosal invasion by
Johnston. 1982. Identification of immunogenic outer membrane pathogenic Neisseria. Rev. Infect. Dis. 5(Suppl. 4):S708-S714.
proteins of Haemophilus influenzae type b in the infant rat 75. McGowan, J. E., Jr., P. J. Chesney, K. B. Crossley, and F. M.
model system. Infect. Immun. 32:1084-1092. LaForce. 1992. Guidelines for the use of systemic glucocorti-
57. Horwitz, S. J., B. Boxerbaum, and J. O'Bell. 1980. Cerebral coids in the management of selected infections. J. Infect. Dis.
herniation in bacterial meningitis in childhood. Ann. Neurol. 165:1-13.
7:524-528. 76. McMenamin, J. B., and J. J. Volpe. 1984. Bacterial meningitis
58. Houssiau, F. A., K. Bukasa, C. J. M. Sindic, J. Van Damme, in infancy: effects on intracranial pressure and cerebral blood
134 TUNKEL AND SCHELD CLIN. MICROBIOL. REV.

flow velocity. Neurology 34:500-504. 1. J. Exp. Med. 163:1363-1375.


77. Meyrick, B., R. Hoover, M. R. Jones, L. C. Berry, Jr., and 95. Nichols, F. C., S. W. Garrison, and H. W. Davis. 1988.
K. L. Brigham. 1989. In vitro effects of endotoxin on bovine Prostaglandin E2 and thromboxane B2 release from human
and sheep lung microvascular endothelial cells. J. Cell. Phys- monocytes treated with bacterial lipopolysaccharide. J. Leu-
iol. 138:165-174. kocyte Biol. 44:376-384.
78. Miossec, P., D. Cavender, and M. Ziff. 1986. Production of 96. Nolan, C. M., R. A. Clark, and H. N. Beaty. 1975. Experimen-
interleukin 1 by human endothelial cells. J. Immunol. 136: tal pneumococcal meningitis. III. Chemotactic activity in ce-
2486-2491. rebrospinal fluid. Proc. Soc. Exp. Biol. Med. 150:134-136.
79. Moller, B., S. C. Mogensen, P. Wendelboe, K. Bendtzen, and 97. Nolan, C. M., C. K. McAllister, E. Walters, and H. N. Beaty.
C. M. Petersen. 1991. Bioactive and inactive forms of tumor 1978. Experimental pneumococcal meningitis. IV. The effect
necrosis factor-a in spinal fluid from patients with meningitis. of methylprednisolone on meningeal inflammation. J. Lab.
J. Infect. Dis. 163:886-889. Clin. Med. 91:979-988.
80. Movat, H. Z. 1987. Tumor necrosis factor and interleukin-1: 98. Nugent, S. K., J. A. Bausher, E. R. Moxon, and M. C. Rogers.
role in acute inflammation and microvascular injury. J. Lab. 1979. Raised intracranial pressure: its management in Neisse-
Clin. Med. 110:668-681. ria meningitidis meningoencephalitis. Am. J. Dis. Child. 133:
81. Moxon, E. R., and P. Anderson. 1979. Meningitis caused by 260-262.
Haemophilus influenzae in infant rats: protective immunity 99. Odio, C. M., I. Faingezicht, M. Paris, M. Nassar, A. Baltodano,
and antibody priming by gastrointestinal colonization with J. Rogers, X. Saez-Llorens, K. D. Olsen, and G. H. McCracken,
Escherichia coli. J. Infect. Dis. 140:471-478. Jr. 1991. The beneficial effects of early dexamethasone admin-
82. Moxon, E. R., M. P. Glode, A. Sutton, and J. B. Robbins. 1977. istration in infants and children with bacterial meningitis. N.
The infant rat as a model of bacterial meningitis. J. Infect. Dis. Engl. J. Med. 324:1525-1531.
136(Suppl.):S186-S190. 100. Ostrow, P. T., E. R. Moxon, N. Vernon, and R. Kapko. 1979.
