Download as pdf or txt
Download as pdf or txt
You are on page 1of 28

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

net/publication/237091401

Bacterial meningitis

Chapter · January 2013


DOI: 10.1007/978-3-642-30144-5_106

CITATION READS

1 19,521

3 authors, including:

Tone Tønjum Birgitta Henriques-Normark


University of Oslo Karolinska Institutet
249 PUBLICATIONS   4,090 CITATIONS    235 PUBLICATIONS   8,388 CITATIONS   

SEE PROFILE SEE PROFILE

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

Microbial genome dynamics View project

Cinobufagin immunomodulation View project

All content following this page was uploaded by Tone Tønjum on 13 September 2015.

The user has requested enhancement of the downloaded file.


18 Meningitis
Tone Tønjum1 . Petter Brandtzæg2 . Birgitta Henriques-Normark3
1
Centre for Molecular Biology and Neuroscience (CMBN), Department of Microbiology, University of
Oslo, Oslo University Hospital, Oslo, Norway
2
Department of Pediatrics, Ullevål University Hospital, University of Oslo, Oslo, Norway
3
Department of Microbiology, Tumor, and Cell Biology, Karolinska Institutet, Stockholm, Sweden

Major Causes of Bacterial Meningitis . . . . . . . . . . . . . . . . . . . . 402 the causative agent. Young children are at highest risk for mor-
Neisseria meningitidis and Meningococcal Disease . . . . . . 403 tality and morbidity, especially those from lower socioeconomic
Streptococcus pneumoniae and Pneumococcal strata in countries with poor medical infrastructure and those
Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 404 infected with Neisseria meningitidis (the meningococcus) or
Pathogenesis of Meningococcal and Pneumococcal Streptococcus pneumoniae (the pneumococcus). Additional risk
Meningitis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 405 factors for poor prognosis after infection include the severity/
Meningococcal and Pneumococcal Colonization and stage of illness on presentation, exposure to an antibiotic-
Carriage . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 405 resistant organism, and the fact that medical professionals lack
Invasive Meningococcal and Pneumococcal Disease . . . . 408 understanding of mechanisms underlying the pathological fea-
Meningococcal Cell Structure and Virulence Factors . . . 408 tures of meningitis. When bacterial meningitis is suspected,
Meningococcal Genome Characteristics and immediate action is imperative to establish a definitive diagno-
Dynamics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410 sis, and antimicrobial treatment must be initiated immediately
as a precautionary measure, because the mortality rate for
Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 410 untreated bacterial meningitis approaches 100 %; even with
Pathogenesis of Meningococcal Meningitis . . . . . . . . . . . . . 410 optimal treatment, mortality and morbidity remain high. Neu-
Pathogenesis of Pneumococcal Meningitis . . . . . . . . . . . . . . 411 rological sequelae are relatively common in meningitis survi-
vors, especially if the agent of disease is a pneumococcal
Clinical Features . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 412 microorganism.
Most pathogenic microbes could potentially cause meningi-
Diagnosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 414 tis in the human brain; however, only two pathogens, N.
Laboratory Diagnosis of Meningitis . . . . . . . . . . . . . . . . . . . . . 414 meningitidis and S. pneumoniae, account for most cases of
Molecular Typing of N. meningitidis . . . . . . . . . . . . . . . . . . . 416 acute bacterial meningitis, when patients in all age groups are
Molecular Typing of S. pneumoniae . . . . . . . . . . . . . . . . . . . . . 416 considered. In contrast, in very young children and neonates,
Host Susceptibility to Meningococcal most cases are caused by group B streptococcus, Escherichia coli,
and Pneumococcal Meningitis . . . . . . . . . . . . . . . . . . . . . . . . . . 417 and Listeria monocytogenes. In developing countries,
Haemophilus influenza type b and Salmonella species are still
Therapy and Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 418 major causes of meningitis in infants and young children. Sal-
Antimicrobial Treatment of Meningococcal monella meningitis has a particularly dismal prognosis. Menin-
and Pneumococcal Meningitis . . . . . . . . . . . . . . . . . . . . . . . . . . 418 gitis is, in the majority of the cases, a consequence of a preceding
Adjunctive Treatment . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 419 bacteremia with encapsulated strains. Although the reasons for
this association are incompletely understood, bacterial agents
Prevention of Meningococcal and Pneumococcal that cause meningitis tend to express surface structures mim-
Disease . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 419 icking structures and epitopes on human cells and a capsule with
Chemoprophylaxis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 421 antiphagocytic properties that protect them from phagocytosis
and normal immune surveillance. Thus, the absence of opsonic
Future Challenges and Opportunities . . . . . . . . . . . . . . . . . . . . . 421 or bactericidal antibodies is considered a major risk factor for
meningitis. In this regard, age-related incidence of meningococ-
cal and pneumococcal disease is inversely related to prevalence
Abstract of serum bactericidal activity. Successful identification of micro-
Bacterial meningitis is an inflammation of the meninges, includ- bial epitopes that induce opsonic or bactericidal antibodies and
ing the pia, arachnoid, and subarachnoid space, that occurs in successful vaccination of infants and children using antigenic
response to infection with bacteria and/or bacterial products. agents based on these epitopes has changed the epidemiology of
Bacterial meningitis is a significant cause of mortality and mor- bacterial meningitis, particularly due to reduced incidence of
bidity worldwide, with considerable variation in incidence Haemophilus influenzae type b-induced meningitis more so in
depending on age and geographic location of the patient and industrialized countries. However, antigenic epitopes suitable

E. Rosenberg et al. (eds.), The Prokaryotes – Human Microbiology, DOI 10.1007/978-3-642-30144-5_106,


# Springer-Verlag Berlin Heidelberg 2013
402 18 Meningitis

for this preventive approach have not been identified in all . Table 18.1
organisms that cause meningitis with significant frequency Meningococcal virulence factors
today.
Virulence factor Function
Lipopolysaccharide Lipooligosaccharide (LOS) has endotoxin
Major Causes of Bacterial Meningitis (LPS/LOS) activity and is released as bacterial outer
membrane vesicles (blebs) or through
Bacterial meningitis, an inflammation of the meninges affecting cellular lysis. LOS is responsible for toxic
damage to the human tissue, development
the pia, arachnoid, and subarachnoid space in response to
of septic shock, and disseminated
bacteria and bacterial products, continues to be an important
intravascular coagulation (DIC) through
cause of mortality and morbidity worldwide (WHO 2012). interactions with Toll-like receptors (TLR4)
However, mortality and morbidity vary by age and geographic and cytokine induction
location of the patient and the causative organism. Patients Polysaccharide Polysaccharide surface component which
at risk for high mortality and morbidity include infants capsule works as a protective shell and blocks the
and young children, those living in low-income countries insertion of the membrane attack complex
[or in low socioeconomic strata], HIV-infected patients in of the complement system and protects
developing countries, those infected with Neisseria meningitidis the bacteria from phagocytosis. The
(the meningococcus, Mc) or Streptococcus pneumoniae capsule is the main component enabling
(the pneumococcus, Pc), and infants living in resource-poor bacterial survival in blood and resisting
countries infected with Salmonella species (Chang et al. 2004; bactericidal antibodies. The serogroup
Davidsen et al. 2007; Molyneux et al. 2006). In the pre-vaccine B capsule can also mimic human antigens
era, Haemophilus influenzae was the most common bacterial Type 4 pili Major adhesins that mediate initial
pathogen causing meningitis in young children, and attachment to nonciliated human cells.
H. influenzae type b caused approximately 70 % of bacterial Also required for efficient transformation
of DNA
meningitis in children younger than 5 years of age (Dery and
Hasbun 2007). However, in the early 1990s, a conjugated vaccine Outer membrane Dominant antigens. Porin protein
proteins (OMP) promotes intracellular survival. Opacity
against H. influenzae type b was developed, and after a program
proteins mediate firm attachment to
to vaccinate against H. influenza type b was implemented, the
eukaryotic cells. Rmp protein can protect
global incidence of H. influenzae-induced meningitis decreased other antigens from bactericidal
dramatically (55 % reduction in the annual number of cases interactions with antibodies. Frequent
in the United States alone) (Thigpen et al. 2011). Subsequently, antigenic variation makes it difficult for the
S. pneumoniae and N. meningitidis have emerged as the host immune system to recognize these
pathogens responsible for most cases of bacterial meningitis antigens
(Dery and Hasbun 2007; Brouwer et al. 2010). Iron-binding Transferrin-, lactoferrin-, and hemoglobin-
Almost all microbes that are pathogenic to human beings proteins binding proteins. Pathogenic Neisseria
have the potential to cause meningitis, but a relatively small spp. are dependent on a constant iron
number of pathogens (primarily N. meningitidis, S. pneumoniae, supply for growth
H. influenzae type b, group B streptococcal disease, Escherichia IgA1 protease Destroys mucosal IgA which is a part of the
coli, Salmonella species, and Listeria monocytogenes) account local immune system
for most cases of acute bacterial meningitis in children and Beta-lactamase An enzyme that hydrolyzes the b-lactam
neonates, although the reasons for this remain incompletely ring of penicillin. Important for antibiotic
understood (> Table 18.1). One common denominator among resistance development
bacterial agents that cause meningitis is the presence of an
antiphagocytic capsule and the related fact that opsonic or
bactericidal antibody is absent in most cases of meningitis
(Gotschlich et al. 1969). As a correlate, age-related incidence
of N. meningitidis, S. pneumoniae, and H. influenzae type b
disease is inversely related to prevalence of serum bactericidal Miller 2006; Tsai et al. 2008). However, antigenic epitopes suit-
activity/antibodies (Gotschlich et al. 1969; Chudwin et al. 1983), able for this preventive approach have not been identified for all
and the lack of type-specific antibody is a major risk factor for organisms that cause meningitis with significant frequency.
neonatal group B streptococcal disease (Baker and Kasper 1976). Currently, the most agents that cause severe meningitis most
Successful identification of microbial epitopes that induce frequently in all age groups worldwide are N. meningitidis and
opsonic or bactericidal antibodies and successful vaccination S. pneumoniae. This chapter reviews the extensive knowledge
of infants and children using antigenic compounds based base, accumulated over many years by many researchers,
on these epitopes have changed the epidemiology of bacterial on these organisms and their pathological effects on their
meningitis (Peltola 2000; Whitney et al. 2003; Borrow and human host.
Meningitis 18 403

epithelium CSF

mucus blood

The meningococcus The pneumococcus


Occasional tissue
invasion
and dissemination to
blood
and CSF

CSF

Oropharyngeal colonization

. Fig. 18.1
Stages in microbial CNS pathogenesis. Neisseria meningitidis and Streptococcus pneumoniae are causative agents of meningitis.
Primary adherence to mucosal epithelial cells occurs via pili and other surface components. The bacteria can then establish an intimate
contact with the host cells via outer membrane proteins such as the Opa protein(s), an interaction that might allow bacterial
transcytotic passage to subepithelial tissues. Bacterial interactions with the mucosal cells, submucous tissue and endothelial cells might
result in its entry into the bloodstream and subsequent entry/passage of the blood–brain-barrier to cause meningitis/CNS infection

Neisseria meningitidis and Meningococcal Disease countries, and when patients survive, they often suffer limb loss,
hearing loss, cognitive dysfunction, visual impairment,
N. meningitidis, the meningococcus, is a Gram-negative educational difficulties, developmental delays, motor nerve
diplococcus in b-proteobacterium (> Fig. 18.1) that causes deficits, seizure disorders, and behavioral problems (Kim 2003;
endemic and epidemic meningitis and/or septicemia worldwide. Roine et al. 2008). Curiously, certain geographic and temporal
Epidemic meningococcal meningitis was first described by anomalies exist in the natural history of the disease;
Vieusseux in 1805 in Geneva (Vieusseux 1805). Throughout these include the fact that no outbreaks of epidemic
the nineteenth century, periodic epidemics occurred, involving meningococcemia or meningitis prior to 1805 have been
primarily young children and adolescents, as well as military reported, as well as no reported epidemics in the meningitis
recruits. The genus Neisseria was named after Albert Neisser, belt of sub-Saharan Africa prior to 1900 (Cartwright 1995).
who observed gonococci (Neisseria gonorrhoeae) in leukocytes in Epidemiology of Meningococcal Disease. Meningococcal
urethral exudates from patients with gonorrhea in 1879. disease is a major global health problem (> Fig. 18.1)
Marchiafava and Celli (1884) described intracellular oval (Stephens 2007) that causes endemic, hyperendemic, epidemic,
micrococci in a sample of cerebrospinal fluid (CSF), and and pandemic outbreaks at a rate that varies according to
Anton Weichselbaum (1887) isolated the organism from six geographic region, population demographics, host susceptibil-
of eight cases of primary sporadic community-acquired ity, and infectious agent/strain. In 2010, approximately 170,000
meningitis, identified features that distinguish pneumococci individuals died from meningococcal disease worldwide.
from meningococci, and gave it the name Diplococcus The case fatality rate is 5–10 % in industrialized countries, and
intracellularis meningitidis (Marchiafava and Celli 1884; of those who survive, 10–20 % develop permanent sequelae.
Weichselbaum 1887). The fact that N. meningitidis enters Transmission of meningococci occurs by respiratory droplets
human cells is an important feature of meningococcal patho- or kissing, requiring close contact. They colonize nonciliated
genesis. In 1896, Kiefer reported that healthy individuals can be epithelial cells in nasopharynx and the tonsils (Stephens 1982).
asymptomatic ‘‘carriers’’ of nasopharyngeal meningococci. Infection, which occurs within 2–10 days, leads to invasive
Meningococci are recognized as agents that cause endemic disease in individuals who lack bactericidal antibodies that
cases, case clusters, epidemics and pandemics of meningitis, recognize the invading strain and in complement-factor-
devastating septicemia, and, less commonly, pneumonia, septic deficient individuals (Gotschlich et al. 1969; Stephens et al.
arthritis, pericarditis, chronic bacteremia, and conjunctivitis in 2007). Concurrent viral or mycoplasmal respiratory tract infec-
hundreds of thousands of individuals worldwide each year tions increase susceptibility to systemic invasion by the patho-
(WHO 2012). Mortality can be 10 % or higher in developing genic bacteria.
404 18 Meningitis

The polysaccharide capsule is the primary determinant of are or are soon to be available, there remain many challenges
the relative virulence of disease-causing meningococci. Most before it will be possible to optimize and deliver effective preven-
infections are caused by strains belonging to serogroups A, B, tive and therapeutic approaches for meningococcal disease.
C, X, Y, and W-135 (Stephens et al. 2007; Khatami and Pollard
2011). In Western Europe, North America, and South America,
serogroups B and C are the primary disease-causing pathogens, Streptococcus pneumoniae and Pneumococcal
and these strains are endemic, causing disease at an incidence Disease
of 1–3/100,000. Periodically, local hyperendemic outbreaks
occur when new lineages spread through the population. The Gram-positive diplococcus S. pneumoniae (> Fig. 18.1) is
In 2001–2006, serogroup B infection spread worldwide, a eubacterium belonging to phylum Firmicutes and order
culminating in disease outbreaks in Australia and New Zealand Lactobacillales. S. pneumoniae is a major cause of mild respira-
(Stephens 2007; Stephens et al. 2007). In China, the Middle East, tory tract infections (i.e., otitis media and sinusitis) and is also
and parts of Africa, serogroups A and C predominate. the worldwide leading cause of the much more severe diseases,
Large epidemics are attributed predominantly to serogroup community-acquired pneumonia, septicemia, and meningitis.
A strains. In the African ‘‘meningitis belt,’’ major periodic Epidemiology of S. pneumoniae. It is estimated that between
epidemics of serogroup A disease occur every 5–12 years, 1.5 and 2 million people die from pneumococcal infection every
with attack rates of 500/100,000 population or higher year, a rate similar to mortality from tuberculosis. Young
(Achtman 1995). The emergence and global importance of children, the elderly, and immunocompromised individuals
serogroups W-135, X, and Y were recognized only in the last (i.e., splenectomized individuals, HIV patients, and chronically
10 years. Serogroup W-135 was identified in 2002–2003 as ill patients suffering from renal or liver disease, alcoholism,
a major threat, and it was the primary pathogen responsible diabetes mellitus, skull fracture, or cochlear implants) (Biernath
for outbreaks in Africa. An unprecedented increase in incidence et al. 2006; Weisfelt et al. 2006; Brouwer et al. 2010). Past history
of serogroup X meningitis was observed in Niger in 2006 of viral infection, especially influenza A virus (IAV), also
(Boisier et al. 2007). Occasionally, particularly virulent strains sensitizes the host for pneumococcal infection. Coinfection
arise that cause pandemic outbreaks that manifest across conti- with pneumococcal influenza is the most important factor
nents (Stephens et al. 2007). In the USA, Israel, and Sweden, contributing to increased morbidity and/or mortality from
disease due to serogroup Y strains has increased (Rosenstein S. pneumoniae worldwide, even accounting for the severity and
et al. 2001). increased mortality associated with the 1918 influenza
Meningococcal disease can occur when a pathogenic organ- pandemic. Based on 156 studies published in 2000, O’Brien
ism infects a susceptible host. Specific factors that increase risk et al. estimated the global burden of pneumococcal disease
of meningococcal disease include climate, age, social behavior, in children younger than 5 years of age (2009) as close to
health status including preexisting or coinfection with other 14.5 million serious pneumococcal infections and 826,000
microorganisms (MacLennan et al. 2006), and hereditary factors deaths, of which 91,000 were in individuals positive for HIV
(Schneider et al. 2007; Stephens et al. 2007). Additional risk (O’Brien et al. 2009). More than 61 % of the deaths occurred in
factors for invasive meningococcal and pneumococcal disease 10 African and Asian countries. On a global level, it was estimated
include smoking, living in crowded conditions, exposure to that infection with S. pneumoniae accounts for approximately
pathogen by travel to epidemic areas, deficiency in terminal 11 % of mortality in this age group, when deaths due to infection
complement components, and asplenia (Yazdankhah and with HIV were excluded. Europe had the lowest incidence rate,
Caugant 2004). Meningococcal and pneumococcal disease can 6 per 100,000, while the highest rate was 38 per 100,000 in Africa.
affect persons of all age groups, but higher rates of invasive The incidence of pneumococcal meningitis in the United States
disease in developed countries are seen in infants and children in children and adults decreased from 1.09 per 100,000 in
less than 4 years old, adolescents, military recruits, and individ- 1998–1999 to 0.81 in 2006–2007, likely reflecting implementation
uals living among a transient population (e.g., college students of a childhood vaccination program in 2000 (see below) (Thigpen
in dormitories) (Stephens et al. 2007; Rosenstein et al. 2001). et al. 2011). In children 2–23 months of age, incidence decreased
Elderly individuals are also at risk for pneumococcal disease. from 9.68 to 3.67 in year 2006–2007 (Thigpen et al. 2011).
Meningococcal serogroup A and C disease increases during the The case fatality rate in patients with pneumococcal menin-
dry season in Africa. The early stages of disease can mimic a viral gitis is equally high in developed and undeveloped regions of the
infection such as influenza, but the disease course can be fulmi- world (Molyneux et al. 2006). In the youngest children, annual
nant. Thus, it can be difficult to identify and treat meningococcal global case fatality for the year 2000 was estimated to be 59 %,
disease quickly. Rapid progression from bacteremia and/or men- ranging from 29 % in the western Pacific to 73 % in Africa
ingitis to life-threatening septic shock can occur within the first (O’Brien et al. 2009). In the United States, the mortality rate
few hours after initial symptoms appear. Because of these factors, in all patients with pneumococcal meningitis was 17.9 %
vaccination is generally the best preventive option for controlling in 1998–1999 and 14.7 % in 2006–2007, hence not changing
this disease. Although significant progress has been made in significantly after vaccine introduction (Thigpen et al. 2011).
understanding meningococcal pathogenesis, and effective Moreover, during 2003–2007, according to the Emerging
meningococcal vaccines along with strategies for vaccination Infections Programs network in the United States, the case
Meningitis 18 405

