You are on page 1of 10

See discussions, stats, and author profiles for this publication at: https://www.researchgate.

net/publication/255955616

Pathophysiology of neonatal acute bacterial meningitis

Article  in  Journal of Medical Microbiology · August 2013


DOI: 10.1099/jmm.0.059840-0 · Source: PubMed

CITATIONS READS

30 432

6 authors, including:

Tatiana Barichello Glauco Danielle Fagundes


Universidade do Extremo Sul Catarinense (UNESC) Universidade do Extremo Sul Catarinense (UNESC)
154 PUBLICATIONS   2,317 CITATIONS    5 PUBLICATIONS   69 CITATIONS   

SEE PROFILE SEE PROFILE

Jaqueline S Generoso Samuel Galvão Elias


Universidade do Extremo Sul Catarinense (UNESC) University of Brasília
52 PUBLICATIONS   562 CITATIONS    8 PUBLICATIONS   107 CITATIONS   

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

MIND.Funga - Monitoring and Inventorying Neotropical Diversity of Fungi View project

All content following this page was uploaded by Samuel Galvão Elias on 10 December 2014.

The user has requested enhancement of the downloaded file.


Journal of Medical Microbiology (2013), 62, 1781–1789 DOI 10.1099/jmm.0.059840-0

Review Pathophysiology of neonatal acute bacterial


meningitis
Tatiana Barichello,1,2,3,4 Glauco D. Fagundes,1,2,3
Jaqueline S. Generoso,1,2,3 Samuel Galvão Elias,1,2,3 Lutiana R. Simões1,2,3
and Antonio Lucio Teixeira5
Correspondence 1
Laboratório de Microbiologia Experimental e Instituto Nacional de Ciência e Tecnologia
Tatiana Barichello Translacional em Medicina, Programa de Pós-Graduação em Ciências da Saúde,
tba@unesc.net Universidade do Extremo Sul Catarinense, Criciúma, SC, Brazil
2
Instituto Nacional de Ciência e Tecnologia Translacional em Medicina (INCT-TM),
Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde,
Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
3
Núcleo de Excelência em Neurociências Aplicadas de Santa Catarina (NENASC),
Programa de Pós-Graduação em Ciências da Saúde, Unidade Acadêmica de Ciências da Saúde,
Universidade do Extremo Sul Catarinense, 88806-000 Criciúma, SC, Brazil
4
Center for Experimental Models in Psychiatry, Department of Psychiatry and Behavioral Sciences,
Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
5
Laboratório de Imunofarmacologia, Departamento de Bioquı́mica e Imunologia, Instituto de
Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, MG, Brazil

Neonatal meningitis is a severe acute infectious disease of the central nervous system and an
important cause of morbidity and mortality worldwide. The inflammatory reaction involves the
meninges, the subarachnoid space and the brain parenchymal vessels and contributes to neuronal
injury. Neonatal meningitis leads to deafness, blindness, cerebral palsy, seizures, hydrocephalus or
cognitive impairment in approximately 25–50 % of survivors. Bacterial pathogens can reach the
blood–brain barrier and be recognized by antigen-presenting cells through the binding of Toll-like
receptors. They induce the activation of NFkB or mitogen-activated protein kinase pathways and
subsequently upregulate leukocyte populations and express numerous proteins involved in
inflammation and the immune response. Many brain cells can produce cytokines, chemokines and
other pro-inflammatory molecules in response to bacterial stimuli, and polymorphonuclear
leukocytes are attracted, activated and released in large amounts of superoxide anion and nitric
oxide, leading to peroxynitrite formation and generating oxidative stress. This cascade leads to lipid
peroxidation, mitochondrial damage and breakdown of the blood–brain barrier, thus contributing to
cell injury during neonatal meningitis. This review summarizes information on the pathophysiology
and adjuvant treatment of acute bacterial meningitis in neonates.

Introduction mortality worldwide, principally in neonates and children


(Grandgirard & Leib, 2010), with the impact varying
Meningitis is the most frequent and serious infection of
according to the geographical location of the patient and
the central nervous system (CNS) and affects the pia
the causative organism (Kim, 2003).
matter, arachnoids and subarachnoid space (van de Beek
et al., 2004). It is an important cause of morbidity and Neonatal meningitis occurs beyond 28 days of life (Nizet &
Klein, 2011). Several studies conceptualize the neonatal
Abbreviations: AIM2, absent in melanoma 2; BBB, blood–brain barrier; period as up to 30- or 90-days-old (Furyk et al., 2011).
BDNF, brain-derived neurotrophic factor; CNS, central nervous system; Bacterial meningitis is a severe infectious disease, with
CSF, cerebrospinal fluid; ICAM, intercellular adhesion molecule; IRAK, mortality rates varying between 10 % and 15 %
IL-1 receptor-associated kinase; LFA-1, lymphocyte function-associated
antigen 1; MyD88, myeloid differentiation factor 88; NLR, nucleotide-
(Gaschignard et al., 2011), and the incidence is described
binding domain leucine-rich repeat; NLRP3, NLR family, pyrin-domain- as 0.25–6.1 per 1000 live births (developed countries 0.3;
containing 3; NOD2, nucleotide-binding oligomerization domain-2; TLR, Asia 0.48–2.4; Africa and South Africa 0.81–6.1) (Heath &
Toll-like receptor. Okike, 2010). In a systematic analysis of mortality estimates