83. Moxon, E. R., and P. T. Ostrow. 1977. Haemophilus influenzae Pathogenesis of bacterial meningitis. Studies on the route of
bacteremia in infant rats: role of bacteremia in pathogenesis of meningeal invasion following Haemophilus influenzae inocula-
age dependent inflammatory response in cerebrospinal fluid. J. tion in infant rats. Lab. Invest. 40:678-685.
Infect. Dis. 135:303-307. 101. Parkkinen, J., T. K. Korhonen, A. Pere, J. Hacker, and S.
84. Moxon, E. R., A. L. Smith, D. R. Averill, and D. H. Smith. Soinila. 1988. Binding sites in the rat brain for Escherichia coli
1974. Haemophilus influenzae meningitis in infant rats after S fimbriae associated with neonatal meningitis. J. Clin. Invest.
intranasal inoculation. J. Infect. Dis. 129:154-162. 81:860-865.
85. Moxon, E. R., and K. A. Vaughn. 1981. The type b capsular 102. Patrick, D., J. Betts, E. A. Frey, R. Prameya, K. Dorovini-Zis,
polysaccharide as a virulence determinant of Haemophilus and B. B. Finlay. 1992. Haemophilus influenzae lipopolysac-
influenzae: studies using clinical isolates and laboratory trans- charide disrupts confluent monolayers of bovine brain endo-
formants. J. Infect. Dis. 143:517-524. thelial cells via a serum-dependent cytotoxic pathway. J.
86. Mulks, M. H., S. J. Kornfeld, B. Frangione, and A. G. Plaut. Infect. Dis. 165:865-872.
1982. Relationship between the specificity of IgA proteases and 103. Pearson, H. 1977. Sickle cell anemia and severe infections due
serotypes in Haemophilus influenzae. J. Infect. Dis. 146:266- to encapsulated bacteria. J. Infect. Dis. 136:S25-S30.
274. 104. Petersdorf, R. G., and C. N. Luttrell. 1962. Studies on the
87. Mustafa, M. M., M. H. Lebel, 0. Ramilo, K. D. Olsen, J. S. pathogenesis of meningitis. I. Intrathecal injection. J. Clin.
Reisch, B. Beutler, and G. H. McCracken, Jr. 1989. Correlation Invest. 41:311-319.
of interleukin-1l3 and cachectin concentrations in cerebrospinal 105. Pfister, H. W., U. Koedel, R. L. Haberl, U. Dinargl, W. Feiden,
fluid and outcome from bacterial meningitis. J. Pediatr. 115: G. Ruckdeschel, and K. M. Einhaupl. 1990. Microvascular
208-213. changes during the early phase of experimental bacterial men-
88. Mustafa, M. M., J. Mertsola, 0. Ramilo, X. Saez-Llorens, R. C. ingitis. J. Cerebral Blood Flow Metab. 10:914-922.
Risser, and G. H. McCracken, Jr. 1989. Increased endotoxin 106. Pichichero, M. E., M. Loeb, P. Anderson, and D. H. Smith.
and interleukin-1lB concentrations in cerebrospinal fluid of 1982. Do pili play a role in pathogenicity of Haemophilus
infants with coliform meningitis and ventriculitis associated influenzae type b? Lancet ii:960-962.
with intraventricular gentamicin therapy. J. Infect. Dis. 160: 107. Pike, M. G., P. K. H. Wong, R. Bencivenga, 0. Flodmark,
891-895. D. A. Cabral, D. P. Speert, and K. Farrell. 1990. Electrophys-
89. Mustafa, M. M., 0. Ramilo, J. Mertsola, R. C. Risser, B. iologic studies, computed tomography, and neurologic out-
Beutler, E. J. Hansen, and G. H. McCracken, Jr. 1989. Mod- come in acute bacterial meningitis. J. Pediatr. 116:702-706.
ulation of inflammation and cachectin activity in relation to 108. Plaut, A. G. 1983. The IgAl proteases of pathogenic bacteria.
treatment of experimental Haemophilus influenzae type b Annu. Rev. Microbiol. 37:603-622.
meningitis. J. Infect. Dis. 160:818-825. 109. Pomeroy, S. L., S. J. Holmes, P. R. Dodge, and R. D. Feigin.