fatality rate in pediatric patients was 9.4 % and 17.5 % in the (laminin-binding protein), FbsA (fibrinogen-binding protein),
adult population (Thigpen et al. 2011). The most common pili, and IagA (via lipoteichoic acid anchoring) (Stephens et al.
cause of death in patients with pneumococcal meningitis was 2007; Orihuela et al. 2009), but whether these structures are
cardiorespiratory failure, stroke, status epilepticus, or brain unique to meningitis pathogens is unclear. In N. meningitidis,
herniation (Brouwer et al. 2010). the outer membrane protein Opc binds to fibronectin, thereby
Sequelae in survivors of pneumococcal meningitis are anchoring the bacteria to the integrin a5b1 receptor on the cell
present in up to 50 % of cases (Weisfelt et al. 2006; Edmond surface (Orihuela et al. 2009). In addition, N. meningitidis pili
et al. 2010; Jit 2010). In a meta-analysis of 48 studies of bind to CD46 on HBMEC (Johansson et al. 2003), and
pneumococcal meningitis in affluent populations, the pooled lipooligosaccharides contribute to a high degree of bacteremia
prevalence of individual sequelae was 31.7 % (Jit 2010). and subsequent penetration into the CNS (Plant et al. 2006).
The pooled prevalence of hearing loss, seizures, hydrocephalus, CD46 is also a receptor for measles, adenovirus, and human
spasticity/paresis, cranial palsies, and visual impairment was herpesvirus 6 (Manchester et al. 2000; Gaggar et al. 2003;
20.9 %, 6.5 %, 6.8 %, 8.7 %, 12.2 %, and 2.4 %, respectively. Santoro et al. 2003). S. pneumoniae crosses the blood–brain
However, cerebral infarction was found in one study in 36 % of barrier partly through interaction between cell-wall phosphor-
adult patients with pneumococcal meningitis (Schut et al. 2012). ylcholine and the platelet-activating factor receptor (PAFR), as
shown by partial inhibition of pneumococcal invasion of
HBMEC by a PAFR antagonist (Cundell et al. 1995) and delayed
Pathogenesis of Meningococcal translocation of pneumococci from the lung to the blood
and Pneumococcal Meningitis and from the blood to the CSF in PAFR-knockout mice
(Radin et al. 2005).
Experimental animal models indicate that E. coli and Of note, the mechanisms involved in microbial invasion of
group B streptococcus initially penetrate the brain via the the blood–brain barrier differ from those involved in the release
cerebral vasculature (Ferrieri et al. 1980). The blood–brain of cytokines and chemokines in response to meningitis-causing
barrier is a structural and functional barrier formed by brain pathogens. For example, interleukin-8 is secreted from HBMEC
microvascular endothelial cells (Rubin and Staddon 1999; infected with E. coli K1, but infected non-brain endothelial cells
Amiry-Moghaddam et al. 2004; Davidsen et al. 2007) that (i.e., human umbilical vein endothelial cells) do not secrete IL-8,
protects the brain from microbes and toxins in the and IL-8 secretion does not involve the same E. coli proteins that
blood. However, meningitis-causing pathogens, including mediate invasion of HBMEC (Galanakis et al. 2006). In addi-
N. meningitidis and S. pneumoniae, cross the blood–brain tion, c-Jun kinases 1 and 2 promote HBMEC invasion by
barrier as live bacteria (Ring et al. 1998; Hsu et al. 2009). N. meningitidis, and in this context, the p38 mitogen-activated
Meningitis-causing pathogens cross the blood–brain barrier protein kinase (MAPK) pathway promotes release of interleu-
transcellularly, paracellularly, or by means of infected kins 6 and 8. These findings suggest that different proteins and
phagocytes (the ‘‘Trojan horse’’ mechanism) (Kim 2009; distinct mechanisms mediate penetration of host cells into the
Coureuil et al. 2012). For most meningitis-causing pathogens, CNS and the inflammatory process, leading to meningitis.
including E. coli, group B streptococcus, and S. pneumoniae,
transcellular traversal of the blood–brain barrier in infants and
children (Ring et al. 1998; Hsu et al. 2009) is mediated Meningococcal and Pneumococcal Colonization
by physical interaction with a host cell surface receptor and Carriage
(Unkmeir et al. 2002; Kim 2009). For example, meningococcal
and pneumococcal organisms bind to and invade human brain Meningococci and pneumococci are commensal pathogens
microvascular endothelial cells (HBMEC) (Chudwin et al. 1983; (Yazdankhah and Caugant 2004), and even though they cause
Doulet et al. 2006; Banerjee et al. 2010). The bacterial Opa invasive diseases such as septicemia and meningitis, they also
and neuraminidase NanA proteins interact with CD48 and colonize the upper respiratory tract in 60–70 % of healthy
endoplasmin on HBMEC (Cundell et al. 1995). Invasion of children who attend day-care centers and in approximately
HBMEC by meningococcal organisms also occurs through 10 % of the general population. In children under 4 years of
other host–pathogen receptors such as the laminin receptor age, the carriage rate of meningococci is <5 %, progressively
(Huang and Jong 2009; Orihuela et al. 2009), a cell surface increasing to a maximum of 20–25 % in the second and third
membrane receptor for the adhesive basement membrane decades of life. The pathogens colonize the nasopharyngeal
protein laminin. Ribosomal protein SA is also a cell mucosa, subsequently spreading to the lower respiratory tract,
surface ligand for various CNS-infecting microorganisms, where an acute inflammatory response is evoked, and clinical
including N. meningitidis, S. pneumoniae, H. influenza type b, symptoms ensue. Meningococci have also been detected in
dengue virus, adeno-associated virus, Venezuelan equine tonsillar tissues in up to 45 % of patients hospitalized for
encephalitis virus, and prion protein (Orihuela et al. 2009). tonsillectomy (Sim et al. 2000).
It is not currently understood how the same receptor promotes Asymptomatic individuals harboring these pathogens in the
penetration of different microorganisms into the CNS. upper respiratory are considered to be carriers for invasive
Meningitis-causing pathogens binding to HBMEC via Lmb meningococcal and pneumococcal disease with subsequent
406 18 Meningitis

transmission occurring largely through respiratory droplets and recombination. Nevertheless, carriage studies suggest that most
secretions. The size of the inoculum required for transmission individuals are colonized with a single meningococcal strain,
from one host to another is not known. Individuals harboring N. a fact that constrains the opportunity for genetic exchange
meningitidis and S. pneumoniae in the upper respiratory tract between heterologous strains and emphasizes genetic variation
display pathology of variable severity, ranging from local inflam- arising by spontaneous mutations and recombination/gene
mation to invasion of mucosal surfaces, fulminant sepsis, or conversion within a single strain. Meningococcal adhesins in
focal infection (Apicella 2005). Meningococcal and pneumococ- other commensal Neisseria sp. have not been well studied.
cal disease usually occurs 1–14 days after acquisition of the Thus, it is not clear how allelic meningococcal diversity
pathogen (van Deuren et al. 2000). However, in some cases, occurs with such frequency. One possibility is that it reflects
the carrier state can persist for months or even years. intragenomic recombination in combination with strong
The relationship between meningococcal and pneumococcal selection of events providing improved fitness. Alternatively,
carriage and meningococcal and pneumococcal disease has been we may be vastly underestimating the number of carriers who
studied to some extent, and carriage prevalence has even been are colonized by multiple distinct strains of meningococci and
used as a proxy for predicting outbreaks of meningococcal and pneumococci or similarly underestimate the size and diversity of
pneumococcal disease. For example, carriage of pneumococci the genetic pool available to meningococci and pneumococci in
appears to be a risk factor for meningococcal carriage (Ridda the nasopharynx.
et al. 2010). The important parameter is the rate of acquisition of Meningococcal and pneumococcal carriage and transmission,
hypervirulent meningococci or pneumococci, not the overall not disease, determine the global variation and composition of the
meningococcal and pneumococcal carriage. The probability of natural population of these bacterial entities. Conjugate vaccines
progression to meningococcal or pneumococcal disease declines against a variety of encapsulated bacteria including serogroup
very sharply 10–14 days after acquisition of the pathogen. C meningococci and multiple serotypes of pneumococci have
The extent to which N. meningitidis and S. pneumoniae interact been a powerful tool towards preventing or reducing the number
with other commensals/pathogens that reside in the upper respi- of outbreaks of meningococcal and pneumococcal disease,
ratory tract is an important area for future study. Meningococcal providing a compelling rationale for continued study of the
carriage and its consequences are understood in the context of biology of the commensal behavior of meningococci. From an
a dynamic model. Cross-sectional studies of the microbiome in evolutionary perspective, the interactions between meningococci
meningococcal carriers can provide an incomplete ‘‘snapshot’’ and pneumococci and the nonpathogenic flora in the human
of the coexisting flora that colonize the nasopharyngeal mucosa, nasopharynx are key denominators.
especially if some flora localize primarily to intracellular or Meningococcal Adhesion and Cell Invasion. Adhesion to
submucosal tissue in the nasopharynx. With regard to the poly- human mucosal surfaces is essential for meningococcal survival,
saccharide capsule, its role during carriage/transmission is and adhesins are the bacterial proteins that mediate binding to
not well understood. Capsule-deficient strains are carried and cell surface receptors on target host cells. Furthermore, adhesin
transmitted efficiently, and the ideas that the capsule increases redundancy is a hallmark of the meningococcus. Recognized
resistance to desiccation during transit or that it reduces adhe- adhesins include pili, PilC, PilQ, Opa, Opc, LOS, factor
siveness are not well supported. It seems likely that the ability to H-binding protein, PorA, HrpA, PorB, and NadA (Merz and So
switch between capsulate and non-capsulate forms confers 2000; Hill and Virji 2012). Proposed or demonstrated receptors
adaptive and/or fitness advantage, possibly by increasing include platelet-activating factor, CD46, CEACAM1, vitronectin
capacity for cell invasion. In this regard, it has been proposed and a-actinin/integrins, complement receptor 3, laminin, and
that propensity for carriage differs in strains expressing different the GP96 scavenger receptor (Hill and Virji 2012).
capsular polysaccharides (i.e., according to serogroups). Initial contact of meningococci with nasopharyngeal epithelial
Meningococcal carriage induces bactericidal antibodies cells is mediated by type IV pili, the receptor for which may be the
within 1–2 weeks after colonization that persist for several I-domain of integrin a-chains or possibly CD46 (Bourdoulous
months. Bactericidal antibodies to N. lactamica cross-react and Nassif 2006; Doulet et al. 2006). Meningococci proceed to
with antigens from various meningococcal serogroups and proliferate on the surface of human nonciliated epithelial cells,
serotypes. As carriage of N. lactamica is approximately 4 % by forming small microcolonies at the site of initial attachment.
3 months of age and peaks at 21 % by 18–24 months of age, this Capsule blocks close adhesins other than pili and thus may
is much higher than carriage of N. meningitidis at this age aid meningococcal transmission from mucosal surfaces.
(Yazdankhah and Caugant 2004). N. lactamica can protect Attachment can activate two-component regulatory systems,
against meningococcal disease. Development of invasive menin- leading to loss or downregulation of capsule. Close adherence
gococcal disease correlates with the absence of bactericidal of meningococci to the host epithelial cells results in the
antibodies (Goldschneider et al. 1969). formation of cortical plaques and leads to the recruitment
Meningococci express multiple adhesins (i.e., pilus, Opa, of factors ultimately responsible for the formation and extension
NadA) characterized by an impressive and high degree of allelic of epithelial cell pseudopodia that engulf the meningococcus
variation, a property that likely reflects their capacity to import (Doulet et al. 2006). Intimate association is mediated by the
and incorporate genetic information laterally via genetic bacterial opacity proteins, Opa and Opc with CD66/CEACAMs
transformation in an addition to extensive intragenomic and integrins, respectively, on the surface of the epithelial
Meningitis 18 407

. Fig. 18.2
(a) Petechiae on a man with mild systemic meningococcemia caused by Neisseria meningitidis serogroup B. (b) Large petechiae and small
ecchymoses in a patient with bacteriologically confirmed N. meningitidis infection. (c) An 18-month-old boy with lethal cardiovascular
collapse caused by N. meningitidis serogroup B (Petter Brandtzæg)

cell and is one trigger of meningococcal internalization indicated that high amounts of capsular polysaccharide prevent
(Gray-Owen and Blumberg 2006). However, the roles of attachment to host cells, probably by masking underlying
meningococcal adhesins NadA and LOS are less well defined virulence determinants. Interestingly, the amount of capsule
(Doulet et al. 2006). In this complex process, large molecular is substantially reduced upon contact with epithelial cells
complexes involving the molecular linkers ezrin and moesin (Hammerschmidt et al. 2005). In addition, the virulence factor
(known as ERM [ezrin–radixin–moesin] proteins) cluster with pneumolysin plays a significant role in pathogenesis (Paterson
integral membrane proteins, including CD44 and intracellular and Mitchell 2006). The pneumococcal outer cell wall is
adhesion molecule (ICAM) 1, followed by formation of cortical composed of peptidoglycan, teichoic acid, and lipoteichoic
actin polymers (Hoffmann et al. 2001; Lambotin et al. 2005), acid, which differ only in their attachment to the pneumococcal
which ultimately lead to cortical plaques and cell membrane cell wall, as well as phosphorylcholine. Phosphorylcholine is not
protrusions. The latter step requires phosphorylated cortactin. only targeted by the choline-binding domain of choline-binding
Consistent with this, some meningococcal mutants that lack proteins but functions itself as an adhesin by recognizing
functional LOS demonstrate reduced invasiveness and aberrant the platelet-activating factor receptor of host cells (Cundell
actin polymers/polymerization and fail to recruit and/or et al. 1995). Through genome mining, it has been predicted
phosphorylate cortactin (Hoffmann et al. 2001). that S. pneumoniae has approximately 200 proteins with
The next steps of a meningococcal infection include a leader peptide (Bergmann and Hammerschmidt 2006).
internalization, intracellular survival, transcytosis through the The leader peptide is recognized by complex secretion machin-
basolateral tissues, and dissemination into the bloodstream; eries known as translocons and is required for protein traversal
these processes are not yet thoroughly studied or understood. across the membranes. SecA is the main factor of the general
Intracellular meningococci reside within a membranous vacuole secretory pathway, and the ATPase activity of this protein is the
and are capable of translocating through the epithelial layers molecular motor of protein translocation across the mem-
within 18–40 h after internalization. Intracellular survival branes. Three clusters of pneumococcal surface proteins can be
requires IgA1 protease, which degrades lysosome-associated distinguished by genome analysis: lipoproteins, the choline-
membrane proteins (LAMPs), thus preventing phagosomal binding protein family, and proteins with lipoteichoic acid
maturation. IgA1 protease induces a dose-dependent T-cell motifs that are covalently anchored in the cell wall after cleavage
response, which is mainly a Th1-based proinflammatory by a transpeptidase, which is a sortase. Bioinformatics analysis of
immune response (Tsirpouchtsidis et al. 2002). Meningococci the pneumococcal genomes also indicates the presence of
can replicate intracellularly, by a process that requires incomplete biosynthetic pathways, which is consistent with the
sequestration and utilization of cellular iron through specialized inability of this pathogen to carry out respiratory metabolism,
transport systems. This process involves host factors such as the and also explains the high number of ATP-binding cassette
hemoglobin-binding receptor (HmbR), transferrin-binding (ABC) transporters produced by S. pneumoniae. In addition to
protein (TbpAB), and lactoferrin-binding protein (LbpAB) these predicted surface proteins, nonclassical surface proteins
(Perkins-Balding et al. 2004) (> Fig. 18.2). that lack a classical leader peptide and membrane-anchoring
Pneumococcal Adhesion and Cell Invasion. Pneumococci are motifs have been identified on the pneumococcal surface,
encased by a capsular polysaccharide which has been recognized contributing to the virulence of pneumococci and other
as a sine qua non of virulence. However, several studies have pathogenic bacteria.
408 18 Meningitis