059840 G 2013 SGM Printed in Great Britain 1781


T. Barichello and others

in 2010 with time trends since 2000, 7.6 million deaths were continues through the postnatal period, and it may be a
reported to occur in children younger than 5 years old in time of increased permeability that renders the developing
2010, with 64.0 % (4.879 million) of these attributable to brain more vulnerable (Zeng et al., 2011). Other authors
infectious causes and 40.3 % (3.072 million) occurring in have affirmed that adult mechanisms and functionally
neonates. Of these neonatal deaths, 5.2 % (0.393 million) effective tight junctions are present in the embryonic brain
were a result of sepsis or meningitis (Liu et al., 2012). and that some transporters are more active during
The major pathogens are Streptococcus agalactiae and development than in the adult brain (Ek et al., 2012).
Escherichia coli K1, which cause at least two-thirds of all Developing cerebral vessels appear to be more fragile than
deaths from neonatal meningitis in Europe (Heath et al., in adults and more susceptible to drugs, toxins and
2011; Holt et al., 2001). Other Gram-negative bacteria pathological conditions, thus contributing to cerebral
implicated include Klebsiella, Enterobacter, Citrobacter and damage and later neurological disorders. After birth, the
Serratia species. (Heath et al., 2011). The remaining causes loss of defence by efflux transporters in the placenta can
include Streptococcus pneumoniae (6 % of cases) and Listeria contribute to cerebral damage and later neurological
monocytogenes (5 % of cases) (Heath & Okike, 2010). disorders in the neonatal brain because it is more
The early- and late-onset patterns of the disease have been defenceless than the fetal brain (Saunders et al., 2012).
associated with systemic bacterial infections during the first Because of these neonatal particularities, bacterial men-
month of life. Many studies define early-onset bacterial ingitis can cause acute complications involving the brain
infection as an infection that occurs in the first 72 h of life; parenchymal vessels (vasculitis), ventriculitis, systemic
others define it as an infection that occurs in the first 5 or complications (including pneumonia) and septic shock,
6 days of life (Remington, 2011). Group B streptococcal which contribute to an unfavourable outcome (Sellner
infection in neonates and young infants is classified by et al., 2010). Long-term neurological sequelae, including
age at onset (Kim, 2003). Early-onset infection with deafness, blindness, cerebral palsy, seizures, hydrocephalus
Streptococcus agalactiae generally presents at or within or cognitive impairment, present in approximately 25–
24 h of birth (Heath & Okike, 2010), but can occur 50 % of survivors (de Louvois et al., 2005).
through day 6 of life (Puopolo & Baker, 2012). Late-onset
disease has been variably defined for epidemiological
purposes as occurring after 72 h to 6 days of life; late- CNS bacterial invasions
onset infection can manifest after the first week to months Initial bacterial entry into the CNS
after birth as sepsis and meningitis or other focal infections Early-onset infections are acquired vertically through
(Remington, 2011). Late-onset infection with Streptococcus exposure to the micro-organism via the mother’s genital
agalactiae usually occurs at 4–5 weeks of age (range: 7– tract (Remington, 2011). Neonatal infection occurs prim-
89 days) (Puopolo & Baker, 2012). Early-onset meningitis arily when micro-organisms ascend from the vagina to the
is more likely to be caused by Streptococcus agalactiae, E. amniotic fluid after the onset of labour or rupture of the
coli and L. monocytogenes, while late-onset meningitis may membranes, although Streptococcus agalactiae can invade
be caused by Streptococcus agalactiae, E. coli, L. mono- through intact membranes (Verani et al., 2010). Strepto-
cytogenes, Gram-negative organisms, Staphylococcus spp.
coccus agalactiae can be aspirated into the fetal lungs and
(Heath et al., 2003) and Streptococcus pneumoniae (Kim,
might be transferred haematogenously into the CNS (Pong
2008).
& Bradley, 1999). The bacteria can cross the BBB by different
Newborns are especially vulnerable to infection. The mechanisms: via transcellular or paracellular traversal and in
cellular and humoral immunities are immature, including infected phagocytes. Transcellular traversal occurs when the
the phagocytic function (Filias et al., 2011; Pong & Bradley, micro-organism penetrates the cells without any evidence in
1999). A full-term newborn has a distinct innate immune the cells or intracellular tight-junction disruption (Kim,
system that is biased towards T-helper type 2/T-helper type 2008). Streptococcus pneumoniae, Streptococcus agalactiae
17-polarizing and anti-inflammatory cytokine production, and E. coli can all cross the BBB via this mechanism (Kim,
with relative impairment in T-helper type 1-polarizing 2003). To cross the BBB, Streptococcus pneumoniae must
and cytokine production. The deficient expression of interact with cell wall phosphorylcholine and platelet-
complement and of antimicrobial proteins and peptides activating factor receptor (Kim, 2008). Streptococcus aga-
likely contributes to a newborn’s susceptibility to pyogenic lactiae crosses the BBB with the aid of the lipoprotein
bacteria (Cuenca et al., 2013). The phagocytic ability of the laminin-binding protein (Tenenbaum et al., 2007). L.
neutrophils and monocytes of 42 neonates has been monocytogenes crosses the BBB by microbial penetration
evaluated by the intake of E. coli by phagocytes (Filias using transmigration within infected phagocytes; this
et al., 2011). The phagocytic ability was impaired at birth in mechanism is known as a Trojan horse (Kim, 2008).
pre- and full-term neonates compared with adults. This
defect was transient, with the phagocytic ability in neonates
reaching the same level as that of adults 3 days after birth. Bacterial recognition and immune response
It is widely believed that the development of the blood– Micro-organisms can replicate within the subarachnoid
brain barrier (BBB) proceeds from late gestation and space concomitantly with the release of bacterial products,