90. Mustafa, M. M., 0. Ramilo, K. D. Olsen, P. S. Franklin, E. J. 1990. Seizures and other neurologic sequelae of bacterial
Hansen, B. Beutler, and G. H. McCracken, Jr. 1989. Tumor meningitis in children. N. Engl. J. Med. 323:1651-1657.
necrosis factor in mediating experimental Haemophilus influ- 110. Quagliarello, V. J., W. J. Long, Jr., and W. M. Scheld. 1986.
enzae type b meningitis. J. Clin. Invest. 84:1253-1259. Morphologic alterations of the blood-brain barrier with exper-
91. Mustafa, M. M., 0. Ramilo, X. Saez-Llorens, K. D. Olsen, imental meningitis in the rat. Temporal sequence and role of
R. R. Magness, and G. H. McCracken, Jr. 1990. Cerebrospinal encapsulation. J. Clin. Invest. 77:1084-1095.
fluid prostaglandins, interleukin 1l, and tumor necrosis factor 111. Quagliarello, V. J., A. Ma, H. Stukenbrok, and G. E. Palade.
in bacterial meningitis. Clinical and laboratory correlations in 1991. Ultrastructural localization of albumin transport across
placebo-treated and dexamethasone-treated patients. Am. J. the cerebral microvasculature during experimental meningitis
Dis. Child. 144:883-887. in the rat. J. Exp. Med. 174:657-672.
92. Mustafa, M. M., 0. Ramilo, G. A. Syrogiannopoulos, K. D. 112. Quagliarello, V. J., B. Wispelwey, W. J. Long, Jr., and W. M.
Olsen, G. H. McCracken, Jr., and E. J. Hansen. 1989. Induc- Scheld. 1991. Recombinant human interleukin-1 induces men-
tion of meningeal inflammation by outer membrane vesicles of ingitis and blood-brain barrier injury in the rat. Characteriza-
Haemophilus influenzae type b. J. Infect. Dis. 159:917-922. tion and comparison with tumor necrosis factor. J. Clin.
93. Nadal, D., D. Leppert, K. Frei, P. Gallo, H. Lamche, and A. Invest. 87:1360-1366.
Fontana. 1989. Tumor necrosis factor-ax in infectious meningi- 113. Quinn, P. H., F. J. Crosson, Jr., J. A. Winkelstein, and E. R.
tis. Arch. Dis. Child. 64:1274-1279. Moxon. 1977. Activation of the alternative complement path-
94. Nawroth, P. P., I. Bank, D. Handley, J. Cassimeris, L. Chess, way by Haemophilus influenzae type b. Infect. Immun. 16:
and D. Stern. 1986. Tumor necrosis factor/cachectin interacts 400-402.
with endothelial cell receptors to induce release of interleukin 114. Raff, H. V., D. Devereux, W. Schuford, D. Abbott-Brown, and
VOL. 6, 1993 PATHOGENESIS OF BACTERIAL MENINGITIS 135

G. Maloney. 1988. Human monoclonal antibody with protec- nisolone on entry of ampicillin and gentamicin into cerebrospi-
tive activity for Escherichia coli Kl and Neisseria meningitidis nal fluid in experimental pneumococcal and Escherichia coli
group B infections. J. Infect. Dis. 157:118-126. meningitis. Antimicrob. Agents Chemother. 23:108-112.