Invasive Meningococcal and Pneumococcal meningococci are covered with a loosely adherent capsular-like
Disease structure containing high-molecular-weight polyphosphate.
This layer protects against environmental stress (Zhang
Invasive Meningococcal Disease and Meningitis. Once infection is et al. 2010).
established, organisms enter the bloodstream, cause bacteremia, Pili. Pili are filamentous hairlike fibers consisting of
cross the blood–brain barrier, and, ultimately, lead to meningi- thousands of protein subunits (pilin, 16–20 kDa) (Tonjum and
tis. In some instances, untreated meningococcal infections Koomey 1997). Meningococci express long (up to 4,300 nm in
progress rapidly, leading to death within 12–24 h. As discussed length) type IV pili that protrude from the bacterial surface
above, the prognosis after a non-immunized individual is (> Fig. 18.1). Type IV pili confer bacterial cell-to-cell interac-
infected is highly variable, ranging from healthy colonization tions and twitching motility—a form of locomotion that
to serious or fatal clinical disease, and the exact outcome requires extension and retraction of the pilus filament
depends on characteristics of both the infectious agent and (Henrichsen 1983). Pili are essential for adhesion to epithelial
the host. Some of the most important factors that influence and endothelial cells adherence of bacteria to human cells and
disease outcome are discussed below. for DNA transformation (Swanson 1973; Stephens and McGee
1981) and impart tissue tropism (Meyer et al. 1994; Merz and So
2000). Expression of type IV pili is also required for efficient
Meningococcal Cell Structure and Virulence DNA uptake in transformation (Jyssum and Lie 1965; Sparling
Factors 1966; Davidsen and Tonjum 2006; Hamilton and Dillard 2006).
Pili as well as capsule and PorA are expressed during human
N. meningitidis, like other Gram-negative bacteria, has a cell wall infection as documented by skin biopsies (Harrison et al. 2002).
that consists of two membranes separated by a thin peptidogly- Several proteins required for the assembly, extrusion, and
can layer. The inner cytoplasmic membrane consists of proteins retraction of meningococcal type IV pili have been identified
embedded in a phospholipid bilayer that is impermeable to (Carbonnelle et al. 2006). These include PilE (Parge et al. 1995),
hydrophilic compounds. The outer membrane is an asymmet- ComP (Wolfgang et al. 1999), PilQ (Tonjum et al. 1998; Collins
rical bilayer composed of phospholipids in the inner leaflet and et al. 2004; Assalkhou et al. 2007), lipoproteins PilP
lipooligosaccharide (LOS) in the outer leaflet. The LOS renders (Balasingham et al. 2007) and PilW (Trindade et al. 2008), the
the outer membrane relatively resistant to detergents and is prepilin peptidase PilD (Strom et al. 1993), the ATPase PilT
semipermeable due to the presence of protein channels, called (driving pilus retraction) (Wolfgang et al. 1998; Forest et al.
porins. Other surface-exposed outer membrane proteins and 2004), and the adhesin PilC (Rudel et al. 1995).
extracellular appendages such as capsular structures and type The main structural constituent of the type IV pilus fiber is
IV pili particularly contribute to neisserial survival and virulence pilin subunit, PilE. During infection, pilins undergo rapid phase
(Meyer et al. 1994; Merz and So 2000). The neisserial outer shifts and antigenic variation. PilE is encoded by a single gene;
membrane continuously sheds vesicles (blebs) that contain however, expression of the PilE gene requires unidirectional
DNA, protein/peptides, and high levels of LOS. donation of coding sequences from multiple silent partial
Capsules. Neisseria meningitidis produces a polysaccharide pilS genes in a process similar to gene conversion. During
capsule (text box). On the basis of structural differences in this process, an extensive repertoire of antigenic variants of
capsule, meningococci are divided into at least 13 serogroups PilE is generated (Tonjum and Koomey 1997). The frequency
(A, B, C, D, 29E, H, I, K, L, W-135, X, Y, and Z). Serogroups A, B, of antigenic pili variation can be as high as 10 3. Pilin is
C, Y, and W-135 cause more than 90 % of meningococcal also posttranslationally modified with phosphorylcholine,
disease. Capsular types are normally stable, but strains can phosphoethanolamine, and variable acetylated O-linked glycans
acquire variant alleles of capsule gene (Vogel et al. 2000). (Power and Jennings 2003; Aas et al. 2006). N. meningitidis
For example, serogroup B can switch to C and vice versa. expresses class I or class II pili, which are antigenically and
The serogroup A capsule contains N-acetyl-mannosamine-1- structurally distinct. Class II pili are encoded by a different pilE
phosphate. The capsules of serogroups B, C, Y, and W-135 gene that has no silent cassette counterparts.
consist of polymers of N-acetylneuraminic (sialic) acid. Surface Proteins. The repertoire of the meningococcal surface
The B-polysaccharide resembles structures present in human proteins is substantial (Meyer et al. 1994; Merz and So 2000).
neural tissues, limiting its immunogenicity and vaccine Trimeric protein channels (porins) transport low-molecular-
potential. The carbohydrates can be variably O-acetylated. weight nutrients across the outer membrane. N. meningitidis
The capsule polymers are anchored in the outer membrane can express two types of porins simultaneously: PorA and
through a 1,2-dipalmitoyl glycerol moiety. Capsule biosynthesis PorB. PorA (class 1 protein, 44–47 kDa) is variably expressed,
can vary and is subject to regulation. Isolates from healthy and in some patients, the level of expression is below the
carriers are frequently unencapsulated due to lack of capsule detection limit, implying that the gene is highly repressed,
gene expression. A substantial proportion of meningococcal inactivated by mutation, or that the protein is expressed
isolates from carriers carry inactivating mutations in or but sequestered and/or modified. Antigenic differences in PorA
deletions of capsule genes. Isolates from the bloodstream or (and PorB) are used to classify meningococci into serological
CSF are invariably encapsulated. In addition to capsule, subtypes. Meningococcal PorB is equivalent to the gonococcal
Meningitis 18 409

PorB protein and is represented by one of two isoforms, PorB-IA The core oligosaccharide of neisserial LOS is divided into
(class 2 protein, 40–42 kDa) or PorB-IB (class 3 protein, an inner and outer core region. The composition of the
37–39 kDa) (Meyer et al. 1994; Merz and So 2000). inner core is heterogeneous due to variable substitutions
Meningococci fail to survive unless they express PorA or PorB. (phosphoethanolamine, glycine, glucose, O-acetyl groups)
The neisserial RmpM protein (formerly protein III or class 4 (Kahler et al. 2005), which occur in response to environmental
protein) forms a complex with and likely stabilizes outer cues. The outer core is also variable and undergoes
membrane protein complexes including porins. The protein is high-frequency phase and antigenic variation due to frequent
stably expressed by gonococci and meningococci. Its C-terminal targeted mutagenesis during replication of LOS biosynthesis
periplasmic region resembles the analogous protein domain in genes as well as horizontal gene transfer (Kahler and Stephens
E. coli OmpA, whose role involves binding peptidoglycan. 1998). A single strain can simultaneously express up to six
RmpM-specific antibodies interfere with the bactericidal related LOS. The terminal structure of neisserial LOS is sialylated
activity of antibodies against other surface antigens, thereby by bacterial sialyltransferase. Gonococci modify LOS using host
increasing the risk of infection (Plummer et al. 1993). sialic acid (CMP-NeuNAc). In contrast, meningococci use an
Meningococci express opacity (Opa) proteins (20–28 kDa) endogenous source of CMP-NeuNAc. The terminal LOS of
(Meyer et al. 1994; Merz and So 2000). Opa proteins are the pathogenic Neisseria spp. often shares epitopes with host
structurally similar to one another but display considerable glycolipids (Kahler and Stephens 1998). In this manner,
intra- and interstrain variation in surface-exposed regions molecular mimicry is exploited by the pathogens, which gain
and in level of expression. Intrastrain antigenic variation arises the ability to bind to host cell lectin receptors. This limits the
by intragenomic recombination and horizontal gene transfer, usefulness of LOS as a vaccine target.
and variants are subject to selective pressure during infection. Meningococcal LOS initially react with CD14 and
High-frequency phase variation is due to translational subsequently with TLR4 receptor which is critical to the
frame-shifting involving a pentameric repeat in the opa genes. innate immune responses to bacterial endotoxins including
By this mechanism, Opa proteins are switched on and off, meningococcal LOS (Akira and Takeda 2004). Activation of
independent of one another, enabling simultaneous expression TLR4 by endotoxin requires association with the accessory
of multiple proteins (Meyer et al. 1994). The meningococcal protein MD-2, an N-glycosylated (Viriyakosol et al. 2001)
genome contains 3–4 opa genes. Opa proteins play an important 19–27-kDa protein that is expressed in both a soluble and
role in promoting adherence to and invasion of eukaryotic cells. a membrane-bound form. Binding of endotoxin LOS to MD-2
Because Opa proteins are subject to a high degree of phase in association with TLR4 can lead to dimerization or oligomer-
and antigenic variation, they have limited usefulness as ization of TLR4 receptors and subsequent cellular activation.
targets for vaccine-based interventions. Approximately 70 % of MD-2 directly interacts with lipid A of meningococcal
meningococcal strains express the Opc protein. This protein also endotoxin.
confers colonial opacity and is functionally similar to Opa. Peptidoglycan. Neisserial peptidoglycan consists of long
In addition to the aforementioned major outer membrane chains of repeated disaccharide units cross-linked via peptide
proteins, meningococci express >80 other outer membrane bridges. In E. coli, peptidoglycans are found covalently linked to
proteins. Among these, the pilus-related secretin complex PilQ lipoproteins, but this is not thought to occur in meningococci.
is one of the most abundant, representing approximately 10 % of Peptidoglycan metabolism involves both lytic and synthetic
the total mass of the outer membrane protein fraction (Tonjum enzymes. Initial synthesis is carried out using four penicillin-
et al. 1998). The level of expression of PilQ varies under different binding proteins (PBPs) (Dillard and Hackett 2005), followed by
growth conditions. Iron-regulated proteins are also expressed on O-acetylation, which protects against autolysis by endogenous
the surface of meningococci in vivo. Among these, transferrin- lytic transglycosylates and host lysozymes. After release,
binding proteins (Tbp-1 and Tbp-2) and the lactoferrin-binding peptidoglycan fragments activate the innate immune response
proteins (Lbp) facilitate transport and internalization of iron, an through the intracellular NOD1 and NOD2 receptors and
essential nutrient for sustained infection. Additional conserved contribute to inflammatory response.
proteins that provide exposed antigenic targets on the bacterial Secreted Factors. The meningococcal genome is predicted to
cell surface include OMP85, NspA, NadA, GNA1870 encode autotransporter, two-partner, and type I and type II
(factor H-binding protein), and GNA2132 (hypothetical secretion mechanisms (van Ulsen and Tommassen 2006).
lipoprotein); these are considered to be viable targets for vaccine The pathogenic Neisseria spp. secrete immunoglobulin
development. A1 (IgA1) protease. This serine protease directs its own
Lipooligosaccharide. Approximately 50 % of the neisserial transport across the outer membrane into the environment.
surface is covered by lipid-anchored oligosaccharide (LOS). The enzyme cleaves IgA1 in the hinge region, separating
LOS lack repeating carbohydrate units (O-chain) of enterobac- Fab and Fc; this inactivates IgA function. IgA protease also
terial lipopolysaccharide (LPS). Neisserial LOS is composed of cleaves other proteins such as endosomal Lamp1, important
hexa-acylated lipid A, two KDO molecules, and one or more for intracellular vesicle trafficking. The functions of other
carbohydrate chains of 8–12 saccharide units, the core oligosac- secreted proteins including the filamentous hemagglutinin
charide. The lipid A anchors LOS in the outer membrane and is (FHA)-like protein TpsA and FrpA/C are largely unknown.
one of the most potent bacterial endotoxins. A subset (80 %) of meningococcal and gonococcal strains
410 18 Meningitis

secrete DNA via a type IV secretion system Most isolates of N. gonorrhoeae, but not of N. meningitidis,
(Hamilton et al. 2005). The genes encoding this system are carry plasmids (Roberts 1989). Nearly all gonococcal strains
located on the gonococcal genetic island, a DNA region that carry a 4.2-kb cryptic plasmid of unknown function, and many
is acquired by horizontal gene transfer. Unlike many other strains carry plasmids encoding b-lactamase, which confers
bacterial pathogens, Neisseria spp. lack a type III secretion resistance to penicillin. The conjugative plasmid TetM
mechanism. confers tetracycline resistance.
Genome-based phylogenetic reconstruction indicates that
pathogenic N. meningitidis emerged from a common
Meningococcal Genome Characteristics unencapsulated ancestor by acquisition of capsule genes has
and Dynamics several hundred years ago, probably from members of the family
Pasteurellaceae (Schoen et al. 2008).
Neisserial chromosomes are 2.2–2.3 Mb in length with an
average G+C content of 48–56 mol%. Approximately 95 %
of the genic material, excluding intergenic regions, is Pathogenesis
shared between N. meningitidis and N. gonorrhoeae (Claus
et al. 2007). The vast majority of genes are also present in Pathogenesis of Meningococcal Meningitis
nonpathogenic N. lactamica, but the same gene may be
differentially regulated in pathogenic and commensal strains. Humans are the only host for the meningococcus and the
The meningococcal and gonococcal genomes are considered to pneumococcus. N. meningitidis frequently colonizes the
be hyperdynamic (Davidsen and Tonjum 2006), and the high human pharynx as well as buccal mucosa, rectum, urethra,
level of genomic plasticity/instability is thought to contribute to urogenital tract, and dental plaque. The most common natural
pathogenicity and development of hypervirulence. The most habitat of the meningococcus is the epithelial cells of the naso-
important sources of neisserial genome instability are: and posterior pharynx and the tonsils. N. meningitidis is carried
in the pharynx by 4–25 % of the human population. Thus, there
1. Phase variation, reflecting slip mispairing in homopolymer
are hundreds of millions of carriers worldwide, and adolescents
nucleotide runs at or near the promoter or in open reading
are a principal reservoir (Rosenstein et al. 2001).
frames (affect translation)
Invasive strains of meningococci express capsules, and
2. Recombination, integration, or rearrangement of DNA from
pathological meningococcal strains were originally distinguished
external or internal sources
from nonpathological strains by differences in capsular polysac-
3. Horizontal gene transfer via uptake of exogenous or
charide structure (Kim 2003). Virulence determinants include
‘‘foreign’’ DNA, with subsequent RecA-mediated integration
the polysaccharide capsule, outer membrane proteins
into homologous region of the genome
including pili, the porins (PorA and PorB), the adhesion
4. Hypermutation due to error-prone DNA repair, replication
molecule, Opc, iron sequestration mechanisms, and endotoxin
infidelity, or overexpression of error-prone translesion DNA
(lipooligosaccharide) (de Louvois et al. 2005).
polymerases
N. meningitidis is classified into 13 serogroups based on the
Meningococci are naturally competent for DNA uptake immunogenicity and structure of the polysaccharide capsule.
throughout their growth cycle (Jyssum and Lie 1965; Sparling Further classification into serosubtype, serotype, and
1966; Hamilton and Dillard 2006; Ambur et al. 2009). immunotype is based on class 1 outer membrane proteins
The pathogenic Neisseria spp. share several genomic regions (PorA), class 2 or 3 (PorB) outer membrane proteins, and
including up to nine prophage and eight genetic islands that lipopoly[oligo]saccharide structure, respectively (Chang et al.
are absent from N. lactamica (Snyder et al. 2005). In contrast to 2004; Jolley et al. 2012). PorA is an important target for
many other bacterial species, there are no classical pathogenicity bactericidal antibodies. In addition to these specific virulence
islands in N. meningitidis. N. meningitidis-specific DNA factors, N. meningitidis has evolved and exploits genetic
sequences include the cps locus encoding the polysaccharide mechanisms that result in high-frequency phase and antigenic
capsule, genes that encode the RTX family of toxins, and an variation and molecular mimicry. Capsule switching, due to
ortholog of the filamentous hemagglutinin of Bordetella allelic exchange of capsule biosynthesis genes by transformation,
pertussis. The genomes of disease and carriage isolates show no is one mechanism by which meningococci evade immune
consistent differences. Certain hypervirulent lineages contain detection (Fothergill and Wright 1933).
the filamentous prophage Nf1 (Bille et al. 2005). N. meningitidis Infection by N. meningitidis commonly develops in asymp-
strains of serogroups W-135, H, and Z contain the ‘‘gonococcal tomatic individuals carrying bacteria in the oronasopharyngeal
genetic island’’ (GGI, 57 kb) (Snyder and Saunders 2006). This is cavity (Yazdankhah and Caugant 2004). Type IV pili facilitate
an often chromosomally integrated conjugative plasmid that initial adherence and opacity-associated proteins (Opa and
encodes a type IV secretion system involved in DNA secretion. Opc) and PorB trigger uptake of the bacteria into the cells,
The genome of Neisseria spp. has a variable number of largely by similar types of receptors (CEACAM, HSPG). Opa
noncoding repeat arrays and insertion (IS) elements among variants are found in hyperinvasive meningococcal lineages. Opa
which IS1655 appears to be unique to N. meningitidis. and Opc bind to heparin and interact with vitronectin and
Meningitis 18 411