1782 Journal of Medical Microbiology 62


Pathophysiology of neonatal acute bacterial meningitis

such as peptidoglycan and cell wall fragments, which are (MyD88) (Koedel, 2009). MyD88 is associated with IL-1
highly immunogenic and can lead to an increased and IL-1 receptor-associated kinase-4 (IRAK4), which is a
inflammatory response in the host (Sellner et al., 2010). serine/threonine kinase that plays an essential role in signal
These compounds are recognized by antigen-presenting transduction by Toll/IL-1 receptors (Wang et al., 2006).
cells through binding to pattern-recognition receptors such Subsequently, IRAK interacts with TNF and the TNF
as Toll-like receptors (TLRs), a nucleotide-binding domain receptor-associated factor family and provides a link to
leucine-rich repeat (NLR) region-containing family of NFkB-inducing kinase, resulting in the release and nuclear
proteins (Mook-Kanamori et al., 2011). translocation of NFkB (Hanke & Kielian, 2011). The NFkB
family comprises a closely associated family of transcrip-
Nucleotide-binding oligomerization domain-2 (NOD2) is
tion factors, which play a key role in the expression of
activated by cell wall peptidoglycan (Liu et al., 2010; Opitz
genes that have been implicated in the development of
et al., 2004). NOD2 has a critical role in the establishment
accessory cell and leukocyte populations and in the
of the lethal inflammation associated with streptococcal
meningitis through activation of the transcription factor expression of many proteins involved in inflammation
NFkB and production of inflammatory cytokines (Mook- and the immune response (Tato & Hunter, 2002). NFkB is
Kanamori et al., 2011), as have been demonstrated in a transcriptional activator of various genes involved in the
murine microglia and astrocyte cells (Liu et al., 2010). The pathogenesis of meningitis, such as TNF-a, IL-1b, indu-
NLR family, pyrin-domain-containing 3 (NLRP3) and the cible nitric oxide synthase and intercellular adhesion
absent in melanoma 2 (AIM2) inflammasomes are molecules (Fig. 2) (Kastenbauer et al., 2004; Koedel et al.,
activated by exotoxin pneumolysin and DNA bacteria in 2000).
pneumococcal infection, respectively, which mediates the
cleavage of pro-IL-1b into mature IL-1b (Koppe et al., Neuronal inflammation
2012). Streptococcus agalactiae activates NLRP3 through the
Initial immune response
expression of b-haemolysin, an important virulence factor
(Costa et al., 2012). NLRP3 has been implicated in Knowledge of the pathophysiology of bacterial meningitis
responses to Staphylococcus aureus, L. monocytogenes, in neonates is predominantly based on observations of
Klebsiella pneumoniae and E. coli (Davis et al., 2011). human patients and studies with experimental animal
models (Grandgirard & Leib, 2010). A relative comparison
Eleven TLR family members have been described in of the ages and stages of human versus rat development has
humans. These are separated into two broad categories: shown that the neonatal period for rats is from birth until
members of one group are expressed at the cell surface for day 10 of life (Andersen, 2003).
extracellular ligand recognition, while members of the
other group are localized in the endosomal compartment Many brain cells, such as astrocytes, glial cells, endothelial
to recognize pathogen nucleic acids (Hanke & Kielian, cells, ependymal cells and resident macrophages, can
2011). TLRs 1–9 are expressed in microglia cells, whereas produce cytokines and pro-inflammatory molecules in
astrocytes express TLRs 2, 3 and 9. Neurons express TLRs response to bacterial replication and its components
3, 7, 8 and 9. Oligodendrocytes express TLRs 2 and 3. (Kronfol & Remick, 2000). In a study with 54 human
Streptococcus pneumoniae bacterial compounds, including newborns (30 with meningitis and 24 controls), increased
peptidoglycans and lipoteichoic acids, are recognized in the levels of TNF-a, IL-1b and IL-6 were detected in the
CNS by TLR2 (Mitchell et al., 2010). The exotoxin cerebrospinal fluid (CSF) of all of the newborns with
pneumolysin is recognized by TLR4 (Malley et al., 2003) meningitis (Krebs et al., 2005). Elevated levels of pro-
and the bacterial DNA is recognized by TLR9, which is an inflammatory mediators in the CSF are normally found in
intracellular pattern-recognition receptor activated by patients with bacterial meningitis (Lahrtz et al., 1998; van
CpG (cytosine-phosphate-guanine) (Hemmi et al., 2000). Furth et al., 1996). In animal models, administration of
E. coli interacts through toxin lipopolysaccharide with TNF-a into the CSF and intravenously has been reported to
TLR4 (Rivest, 2003). In vitro, L. monocytogenes cell wall cause changes that are characteristic of bacterial meningitis
components such as lipoteichoic acids are recognized by and lead to BBB breakdown (Rosenberg et al., 1995; Tsao
TLR2 with the help of CD14 and TLR6 (Flo et al., 2000; et al., 2002). TNF-a is a marker of an acute inflammatory
Janot et al., 2008; Seki et al., 2002). The protein flagellin response in children with bacterial meningitis (Mukai et al.,
interacts through TLR5 (Hayashi et al., 2001). Streptococcus 2006) and is essential for an adequate host immune
agalactiae interacts with TLR2 through lipoteichoic acid response (Aas et al., 2005). IL-1b is an important cytokine
and through the engagement of TLR7 and/or TLR8 by that has potent stimulatory effects on white blood cells and
microbial RNA (Mook-Kanamori et al., 2011) (Table 1). promotes the adhesion of neutrophils and monocytes in
endothelial cells (Østergaard et al., 2004). In a study of 21
TLRs are essential to trigger the immune response during
patients with bacterial meningitis, endothelial-derived
meningitis, signalling the production of key inflammatory
adhesion molecules were associated with the extent of
mediators (Fig. 1) (Hanke & Kielian, 2011).
CSF pleocytosis and with concentrations of the pro-
The majority of TLRs utilize a common intracellular inflammatory cytokines IL-1b and TNF-a in CSF
adaptor protein known as myeloid differentiation factor 88 (Fassbender et al., 1997).

http://jmm.sgmjournals.org 1783
T. Barichello and others

Table 1. Bacterial components and TLRs

Micro-organism TLR agonist TLR Expression of TLR family members in CNS cells

Streptococcus pneumoniae Peptidoglycan 2 Microglia, astrocytes, oligodendrocytes


Lipoteichoic acid 2 Microglia, astrocytes, oligodendrocytes
Pneumolysin 4 Microglia
Bacterial CpG DNA 9 Microglia, astrocytes, neurons
Triacylated lipoprotein 1/2 Microglia
Diacylated lipoprotein 2/6 Microglia
E. coli Lipopolysaccharide 4 Microglia
Triacylated lipoprotein 1/2 Microglia
Diacylated lipoprotein 2/6 Microglia
L. monocytogenes Lipoteichoic acid 2 Microglia, astrocytes, oligodendrocytes
Flagellin 5 Microglia
Triacylated lipoprotein 1/2 Microglia
Diacylated lipoprotein 2/6 Microglia
Streptococcus agalactiae Lipoteichoic acid 2 Microglia, astrocytes, oligodendrocytes
Microbial RNA 7/8 Microglia, neurons
Triacylated lipoprotein 1/2 Microglia
Diacylated lipoprotein 2/6 Microglia

Streptococcus Listeria Streptococcus


Escherichia coli
pneumoniae monocytogenes agalactiae

PRC Lmb
PAF CD46

CpG
LTA DNA FLY RNA
PLY LPS RNA LTA PLY LTA
β-HLY PLY
PGL β-HLY
TLR4 TLR2
TLR2 TLR4 TLR2 TLR5
NO CD14
D TLR6 NLRP3 TLR7
NLRP3

TLR8
TLR9

Fig. 1. Pathophysiology of acute bacterial meningitis. The micro-organism adheres to endothelial cells and may breach the BBB
and be recognized by antigen-presenting cells through the binding of TLRs. TLRs induce the activation of an inflammatory
cascade and express numerous proteins involved in inflammation and the immune response. FLY, flagellin; b-HLY, b-
haemolysin; Lmb, lipoprotein laminin; LPS, lipopolysaccharide; LTA, lipoteichoic acid; PGL, peptidoglycan; PAF, platelet-
activating factor receptor; PLY, pneumolysin; PRC, phosphorylcholine.