115. Rahal, J. J., and M. S. Simberkoff. 1982. Host defense and 133. Scheld, W. M., R. G. Dacey, H. R. Winn, J. E. Welsh, J. A.
antimicrobial therapy in adult gram-negative bacillary menin- Jane, and M. A. Sande. 1980. Cerebrospinal fluid outflow
gitis. Ann. Intern. Med. 96:468-474. resistance in rabbits with experimental meningitis. Alterations
116. Raimondi, A. J., and C. DiRocco. 1979. The physiopathoge- with penicillin and methylprednisolone. J. Clin. Invest. 66:243-
netic basis for the angiographic diagnosis of bacterial infection 253.
of the brain and its coverings in children. I. Leptomeningitis. 134. Scheld, W. M., C. Giampaolo, J. Boyd, J. Savory, M. R. Wills,
Child's Brain 5:398-413. and M. A. Sande. 1982. Cerebrospinal fluid prognostic indices
117. Ramilo, O., X. Saez-Llorens, J. Mertsola, H. Jafari, K. D. in experimental pneumococcal meningitis. J. Lab. Clin. Med.
Olsen, E. J. Hansen, M. Yoshinaga, S. Ohkawara, H. Nariuchi, 100:218-229.
and G. H. McCracken, Jr. 1990. Tumor necrosis factor 135. Scheld, W. M., T. S. Park, R. G. Dacey, H. R. Winn, J. A.
a/cachectin and interleukin 11 initiate meningeal inflammation. Jane, and M. A. Sande. 1979. Clearance of bacteria from
J. Exp. Med. 172:497-507. cerebrospinal fluid to blood in experimental meningitis. Infect.
118. Riesenfeld-Orn, I., S. Wolpe, J. F. Garcia-Bustos, M. K. Immun. 24:102-105.
Hoffmann, and E. Tuomanen. 1989. Production of interleukin-1 136. Schlech, W. F., III, J. I. Ward, J. D. Band, A. Hightower,
but not tumor necrosis factor by human monocytes stimulated D. W. Fraser, and C. V. Broome. 1985. Bacterial meningitis in
with pneumococcal cell surface components. Infect. Immun. the United States, 1978 through 1981. The national bacterial
57:1890-1893. meningitis surveillance study. JAMA 253:1749-1754.
119. Robbins, J. B., G. H. McCracken, Jr., E. L. Gotschich, F. 137. Schleimer, R. P., and B. K. Rutledge. 1986. Cultured human
Orskov, I. Orskov, and L. A. Hanson. 1974. Eschenchia coli vascular endothelial cells acquire adhesiveness for neutrophils
Kl capsular polysaccharide associated with neonatal meningi- after stimulation with interleukin 1, endotoxin, and tumor-
tis. N. Engl. J. Med. 290:1216-1220. promoting phorbol diesters. J. Immunol. 136:649-654.
120. Roberts, M., T. L. Stull, and A. L. Smith. 1981. Comparative 138. Schneerson, R., and J. B. Robbins. 1975. Induction of serum
virulence of Haemophilus influenzae with a type b or type d Haemophilus influenzae type b capsular antibodies in adult
capsule. Infect. Immun. 32:518-524. volunteers fed cross-reacting Escherichia coli 075:K100:H5.
121. Rodriguez, A. F., S. L. Kaplan, E. P. Hawkins, and E. 0. N. Engl. J. Med. 292:1093-1096.
Mason, Jr. 1991. Hematogenous pneumococcal meningitis in 139. Simberkoff, M. S., H. N. Moldover, and J. J. Rahal, Jr. 1980.
the infant rat: description of a model. J. Infect. Dis. 164:1207- Absence of detectable bactericidal and opsonic activities in
1209. normal and infected human cerebrospinal fluids. A regional
122. Ross, S. C., and P. Densen. 1984. Complement deficiency states host defense deficiency. J. Lab. Clin. Med. 95:362-372.
and infection: epidemiology, pathogenesis and consequences 140. Smith, A. L. 1987. Pathogenesis of Haemophilus influenzae
of neisserial and other infections in an immune deficiency. meningitis. Pediatr. Infect. Dis. J. 6:783-786.