fibronectin, promoting transport into cells via integrin receptors Attracted polymorphonuclear cells aggravate the inflammatory
(Duensing et al. 1999). This is accompanied by response and release cytotoxic mediators.
a downregulation of pili and capsule enabling optimal contact Immunity to invasive meningococcal disease depends upon
between bacterial adhesins and the host mucosa. Transferrin the presence of bactericidal IgG antibodies directed against
(TbpA, TbpB) and hemoglobin (Hbp) bind and sequester iron capsule (except serogroup B), PorA, PorB, Opa, LOS,
to support bacterial growth (Perkins-Balding et al. 2004). iron-regulated proteins, and minor surface proteins. Carriage
Ciliated mucosal cells can be damaged by released peptidogly- of nonpathogenic Neisseria spp. (e.g., N. lactamica) in the
can fragments and LOS. However, the oropharyngeal nasopharynx elicits cross-reactive antibodies that contribute to
region has a relatively tolerance for foreign matter and is rela- development of immunity against N. meningitidis. In vivo phase
tively refractory to the typical inflammatory response charac- and antigen variation of meningococcal surface antigens indi-
teristic of more sterile anatomical niches such as the urethra. cate selective immunological pressure during natural infection.
This may explain why meningococcal (and gonococcal)
colonization of the oropharynx is rarely associated with
clinical disease. Pathogenesis of Pneumococcal Meningitis
The few phylogenetic groups of N. meningitidis that cause
meningococcal disease often carry a filamentous prophage in The mechanisms underlying pneumococcal meningitis are not
their genome that is secreted from the bacteria via the type IV fully understood, but involve bacterial, host, as well as environ-
pilin secretin (Bille et al. 2005). The prophage may promote the mental factors. The route from the initial site of infection to the
development of new epidemic clones. The mechanism by which meninges is believed to occur via a bacteremic phase and
meningococci penetrate and pass through the mucosa is only subsequent traversal of the organism from the circulation across
partially understood (Merz and So 2000). Meningococci survive the blood–brain barrier (BBB) into the subarachnoid space.
and multiply during epithelial cell traversal. The IgA1 protease However, animal experiments suggest direct axonal retrograde
and PorB may promote survival inside epithelial cells. Menin- transport to the brain without detectable bacteremia, in
gococci isolated from the bloodstream invariably produce poly- a process requiring pneumococcal interactions with gangliosides
saccharide capsule. The capsule protects the bacterium from (van Ginkel et al. 2003). There is some epidemiological support
phagocytosis and complement-mediated lysis by preventing for this, in that pneumococcal CSF isolates belong to a multitude
insertion of the terminal complement attack complex. Invasive of serotypes more reflective of the commensal nasopharyngeal
meningococci express sialylated LOS which influences binding flora than to blood isolates (Henriques Normark et al. 2001).
of C4b, while the proteins PorA and GNA1870 recruit the It is believed that pneumococci exploit selective tropism
negative regulators of complement activation C4BP and towards brain endothelial cells to invade into the CSF. The
factor H (Schneider et al. 2007). Individuals with inherited main pathogens causing bacterial meningitis, S. pneumoniae,
deficiencies in the late complement components (C5–C9) have N. meningitidis, and H. influenzae all interact with the laminin
a high risk in developing meningococcal disease. Intriguingly, receptor on rodent as well as HBMECs. In pneumococci, this
they acquire the infection at a much later age and have frequent interaction is mediated by choline-binding protein CbpA
recurrences, and the case fatality rate is much lower than for (PspC) on the pneumococcal surface. The binding site on
normocomplementemic individuals. CbpA for the laminin receptor was localized to a highly
In the blood, N. meningitidis replicates to high levels (up to conserved, surface-exposed loop not involved in other known
108/mL plasma) and sheds outer membrane vesicles (blebs) CbpA–host interactions (Orihuela et al. 2009).
(Stephens and Greenwood 2007). The blebs may subvert the All pneumococcal isolates possess the nanA gene encoding
complement system, and high levels of circulating LOS a cell-wall-anchored neuraminidase that cleaves sialic acid from
overactivate the innate immune system. Circulating levels of host cells and proteins. The lectin moiety of NanA rather than
proinflammatory mediators (TNF-a, IL-1, and IL-6) strongly the sialidase region promotes pneumococcal adherence to and
correlate with development of lethal septic shock (Stephens et al. entry into HBMECs, suggesting that bacterial binding to
2007; Brandtzaeg and van Deuren 2012). sialylated glycoconjugates might represent an early step in the
Meningococci and pneumococci most often enter the CSF BBB translocation process (Uchiyama et al. 2009). In addition,
likely by the hematogenous route via the capillaries and veins in lectin-bound NanA activates brain endothelial chemokine
the subarachnoid space (the blood–CSF barrier) and the choroid expression and recruits neutrophils, promoting a local inflam-
plexi rather than through the brain parenchyma (blood–brain matory response at the site of infection even the absence of
barrier). Encapsulated N. meningitidis invade the CSF probably invasion (Banerjee et al. 2010). This proinflammatory response
via the transcellular route (Nikulin et al. 2006). The absence of of pneumococcal NanA requires unmasking of an inhibitory
non-opsonophagocytosis in CSF initially enables uncontrolled sialic acid-binding receptor on the surface of immune cells
bacterial growth and inflammation of the leptomeninges and (Chang et al. 2012). Other data also suggest that local
subarachnoid space. In the CSF, N. meningitidis produce poly- proinflammatory events that trigger endocytosis and/or locally
saccharide capsule and pili and stimulate proinflammatory cyto- damage HBMECs facilitate transport across the BBB.
kine (Il-6, IL-8, MCP-1) and chemokine (RANTES, GM-CSF) Cell surface phosphorylcholine on pneumococci can bind
production in meningeal cells (Christodoulides et al. 2002). the human platelet-activating factor receptor, activating
412 18 Meningitis

beta-arrestin-mediated uptake of pneumococci into cells within and its receptor (c-Mpl) after intrathecal infection.
the BBB (Radin et al. 2005). Pneumococcal-induced death of Thrombopoietin is known to exhibit proapoptotic effects on
brain endothelial cells can be mediated by cell-wall components neurons, and its upregulation upon pneumococcal infection
via TLR2 signaling and/or via the pore-forming cytotoxin may therefore have neurotoxic effects (Hoffmann et al. 2011).
pneumolysin and the unusually high levels of hydrogen peroxide The high expression of H2O2 is normally attributed to the
produced by this catalase-negative organism (Bermpohl et al. pneumococcal spxB gene encoding a pyruvate oxidase. In vivo
2005). TLR stimulation mediated by pneumococci is sufficient transcriptomic analyses of mouse brain tissue, after induction of
to promote translocation of the organism as well as its inflam- pneumococcal meningitis, revealed an upregulation of the glpO
matory components across the epithelium by downregulation of gene encoding an alpha-glycerophosphate oxidase that was cyto-
genes involved in the formation of tight junctions (Clarke et al. toxic for HBMECs via generation of H2O2. A glpO deletion
2011). It is, however, not known whether innate immune mutant was defective in the progression from the blood to the
activation by pneumococci opens tight junctions in the brain brain during in vivo infection, and mutant bacteria caused
endothelium, even though early work demonstrated that a significantly lower meningeal inflammation and brain pathol-
intracisternal infection in rats by S. pneumoniae serotype 3 caused ogy compared with wild type. Interestingly, GlpO immunization
morphological changes in cerebral endothelium including protected against pneumococcal invasive disease (Mahdi et al.
completely separated cell junctions (Quagliarello et al. 1986). 2012).
Once pneumococci have entered the CSF, they proliferate Even though brain lesions appear to result from local men-
easily and release large quantities of proinflammatory compo- ingeal infection, experimental infection demonstrates that the
nents that are recognized by resident immune cells via cell systemic bacteremic component in pneumococcal meningitis
surface and intracellular pattern recognition receptors such as significantly contributes to clinical disease presentation and the
TLRs. This leads to high production of cytokines and pathophysiology of BBB breakdown and ventricle expansion
chemokines that accumulate in CSF, contributing to inflamma- (Brandt et al. 2008).
tion-mediated brain damage. Several cytokines increase in CSF By comparing host response proteins in the CSF from
from meningitis patients including IL-1beta. In CSF, IL-1beta survivors with non-survivors with pneumococcal invasive
concentration correlates with CSF leukocyte count and clinical disease, it was found that complement C3 levels were fivefold
outcome (Mustafa et al. 1989). IL-1beta activation results from lower in non-survivors, suggesting that C3 depletion in CSF is a
induction of the precursor pro-IL-1beta typically mediated by major factor contributing to death in pneumococcal meningitis.
TLR signaling and inflammasome-controlled activation of Also, transferrin levels in CSF were higher in the group of
caspase 1 cleaving the pro-IL-1beta into active IL-1beta. It has non- survivors suggesting a more extensive damage of the
been demonstrated that the ASC and NLRP3 components of the blood–brain barrier. There were however no differences in the
inflammasome promote inflammatory damage in a murine level of cortical necrosis in the two patient groups as monitored
pneumococcal meningitis model and that pneumococcal by the CSF levels of creatinine kinase BB (Goonetilleke et al.
pneumolysin is the main inducer of IL-1beta expression and 2012). The central role played by the complement system in
inflammasome activation upon pneumococcal challenge protecting the CNS against pneumococcal growth has also
(Hoegen et al. 2011). However, infection of human dendritic been demonstrated in a murine meningitis model. Thus, 24 h
cells revealed that pneumolysin inhibits human dendritic cell after intracisternal infection, bacterial titers in the CNS were
maturation, induction of proinflammatory cytokines, and acti- almost 12- and 20-fold higher in C1q- and C3-deficient mice,
vation of the inflammasome testifying to important differences respectively, than in wild-type mice (Rupprecht et al. 2007).
between human and murine immune cells in their responses to The cell-wall component lipoteichoic acid has been
pneumolysin (Littmann et al. 2009). suggested to be a pattern recognition molecule and inflamma-
Pneumolysin and other cytolysins can also have a direct toxic tory mediator; however, the receptor for this interaction
effect on cortical neurons independent on leukocyte infiltration remains controversial. Interestingly, intrathecal treatment with
and caspase activation as demonstrated in an infant rat antibodies against phosphorylcholine recognizing teichoic and
meningitis model. The damaging effect on cortical neurons lipoteichoic acids decreases neuronal damage in experimental
due to pore-forming cytolysins was shown to be age dependent pneumococcal meningitis.
and more pronounced in 7- as compared to 11-day-old rats
(Reiss et al. 2011). The cytolytic effects of pneumolysin on
brain tissue astroglia were significantly enhanced by reducing Clinical Features
the calcium concentration (Wippel et al. 2011).
The large quantities of H2O2 produced by the catalase- Severity of illness on presentation, infection with antimicrobial-
negative pneumococci besides causing oxidative damage have resistant organisms, and incomplete knowledge of the
also been shown to affect brain tissue in other ways. Thus, H2O2 pathogenesis of meningitis are factors that contribute signifi-
produced by pneumococci contributed to regional hyperemia in cantly to mortality and morbidity associated with bacterial
an experimental meningitis model (Hoffmann et al. 2007). meningitis (Chang et al. 2004; Davidsen et al. 2007). When
Pneumococcal production of H2O2 was also responsible for bacterial meningitis is suspected, immediate action is imperative
the observed transcriptional activation of brain thrombopoietin to establish a definitive diagnosis, and antimicrobial treatment
Meningitis 18 413

must be initiated as soon as possible as a precautionary measure, N. meningitidis has the propensity to invade the meninges
because the mortality rate for untreated bacterial meningitis and will do so in most cases left untreated. Even in the most
approaches 100 %; even with optimal treatment, mortality and fulminant cases of septic shock reaching the hospital in Europe
morbidity remain high. within median 12 h, 59 % had a positive CSF culture. Invasive
However, especially in children, the symptoms and signs meningococcal infections lead to compartmentalized bacterial
depend on the age of the child, the duration of illness, and the proliferation. In patients with clinically distinct meningitis, the
host response to infection (Tonjum et al. 1983). Notably, the levels of meningococci and inflammatory mediators are several
clinical features of bacterial meningitis in infants and children logs higher in the CSF than the blood. Conversely, in patients
can be subtle, variable, nonspecific, or even absent. In infants, presenting with septic shock, the bacterial proliferation and the
they might include fever, hypothermia, lethargy, irritability, poor inflammatory response mainly occur in the circulation with
feeding, vomiting, diarrhea, respiratory distress, seizures, or bacterial load and inflammatory response several logs higher
bulging fontanelles. However, at a certain stage, even infants than the subarachnoid space. Thus, the clinical presentation
develop nuchal rigidity. In older children, clinical features include depends on the velocity of proliferation in the blood. Low-graded
fever, headaches, photophobia, nausea, vomiting, confusion, proliferation leads gradually to meningitis within median 24 h
lethargy, or irritability. Other signs of bacterial meningitis on (van Deuren 2001; Brandtzaeg 2006; Stephens 2007; Brandtzaeg
physical examination include neck and back rigidity, Kernig’s 2012). Bacterial load and level of LOS in the circulation
sign (flexing the hip and extending the knee to elicit pain in the are below the shock (10 endotoxin units/mL) threshold.
back and legs), Brudzinski’s sign (passive flexion of the neck elicits In those developing septic shock, the proliferation is very
flexion of the hips), focal neurological findings, and increased rapid, the bacterial load massive with up to 108/mL, and
intracranial pressure. Signs of meningeal irritation are present in LOS activity as high as 2150 endotoxin unites/mL (van Deuren
75 % of children with bacterial meningitis at the time of 2001; Brandtzaeg 2006; Stephens and Greenwood 2007;
presentation (Levy et al. 1990; Borchsenius et al. 1991). Absence Brandtzaeg 2012).
of meningeal irritation in children with bacterial meningitis is For unknown reasons, patients in the large meningococcal
substantially more common in those younger than 12 months. epidemics in sub-Saharan Africa usually develop meningitis
The constellation of systemic hypertension, bradycardia, without shock and severe DIC, leading to large hemorrhagic
and respiratory depression (Cushing’s triad) is a late sign of skin lesions and thrombosis and subsequent gangrene of
increased intracranial pressure. Neurological sequelae are peripheral extremities.
relatively common in survivors of meningitis, especially in On admission, 60 % of cases have experienced symptoms for
individuals infected by a pneumococcal microorganism less than 24 h and 12–20 % for less than 2 days (Tønjum et al.
(Arditi et al. 1998; Roine et al. 2008). 1986). These symptoms can occur discretely or can blend into
Meningococcal Meningitis. The clinical spectrum of systemic one another during clinical disease progression. The disease
meningococcal disease includes meningitis/meningoencephalitis, usually begins abruptly with headache, meningeal signs includ-
fulminant septic shock, the combination of the two, or mild ing stiffness of the neck, and fever. However, classic signs of
meningococcemia without clinically distinct meningitis. Occasion- meningitis (i.e., confusion, headache, fever, and nuchal rigidity)
ally, the bacteremia leads to localized joint infection, pericarditis, are seen in only about one-half of infected patients. Very young
panophthalmitis, and subchronic or chronic meningococcemia children often have only nonspecific signs including fever,
(Rosenstein et al. 2001; Stephens et al. 2007; Brandtzaeg and abdominal pain, and vomiting. Other signs include reduced
van Deuren 2012). consciousness and photophobia. Mortality approaches 100 %
The most frequent form of meningococcal infection is acute in untreated cases but is around 10 % when appropriate antibi-
pyogenic meningitis due to inflammation of the meninges. otic therapy is instituted. The incidence of neurological sequelae
Based on distinct clinical symptoms of 862 patients in three is low, with hearing deficits, epilepsy, and arthritis most com-
prospective studies with documented meningococcal disease, monly noted. These sequelae are most probably underreported.
37–49 % had meningitis without shock, 13–20 % meningitis In CSF, the number of bacteria is higher than in plasma,
with shock, 10–18 % shock without meningitis, and 18–33 % leading to a large compartmentalized inflammatory response in
mild meningococcemia without meningitis or shock the subarachnoid space, with pronounced increase in the
(Brandtzaeg and van Deuren 2012). Using the same classifica- concentration of endotoxin, tumor necrosis factor-a (TNF-a),
tion of meningococcal patients admitted to a tertiary academic interleukins (IL-1b, IL-6, IL-8, and IL-10), chemokines, and
hospital, the blood culture was positive in 50 % of patients with other mediators. The overall inflammatory response in the
meningitis without shock, 87 % of patients with meningitis systemic vasculature, as indicated by activation of cytokines
and shock, 93 % of patients with shock without clinically dis- and complement, is modest (Turner et al. 1990; Latorre et al.
tinct meningitis, and 77 % of patients without meningitis or 2000; Chang et al. 2004; Dubos et al. 2008). Meningococcemia
shock. Interestingly, the CSF culture was positive in 84 % with can manifest as pink maculopapular petechial eruptions
meningitis without shock, 83 % in those with meningitis and (Stephens et al. 2007). Rapidly progressive infections can be
shock, 59 % in shock without clinically distinct meningitis, and accompanied by in purpuric/petechial or ecchymotic skin
in 47 % in those without clinically distinct meningitis or shock lesions that are hemorrhagic and necrotic. However, skin lesions
(Brandtzaeg 2006). can be atypical, evanescent, or even entirely absent in patients
414 18 Meningitis