1784 Journal of Medical Microbiology 62


Pathophysiology of neonatal acute bacterial meningitis

LPS PGL inflammatory response and decreases vascular permeability


RNA LTA Cytokines
in bacterial meningitis (Paul et al., 2003). In animal model
FLY PLY Chemokines studies, we determined that TNF-a, IL-1b, cytokine-induced
neutrophil chemoattractant 1 (CINC-1) and IL-6 levels were
TLRs
CpG increased prior to BBB breakdown, and that this breakdown
PLY DNA PGL occurred in the hippocampus and in the cortex after the
induction of neonatal pneumococcal meningitis (Barichello
et al., 2012a) and Streptococcus agalactiae meningitis
(Barichello et al., 2011). We suggest that these cytokines
MyD88 are produced at the site of inflammation and play an
important role in neutrophil infiltration.
IRAK4

Later neutrophil-mediated response


The production of cytokines leads to the attraction and
AIM2
activation of polymorphonuclear leukocytes and the
NOD
NLRP3 production of high amounts of reactive oxygen species
TRAF6 (Kastenbauer et al., 2002). Sialyl-LewisX on leukocytes
binds to P- and E-selectin on endothelial cells. This
connection becomes stronger when CXCL8 binds to its
Iκ B
specific receptor on neutrophils, triggering the production
Caspase-1 of integrins lymphocyte function-associated antigen 1
NFκ B (LFA-1) and CX3 (mac-1). Inflammatory cytokines such
as TNF-a are necessary to induce the expression of
intercellular adhesion molecule (ICAM)-1 and -2. The
link between endothelial cells and ICAM-1 allows the
passage of neutrophils in the direction of the gradient of
chemoattractant substances (Fig. 3) (Carlos & Harlan,
1994; Hanna & Etzioni, 2012). This leads to oxidative
stress, which in turn leads to cytokine and chemokine
activation, enhanced neutrophil activation, lipid pero-
xidation, DNA single-strand breaks, mitochondrial
impairment, tyrosine nitration, matrix metalloproteinase
activation and prostaglandin production (Klein et al.,
2006). A study with neonatal rats found that protein
carbonyls and lipid peroxidation were increased in the
hippocampus and cortex during the first hours after
pneumococcal and Streptococcus agalactiae meningitis
induction (Barichello et al., 2011, 2012a).
Fig. 2. Intracellular pathophysiology of bacterial meningitis. TLRs
utilize the common intercellular adaptor MyD88, which is Reactive oxygen species and calcium can result in
associated with IRAK4. IRAK4 interacts with TNF receptor- mitochondrial dysfunction, which leads to the release of
associated factor (TRAF), thus providing a link to NFkB and the apoptosis-inducing factor into the cytosol; apoptosis-
subsequent release of cytokines and chemokines. Alternatively, the inducing factor has been reported to be responsible for
release of caspase-1 for its transcription in the nucleus may be executing the caspase-independent pathway. Polymorpho-
mediated by the stimulation of NLRP3 and AIM2 by pneumolysin nuclear leukocytes can also induce the release of cyto-
(PLY) and bacterial DNA, respectively. FLY, flagellin; IkB, inhibitor chrome c from the mitochondria into the cytosol, resulting
of kB; LPS, lipopolysaccharide; LTA, lipoteichoic acid; PGL, in caspase-3 cleavage in a mouse model of pneumococcal
peptidoglycan; PLY, pneumolysin. meningitis (Mitchell et al., 2004). In children with bacterial
meningitis, acrolein-lysine (a marker of lipid peroxidation)
and nitrite (a marker of nitric oxide production) have been
IL-6 has predominantly pro-inflammatory effects such as reported to be several times higher in during the early
fever, leukocytosis and the potent induction of acute-phase phase of bacterial meningitis compared with children
proteins (Gruol & Nelson, 1997), including the synthesis without meningitis or with aseptic meningitis (Tsukahara
and secretion of C-reactive protein, serum amyloid A, et al., 2002). A positive correlation was found between the
fibrinogen, ar-antitrypsin, q-antichymotrypsin and hapto- nitric oxide index and lipid peroxide with white blood cells
globin (Castell et al., 1989). IL-6 acts as an anti- in the CSF of children with bacterial meningitis (Hamed
inflammatory cytokine, and a lack of IL-6 enhances the et al., 2009). These free radicals are highly reactive and may

http://jmm.sgmjournals.org 1785
T. Barichello and others

Sialyl-LewisX Blood vessel

ICAM-
P- and E-selectin 1 or -2

CNS

Cytokines
Chemokines Gradient of
Macrophage chemoattractant substances

Fig. 3. Leukocyte migration. Leukocytes leave the blood and migrate to sites of infection. Sialyl-LewisX on leukocytes binds to
P- and E-selectin on endothelial cells. This connection becomes stronger when CXCL8 binds to its specific receptor on
neutrophils, triggering the production of the integrins LFA-1 and CX3 (mac-1). Inflammatory cytokines such as TNF-a are
necessary to induce the expression of the adhesion molecules ICAM-1 and -2. The link between endothelial cells and ICAM-1
allows the passage of neutrophils in the direction of the gradient of chemoattractant substances.

cause impairment of lipids, proteins, carbohydrates or meningitis. In a rat model, exogenous administration of
nucleic acids, thus increasing the risk of sequelae. Because the intracerebroventricular BDNF was found to block
of the high lipid content in the brain and low cerebral caspase-3 and reduce neuronal apoptosis in pneumococcal
antioxidant defences, the CNS is particularly susceptible to meningitis and reduce cortical necrosis in meningitis
the deleterious properties of oxidative stress (de Menezes caused by Streptococcus agalactiae (Bifrare et al., 2005). In
et al., 2009). another study with the rat model of bacterial meningitis,
administration of exogenous BDNF protected and increased
Brain-derived neurotrophic factor (BDNF) is widely
the neuronal cell population from inflammatory brain
distributed throughout the brain and has demonstrated
injury (Li et al., 2005, 2007). Furthermore, decreased
the ability to protect neurons against damage caused by
hippocampal BDNF levels were correlated with memory
oxidative, metabolic and excitotoxic stress (Marini et al.,
impairment in adult rats that were infected with pneumo-
2007). BDNF has an important regulatory function in cell
coccal and Streptococcus agalactiae meningitis in the
proliferation and exerts neuroprotective effects via the
neonatal period (Barichello et al., 2010, 2013).
modulation of synaptic plasticity by regulating dendritic
spines and synaptic density and the expression of synaptic Dexamethasone is an anti-inflammatory drug with the
proteins (Abdallah et al., 2013; Tartaglia et al., 2001). ability to suppress inhibitor of kB (IkB) degradation
(Bhattacharyya et al., 2010), which decreases pro-inflam-
matory cytokines and inhibits the reactive oxygen species
Novel therapeutic targets in bacterial meningitis
produced by leukocytes (Dandona et al., 1999). Dexameth-
In neonates with suspected bacterial meningitis, empiric asone aggravated hippocampal apoptosis and learning
antibiotic treatment should be instituted without delay. In deficiency in rat pups infected with Streptococcus pneumo-
Wistar rats, early antibiotic administration (i.e. 8 h after niae by intracisternal injection on postnatal day 11 (Leib
induction), compared with late antibiotic administration et al., 2003) and increased hippocampal neuronal apoptosis
(i.e. 16 h after induction), prevented the cognitive impair- in a rabbit model of E. coli meningitis (Spreer et al., 2006).
ments that are typically induced by pneumococcal In a human study, 52 full-term neonates with bacterial
meningitis (Barichello et al., 2009). Another significant meningitis were alternately assigned to receive or not
factor is the ability of an antimicrobial agent to penetrate receive dexamethasone; both groups showed a similar
into the BBB and CNS (Honda & Warren, 2009). New clinical response and similar frequencies of mortality and
targets are being investigated for adjunctive therapy in sequelae (Daoud et al., 1999). On the other hand,