Medicine (Baltimore) 63:243-273. 141. Smith, A. L., R. S. Daum, D. Scheifele, V. Syriopolou, D. R.
123. Rusconi, F., F. Parizzi, L. Garlaschi, B. M. Assael, M. Sironi, Averill, M. C. Roberts, and T. L. Stull. 1982. Pathogenesis of
P. Ghezzi, A. Mantovani,. and the Collaborative Study on Haemophilus influenzae meningitis, p. 89-109. In S. H. Sell
Meningitis. 1991. Interleukin 6 activity in infants and children and P. F. Wright (ed.), Haemophilus influenzae: epidemiology,
with bacterial meningitis. Pediatr. Infect. Dis. J. 10:117-121. immunology, and prevention of disease. Elsevier Science
124. Saez-Llorens, X., H. S. Jafari, C. Severien, F. Parras, K. D. Publishing Co., New York.
Olsen, E. J. Hansen, I. I. Singer, and G. H. McCracken, Jr. 142. Smith, H., B. Bannister, and M. J. O'Shea. 1973. Cerebrospi-
1991. Enhanced attenuation of meningeal inflammation and nal-fluid immunoglobulins in meningitis. Lancet i:591-593.
brain edema by concomitant administration of anti-CD18 mono- 143. Stephens, D. S., and M. M. Farley. 1991. Pathogenic events
clonal antibodies and dexamethasone in experimental Haemo- during infection of the human nasopharynx with Neisseria
philus meningitis. J. Clin. Invest. 88:2003-2011. meningitidis and Haemophilus influenzae. Rev. Infect. Dis.
125. Saez-Llorens, X., 0. Ramilo, M. M. Mustafa, J. Mertsola, C. de 13:22-33.
Alba, E. Hansen, and G. H. McCracken, Jr. 1990. Pentoxifyl- 144. Stephens, D. S., L. H. Hoffman, and Z. A. McGee. 1983.
line modulates meningeal inflammation in experimental bacte- Interaction of Neisseria meningitidis with human nasopharyn-
rial meningitis. Antimicrob. Agents Chemother. 34:837-843. geal mucosa: attachment and entry into columnar epithelial
126. Saez-Llorens, X., 0. Ramilo, M. M. Mustafa, J. Mertsola, and cells. J. Infect. Dis. 148:369-376.
G. H. McCracken, Jr. 1990. Molecular pathophysiology of 145. Stephens, D. S., and Z. A. McGee. 1981. Attachment of
bacterial meningitis: current concepts and therapeutic implica- Neisseria meningitidis to human mucosal surfaces: influence of
tions. J. Pediatr. 116:671-684. pili and type of receptor cell. J. Infect. Dis. 143:525-532.
127. Saukkonen, K., S. Sande, C. Cioffe, S. Wolfe, B. Sherry, A. 146. Stull, T. L., P. M. Mendelman, J. L. Haas, M. A. Schoenborn,
Cerami, and E. Tuomanen. 1990. The role of cytokines in the K. D. Mack, and A. L. Smith. 1984. Characterization of
generation of inflammation and tissue damage in experimental Haemophilus influenzae type b fimbriae. Infect. Immun. 46:
gram-positive meningitis. J. Exp. Med. 171:439-448. 787-796.
128. Saukkonen, K. M., B. Nowicki, and M. Leinonen. 1988. Role of 147. Sullivan, G. W., H. T. Carper, W. J. Novick, Jr., and G. L.
type 1 and S fimbriae in the pathogenesis of Escherichia coli Mandell. 1988. Inhibition of the inflammatory action of inter-
018:K1 bacteremia and meningitis in the infant rat. Infect. leukin-1 and tumor necrosis factor (alpha) on neutrophil func-
Immun. 56:892-897. tion by pentoxifylline. Infect. Immun. 56:1722-1729.