. Fig. 18.3
(a) A young woman with sepsis and hemorrhagic skin lesions in the face caused by S. pneumoniae. She also had ecchymosis on the
extremities. (b) Hemorrhagic skin lesions in a man with fulminant S. pneumoniae sepsis. (c) Hemorrhagic skin lesions in the face
of a man with fulminant S. pneumoniae sepsis. He also had similar lesions on the body and extremities (Petter Brandtzæg)

who have blood culture-positive meningococcal sepsis. Fulmi- Laboratory Diagnosis of Meningitis
nant shock can dominate the clinical picture in patients with
acute meningococcal sepsis (Stephens et al. 2007; Brandtzaeg It is of paramount importance to examine the CSF in order to
and van Deuren 2012). Sepsis can progress to disseminated properly diagnose all forms of meningitis. Cerebrospinal fluid
intravascular coagulation (DIC) characterized by increasing (CSF), blood, skin biopsies, nasopharyngeal swabs, and aspirates
petechiae or purpura fulminans, resulting in extensive areas of are relevant specimens for the diagnosis of meningococcal and
tissue destruction secondary to coagulopathy, rapid onset of pneumococcal disease. Synovial fluid, sputum, and conjunctival
hypotension, and adrenal hemorrhage (Waterhouse– swabs can also be cultured, if clinically indicated. Because
Friderichsen syndrome). Gangrenous cases in the extremities meningococci and pneumococci are susceptible to desiccation
can occur due to thrombosis, and death is usually caused by and temperature extremes, specimens should be cultured as
cardiovascular collapse (> Fig. 18.3). soon as possible after collection.
For presumptive diagnosis, specimens are examined by Gram
and acridine orange stain. Gram- and acridine orange-stained
Diagnosis smears are made directly from CSF, if the CSF is cloudy or after
centrifugation when the CSF is clear. The majority of the smears
The clinical diagnosis of meningococcal meningitis begins with will show Gram-negative diplococci inside and outside polymor-
recognition of fever, petechial rash, meningeal signs, and altered phonuclear cells when the CSF bacterial count is >105/mL.
mental status and is confirmed by pleocytosis, Gram stain with or Approximately 25 % of smears will stain positively with Gram
without culture of CSF, or blood or skin lesions. The early diag- stain when the bacterial density in the CSF is <103 mL; on
nosis of meningococcemia is difficult when rash and meningeal average, 60–90 % of CSF specimens that are culture positive
signs are not present. General symptoms of sepsis (leg pains, cold are stain positive. Gram-stained smears combined with culture
hands and feet, abnormal skin color) develop first in patients with from disease-related petechial skin lesions detect meningococci
severe meningococcemia (Borchsenius et al. 1991; Tonjum et al. in 62 % of cases (Stephens and Greenwood 2007).
1983). However, these symptoms are not specific to meningo- Meningococcal capsular polysaccharides are detected directly
coccal and pneumococcal disease. Parents and relatives should in CSF by performing latex agglutination and coagglutination
be instructed to undress and inspect a febrile child, adolescent, with polyclonal antibodies for serogroups A, B, C, Y, and W-135
or young adult for rash, and physicians and health-care (Chanteau et al. 2007). These methods can detect 0.02–0.05 mg of
providers should heed the concern of parents or relatives, if and antigen per mL, with a sensitivity of approximately 50 %, com-
when they describe abrupt or rapid deterioration of a patient. pared to 82–90 % for direct detection of meningococci in CSF and
Meningitis 18 415

blood by NAATs/PCR (Taha and Fox 2007). The latter tests are 1992). In a multicenter pneumococcal meningitis surveillance
also useful for confirming the diagnosis in patients treated with study, latex agglutination was positive in 49 (66 %) of 74 CSF
antibiotic prior to sample collection or who tests negative in all samples that grew S. pneumoniae and in four of 14 CSF samples
prior testing (i.e., Gram stain, antigen test, and culture). that were culture negative. The use of standard or sequential-
Positive Gram stain is observed in approximately 90 % of multiplex PCR has been shown to be useful in identification of
children with pneumococcal meningitis, 80 % of children with infecting pathogens in patients who have previously received
meningococcal meningitis, half of patients with Gram-negative antibiotics or in resource-poor settings (Corless et al. 2001;
bacillary meningitis, and a third of patients with Listeria Schuurman et al. 2004; Saha et al. 2008; Chiba et al. 2009).
meningitis (La Scolea and Dryja 1984). When CSF is first Multiplex real-time PCR or broad-range PCR aimed at the 16S
clarified by Cytospin centrifugation, the fraction of samples ribosomal RNA gene of eubacteria is promising for the detection
that stain positively increases (Shanholtzer et al. 1982). CSF of pathogens from CSF. The detection rate was substantially
cell count and differential, and concentrations of protein and higher with PCR than with cultures in patients who had previ-
glucose often help with differential diagnosis of various forms of ously received antibiotics (Chiba et al. 2009). However, the limit
meningitis. The proportion of polymorphonuclear cells in CSF of detection differs between assays. Real-time PCR has been
from patients who have meningitis ranges from 49 % to 98 % shown to detect as few as two copies of N. meningitidis, S.
(mean of 86 %). The prognosis is poor when patients present pneumoniae, and E. coli, 16 copies of L. monocytogenes, and 28
with low white blood cell count and positive Gram staining copies of group B streptococcus, whereas the sensitivity for
in CSF. CSF culture can be negative in children who receive broad-range 16S ribosomal DNA PCR was about 10–200 organ-
antibiotic treatment before CSF examination. During a course isms per mL CSF (Lu et al. 2000; Schuurman et al. 2004). The
of antibiotic treatment, the CSF leukocyte count, glucose and time needed for the whole process from DNA extraction to the
protein concentration, and antigen tests are abnormal for several end of real-time PCR was 1.5 h (Chiba et al. 2009), an attractive
days, even though bacteria might not be evident on smear or by timeframe for its application in clinical practice. A Gram-stain-
CSF culture. Blood cultures test positive in only 50 % of specific probe-based real-time PCR using 16S ribosomal RNA
the patients with meningococcal and pneumococcal disease. has been shown to allow simultaneous detection and discrimi-
In those who have received antibiotics prior to the collection nation of clinically relevant Gram-positive and Gram-negative
of blood for culture, blood cultures are sterile. A nasopharyngeal bacteria directly from blood samples (Wu et al. 2008), which
swab from young children will provide valuable information in might provide more rapid and accurate diagnosis of bacterial
cases of suspected meningococcal and pneumococcal disease. meningitis. In addition, sequential PCR-based serotyping of S.
For isolation of N. meningitidis, the clinical specimen should pneumoniae using serotype-specific primers could improve
be inoculated on selective and nonselective growth media ascertainment of pneumococcal serotype distribution in settings
(Tonjum 2005). Appropriate nonselective media are 5 % sheep in which prior use of antibiotics is high (Saha et al. 2008).
or human blood agar and chocolate agar. Meningococci and A recently developed NAAT, loop-mediated isothermal amplifi-
pneumococci are grown on agar media in a 5–10 % carbon cation, which amplifies DNA under isothermal conditions
dioxide-enriched atmosphere with rather high humidity at (63  C), is a promising tool, particularly in resource-poor set-
35–37  C (95–98.6  F). After 18–24 h, flat, gray–brown, tings, because it does not require a thermocycling apparatus and
translucent, smooth, 1–3 mm colonies of N. meningitidis or the results can be read with the naked eye (based on turbidity or
S. pneumoniae are present which can be analyzed by Gram color development by SYBR Green dye for staining nucleic
stain (Tonjum 2005). The finding of oxidase- and catalase- acids) (Seki et al. 2005). The assay detected ten or more copies
positive Gram-negative diplococci is sufficient to support of S. pneumoniae in oral mucosa swab samples (Seki et al. 2005),
a tentative diagnosis of meningococcal disease. Differentiation but its use in the diagnosis of bacterial meningitis has not been
characteristics are the production of acid from glucose tested. Identification of pathogens by use of a microarray or
and maltose. Optochin-sensitive bacteria with a characteristic biochip involves extraction of genomic DNA from CSF, ampli-
central umbilicus indicate S. pneumoniae. Isolation of fication of targeted DNA, and hybridization of labeled DNA with
N. meningitidis and S. pneumoniae can also be finally confirmed oligonucleotide probes (pathogen-specific or virulence genes)
by nucleic acid amplification technique (NAATs/PCR), DNA immobilized on a microarray. A rapid immunochroma-
sequence, or MALDI/TOF analysis. tographic test for S. pneumoniae was evaluated in 122 children
Non-culture Methods. Non-culture tests are particularly with pneumococcal meningitis (Saha et al. 2005). Compared
important for patients who need rapid identification of with CSF culture (sensitivity of 71 %) and latex agglutination
pathogens or have previously received antibiotics, or whose (86 %), immunochromatography was 100 % sensitive for the
initial CSF Gram stain is negative with negative culture at 72-h diagnosis of pneumococcal meningitis, suggesting that
incubation. Such tests include latex agglutination, PCR, immunochromatography might be useful in the diagnosis of
loop-mediated isothermal amplification method, microarray pneumococcal meningitis.
or biochip, and immunochromatography. Latex agglutination PCR or other non-culture tests can be especially useful for
uses latex beads adsorbed with microbe-specific antibodies. diagnosis in regions where patients frequently receive antibiotics
In the presence of homologous antigen, there is visible aggluti- before reaching the hospital. Recent WHO reports may have
nation of the antibody-coated latex beads (Gray and Fedorko underestimated the real disease burden, because bacteremia and
416 18 Meningitis

severe (fatal) disease are often not reported. Further underesti- (Maiden et al. 1998; Yazdankhah and Caugant 2004). MLST is
mation may be due to limited resources for establishing presently the best method for global typing of meningococci and
a diagnosis. In many, but not all, developing countries, at least for understanding the impact of vaccination. As has been the
one laboratory is available for the surveillance of meningococcal case for the W-135 epidemic in Africa, surveillance can direct the
disease, but limitations in the availability of diagnostic and preparation of vaccines against new clonal strains or specific
typing methods may further result in underestimation of disease serogroups (Khatami and Pollard 2011). However, the
burden. robustness of MLST in predicting antigenic profile needs further
analysis. Especially in the context of new protein-based menin-
gococcal vaccines, the expansion of current typing protocols
Molecular Typing of N. meningitidis needs consideration. Because MLST uses conserved core
sequences, it may not necessarily predict associations between
Phenotypic classification of N. meningitidis is based upon many vaccine antigens because of the lack of concordance
antigenic differences of the major surface antigens which introduced by recombination. Targeted and complete
provides information about the serogroup (capsule, e.g., B), genome sequencing is now the method of choice for genotyping
serotype (PorB porin, e.g., 15), serosubtype (PorA porin, e.g., (Jolley et al. 2012).
P1.7), and LOS immunotype (e.g., L3) of a particular strain.
This results in the classification: B, 15, P1.7, and L3. Multiple
epitopes can be recognized depending upon the presence Molecular Typing of S. pneumoniae
of phase or antigen variants in the bacterial population.
Antigen-based typing is currently only relevant for vaccine S. pneumoniae is highly diverse genetically, due to an efficient
efficacy studies. transformation system. Some of the many clonal lineages in the
A genetic typing system based upon polymorphisms in mul- genus cause severe pneumonia with invasive disease or
tiple housekeeping genes (multilocus sequence typing or MLST) meningitis. On the other hand, other lineages are colonizers
is the gold standard for molecular typing and has defined that are less virulent and being less lethal, and they instead
hypervirulent meningococcal lineages (Maiden et al. 1998). tend to promote the spread of antibiotic resistance. Comparative
Why hypervirulent meningococcal lineages are more pathogenic genomic analyses and functional studies reveal that a significant
has been a subject of considerable interest. Based on sequencing fraction of the pneumococcal genome is variable and not
of eight genomes, the chromosome is between 2.0 and 2.1 Mb in conserved in all strains. These genomic regions encode
size and contains about 2,000 genes (Parkhill et al. 2000; Tettelin nonessential ‘‘accessory’’ gene products, which, at least in
et al. 2000; Schoen et al. 2008). Each new strain sequenced has mouse models, alter virulence (Blomberg et al. 2009).
identified 40–50 new genes, and the meningococcus shares A number of pneumococcal genomes have been sequenced
about 90 % homology at the nucleotide level with either completely, revealing that the core pneumococcal genome, the
N. gonorrhoeae or N. lactamica. Mobile genetic elements includ- portion that is conserved between strains, comprises approxi-
ing IS elements and prophage sequences make up 10 % of the mately 50 % of all pneumococcal genes (Hiller et al. 2007).
genome (Parkhill et al. 2000). Other than the capsule locus, no Hence, the accessory genome is of considerable size and may
core pathogenome has been identified, suggesting that virulence provide different pneumococcal strains (clones) with different
can be clonal group dependent. Given that transformation is an properties. Molecular tools such as pulse field gel electrophoresis
efficient mechanism of genetic exchange and that meningococci (PFGE) and multilocus sequence typing (MLST) (partial
have acquired DNA from commensal Neisseria spp. and other sequencing of seven housekeeping genes) have allowed direct
bacteria (e.g., Haemophilus) as well as phages, the gene pool for strain comparisons from the same or different geographic areas
adaptation and evolution is quite large. Genome plasticity and (Blomberg et al. 2009). Together, these studies show that the
phenotype diversity through gain and loss of DNA or, for exam- capsule and its accessory components are the major virulence
ple, through DNA repeats are characteristics of meningococcal attribute of the pneumococcus. Based on difference in capsular
evolution. This is in contrast to the relatively conserved genomes components, at least 93 capsular serotypes have been identified.
of, for example, Bacillus anthracis. The acquisition of the capsule Epidemiological studies that compared carrier and invasive iso-
locus by horizontal transfer possibly from Pasteurella multocida lates from the same geographic area over the same time period
or P. haemolytica (Schoen et al. 2008) appears to be a major event revealed that different serotypes have different odds ratios (ORs)
in the evolution of the pathogenicity of the meningococcus. of causing invasive disease (Brueggemann et al. 2003; Sandgren
Many molecular methods are being used to characterize et al. 2004). However, strains of different MLST (ST) but with
the structure and evolution of the N. meningitidis genome. the same serotype display different degrees of virulence in mice
These include multilocus enzyme electrophoresis (ET) typing, (Sandgren et al. 2005). Thus, the OR for risk of invasive disease
DNA restriction analysis, randomly amplified polymorphic DNA varies between strains of the same serotype and between strains
(RAPD), restriction fragment length polymorphism (RFLP), of the same ST type as well as by some host characteristics
and ribotyping (Yazdankhah and Caugant 2004). The current including age and health status, as discussed above (Sjostrom
strain typing method, MLST, shows that epidemics are often et al. 2006). Therefore, disease severity and disease outcome are
caused by specific complexes of related hypervirulent lineages affected by the capsular type as well as by other pneumococcal
Meningitis 18 417

properties, especially those encoded by the variable accessory of invasive pneumococcal disease (Roy et al. 2002). In contrast,
genome, and by characteristics of the host. little is known about genetic variation affecting susceptibility to
The serotype and clonal distribution for pneumococcal pneumococcal meningitis, but polymorphisms in Toll-like
meningitis is broader than for cases of septicemia caused by receptors 2, 4, and 9 were recently linked to hearing loss in
the same organism (Henriques et al. 2000; Henriques Normark patients who survived meningococcal or pneumococcal menin-
et al. 2001; Darenberg and Henriques Normark 2009). Clinical gitis (van Well et al. 2012).
presentation and outcome of pneumococcal meningitis also Polymorphisms in genes coding for the Fcg-receptor II
vary for pneumococcal strains of different genotypes (van (CD32), Fcg-receptor III (CD16), mannose-binding lectin, and
Hoek et al. 2012). For example, Burckhardt et al. found in TLR4 are associated with increased risk (Hibberd et al. 1999;
a prospective population-based study that serotype 23F was an Fijen et al. 2000; Read et al. 2001; Smirnova et al. 2003; Faber
independent risk factor for pneumococcal meningitis et al. 2006; Tully et al. 2006). Mannose-binding lectin is a plasma
(Burckhardt et al. 2010). In France et al. showed that despite opsonin that initiates complement activation; specific polymor-
a high vaccination rate, the incidence of pneumococcal menin- phisms in this gene are enriched in children with meningococcal
gitis did not decline between 2001–2002 and 2007–2008, due to disease relative to controls (Hibberd et al. 1999). Expression of
a wide diversity of serotypes causing pneumococcal meningitis plasminogen activator inhibitor (PAI-1) affects severity and
after vaccine introduction (see below) (Levy et al. 2011). How- mortality of meningococcal sepsis, suggesting that impaired
ever, they observed a decline in cases in children younger than fibrinolysis is an important factor in the pathophysiology of
2 years of age, which was offset by an increase in older children. meningococcal sepsis (Emonts et al. 2003). Meningococcal
disease is occasionally linked to immune suppressive disorders
such as the nephrotic syndrome, hypogammaglobulinemia,
Host Susceptibility to Meningococcal splenectomy, and HIV/AIDS (10-fold increased risk for spo-
and Pneumococcal Meningitis radic disease vs 100-fold increased risk for infection with the
pneumococcus or meningococcus in HIV/AIDS). However, the
Genetic Cofactors. Absence of protective bactericidal antibodies risk of an epidemic outbreak of meningococcal disease in coun-
is the most important single predisposing factor for systemic tries with high rates of HIV does not appear to be elevated.
meningococcal and pneumococcal disease, but genetic polymor- Congenital and acquired antibody deficiencies, and possibly
phisms and other host cofactors contribute to disease (Sanders blocking of IgA immunoglobulin, also increase risk.
et al. 2011). Complement deficiency and polymorphisms in Opsonization and phagocytic function are important host
other innate host determinants, such as the FcII receptor, play defense mechanisms that influence disease incidence as well;
important roles as risk factors for meningococcal disease. Com- for example, pp[as shown by disease reduction after polysaccha-
plement is required both for meningococcal bactericidal activity ride vaccination in individuals with complement deficiencies.
and for opsonophagocytosis (Fijen et al. 2000). Individuals Rapidly progressive, fatal meningococcemia can arise in patients
deficient in both the early and late components of the comple- deficient in properdin (Sjoholm et al. 1982), and there is
ment system are at increased risk for meningococcal disease a significant increased risk of recurrent meningococcal infec-
(Sjoholm et al. 1982; Fijen et al. 1999; Emonts et al. 2003). tions for those with defects in the terminal complement pathway
Compared to the general population, these patients usually (C5–C9) (Fijen et al. 1999).
experience less severe, more recurrent disease at an older age Epigenetic Cofactors. There is limited knowledge concerning
with less common serogroups. Ten to 20 % of invasive menin- the cofactors that influence the spread and severity of meningo-
gococcal disease in adults is associated with a complement coccal and pneumococcal disease. Low absolute humidity can
defect. The mannose-binding lectin (MBL) pathway of comple- damage the nasopharyngeal mucosa, allowing meningococci to
ment activation can be genetically variable and is associated with pass the mucosal barrier more easily or be transmitted by
difference in susceptibility to meningococcal disease in one third coughing. In countries with a temperate climate, susceptibility
of all cases (Hibberd et al. 1999). Other genetic polymorphisms to meningococcal and pneumococcal disease is highest in the
affecting the risk of acquiring meningococcal disease have also winter when absolute humidity is low. There is also evidence
been described (TNF, FcgRIIA, FcgRIII, PAI-1, ACE-1, IL-1Ra, that viral (influenza) and mycoplasma respiratory infections can
IL-1b, TLR4) (Stephens et al. 2007). predispose to meningococcal and pneumococcal disease,
It is well known that genetic variation in innate immune perhaps by damaging mucosal surfaces; altering dynamics of
response genes contributes to interindividual differences in adherence, colonization, and spread; and impairing mucosal
meningococcal and pneumococcal disease manifestations immunity. The role of mucosal immunity in preventing
(Sanders et al. 2011). Disappearance of antibody acquired or enhancing meningococcal disease requires more analysis in
from the mother increases the risk for infants and young chil- the future.
dren. For example, homozygotes for codon variants in the man- The association between HIV and meningococcal disease is
nose-binding lectin, an important mediator of host innate not well studied. Population-based studies in the USA indicate
immunity, representing about 5 % of north Europeans and an increased risk (7-fold) (Stephens et al. 1995), but this has
North Americans and larger proportions of populations in not been noted for African outbreaks. However, the high prev-
many developing countries, have a substantially increased risk alence of HIV in African and Asian countries might influence
418 18 Meningitis