1786 Journal of Medical Microbiology 62


Pathophysiology of neonatal acute bacterial meningitis

adjunctive use of dexamethasone was reported to signific- Abdallah, M. W., Mortensen, E. L., Greaves-Lord, K., Larsen, N.,
antly decrease case fatalities and neurological sequelae in a Bonefeld-Jorgensen, E. C., Norgaard-Pedersen, B., Hougaard, D. M.
& Grove, J. (2013). Neonatal levels of neurotrophic factors and risk of
neonatal population in Nigeria (Airede et al., 2008).
autism spectrum disorders. Acta Psychiatr Scand 128, 61–69.
The use of non-bacteriolytic antibiotics has been shown to Airede, K. I., Adeyemi, O. & Ibrahim, T. (2008). Neonatal bacterial
be potentially beneficial in the treatment of bacterial meningitis and dexamethasone adjunctive usage in Nigeria. Niger J
meningitis in infants. In a rat model of pneumococcal Clin Pract 11, 235–245.
meningitis, daptomycin was more efficient than ceftriaxone Andersen, S. L. (2003). Trajectories of brain development: point of
in decreasing the bacterial titre after infection, and only the vulnerability or window of opportunity? Neurosci Biobehav Rev 27, 3–
animals treated with ceftriaxone showed cortical damage 18.
(Grandgirard et al., 2007). In a rat model of Streptococcus Angstwurm, K., Weber, J. R., Segert, A., Bürger, W., Weih, M., Freyer,
D., Einhäpl, K. M. & Dirnagl, U. (1995). Fucoidin, a polysaccharide
agalactiae meningitis, animals treated with a monoclonal
inhibiting leukocyte rolling, attenuates inflammatory responses in
antibody against TNF-a had less hippocampal injury than experimental pneumococcal meningitis in rats. Neurosci Lett 191, 1–4.
controls (Bogdan et al., 1997). One strategy with which to
Auer, M., Pfister, L. A., Leppert, D., Täuber, M. G. & Leib, S. L. (2000).
prevent brain damage is to restrict leukocyte recruitment to Effects of clinically used antioxidants in experimental pneumococcal
the CSF. Fucoidin is a polysaccharide that blocks the meningitis. J Infect Dis 182, 347–350.
leukocyte receptor L-selectin. Intravenous treatment of rats Barichello, T., Silva, G. Z., Batista, A. L., Savi, G. D., Feier, G., Comim,
with fucoidin at 0, 2, 4 and 6 h after pneumococcal C. M., Quevedo, J. & Dal-Pizzol, F. (2009). Early antibiotic
meningitis induction reduced intracranial pressure and administration prevents cognitive impairment induced by meningitis
pleocytosis in the CSF (Angstwurm et al., 1995). in rats. Neurosci Lett 465, 71–73.
Furthermore, fucoidin injected intracisternally inhibited Barichello, T., Belarmino, E., Jr, Comim, C. M., Cipriano, A. L.,
the accumulation of CSF leukocytes upon the administra- Generoso, J. S., Savi, G. D., Stertz, L., Kapczinski, F. & Quevedo, J.
tion of antibiotics in experimental meningitis (Granert (2010). Correlation between behavioral deficits and decreased brain-
derived neurotrophic [correction of neurotrofic] factor in neonatal
et al., 1998).
meningitis. J Neuroimmunol 223, 73–76.
Reactive oxygen species are produced by resident immune Barichello, T., Lemos, J. C., Generoso, J. S., Cipriano, A. L., Milioli,
cells in the brain and by leukocyte recruitment, as part of G. L., Marcelino, D. M., Vuolo, F., Petronilho, F., Dal-Pizzol, F. & other
the host’s reaction to invasive bacterial infection. To inhibit authors (2011). Oxidative stress, cytokine/chemokine and disruption
the production of oxidative stress, the use of the N- of blood-brain barrier in neonate rats after meningitis by Streptococcus
agalactiae. Neurochem Res 36, 1922–1930.
acetylcysteine and deferoxamine as adjuvant treatment
prevented cognitive impairment in animals submitted to Barichello, T., Fagundes, G. D., Generoso, J. S., Paula Moreira, A.,
Costa, C. S., Zanatta, J. R., Simões, L. R., Petronilho, F., Dal-Pizzol, F.
pneumococcal meningitis (Barichello et al., 2012b). In & other authors (2012a). Brain–blood barrier breakdown and pro-
another study, infant rats treated with antioxidant drugs, inflammatory mediators in neonate rats submitted meningitis by
including N-acetylcysteine, deferoxamine and trylizad- Streptococcus pneumoniae. Brain Res 1471, 162–168.
mesylate, showed reduced cortical injury and mortality Barichello, T., Santos, A. L., Savi, G. D., Generoso, J. S., Otaran, P.,
(Auer et al., 2000). Michelon, C. M., Steckert, A. V., Mina, F., Comim, C. M. & other
authors (2012b). Antioxidant treatment prevents cognitive impair-
ment and oxidative damage in pneumococcal meningitis survivor
Conclusion rats. Metab Brain Dis 27, 587–593.
Despite important advances in treatment, neonatal men- Barichello, T., Lemos, J. C., Generoso, J. S., Carradore, M. M.,
Moreira, A. P., Collodel, A., Zanatta, J. R., Valvassori, S. S. &
ingitis is one of the most significant infectious diseases of
Quevedo, J. (2013). Evaluation of the brain-derived neurotrophic
the CNS, with high mortality and morbidity. Experimental factor, nerve growth factor and memory in adult rats survivors of the
animal models, despite limitations, provide an under- neonatal meningitis by Streptococcus agalactiae. Brain Res Bull 92, 56–
standing of the complex pathophysiology of this disease 59.
and suggest new adjunctive therapies. Bhattacharyya, S., Ratajczak, C. K., Vogt, S. K., Kelley, C., Colonna,
M., Schreiber, R. D. & Muglia, L. J. (2010). TAK1 targeting by
glucocorticoids determines JNK and IkappaB regulation in Toll-like
Acknowledgements receptor-stimulated macrophages. Blood 115, 1921–1931.