129. Schaad, U. B., S. Suter, A. Gianella-Bonadori, J. Pfenninger, R. 148. Swartz, M. N. 1984. Bacterial meningitis. More involved than
Auckenthaler, 0. Bernath, J. J. Chesaux, and J. Wedgwood. just the meninges. N. Engl. J. Med. 311:912-914.
1990. A comparison of ceftriaxone and cefuroxime for the 149. Syrogiannopoulos, G. A., E. J. Hansen, A. L. Erwin, R. S.
treatment of bacterial meningitis in children. N. Engl. J. Med. Munford, J. Rutledge, J. S. Reisch, and G. H. McCracken, Jr.
322:141-146. 1988. Haemophilus influenzae type b lipooligosaccharide in-
130. Scheifele, D. W., R. S. Daum, V. P. Syriopoulou, D. R. Averill, duces meningeal inflammation. J. Infect. Dis. 157:237-244.
and A. L. Smith. 1980. Haemophilus influenzae bacteremia and 150. Syrogiannopoulos, G. A., K. D. Olsen, J. S. Reisch, and G. H.
meningitis in infant primates. J. Lab. Clin. Med. 95:450-462. McCracken, Jr. 1987. Dexamethasone in the treatment of
131. Scheld, W. M. 1984. Bacterial meningitis in the patient at risk: experimental Haemophilus influenzae type b meningitis. J.
intrinsic risk factors and host defense mechanisms. Am. J. Infect. Dis. 155:213-219.
Med. 76:(Suppl. SA):193-207. 151. Takala, A. K., L. van Alphen, J. Eskola, J. Palmgren, P. Bol,
132. Scheld, W. M., and J. P. Brodeur. 1983. Effect of methylpred- and P. H. Makela. 1987. Haemophilus influenzae type b strains
136 TUNKEL AND SCHELD CLIN. MICROBIOL. REV.

of outer membrane subtypes 1 and lc cause different types of 169. Tuomanen, E., A. Tomasz, B. Hengstler, and 0. Zak. 1985. The
invasive disease. Lancet ii:647-650. relative role of bacterial cell wall and capsule in the induction
152. Tauber, M. G., U. Borschberg, and M. A. Sande. 1988. of inflammation in pneumococcal meningitis. J. Infect. Dis.
Influence of granulocytes on brain edema, intracranial pres- 151:535-540.
sure, and cerebrospinal fluid concentrations of lactate and 170. Tuomanen, E. I., K. Saukkonen, S. Sande, C. Cioffe, and S. D.
protein in experimental meningitis. J. Infect. Dis. 157:456-464. Wright. 1989. Reduction of inflammation, tissue damage, and
153. Tauber, M. G., M. Burroughs, U. M. Niemoller, H. Kuster, U. mortality in bacterial meningitis in rabbits treated with mono-
Borschberg, and E. Tuomanen. 1991. Differences of pathophys- clonal antibodies against adhesion-promoting receptors of leu-
iology in experimental meningitis caused by three strains of kocytes. J. Exp. Med. 170:959-968.
Streptococcus pneumoniae. J. Infect. Dis. 163:806-811. 171. Tureen, J. H., R. J. Dworkin, S. L. Kennedy, M. Sachdeva, and
154. Tauber, M. G., H. Khayam-Bashi, and M. A. Sande. 1985. M. A. Sande. 1990. Loss of cerebrovascular autoregulation in
Effects of ampicillin and corticosteroids on brain water con- experimental meningitis in rabbits. J. Clin. Invest. 85:577-581.
tent, cerebrospinal fluid pressure, and cerebrospinal fluid lac- 172. Tureen, J. H., M. G. Tauber, and M. A. Sande. 1991. Effect of
tate levels in experimental pneumococcal meningitis. J. Infect. indomethacin on the pathophysiology of experimental menin-
Dis. 151:528-534. gitis in rabbits. J. Infect. Dis. 163:647-649.