carriage or susceptibility to and severity of meningococcal disease, continued surveillance is necessary. Cefotaxime or ceftriaxone is
as demonstrated for pneumococcal infections. used when relatively penicillin-resistant strains are isolated.
The ability of an antimicrobial agent to penetrate the blood–
brain barrier is the most important factor that determines
Therapy and Management whether efficient bacterial killing in the CSF will happen.
Blood–brain barrier penetration is affected by lipophilic prop-
Antimicrobial Treatment of Meningococcal erty, molecular weight, and protein-binding ability of drugs,
and Pneumococcal Meningitis inflammation of the meninges, water transport, and efflux
transporters (Loscher and Potschka 2005). Lipophilic agents
Eradication of the infecting organism from the CSF is entirely (i.e., fluoroquinolones and rifampicin) penetrate relatively well
dependent on antibiotics, and bactericidal antibiotics should be into the CSF even if the meninges are not inflamed, whereas
administered intravenously at the highest clinically validated hydrophilic agents (i.e., b-lactams and vancomycin) have
doses to patients with suspected bacterial meningitis (Tunkel decreased penetration into CSF in the absence of meningeal
et al. 2004). Several retrospective and prospective studies showed inflammation (Ahmed et al. 1999). An important factor in the
that delay in antibiotic treatment was associated with adverse choice of empirical antimicrobial agents is the emergence of
outcomes (Miner et al. 2001; Auburtin et al. 2006). In patients antimicrobial-resistant organisms, including N. meningitidis
with suspected bacterial meningitis for whom immediate and S. pneumoniae that is resistant to penicillin or third-
lumbar puncture is delayed due to pending brain imaging generation cephalosporins, and Gram-negative bacilli that are
study or the presence of disseminated intravascular coagulation, resistant to many b-lactam drugs. For example, the prevalence of
blood cultures must be obtained, and antimicrobial treatment S. pneumoniae strains that are relatively resistant to penicillin
should be initiated immediately. Selection of empirical antimi- (minimum inhibitory concentration [MIC] 0.1–1.0 mg/mL) or
crobial regimens is designed to cover the likely pathogens, based highly resistant to penicillin (MIC greater than 1.0 mg/mL) is
on age of the patient and specific risk factors, with modifications increasing, and many of the penicillin-resistant pneumococci
if CSF Gram stain is positive. have reduced susceptibility to third-generation cephalosporins
The recommended treatment for patients who have meningo- (i.e., cefotaxime and ceftriaxone) (Tunkel et al. 2004). Treatment
coccal or pneumococcal meningitis is benzylpenicillin or a third- failures in bacterial meningitis as a result of multiresistant
generation cephalosporin (e.g., ceftriaxone) (Stephens et al. 2007). organisms have been reported (John 1994). Therefore, empirical
For most cases of uncomplicated bacterial meningitis, 7-day treatment for patients with bacterial meningitis in areas where
treatment is adequate. When the etiology is not known at resistant S. pneumoniae strains are prevalent must include the
admission, ceftriaxone or cefotaxime is used for the first addition of vancomycin. However, penetration of vancomycin
24–48 h to cover the possibility of other bacterial pathogens into the CSF can be reduced in the absence of meningeal inflam-
(Stephens et al. 2007). Beta-lactamase-producing strains have mation and also in patients who receive adjunctive dexametha-
occasionally been recovered, harboring a penicillinase-encoding sone treatment.
plasmid. In addition, there are N. meningitidis strains that are Before passive immune or antibiotic treatment was available,
not b-lactamase positive, but have decreased sensitivity to the mortality of systemic meningococcal and pneumococcal
penicillin due to reduced affinity of penicillin to penicillin- disease was 70–90 %. The case fatality rate is now around 10 %
binding proteins 2 and 3, resulting from an altered penA gene in many countries. However, early recognition by parents and
(Spratt et al. 1989; Bowler et al. 1994). Studies have reported health professionals of the importance of fever and headache
isolates of S. pneumoniae with penicillin MICs of 0.12–1.0 mg/ with a non-blanching rash (the glass test), prehospital antibiotic
mL that had mutations in the target penicillin-binding proteins treatment, rapid transportation to a local hospital, and stabili-
(Spratt et al. 1989; Bowler et al. 1994; Dowson et al. 1989). zation in an intensive care unit has substantially reduced the case
Similarly, penicillin has been the standard treatment for fatality rate in children (Levy et al. 1990; Borchsenius et al.
meningococcal meningitis, but penicillin resistance has evolved, 1991). For patients in intensive care, recognition of the different
with an implication of treatment failures. An increased pathophysiological processes associated with meningococcal
incidence in penicillin non-susceptible strains of N. meningitidis meningitis (which causes death predominantly by cerebral
(e.g., MICs 0.1–0.5 mg/mL) from 9.1 % in 1986 to 81.4 % in edema) and meningococcal septic shock (which causes death
Spain and South America (Latorre et al. 2000; Ibarz-Pavon predominantly through hypovolemia, capillary leak, myocardial
et al. 2012). By contrast, relative resistance to penicillin dysfunction, and multiorgan failure) has led to major changes in
(MIC 0.1 mg/mL) has been shown to occur in 3–4 % of the treatment and management strategies for these two different
meningococcal isolates in the USA and in 2 % of the 137 isolates forms of disease (Stephens et al. 2007; Brandtzaeg and van
recovered between 2000 and 2006 from equatorial sub-Saharan Deuren 2012). Aggressive management of raised intracranial
Africa (Brigham and Sandora 2009). These findings support the pressure reduces mortality.
use of a third-generation cephalosporin for meningococcal Prehospital antibiotic treatment is advocated in many coun-
meningitis in areas where penicillin resistance is prevalent, tries, to reduce the case fatality rate for patients with fulminant
at least until penicillin susceptibility is known. Although the meningococcal or pneumococcal disease. If antibiotic treatment
frequency of relatively penicillin-resistant meningococci is low, is initiated before admission, benzylpenicillin, ceftriaxone, or
Meningitis 18 419

another effective antibiotic should be injected intravenously beneficial effects on hearing and other neurological sequelae are
or intramuscularly in adults and intramuscularly in children not as clear against meningitis caused by other organisms (van
(Stephens et al. 2007). During epidemics in developing de Beek et al. 2010). Dexamethasone treatment might be con-
countries, a single injection of long-acting chloramphenicol sidered for infants and children older than 6 weeks with pneu-
injected intramuscularly can be sufficient treatment for patients mococcal meningitis after considering the potential benefits and
with meningitis, and this simple treatment has saved possible risks. There is, however, no evidence from randomized
many thousands of lives. A single injection of ceftriaxone is controlled clinical trials that dexamethasone reduces death
equally effective and could become the preferred treatment caused by brain edema, which can take place in patients with
for epidemic meningitis. meningococcal meningitis. The widespread use of dexametha-
Pneumococcal infections have been treated with penicillin sone in children with bacterial meningitis needs careful moni-
since decades. However, an emerging increase in resistance rates toring of clinical (e.g., fever curve, resolution of symptoms and
to penicillin and to other common antibiotics is now being signs) and bacteriological responses to antimicrobial treatment,
observed, affecting treatment outcome. So far resistance has particularly for patients with meningitis caused by pneumococci
been found to most antibiotic drugs except for vancomycin. that are resistant to third-generation antibiotics, in whom bac-
Resistance rates to penicillin and macrolides can be high and teriological killing in the CSF depends on vancomycin. Moni-
above 50 % in some areas (Prymula et al. 2011). Also, pneumo- toring of the clinical response (e.g., fever curve) can be
cocci with reduced susceptibility to penicillin often carry complicated by the use of dexamethasone. In addition, concom-
other resistance determinants. In countries with low antibiotic itantly given dexamethasone and vancomycin can reduce
resistance rates such as Norway and Sweden, pneumococcal penetration of vancomycin into the CSF by virtue of the anti-
isolates with reduced susceptibility to penicillin are inflammatory activity of dexamethasone, resulting in treatment
multiresistant (resistant to more than two antibiotic classes) in failure. However, CSF bactericidal activity has been shown in
30–60 % of cases. The spread of antibiotic resistance is mainly children who have meningitis due to cephalosporin-resistant
due to the spread of successful international clones carrying pneumococci, and such cases should be treated with dexameth-
resistance traits. The wide use of antibiotics also influences asone as well as vancomycin and ceftriaxone (Klugman et al.
resistance rates. Milder infections caused by pneumococci with 1995). Another issue with adjunctive dexamethasone treatment
low MIC values to penicillin can usually be treated with a higher is the possibility of neuronal injury, including hippocampal
dose of penicillin. However, pneumococcal meningitis caused by apoptosis in experimental animals with pneumococcal menin-
pneumococci with a reduced susceptibility to penicillin needs to gitis who received dexamethasone (Leib et al. 2003). Long-term
be treated with other antibiotics than penicillin. follow-up studies are thus needed to address the effect of dexa-
methasone treatment on any cognitive and neuropsychological
outcomes in patients with bacterial meningitis.
Adjunctive Treatment

Neurological sequelae are common in survivors of meningitis Prevention of Meningococcal


and include hearing loss, cognitive impairment, and develop- and Pneumococcal Disease
mental delay. For example, bacterial meningitis has been iden-
tified as the leading postnatal cause of developmental Prevention of meningococcal and pneumococcal disease is based
disabilities, including cerebral palsy and mental retardation on chemoprophylaxis and vaccination (Rosenstein et al. 2001).
(Kim 2012). Hearing loss happens in 22–30 % of survivors The advancement of vaccine design in enhancing immunoge-
of pneumococcal meningitis compared to 1–8 % after nicity has been shown to be important in preventing meningitis
meningococcal meningitis (Andersen et al. 1997). In a recent caused by N. meningitidis and S. pneumoniae. Protein-
meta-analysis, adjunctive treatment with dexamethasone was conjugated capsular polysaccharide vaccines have almost
associated with lower case mortality, and lower rates of severe completely eliminated meningitis caused by vaccine serotypes.
hearing loss and long-term neurological sequelae (van de Beek Meningococcal Capsular Vaccines. Meningococcal polysac-
et al. 2010). The beneficial effect of adjunctive dexamethasone charide vaccines reduce the incidence of infection among mili-
treatment was also evident in adults with bacterial meningitis. tary recruits, reduce the progress of epidemics of serogroup
The outcome of bacterial meningitis has been suggested to A disease, and protect susceptible complement-factor-deficient
be related to inflammation of the subarachnoid space. Hence, individuals (Stephens et al. 2007). Capsule polysaccharide vac-
it has also been suggested that in addition to antibiotics, menin- cines are available for the pathogenic meningococcal serogroups
gococcal and pneumococcal meningitis can be treated with A, C, Y, and W-135. These vaccines are safe with mild local
corticosteroids. Data in the literature are controversial where adverse events and have good efficacy (>85 %) in older children
some studies show an effect on sequelae such as hearing loss and and adults. However, due to lack of a T-helper response, the
mortality, while others do not. vaccines are poorly immunogenic below 2 years of age, fail to
Dexamethasone given shortly before or when antibiotics induce immunological memory, and provide protection for only
were first given has been shown to reduce the rate of hearing 3–5 years. Polysaccharide vaccines are used by travelers visiting
loss in children with H. influenzae type b meningitis, but its countries with a high incidence of meningococcal disease.
420 18 Meningitis

A polysaccharide vaccine against serogroup B meningococci is safe and immunogenic, are anticipated to provide long duration
not available due to carbohydrate mimicry and poor of protection (as they induce a T-cell-dependent response), and
immunogenicity. are effective in young children (Bilukha and Rosenstein 2005).
Capsular polysaccharide vaccines to decrease A, C, Y, and W- Introduction of the C conjugate meningococcal vaccines in 2000
135 meningococcal disease were introduced in the 1970s and markedly reduced the incidence of serogroup C disease in the
1980s on the basis of Gotschlich, Gold, Goldschneider, and UK with estimated vaccine efficacies of 88 % in young children
Artenstein’s classic studies (Snape and Pollard 2005). These and 95 % in young adolescents. Immunization also decreased
vaccines are safe with mild local adverse events, are effective nasopharyngeal carriage by 66 % and transmission of the path-
(>85 %) in children (older than 2 years) and adults, but are ogen (herd immunity) (Snape and Pollard 2005; Vipond et al.
less immunogenic (C less than A) in children younger than 24 2012). However, widespread use of monovalent serogroup con-
months. Immunity to the polysaccharide vaccines is limited to jugate vaccines can become ineffective when the capsule types
3–5 years of protection, and immunological hyporesponsiveness switch due to genetic exchange or strains arise that show reduced
is induced by repeated doses of the group C and possibly group capsule expression.
A polysaccharides. Polysaccharide vaccines do not induce Additional research on meningococcal conjugate vaccines
immunological memory and have little or no effect on nasopha- holds great potential for control of meningococcal disease in
ryngeal carriage. Despite their limitations, meningococcal poly- areas (e.g., sub-Saharan Africa) where epidemics are frequent.
saccharide vaccines have been used extensively to control A serogroup A conjugate vaccine has been carried out by
epidemics in countries of the African meningitis belt, and they a nonprofit organization, the Meningitis Vaccine Program,
have saved many lives. However, they have often been deployed which is supported by the Bill & Melinda Gates Foundation
too late in the course of an outbreak to achieve maximum effect. (Borrow 2012; Caugant et al. 2012). The results of the first trials
A major advance in the prevention of meningococcal disease of this vaccine in Africa are promising, and researchers hope that
has been the development of meningococcal polysaccharide and this vaccine shortly will be ready for widespread deployment.
protein conjugate vaccines and their introduction into the UK, A combined pediatric vaccine that contains serogroup A and
other parts of Europe, Canada, and the USA (Snape and Pollard C meningococcal conjugates has also been tested in Africa
2005; Borrow 2012). These vaccines are safe and immunogenic (Borrow 2012).
in young children, induce immunological memory, and decrease Pneumococcal Capsular Vaccines. Year 2000, a pneumococcal
nasopharyngeal carriage of meningococci. In the UK, introduc- vaccine (PCV7) based on 7 out of the 93 capsular serotypes,
tion of the C conjugate meningococcal vaccines in 2000 to all associated to a protein in a so-called conjugated vaccine, was
children and young adults greatly reduced the rate of serogroup introduced in the United States. The 7 capsular types (types 4,
C disease (90 % vaccine effectiveness at 3 years for patients aged 6B, 9V, 14, 18C, 19F, and 23F) were chosen because they were the
11–18 years) (Vipond et al. 2012). A major protective effect of most prominent causing invasive pneumococcal disease in the
the C conjugate vaccine is mediated through herd immunity. United States. The vaccine introduction led to a decrease of
Rates of serogroup C carriage and disease in non-vaccinated invasive pneumococcal disease among infants and children
individuals are reduced by more than 50 %. However, the younger than 5 years and also a herd immunity effect in the
three-dose schedule of group C immunization in infancy orig- adult population (Hsu et al. 2009). Here, Hsu et al. showed
inally used in the UK provided only transitory protection, and a decline of pneumococcal meningitis in eight sites in the Unites
a booster dose or alternative immunization schedule was States between 1998–1999 and 2005 from 1.13 cases to 0.79 cases
needed. Thus, the UK has changed to a schedule of two doses per 100, 000. In children younger than 2 years of age and in those
of meningococcal C conjugate vaccine given at 3 and 4 months 65 years of age or older, the incidence of pneumococcal menin-
of age, followed by a booster at 12 months. In the Netherlands, gitis decreased by 64 % and 54 % respectively during the study
meningococcal C conjugate vaccination is not started until the period. Furthermore, importantly since pneumococcal carriage
second year of life. A serogroup A, C, Y, and W-135 polysaccha- is a prerequisite for a pneumococcal invasive disease, a reduction
ride-conjugate meningococcal vaccine has been introduced in of vaccine type carriage was observed. Use of these protein-
the USA for adolescents (Vipond et al. 2012). In addition to conjugated vaccines has also reduced H. influenzae type b and
children and adolescents, populations who should benefit from pneumococcal meningitis among unvaccinated populations
the new conjugate vaccines are military recruits, patients with through herd immunity. At present, limitations with PCV7
complement or other immune deficiencies, microbiologists who conjugate vaccines include an apparent increase in the incidence
are routinely exposed to isolates of N. meningitidis, and people of invasive pneumococcal disease, including meningitis caused
who travel to or reside in countries where N. meningitidis is by non-PCV7 serotypes, such as serotype 19A (a penicillin and
epidemic. third-generation cephalosporin-resistant non-PCV7 serotype),
The immunogenicity of polysaccharide vaccines is greatly and an apparent decline in bactericidal antibody against
improved by chemical conjugation to a protein carrier. N. meningitidis in infants, requiring a booster immunization in
The resulting polysaccharide-conjugate vaccines are safe the second year of life (Borrow 2012). The conjugated pneumo-
and immunogenic in young infants and induce long-term coccal vaccines have now been introduced into the childhood
memory. Conjugate polysaccharide vaccines against serogroups vaccination program in several countries worldwide, and
A, C, W-135, and Y are now available. These vaccines are so far a decrease of invasive disease has been noticed in most countries.
Meningitis 18 421