This research was supported by grants from CNPq, FAPESC, Bifrare, Y. D., Kummer, J., Joss, P., Täuber, M. G. & Leib, S. L. (2005).
FAPEMIG, UNESC, INCT-TM and the L’Oréal-UNESCO Brazil Brain-derived neurotrophic factor protects against multiple forms of
Fellowship for Women in Science 2011. The authors declare that they brain injury in bacterial meningitis. J Infect Dis 191, 40–45.
have no conflicts of interest. Bogdan, I., Leib, S. L., Bergeron, M., Chow, L. & Täuber, M. G. (1997).
Tumor necrosis factor-alpha contributes to apoptosis in hippocampal
neurons during experimental group B streptococcal meningitis.
J Infect Dis 176, 693–697.
References
Carlos, T. M. & Harlan, J. M. (1994). Leukocyte-endothelial adhesion
Aas, J. A., Paster, B. J., Stokes, L. N., Olsen, I. & Dewhirst, F. E. molecules. Blood 84, 2068–2101.
(2005). Defining the normal bacterial flora of the oral cavity. J Clin Castell, J. V., Gómez-Lechón, M. J., David, M., Andus, T., Geiger, T.,
Microbiol 43, 5721–5732. Trullenque, R., Fabra, R. & Heinrich, P. C. (1989). Interleukin-6 is the

http://jmm.sgmjournals.org 1787
T. Barichello and others

major regulator of acute phase protein synthesis in adult human Hanke, M. L. & Kielian, T. (2011). Toll-like receptors in health and
hepatocytes. FEBS Lett 242, 237–239. disease in the brain: mechanisms and therapeutic potential. Clin Sci
Costa, A., Gupta, R., Signorino, G., Malara, A., Cardile, F., Biondo, C., (Lond) 121, 367–387.
Midiri, A., Galbo, R., Trieu-Cuot, P. & other authors (2012). Hanna, S. & Etzioni, A. (2012). Leukocyte adhesion deficiencies. Ann
Activation of the NLRP3 inflammasome by group B streptococci. N Y Acad Sci 1250, 50–55.
J Immunol 188, 1953–1960. Hayashi, F., Smith, K. D., Ozinsky, A., Hawn, T. R., Yi, E. C., Goodlett,
Cuenca, A. G., Wynn, J. L., Moldawer, L. L. & Levy, O. (2013). Role of D. R., Eng, J. K., Akira, S., Underhill, D. M. & Aderem, A. (2001). The
innate immunity in neonatal infection. Am J Perinatol 30, 105–112. innate immune response to bacterial flagellin is mediated by Toll-like
Dandona, P., Mohanty, P., Hamouda, W., Aljada, A., Kumbkarni, Y. & receptor 5. Nature 410, 1099–1103.
Garg, R. (1999). Effect of dexamethasone on reactive oxygen species Heath, P. T. & Okike, I. O. (2010). Neonatal bacterial meningitis: an
generation by leukocytes and plasma interleukin-10 concentrations: a update. Paediatr Child Health (Oxford) 20, 526–530.
pharmacodynamic study. Clin Pharmacol Ther 66, 58–65. Heath, P. T., Nik Yusoff, N. K. & Baker, C. J. (2003). Neonatal
Daoud, A. S., Batieha, A., Al-Sheyyab, M., Abuekteish, F., Obeidat, A. meningitis. Arch Dis Child Fetal Neonatal Ed 88, F173–F178.
& Mahafza, T. (1999). Lack of effectiveness of dexamethasone in Heath, P. T., Okike, I. O. & Oeser, C. (2011). Neonatal meningitis: can
neonatal bacterial meningitis. Eur J Pediatr 158, 230–233. we do better? Adv Exp Med Biol 719, 11–24.
Davis, B. K., Wen, H. & Ting, J. P. (2011). The inflammasome NLRs in Hemmi, H., Takeuchi, O., Kawai, T., Kaisho, T., Sato, S., Sanjo, H.,
immunity, inflammation, and associated diseases. Annu Rev Immunol Matsumoto, M., Hoshino, K., Wagner, H. & other authors (2000). A
29, 707–735. Toll-like receptor recognizes bacterial DNA. Nature 408, 740–745.
de Louvois, J., Halket, S. & Harvey, D. (2005). Neonatal meningitis in Holt, D. E., Halket, S., de Louvois, J. & Harvey, D. (2001). Neonatal
England and Wales: sequelae at 5 years of age. Eur J Pediatr 164, 730– meningitis in England and Wales: 10 years on. Arch Dis Child Fetal
734. Neonatal Ed 84, F85–F89.
de Menezes, C. C., Dorneles, A. G., Sperotto, R. L., Duarte, M. M., Honda, H. & Warren, D. K. (2009). Central nervous system infections:
Schetinger, M. R. & Loro, V. L. (2009). Oxidative stress in meningitis and brain abscess. Infect Dis Clin North Am 23, 609–623.
cerebrospinal fluid of patients with aseptic and bacterial meningitis.
Neurochem Res 34, 1255–1260. Janot, L., Secher, T., Torres, D., Maillet, I., Pfeilschifter, J.,
Quesniaux, V. F., Landmann, R., Ryffel, B. & Erard, F. (2008). CD14