155. Tauber, M. G., A. M. Shibl, C. J. Hackbarth, J. W. Larrick, 173. Tureen, J. H., M. G. Tauber, and M. A. Sande. 1992. Effect of
and M. A. Sande. 1987. Antibiotic therapy, endotoxin concen- hydration status on cerebral blood flow and cerebrospinal fluid
tration in cerebrospinal fluid, and brain edema in experimental lactic acidosis in rabbits with experimental meningitis. J. Clin.
Escherichia coli meningitis in rabbits. J. Infect. Dis. 156:456- Invest. 89:947-953.
462. 174. Waage, A., P. Brandtzaeg, A. Halstensen, P. Kierulf, and T.
156. Taylor, H. G., E. L. Mills, A. Ciampi, R. du Berger, G. V. Espevik. 1989. The complex pattern of cytokines in serum from
Watters, R. Gold, N. MacDonald, and R. H. Michaels. 1990. patients with meningococcal septic shock. Association be-
The sequelae of Haemophilus influenzae meningitis in school- tween interleukin 6, interleukin 1, and fatal outcome. J. Exp.
age children. N. Engl. J. Med. 323:1657-1663. Med. 169:333-338.
157. Tofte, R. W., P. K. Peterson, Y. Kim, and P. G. Quie. 1979. 175. Waage, A., A. Halstensen, R. Shalaby, P. Brandtzaeg, P.
Opsonic activity of normal human cerebrospinal fluid for Kierulf, and T. Espevik. 1989. Local production of tumor
selected bacterial species. Infect. Immun. 26:1093-1098. necrosis factor a, interleukin 1, and interleukin 6 in meningo-
158. Tonnesen, M. G., L. A. Smedley, and P. M. Henson. 1984. coccal meningitis. Relation to the inflammatory response. J.
Neutrophil-endothelial cell interaction. Modulation of neutro- Exp. Med. 170:1859-1867.
phil adhesiveness induced by complement fragments C5a and 176. Waggener, J. D. 1974. The pathophysiology of bacterial men-
CSa des arg and formyl-methionyl-leucyl-phenylalanine in ingitis and cerebral abscesses: an anatomical interpretation.
vitro. J. Clin. Invest. 74:1581-1592. Adv. Neurol. 6:1-17.
159. Tracey, K. J., S. F. Luwry, and A. Cerami. 1988. Cachectin: a 177. Weber, A., K. Harris, S. Lohrke, L. Forney, and A. L. Smith.
hormone that triggers acute shock and chronic cachexia. J. 1991. Inability to express fimbriae results in impaired ability of
Infect. Dis. 157:413-420. Haemophilus influenzae b to colonize the nasopharynx. Infect.
160. Tunkel, A. R., M. T. Kinter, S. W. Rosser, M. A. Aubin, E. J. Immun. 59:4724-4728.
Hansen, and W. M. Scheld. 1990. Haemophilus infiuenzae type 178. Wenger, J. D., A. W. Hightower, R. R. Facklam, S. Gaventa,
b lipopolysaccharide (LPS)-induced synthesis of prostaglandin
E2 (PGE2) and interleukin-6 (IL-6) by cerebral microvascular C. V. Broome, and the Bacterial Meningitis Study Group. 1990.
endothelial cells (MECs), abstr. 74, p. 99. Program Abstr. 30th Bacterial meningitis in the United States, 1986: report of a
Intersci. Conf. Antimicrob. Agents Chemother. 1990. Ameri- multistate surveillance study. J. Infect. Dis. 162:1316-1323.
can Society for Microbiology, Washington, D.C. 179. Whittle, H. C., and B. M. Greenwood. 1977. Cerebrospinal
161. Tunkel, A. R., S. W. Rosser, E. J. Hansen, and W. M. Scheld. fluid immunoglobulins and complement in meningococcal men-
1990. Haemophilus influenzae type b lipopolysaccharide ingitis. J. Clin. Pathol. 30:720-722.
(LPS)-induced formation of cAMP and cGMP in cerebral 180. Williams, A. E., and W. F. Blakemore. 1990. Pathogenesis of
microvascular endothelial cells (MECs), abstr. 73, p. 99. meningitis caused by Streptococcus suis type 2. J. Infect. Dis.