However, recently, it was shown that serotypes not included in following recognition of the index case but extends for several
the vaccine are increasing as well as certain lineages harboring weeks.
antibiotic resistance determinants (serotype replacement and Many antibiotics used for therapy do not effectively eradi-
serotype shift) (Grijalva and Pelton 2011). An increase has cate or prevent carriage of meningococci because of inadequate
been seen, for example, of serotype 19A causing invasive disease levels in oropharyngeal secretions (Deghmane et al. 2009).
and carrying antibiotic resistance markers creating treatment Rifampicin, ceftriaxone, azithromycin, and the quinolones are
problems (McGee 2007). In some countries, the decline in effective against meningococci in the naso- and oropharynx
invasive disease has been hampered because of an increase of (Stephens et al. 2007). However, rifampicin resistance can
non-vaccine types. Moreover, some studies show that the decline develop rapidly, and quinolone resistance in meningococci has
in colonization has been hampered by an increase of non- recently been reported. Ceftriaxone as a single intramuscular
vaccine-type carriage (Tocheva et al. 2011). Recently, second- dose is 97 % effective in household contacts 1–2 weeks after
generation conjugated vaccines including 10 (PCV10 including infection. The advantage of ceftriaxone is that it can be used in
also serotypes 1, 5, and 7F) or 13 (PCV13 including in addition pregnancy and in small children.
types 3, 6A, and 19A) serotypes have been launched. Chemoprophylaxis can be helpful in the control of localized
Outer Membrane Protein Vaccines. The development of vac- outbreaks in residential schools, barracks, etc., but is generally
cines for serogroup B N. meningitidis remains a challenge not recommended for the control of epidemics because of cost
(Borrow 2012). The serogroup B capsule has an identical struc- and drug resistance. For example, widespread distribution of
ture to polysialic structures expressed in fetal neural tissue and rifampicin would be unwise in communities in which tubercu-
does not induce a protective IgG response. Thus, strategies have losis is prevalent. Many meningococcal strains are sulfur
focused on non-capsular antigens such as outer membrane porins resistant (folP mutation), and so sulfur drugs, once highly
and vesicles and lipooligosaccharides. The diversity of major effective, can no longer be used for chemoprophylaxis. Rifam-
outer membrane structures in meningococci has, however, lim- picin, ceftriaxone, azithromycin, and quinolones all have activity
ited these approaches (Snape and Pollard 2005; Bjune et al. 1991). against meningococci in the nasopharynx. However, resistance
Complement-mediated killing of encapsulated strains is also to rifampicin can develop rapidly, and quinolone resistance
achieved with cross-reactive antibodies directed against outer in meningococci has been reported (Gorla et al. 2011).
membrane components. Developed outer membrane vesicle Chemoprophylaxis is sometimes recommended for patients
(OMV) vaccines with a low LOS composition show efficacies given penicillin or chloramphenicol for treatment since
of 50–80 % in clinical trials, but do not protect young children pharyngeal carriage may not be eliminated by intravenous
and are in general too strain specific; that is, the vaccines can be administration of these antibiotics and the patient could remain
used against clonal disease outbreaks but not for prevention of colonized with a virulent strain.
sporadic diseases caused by diverse strains. Multivalent vaccine
strains based on common variants of PorA (a major inducer and
target of bactericidal antibodies) may provide protection against Future Challenges and Opportunities
multiple subtypes of N. meningitidis. Recently, novel conserved
candidate vaccine antigens have been identified using a ‘‘reverse Bacterial meningitis continues to be an important cause of mor-
vaccinology’’ approach (Rappuoli 2000; Giuliani et al. 2006; tality and morbidity throughout the world, with differential risk
Palumbo et al. 2012). First, the lipoproteins GNA1870 and for disease among small children, individuals living in low-income
GNA2132; the conserved surface proteins OMP85, NspA, and countries, and due to infection with antimicrobial- or multidrug-
NadA; but also PorA, pilin, and LOS conjugates were evaluated resistant pathogen. Vaccination with protein-conjugated
for their vaccine potential. The resulting vaccine, referred to as H. influenzae type b, S. pneumococcus PCV, and N. meningitidis
the four-component MenB (4CMenB) vaccine, currently con- Mencevax ACWY has successfully reduced worldwide incidence
tains the OMV (PorA) from the New Zealand vaccine along with of meningitis; this raises the hope that other conserved
three recombinant proteins identified by reverse vaccinology: bactericidal epitopes exist and can be identified and exploited
factor H-binding protein (fHbp), neisserial adhesin A (NadA), in a similar manner. Unfortunately, conjugated vaccines have so
and Neisseria heparin-binding antigen (NHBA) (Major et al. far been introduced only in the developed world, even though
2011). the highest incidence of meningococcal and pneumococcal
invasive diseases occurs in less affluent countries.
Host receptors and signal transduction pathways involved in
Chemoprophylaxis the microbial invasion of the BBB might represent potential
targets for novel therapeutic approaches for meningococcal
The aim of chemoprophylaxis is to reduce secondary cases of and pneumococcal disease (Huang and Jong 2009). Using
meningococcal and pneumococcal disease and to arrest out- a model system that analyzed penetration of the BBB by E. coli,
breaks. The risk of a secondary case among close contacts in a proof-of-concept study suggested that the HBMEC receptor
the household setting is 150–1,000 times higher than that in the for CNF1 (RPSA) and cytosolic phospholipase A2a might
general population. Children are at greatest risk, but secondary represent such ‘‘druggable’’ targets, at least for E. coli,
disease can occur at all ages. Risk is maximal in the week but potentially also for meningococci and pneumococci
422 18 Meningitis

(Plant et al. 2006; Orihuela et al. 2009). Other studies suggest Ahmed A, Jafri H, Lutsar I, McCoig CC, Trujillo M, Wubbel L, Shelton S,
McCracken GH Jr (1999) Pharmacodynamics of vancomycin for the treat-
that the cell-wall component lipoteichoic acid is a pattern rec-
ment of experimental penicillin- and cephalosporin-resistant pneumococcal
ognition molecule and inflammatory mediator that plays a role meningitis. Antimicrob Agents Chemother 43:876–881
in pneumococcal disease; however, the identity of the receptor Akira S, Takeda K (2004) Toll-like receptor signalling. Nat Rev Immunol 4:499–511
for lipoteichoic acid remains controversial. Interestingly, intra- Ambur OH, Davidsen T, Frye SA, Balasingham SV, Lagesen K, Rognes T, Tonjum T
thecal treatment with antibodies that recognize phosphor- (2009) Genome dynamics in major bacterial pathogens. FEMS Microbiol Rev
33:453–470
ylcholine, teichoic acid, and lipoteichoic acid was effective in
Amiry-Moghaddam M, Frydenlund DS, Ottersen OP (2004) Anchoring of
reducing neuronal damage in experimental pneumococcal aquaporin-4 in brain: molecular mechanisms and implications for the phys-
meningitis (Gerber et al. 2012). iology and pathophysiology of water transport. Neuroscience 129:999–1010
Basic understanding of the molecular mechanisms of Andersen J, Backer V, Voldsgaard P, Skinhoj P, Wandall JH (1997) Acute menin-
meningococcal and pneumococcal pathogenesis is still lacking gococcal meningitis: analysis of features of the disease according to the age of
255 patients. Copenhagen Meningitis Study Group. J Infect 34:227–235
and is urgently needed to support advances on novel therapeutic
Apicella MA (2005) Neisseria meningitidis. In: Mandell GL, Bennett JE, Dolin R
and preventive approaches for meningitis. Such basic research (eds) Principles and practice of infectious diseases. Elsevier/Churchill Liv-
will enhance our understanding of bacterial emergence, ingstone, Philadelphia, pp 2498–2513
pathogenic genome structure, horizontal genetic exchange, and Arditi M, Mason EO Jr, Bradley JS, Tan TQ, Barson WJ, Schutze GE, Wald ER,
innate and adaptive immune responses. N. meningitidis and Givner LB, Kim KS, Yogev R, Kaplan SL (1998) Three-year multicenter
surveillance of pneumococcal meningitis in children: clinical characteristics,
S. pneumoniae are valuable low-complexity model organisms
and outcome related to penicillin susceptibility and dexamethasone use.
for such studies, at least in part because they only colonize and Pediatrics 102:1087–1097
infect human beings. Further studies of the genetics and patho- Assalkhou R, Balasingham S, Collins RF, Frye SA, Davidsen T, Benam AV,
genicity of N. meningitidis and S. pneumoniae should reveal Bjoras M, Derrick JP, Tonjum T (2007) The outer membrane secretin PilQ
much about how they evolved, spread worldwide, and how from Neisseria meningitidis binds DNA. Microbiology 153:1593–1603
Auburtin M, Wolff M, Charpentier J, Varon E, Le Tulzo Y, Girault C, Mohammedi I,
and why they cause disease only in humans. This research is
Renard B, Mourvillier B, Bruneel F, Ricard JD, Timsit JF (2006) Detrimental
important in the quest to define fundamental mechanisms of role of delayed antibiotic administration and penicillin-nonsusceptible strains
microbial pathogenesis and to facilitate design of novel strategies in adult intensive care unit patients with pneumococcal meningitis: the
for managing emerging or reemerging microbial threats. PNEUMOREA prospective multicenter study. Crit Care Med 34:2758–2765
In summary, earlier clinical recognition and more effective Baker CJ, Kasper DL (1976) Correlation of maternal antibody deficiency with
susceptibility to neonatal group B streptococcal infection. N Engl J Med
treatment of meningococcal and pneumococcal disease will be
294:753–756
critical to further reduce morbidity and mortality associated Balasingham SV, Collins RF, Assalkhou R, Homberset H, Frye SA, Derrick JP,
with these diseases. Improved understanding of the pathophys- Tonjum T (2007) Interactions between the lipoprotein PilP and the secretin
iology of infection by meningitis-causing bacteria will PilQ in Neisseria meningitidis. J Bacteriol 189:5716–5727
undoubtedly lead to innovative new approaches for the Banerjee A, Van Sorge NM, Sheen TR, Uchiyama S, Mitchell TJ, Doran KS
(2010) Activation of brain endothelium by pneumococcal neuraminidase
management also of patients with meningococcal and
NanA promotes bacterial internalization. Cell Microbiol 12:1576–1588
pneumococcal septicemia, which could further reduce case Bergmann S, Hammerschmidt S (2006) Versatility of pneumococcal surface
fatality and/or case morbidity. The ultimate control of menin- proteins. Microbiology 152:295–303
gococcal and pneumococcal disease will require widespread and Bermpohl D, Halle A, Freyer D, Dagand E, Braun JS, Bechmann I, Schroder NW,
expanded use of effective vaccines. Worldwide surveillance, the Weber JR (2005) Bacterial programmed cell death of cerebral endothelial
cells involves dual death pathways. J Clin Invest 115:1607–1615
expanded use of polysaccharide-conjugate vaccines, and the
Biernath KR, Reefhuis J, Whitney CG, Mann EA, Costa P, Eichwald J, Boyle C
development of broadly effective serogroup B vaccines could (2006) Bacterial meningitis among children with cochlear implants beyond
eliminate N. meningitidis and S. pneumoniae as major threats 24 months after implantation. Pediatrics 117:284–289
to human health in industrialized countries within the next Bille E, Zahar JR, Perrin A, Morelle S, Kriz P, Jolley KA, Maiden MC, Dervin C,
decade. However, strong, sustained, and coordinated Nassif X, Tinsley CR (2005) A chromosomally integrated bacteriophage in
invasive meningococci. J Exp Med 201:1905–1913
support will be needed from the international community, if
Bjune G, Hoiby EA, Gronnesby JK, Arnesen O, Fredriksen JH, Halstensen A,
meningococcal and pneumococcal disease is to be controlled in Holten E, Lindbak AK, Nokleby H, Rosenqvist E et al (1991) Effect of outer
Africa and other developing areas, where the infection poses the membrane vesicle vaccine against group B meningococcal disease in Norway.
greatest persistent threat. Lancet 338:1093–1096
Blomberg C, Dagerhamn J, Dahlberg S, Browall S, Fernebro J, Albiger B, Morfeldt E,
Normark S, Henriques-Normark B (2009) Pattern of accessory regions
and invasive disease potential in Streptococcus pneumoniae. J Infect Dis
References 199:1032–1042
Boisier P, Nicolas P, Djibo S, Taha MK, Jeanne I, Mainassara HB, Tenebray B,
Aas FE, Egge-Jacobsen W, Winther-Larsen HC, Lovold C, Hitchen PG, Dell A, Kairo KK, Giorgini D, Chanteau S (2007) Meningococcal meningitis:
Koomey M (2006) Neisseria gonorrhoeae type IV pili undergo multisite, unprecedented incidence of serogroup X-related cases in 2006 in Niger.
hierarchical modifications with phosphoethanolamine and phosphocholine Clin Infect Dis 44:657–663
requiring an enzyme structurally related to lipopolysaccharide phosphoetha- Borchsenius F, Bruun JN, Tonjum T (1991) Systemic meningococcal disease: the
nolamine transferases. J Biol Chem 281:27712–27723 diagnosis on admission to hospital. NIPH Ann 14:11–22
Achtman M (1995) Epidemic spread and antigenic variability of Neisseria Borrow R (2012) Advances with vaccination against Neisseria meningitidis. Trop
meningitidis. Trends Microbiol 3:186–192 Med Int Health 1365–3156
Meningitis 18 423