Ek, C. J., Dziegielewska, K. M., Habgood, M. D. & Saunders, N. R. works with toll-like receptor 2 to contribute to recognition and
(2012). Barriers in the developing brain and neurotoxicology. control of Listeria monocytogenes infection. J Infect Dis 198, 115–
Neurotoxicology 33, 586–604. 124.
Fassbender, K., Schminke, U., Ries, S., Ragoschke, A., Kischka, U., Kastenbauer, S., Koedel, U., Becker, B. F. & Pfister, H. W. (2002).
Fatar, M. & Hennerici, M. (1997). Endothelial-derived adhesion Pneumococcal meningitis in the rat: evaluation of peroxynitrite
molecules in bacterial meningitis: association to cytokine release and scavengers for adjunctive therapy. Eur J Pharmacol 449, 177–181.
intrathecal leukocyte-recruitment. J Neuroimmunol 74, 130–134.
Kastenbauer, S., Koedel, U., Weih, F., Ziegler-Heitbrock, L. & Pfister,
Filias, A., Theodorou, G. L., Mouzopoulou, S., Varvarigou, A. A., H. W. (2004). Protective role of NF-kappaB1 (p50) in experimental
Mantagos, S. & Karakantza, M. (2011). Phagocytic ability of pneumococcal meningitis. Eur J Pharmacol 498, 315–318.
neutrophils and monocytes in neonates. BMC Pediatr 11, 29.
Kim, K. S. (2003). Pathogenesis of bacterial meningitis: from
Flo, T. H., Halaas, O., Lien, E., Ryan, L., Teti, G., Golenbock, D. T., bacteraemia to neuronal injury. Nat Rev Neurosci 4, 376–385.
Sundan, A. & Espevik, T. (2000). Human toll-like receptor 2 mediates
monocyte activation by Listeria monocytogenes, but not by group B Kim, K. S. (2008). Mechanisms of microbial traversal of the blood–
streptococci or lipopolysaccharide. J Immunol 164, 2064–2069. brain barrier. Nat Rev Microbiol 6, 625–634.
Furyk, J. S., Swann, O. & Molyneux, E. (2011). Systematic review: Klein, M., Koedel, U. & Pfister, H. W. (2006). Oxidative stress in
neonatal meningitis in the developing world. Trop Med Int Health 16, pneumococcal meningitis: a future target for adjunctive therapy? Prog
672–679. Neurobiol 80, 269–280.
Gaschignard, J., Levy, C., Romain, O., Cohen, R., Bingen, E., Aujard, Koedel, U. (2009). Toll-like receptors in bacterial meningitis. Curr
Y. & Boileau, P. (2011). Neonatal bacterial meningitis: 444 cases in 7 Top Microbiol Immunol 336, 15–40.
years. Pediatr Infect Dis J 30, 212–217. Koedel, U., Bayerlein, I., Paul, R., Sporer, B. & Pfister, H. W. (2000).
Grandgirard, D. & Leib, S. L. (2010). Meningitis in neonates: bench to Pharmacologic interference with NF-kappaB activation attenuates
bedside. Clin Perinatol 37, 655–676. central nervous system complications in experimental Pneumococcal
meningitis. J Infect Dis 182, 1437–1445.
Grandgirard, D., Schürch, C., Cottagnoud, P. & Leib, S. L. (2007).
Prevention of brain injury by the nonbacteriolytic antibiotic Koppe, U., Suttorp, N. & Opitz, B. (2012). Recognition of
daptomycin in experimental pneumococcal meningitis. Antimicrob Streptococcus pneumoniae by the innate immune system. Cell
Agents Chemother 51, 2173–2178. Microbiol 14, 460–466.
Granert, C., Raud, J. & Lindquist, L. (1998). The polysaccharide Krebs, V. L., Okay, T. S., Okay, Y. & Vaz, F. A. (2005). Tumor necrosis
fucoidin inhibits the antibiotic-induced inflammatory cascade in factor-alpha, interleukin-1beta and interleukin-6 in the cerebrospinal
experimental pneumococcal meningitis. Clin Diagn Lab Immunol 5, fluid of newborn with meningitis. Arq Neuropsiquiatr 63, 7–13.
322–324. Kronfol, Z. & Remick, D. G. (2000). Cytokines and the brain:
Gruol, D. L. & Nelson, T. E. (1997). Physiological and pathological implications for clinical psychiatry. Am J Psychiatry 157, 683–694.
roles of interleukin-6 in the central nervous system. Mol Neurobiol 15, Lahrtz, F., Piali, L., Spanaus, K. S., Seebach, J. & Fontana, A. (1998).
307–339. Chemokines and chemotaxis of leukocytes in infectious meningitis.
Hamed, S. A., Hamed, E. A. & Zakary, M. M. (2009). Oxidative stress J Neuroimmunol 85, 33–43.
and S-100B protein in children with bacterial meningitis. BMC Neurol Leib, S. L., Heimgartner, C., Bifrare, Y. D., Loeffler, J. M. & Täuber,
9, 51. M. G. (2003). Dexamethasone aggravates hippocampal apoptosis and