Program Abstr. 30th Intersci. Conf. Antimicrob. Agents 162:474-481.
Chemother. 1990. American Society for Microbiology, Wash- 181. Winkelstein, J. A., and A. Tomasz. 1977. Activation of the
ington, D.C. alternative pathway by pneumococcal cell walls. J. Immunol.
162. Tunkel, A. R., S. W. Rosser, E. J. Hansen, and W. M. Scheld. 118:451-454.
1991. Blood-brain barrier alterations in bacterial meningitis: 182. Wispelwey, B., E. J. Hansen, and W. M. Scheld. 1989. Haemo-
development of an in vitro model and observations on the philus influenzae outer membrane vesicle-induced blood-brain
effects of lipopolysaccharide. In Vitro Cell. Dev. Biol. 27A: barrier permeability during experimental meningitis. Infect.
113-120. Immun. 57:2559-2562.
163. Tunkel, A. R., and W. M. Scheld. 1989. Applications of therapy 183. Wispelwey, B., A. J. Lesse, E. J. Hansen, and W. M. Scheld.
in animal models to bacterial infection in human disease. 1988. Haemophilus influenzae lipopolysaccharide-induced
Infect. Dis. Clin. North Am. 3:441-459. blood brain barrier permeability during experimental meningi-
164. Tunkel, A. R., and W. M. Scheld. 1989. Alterations in the tis in the rat. J. Clin. Invest. 82:1339-1346.
blood-brain barrier in bacterial meningitis: in vivo and in vitro 184. Wyler, D. J., S. I. Wasserman, and A. W. Karchmer. 1979.
models. Pediatr. Infect. Dis. J. 8:911-913. Substances which modulate leukocyte migration are present in
165. Tunkel, A. R., B. Wispelwey, and W. M. Scheld. 1990. Bacte- CSF during meningitis. Ann. Neurol. 5:322-326.
rial meningitis: recent advances in pathophysiology and treat- 185. Yamashima, T., K. Kashihara, K. Ikeda, T. Kubota, and S.
ment. Ann. Intern. Med. 112:610-623. Yamamoto. 1985. Three phases of cerebral arteriography in
166. Tunkel, A. R., B. Wispelwey, and W. M. Scheld. 1990. Patho- meningitis: vasospasm and vasodilatation followed by organic
genesis and pathophysiology of meningitis. Infect. Dis. Clin. stenosis. Neurosurgery 16:546-553.
North Am. 4:555-581. 186. Zwahlen, A., U. E. Nydegger, P. Vaudaux, P. H. Lambert, and
167. Tuomanen, E., B. Hengstler, R. Rich, M. A. Bray, 0. Zak, and F. A. Waldvogel. 1982. Complement-mediated opsonic activity
A. Tomasz. 1987. Nonsteroidal anti-inflammatory agents in the in normal and infected human cerebrospinal fluid: early re-
therapy for experimental pneumococcal meningitis. J. Infect. sponse during bacterial meningitis. J. Infect. Dis. 145:635-646.
Dis. 155:985-990. 187. Zwahlen, A., J. A. Winkelstein, and E. R. Moxon. 1983.
168. Tuomanen, E., H. Liu, B. Hengstler, 0. Zak, and A. Tomasz. Surface determinants of Haemophilus influenzae pathogenic-
1985. The induction of meningeal inflammation by components ity: comparative virulence of capsular transformants in normal
of the pneumococcal cell wall. J. Infect. Dis. 151:859-868. and complement-depleted rats. J. Infect. Dis. 148:385-394.

Das könnte Ihnen auch gefallen