1788 Journal of Medical Microbiology 62


Pathophysiology of neonatal acute bacterial meningitis

learning deficiency in pneumococcal meningitis in infant rats. Pediatr Remington, J. S. (2011). Infectious Diseases of the Fetus and Newborn
Res 54, 353–357. Infant, 7th edn. Philadelphia, PA: Saunders/Elsevier.
Li, L., Shui, Q. X. & Li, X. (2005). Neuroprotective effects of brain- Rivest, S. (2003). Molecular insights on the cerebral innate immune
derived neurotrophic factor (BDNF) on hearing in experimental system. Brain Behav Immun 17, 13–19.
pneumococcal meningitis. J Child Neurol 20, 51–56. Rosenberg, G. A., Estrada, E. Y., Dencoff, J. E. & Stetler-Stevenson,
Li, L., Shui, Q. X., Liang, K. & Ren, H. (2007). Brain-derived W. G. (1995). Tumor necrosis factor-alpha-induced gelatinase B
neurotrophic factor rescues neurons from bacterial meningitis. causes delayed opening of the blood-brain barrier: an expanded
Pediatr Neurol 36, 324–329. therapeutic window. Brain Res 703, 151–155.
Liu, X., Chauhan, V. S., Young, A. B. & Marriott, I. (2010). NOD2 Saunders, N. R., Liddelow, S. A. & Dziegielewska, K. M. (2012).
mediates inflammatory responses of primary murine glia to Barrier mechanisms in the developing brain. Front Pharmacol 3, 46.
Streptococcus pneumoniae. Glia 58, 839–847.
Seki, E., Tsutsui, H., Tsuji, N. M., Hayashi, N., Adachi, K., Nakano, H.,
Liu, L., Johnson, H. L., Cousens, S., Perin, J., Scott, S., Lawn, J. E., Futatsugi-Yumikura, S., Takeuchi, O., Hoshino, K. & other authors
Rudan, I., Campbell, H., Cibulskis, R. & other authors (2012). Global, (2002). Critical roles of myeloid differentiation factor 88-dependent
regional, and national causes of child mortality: an updated proinflammatory cytokine release in early phase clearance of Listeria
systematic analysis for 2010 with time trends since 2000. Lancet monocytogenes in mice. J Immunol 169, 3863–3868.
379, 2151–2161.
Sellner, J., Täuber, M. G. & Leib, S. L. (2010). Pathogenesis and
Malley, R., Henneke, P., Morse, S. C., Cieslewicz, M. J., Lipsitch, M., pathophysiology of bacterial CNS infections. In Handbook of Clinical
Thompson, C. M., Kurt-Jones, E., Paton, J. C., Wessels, M. R. & Neurology, vol. 96, pp. 1–16. Edited by K. L. Roos & A. R. Tunkel.
Golenbock, D. T. (2003). Recognition of pneumolysin by Toll-like Philadelphia, PA: Elsevier.
receptor 4 confers resistance to pneumococcal infection. Proc Natl
Acad Sci U S A 100, 1966–1971. Spreer, A., Gerber, J., Hanssen, M., Schindler, S., Hermann, C.,
Lange, P., Eiffert, H. & Nau, R. (2006). Dexamethasone increases
Marini, A. M., Jiang, X., Wu, X., Pan, H., Guo, Z., Mattson, M. P., hippocampal neuronal apoptosis in a rabbit model of Escherichia coli
Blondeau, N., Novelli, A. & Lipsky, R. H. (2007). Preconditioning and meningitis. Pediatr Res 60, 210–215.
neurotrophins: a model for brain adaptation to seizures, ischemia and
other stressful stimuli. Amino Acids 32, 299–304. Tartaglia, N., Du, J., Tyler, W. J., Neale, E., Pozzo-Miller, L. & Lu, B.
(2001). Protein synthesis-dependent and -independent regulation of
Mitchell, L., Smith, S. H., Braun, J. S., Herzog, K. H., Weber, J. R. &
hippocampal synapses by brain-derived neurotrophic factor. J Biol
Tuomanen, E. I. (2004). Dual phases of apoptosis in pneumococcal
Chem 276, 37585–37593.
meningitis. J Infect Dis 190, 2039–2046.
Tato, C. M. & Hunter, C. A. (2002). Host–pathogen interactions:
Mitchell, D., Yong, M., Schroder, W., Black, M., Tirrell, M. & Olive, C.
subversion and utilization of the NF-kappa B pathway during
(2010). Dual stimulation of MyD88-dependent Toll-like receptors
infection. Infect Immun 70, 3311–3317.
induces synergistically enhanced production of inflammatory cytokines
in murine bone marrow-derived dendritic cells. J Infect Dis 202, 318–329. Tenenbaum, T., Spellerberg, B., Adam, R., Vogel, M., Kim, K. S. &
Schroten, H. (2007). Streptococcus agalactiae invasion of human brain
Mook-Kanamori, B. B., Geldhoff, M., van der Poll, T. & van de Beek,
microvascular endothelial cells is promoted by the laminin-binding
D. (2011). Pathogenesis and pathophysiology of pneumococcal
protein Lmb. Microbes Infect 9, 714–720.
meningitis. Clin Microbiol Rev 24, 557–591.
Mukai, A. O., Krebs, V. L., Bertoli, C. J. & Okay, T. S. (2006). TNF- Tsao, N., Chang, W. W., Liu, C. C. & Lei, H. Y. (2002). Development of
alpha and IL-6 in the diagnosis of bacterial and aseptic meningitis in hematogenous pneumococcal meningitis in adult mice: the role of
children. Pediatr Neurol 34, 25–29. TNF-alpha. FEMS Immunol Med Microbiol 32, 133–140.
Nizet, V. & Klein, J. O. (2011). Chapter 6 - Bacterial sepsis and Tsukahara, H., Haruta, T., Todoroki, Y., Hiraoka, M., Noiri, E., Maeda,
meningitis. In Infectious Diseases of the Fetus and Newborn, 7th edn, M. & Mayumi, M. (2002). Oxidant and antioxidant activities in
pp. 222–275. Edited by J. S. Remington, J. O. Klein, C. B. Wilson, childhood meningitis. Life Sci 71, 2797–2806.
V. Nizet & Y. A. Maldonado. Philadelphia: W. B. Saunders. van de Beek, D., de Gans, J., Spanjaard, L., Weisfelt, M., Reitsma,
Opitz, B., Püschel, A., Schmeck, B., Hocke, A. C., Rosseau, S., J. B. & Vermeulen, M. (2004). Clinical features and prognostic factors
Hammerschmidt, S., Schumann, R. R., Suttorp, N. & Hippenstiel, S. in adults with bacterial meningitis. N Engl J Med 351, 1849–1859.
(2004). Nucleotide-binding oligomerization domain proteins are van Furth, A. M., Roord, J. J. & van Furth, R. (1996). Roles of
innate immune receptors for internalized Streptococcus pneumoniae. proinflammatory and anti-inflammatory cytokines in pathophys-
J Biol Chem 279, 36426–36432. iology of bacterial meningitis and effect of adjunctive therapy. Infect
Østergaard, C., Brandt, C., Konradsen, H. B. & Samuelsson, S. Immun 64, 4883–4890.
(2004). Differences in survival, brain damage, and cerebrospinal fluid Verani, J. R., McGee, L., Schrag, S. J. & Division of Bacterial
cytokine kinetics due to meningitis caused by 3 different Streptococcus Diseases, National Center for Immunization and Respiratory
pneumoniae serotypes: evaluation in humans and in 2 experimental Diseases, Centers for Disease Control and Prevention (CDC)
models. J Infect Dis 190, 1212–1220. (2010). Prevention of perinatal group B streptococcal disease–revised
Paul, R., Koedel, U., Winkler, F., Kieseier, B. C., Fontana, A., Kopf, M., guidelines from CDC, 2010. MMWR Recomm Rep 59 (RR-10), 1–36.
Hartung, H. P. & Pfister, H. W. (2003). Lack of IL-6 augments Wang, H., Zhang, J., Wu, H., Jiang, C., Zheng, Q. & Li, Z. (2006).
inflammatory response but decreases vascular permeability in Inhibition of RAW264.7 macrophage inflammatory cytokines release
bacterial meningitis. Brain 126, 1873–1882. by small haparin RNAi targeting TLR4. J Huazhong Univ Sci
Pong, A. & Bradley, J. S. (1999). Bacterial meningitis and the Technolog Med Sci 26, 500–503.
newborn infant. Infect Dis Clin North Am 13, 711–733, viii. Zeng, H. C., Zhang, L., Li, Y. Y., Wang, Y. J., Xia, W., Lin, Y., Wei, J. &
Puopolo, K. M. & Baker, C. J. (2012). Group B Streptococcal Infection in Xu, S. Q. (2011). Inflammation-like glial response in rat brain
Neonates and Young Infants, 29th edn. Elk Grove Village: Pickering LK. induced by prenatal PFOS exposure. Neurotoxicology 32, 130–139.

http://jmm.sgmjournals.org 1789

View publication stats

You might also like