Borrow R, Miller E (2006) Long-term protection in children with meningococcal Collins RF, Frye SA, Kitmitto A, Ford RC, Tonjum T, Derrick JP (2004) Structure
C conjugate vaccination: lessons learned. Expert Rev Vaccines 5:851–857 of the Neisseria meningitidis outer membrane PilQ secretin complex at 12
Bourdoulous S, Nassif X (2006) Mechanisms of attachment and invasion. In: A resolution. J Biol Chem 279:39750–39756
Frosch M, Maiden MCJ (eds) Handbook of meningococcal disease. Wiley, Corless CE, Guiver M, Borrow R, Edwards-Jones V, Fox AJ, Kaczmarski EB
Weinheim, pp 257–272 (2001) Simultaneous detection of Neisseria meningitidis, Haemophilus
Bowler LD, Zhang QY, Riou JY, Spratt BG (1994) Interspecies recombination influenzae, and Streptococcus pneumoniae in suspected cases of meningitis
between the penA genes of Neisseria meningitidis and commensal Neisseria and septicemia using real-time PCR. J Clin Microbiol 39:1553–1558
species during the emergence of penicillin resistance in N. meningitidis: Coureuil M, Join-Lambert O, Lecuyer H, Bourdoulous S, Marullo S, Nassif X
natural events and laboratory simulation. J Bacteriol 176:333–337 (2012) Mechanism of meningeal invasion by Neisseria meningitidis. Viru-
Brandt CT, Holm D, Liptrot M, Ostergaard C, Lundgren JD, Frimodt-Moller N, lence 3:164–172
Skovsted IC, Rowland IJ (2008) Impact of bacteremia on the pathogenesis of Cundell DR, Gerard NP, Gerard C, Idanpaan-Heikkila I, Tuomanen EI
experimental pneumococcal meningitis. J Infect Dis 197:235–244 (1995) Streptococcus pneumoniae anchor to activated human cells by the
Brandtzaeg P (2006) Pathogenesis and pathophysiology of invasive meningococ- receptor for platelet-activating factor. Nature 377:435–438
cal disease. In: Frosch M, Maiden MCJ (eds) Handbook of meningococcal Darenberg J, Henriques Normark B (2009) The epidemiology of pneumococcal
disease. Wiley, Weinheim, pp 427–480 infections—the Swedish experience. Vaccine 27(Suppl 6):G27–G32
Brandtzaeg P, van Deuren M (2012) Classification and pathogenesis of menin- Davidsen T, Tonjum T (2006) Meningococcal genome dynamics. Nat Rev
gococcal infections. Methods Mol Biol 799:21–35 Microbiol 4:11–22
Brigham KS, Sandora TJ (2009) Neisseria meningitidis: epidemiology, treatment Davidsen T, Koomey M, Tonjum T (2007) Microbial genome dynamics in CNS
and prevention in adolescents. Curr Opin Pediatr 21:437–443 pathogenesis. Neuroscience 145:1375–1387
Brouwer MC, Tunkel AR, van de Beek D (2010) Epidemiology, diagnosis, and de Greeff SC, de Melker HE, Schouls LM, Spanjaard L, van Deuren M (2008) Pre-
antimicrobial treatment of acute bacterial meningitis. Clin Microbiol Rev admission clinical course of meningococcal disease and opportunities for the
23:467–492 earlier start of appropriate intervention: a prospective epidemiological study
Brueggemann AB, Griffiths DT, Meats E, Peto T, Crook DW, Spratt BG on 752 patients in the Netherlands 2003–2005. Eur J Clin Microbiol Infect
(2003) Clonal relationships between invasive and carriage Streptococcus Dis 27:985–992
pneumoniae and serotype- and clone-specific differences in invasive disease de Louvois J, Halket S, Harvey D (2005) Neonatal meningitis in England and
potential. J Infect Dis 187:1424–1432 Wales: sequelae at 5 years of age. Eur J Pediatr 164:730–734
Burckhardt I, Burckhardt F, Van Der Linden M, Heeg C, Reinert RR (2010) Risk Deghmane AE, Alonso JM, Taha MK (2009) Emerging drugs for acute bacterial
factor analysis for pneumococcal meningitis in adults with invasive pneu- meningitis. Expert Opin Emerg Drugs 14:381–393
mococcal infection. Epidemiol Infect 138:1353–1358 Dery MA, Hasbun R (2007) Changing epidemiology of bacterial meningitis. Curr
Carbonnelle E, Helaine S, Nassif X, Pelicic V (2006) A systematic genetic analysis Infect Dis Rep 9:301–307
in Neisseria meningitidis defines the Pil proteins required for assembly, Dillard JP, Hackett KT (2005) Mutations affecting peptidoglycan acetylation
functionality, stabilization and export of type IV pili. Mol Microbiol in Neisseria gonorrhoeae and Neisseria meningitidis. Infect Immun
61:1510–1522 73:5697–5705
Cartwright K (1995) Introduction and historical aspect. In: Cartwright K (ed) Doulet N, Donnadieu E, Laran-Chich MP, Niedergang F, Nassif X, Couraud PO,
Meningococcal disease. Wiley, Chichester, pp 1–19 Bourdoulous S (2006) Neisseria meningitidis infection of human endothelial
Caugant DA, Kristiansen PA, Wang X, Mayer LW, Taha MK, Ouedraogo R, cells interferes with leukocyte transmigration by preventing the formation of
Kandolo D, Bougoudogo F, Sow S, Bonte L (2012) Molecular characteriza- endothelial docking structures. J Cell Biol 173:627–637
tion of invasive meningococcal isolates from countries in the African men- Dowson CG, Hutchison A, Brannigan JA, George RC, Hansman D, Linares J,
ingitis belt before introduction of a serogroup A conjugate vaccine. PLoS Tomasz A, Smith JM, Spratt BG (1989) Horizontal transfer of penicillin-
One 7:e46019 binding protein genes in penicillin-resistant clinical isolates of Streptococcus
Chang CJ, Chang WN, Huang LT, Huang SC, Chang YC, Hung PL, Lu CH, Chang pneumoniae. Proc Natl Acad Sci USA 86:8842–8846
CS, Cheng BC, Lee PY, Wang KW, Chang HW (2004) Bacterial meningitis in Dubos F, De la Rocque F, Levy C, Bingen E, Aujard Y, Cohen R, Breart G, Gendrel D,
infants: the epidemiology, clinical features, and prognostic factors. Brain Dev Chalumeau M (2008) Sensitivity of the bacterial meningitis score in 889
26:168–175 children with bacterial meningitis. J Pediatr 152:378–382
Chang YC, Uchiyama S, Varki A, Nizet V (2012) Leukocyte inflammatory Duensing TD, Wing JS, van Putten JP (1999) Sulfated polysaccharide-directed
responses provoked by pneumococcal sialidase. MBio 3:00220–00211 recruitment of mammalian host proteins: a novel strategy in microbial
Chanteau S, Rose AM, Djibo S, Nato F, Boisier P (2007) Biological diagnosis of pathogenesis. Infect Immun 67:4463–4468
meningococcal meningitis in the African meningitis belt: current epidemic Edmond K, Clark A, Korczak VS, Sanderson C, Griffiths UK, Rudan I (2010)
strategy and new perspectives. Vaccine 25(Suppl 1):A30–A36 Global and regional risk of disabling sequelae from bacterial meningitis:
Chiba N, Murayama SY, Morozumi M, Nakayama E, Okada T, Iwata S, Sunakawa K, a systematic review and meta-analysis. Lancet Infect Dis 10:317–328
Ubukata K (2009) Rapid detection of eight causative pathogens for the Emonts M, Hazelzet JA, de Groot R, Hermans PW (2003) Host genetic determi-
diagnosis of bacterial meningitis by real-time PCR. J Infect Chemother nants of Neisseria meningitidis infections. Lancet Infect Dis 3:565–577
15:92–98 Faber J, Meyer CU, Gemmer C, Russo A, Finn A, Murdoch C, Zenz W,
Christodoulides M, Makepeace BL, Partridge KA, Kaur D, Fowler MI, Weller RO, Mannhalter C, Zabel BU, Schmitt HJ, Habermehl P, Zepp F, Knuf M (2006)
Heckels JE (2002) Interaction of Neisseria meningitidis with human menin- Human toll-like receptor 4 mutations are associated with susceptibility to
geal cells induces the secretion of a distinct group of chemotactic, invasive meningococcal disease in infancy. Pediatr Infect Dis J 25:80–81
proinflammatory, and growth-factor cytokines. Infect Immun 70:4035–4044 Ferrieri P, Burke B, Nelson J (1980) Production of bacteremia and meningitis
Chudwin DS, Wara DW, Lameris-Martin NB, Ammann AJ (1983) Effect of antibody in infant rats with group B streptococcal serotypes. Infect Immun
concentration on opsonic requirements for phagocytosis in vitro of Strepto- 27:1023–1032
coccus pneumoniae types 7 and 19. Proc Soc Exp Biol Med 172:178–186 Fijen CA, Kuijper EJ, te Bulte MT, Daha MR, Dankert J (1999) Assessment
Clarke TB, Francella N, Huegel A, Weiser JN (2011) Invasive bacterial pathogens of complement deficiency in patients with meningococcal disease in
exploit TLR-mediated downregulation of tight junction components to The Netherlands. Clin Infect Dis 28:98–105
facilitate translocation across the epithelium. Cell Host Microbe 9:404–414 Fijen CA, Bredius RG, Kuijper EJ, Out TA, De Haas M, De Wit AP, Daha MR,
Claus H, Vogel U, Swiderek H, Frosch M, Schoen C (2007) Microarray analyses of De Winkel JG (2000) The role of Fcg receptor polymorphisms and C3 in the
meningococcal genome composition and gene regulation: a review of the immune defence against Neisseria meningitidis in complement-deficient
recent literature. FEMS Microbiol Rev 31:43–51 individuals. Clin Exp Immunol 120:338–345
424 18 Meningitis

Forest KT, Satyshur KA, Worzalla GA, Hansen JK, Herdendorf TJ (2004) The Hibberd ML, Sumiya M, Summerfield JA, Booy R, Levin M (1999) Association of
pilus-retraction protein PilT: ultrastructure of the biological assembly. Acta variants of the gene for mannose-binding lectin with susceptibility to menin-
Crystallogr D Biol Crystallogr 60:978–982 gococcal disease. Meningococcal Research Group. Lancet 353:1049–1053
Fothergill LD, Wright J (1933) Influenzal meningitis: the relation of age incidence Hill DJ, Virji M (2012) Meningococcal ligands and molecular targets of the host.
to the bactericidal power of blood against the causal organism. J Immunol Methods Mol Biol 799:143–152
24:273–284 Hiller NL, Janto B, Hogg JS, Boissy R, Yu S, Powell E, Keefe R, Ehrlich NE, Shen K,
Gaggar A, Shayakhmetov DM, Lieber A (2003) CD46 is a cellular receptor for Hayes J, Barbadora K, Klimke W, Dernovoy D, Tatusova T, Parkhill J, Bentley
group B adenoviruses. Nat Med 9:1408–1412 SD, Post JC, Ehrlich GD, Hu FZ (2007) Comparative genomic analyses of
Galanakis E, Di Cello F, Paul-Satyaseela M, Kim KS (2006) Escherichia coli K1 seventeen Streptococcus pneumoniae strains: insights into the pneumococcal
induces IL-8 expression in human brain microvascular endothelial cells. Eur supragenome. J Bacteriol 189:8186–8195
Cytokine Netw 17:260–265 Hoegen T, Tremel N, Klein M, Angele B, Wagner H, Kirschning C, Pfister HW,
Gerber J, Redlich S, Ribes S, Tauber SC, Schmidt H, Nau R (2012) Intrathecal Fontana A, Hammerschmidt S, Koedel U (2011) The NLRP3 inflammasome
treatment with the anti-phosphorylcholine monoclonal antibody TEPC-15 contributes to brain injury in pneumococcal meningitis and is activated
decreases neuronal damage in experimental pneumococcal meningitis. Che- through ATP-dependent lysosomal cathepsin B release. J Immunol
motherapy 58:212–216 187:5440–5451
Giuliani MM, Adu-Bobie J, Comanducci M, Arico B, Savino S, Santini L, Brunelli B, Hoffmann I, Eugene E, Nassif X, Couraud PO, Bourdoulous S (2001) Activation
Bambini S, Biolchi A, Capecchi B, Cartocci E, Ciucchi L, Di Marcello F, of ErbB2 receptor tyrosine kinase supports invasion of endothelial cells by
Ferlicca F, Galli B, Luzzi E, Masignani V, Serruto D, Veggi D, Contorni M, Neisseria meningitidis. J Cell Biol 155:133–143
Morandi M, Bartalesi A, Cinotti V, Mannucci D, Titta F, Ovidi E, Welsch JA, Hoffmann OM, Becker D, Weber JR (2007) Bacterial hydrogen peroxide contrib-
Granoff D, Rappuoli R, Pizza M (2006) A universal vaccine for serogroup utes to cerebral hyperemia during early stages of experimental pneumococcal
B meningococcus. Proc Natl Acad Sci USA 103:10834–10839 meningitis. J Cereb Blood Flow Metab 27:1792–1797
Goldschneider I, Gotschlich EC, Artenstein MS (1969) Human immunity to Hoffmann O, Rung O, Im AR, Freyer D, Zhang J, Held J, Stenzel W, Dame C
the meningococcus. II. Development of natural immunity. J Exp Med (2011) Thrombopoietin contributes to neuronal damage in experimental
129:1327–1348 bacterial meningitis. Infect Immun 79:928–936
Goonetilleke UR, Scarborough M, Ward SA, Hussain S, Kadioglu A, Gordon SB Hsu HE, Shutt KA, Moore MR, Beall BW, Bennett NM, Craig AS, Farley MM,
(2012) Death is associated with complement C3 depletion in cerebrospinal Jorgensen JH, Lexau CA, Petit S, Reingold A, Schaffner W, Thomas A,
fluid of patients with pneumococcal meningitis. MBio 3:00272–00211 Whitney CG, Harrison LH (2009) Effect of pneumococcal conjugate vaccine
Gorla MC, de Paiva MV, Salgueiro VC, Lemos AP, Brandao AP, Vazquez JA, on pneumococcal meningitis. N Engl J Med 360:244–256
Brandileone MC (2011) Antimicrobial susceptibility of Neisseria Huang SH, Jong A (2009) Evolving role of laminin receptors in microbial
meningitidis strains isolated from meningitis cases in Brazil from 2006 to pathogenesis and therapeutics of CNS infection. Future Microbiol 4:959–962
2008. Enferm Infecc Microbiol Clin 29:85–89 Ibarz-Pavon AB, Lemos AP, Gorla MC, Regueira M, Group SW 2nd, Gabastou JM
Gotschlich EC, Goldschneider I, Artenstein MS (1969) Human immunity to the (2012) Laboratory-based surveillance of Neisseria meningitidis isolates
meningococcus. IV. Immunogenicity of group A and group from disease cases in Latin American and Caribbean countries, SIREVA II
C meningococcal polysaccharides in human volunteers. J Exp Med 2006–2010. PLoS One 7:e44102
129:1367–1384 Jit M (2010) The risk of sequelae due to pneumococcal meningitis in high-income
Gray LD, Fedorko DP (1992) Laboratory diagnosis of bacterial meningitis. Clin countries: a systematic review and meta-analysis. J Infect 61:114–124
Microbiol Rev 5:130–145 Johansson L, Rytkonen A, Bergman P, Albiger B, Kallstrom H, Hokfelt T,
Gray-Owen SD, Blumberg RS (2006) CEACAM1: contact-dependent control of Agerberth B, Cattaneo R, Jonsson AB (2003) CD46 in meningococcal dis-
immunity. Nat Rev Immunol 6:433–446 ease. Science 301:373–375
Grijalva CG, Pelton SI (2011) A second-generation pneumococcal conjugate John CC (1994) Treatment failure with use of a third-generation cephalosporin
vaccine for prevention of pneumococcal diseases in children. Curr Opin for penicillin-resistant pneumococcal meningitis: case report and review.
Pediatr 23:98–104 Clin Infect Dis 18:188–193
Hamilton HL, Dillard JP (2006) Natural transformation of Neisseria gonorrhoeae: Jolley KA, Hill DM, Bratcher HB, Harrison OB, Feavers IM, Parkhill J, Maiden
from DNA donation to homologous recombination. Mol Microbiol MC (2012) Resolution of a meningococcal disease outbreak from whole-
59:376–385 genome sequence data with rapid web-based analysis methods. J Clin
Hamilton HL, Dominguez NM, Schwartz KJ, Hackett KT, Dillard JP Microbiol 50:3046–3053
(2005) Neisseria gonorrhoeae secretes chromosomal DNA via a novel type Jyssum K, Lie S (1965) Genetic factors determining competence in transforma-
IV secretion system. Mol Microbiol 55:1704–1721 tion of Neisseria Meningitidis. 1. A permanent loss of competence. Acta
Hammerschmidt S, Wolff S, Hocke A, Rosseau S, Muller E, Rohde M (2005) Pathol Microbiol Scand 63:306–316
Illustration of pneumococcal polysaccharide capsule during adherence and Kahler CM, Stephens DS (1998) Genetic basis for biosynthesis, structure, and
invasion of epithelial cells. Infect Immun 73:4653–4667 function of meningococcal lipooligosaccharide (endotoxin). Crit Rev
Harrison OB et al (2002) Analysis of pathogen-host cell interactions in purpura Microbiol 24:281–334
fulminans: expression of capsule, type IV pili, and por A by Neisseria Kahler CM, Datta A, Tzeng YL, Carlson RW, Stephens DS (2005) Inner core
meningitides in vivo. Infect Immun 70:5193–5201 assembly and structure of the lipooligosaccharide of Neisseria meningitidis:
Heckenberg SGB, de Gans J, Brouwer MC, Weisfelt M, Piet JR, Spanjarard L, van der capacity of strain NMB to express all known immunotype epitopes.
Ende A, van de Beek D (2008) Clinical features, outcome, and meningococcal Glycobiology 15:409–419
genotype in 258 adults with meningococcal meningitis. Medicine 87:185–192 Khatami A, Pollard AJ (2011) The epidemiology of meningococcal disease and
Henrichsen J (1983) Twitching motility. Annu Rev Microbiol 37:81–93 the impact of vaccines. Expert Rev Vaccines 9:285–298
Henriques Normark B, Kalin M, Ortqvist A, Akerlund T, Liljequist BO, Hedlund J, Kim KS (2003) Pathogenesis of bacterial meningitis: from bacteraemia to neuro-
Svenson SB, Zhou J, Spratt BG, Normark S, Kallenius G (2001) Dynamics of nal injury. Nat Rev Neurosci 4:376–385
penicillin-susceptible clones in invasive pneumococcal disease. J Infect Dis Kim KS (2009) Treatment strategies for central nervous system infections. Expert
184:861–869 Opin Pharmacother 10:1307–1317
Henriques B, Kalin M, Ortqvist A, Olsson Liljequist B, Almela M, Marrie TJ, Kim KS (2012) Current concepts on the pathogenesis of Escherichia coli meningitis:
Mufson MA, Torres A, Woodhead MA, Svenson SB, Kallenius G (2000) implications for therapy and prevention. Curr Opin Infect Dis 25:273–278
Molecular epidemiology of Streptococcus pneumoniae causing invasive dis- Klugman KP, Friedland IR, Bradley JS (1995) Bactericidal activity against ceph-
ease in 5 countries. J Infect Dis 182:833–839 alosporin-resistant Streptococcus pneumoniae in cerebrospinal fluid of
Meningitis 18 425

children with acute bacterial meningitis. Antimicrob Agents Chemother Nikulin J, Panzner U, Frosch M, Schubert-Unkmeir A (2006) Intracellular sur-
39:1988–1992 vival and replication of Neisseria meningitidis in human brain microvascular
La Scolea LJ Jr, Dryja D (1984) Quantitation of bacteria in cerebrospinal fluid and endothelial cells. Int J Med Microbiol 296:553–558
blood of children with meningitis and its diagnostic significance. J Clin O’Brien KL, Wolfson LJ, Watt JP, Henkle E, Deloria-Knoll M, McCall N, Lee E,
Microbiol 19:187–190 Mulholland K, Levine OS, Cherian T (2009) Burden of disease caused by
Lambotin M, Hoffmann I, Laran-Chich MP, Nassif X, Couraud PO, Bourdoulous S Streptococcus pneumoniae in children younger than 5 years: global estimates.
(2005) Invasion of endothelial cells by Neisseria meningitidis requires Lancet 374:893–902
cortactin recruitment by a phosphoinositide-3-kinase/Rac1 signalling path- Orihuela CJ, Mahdavi J, Thornton J, Mann B, Wooldridge KG, Abouseada N,
way triggered by the lipo-oligosaccharide. J Cell Sci 118:3805–3816 Oldfield NJ, Self T, Ala’Aldeen DA, Tuomanen EI (2009) Laminin receptor
Latorre C, Gene A, Juncosa T, Munoz C, Gonzalez-Cuevas A (2000) Neisseria initiates bacterial contact with the blood brain barrier in experimental
meningitidis: evolution of penicillin resistance and phenotype in a children’s meningitis models. J Clin Invest 119:1638–1646
hospital in Barcelona, Spain. Acta Paediatr 89:661–665 Palumbo E, Fiaschi L, Brunelli B, Marchi S, Savino S, Pizza M (2012) Antigen
Leib SL, Heimgartner C, Bifrare YD, Loeffler JM, Taauber MG (2003) Dexameth- identification starting from the genome: a ‘‘Reverse Vaccinology’’ approach
asone aggravates hippocampal apoptosis and learning deficiency in pneu- applied to MenB. Methods Mol Biol 799:361–403
mococcal meningitis in infant rats. Pediatr Res 54:353–357 Parge HE, Forest KT, Hickey MJ, Christensen DA, Getzoff ED, Tainer JA
Levy M, Wong E, Fried D (1990) Diseases that mimic meningitis. Analysis of 650 (1995) Structure of the fibre-forming protein pilin at 2.6 A resolution.
lumbar punctures. Clin Pediatr (Phila) 29(254–255):258–261 Nature 378:32–38
Levy C, Varon E, Bingen E, Lecuyer A, Boucherat M, Cohen R (2011) Pneumo- Parkhill J, Achtman M, James KD, Bentley SD, Churcher C, Klee SR, Morelli G,
coccal meningitis in French children before and after the introduction of Basham D, Brown D, Chillingworth T, Davies RM, Davis P, Devlin K,
pneumococcal conjugate vaccine. Pediatr Infect Dis J 30:168–170 Feltwell T, Hamlin N, Holroyd S, Jagels K, Leather S, Moule S, Mungall K,
Littmann M, Albiger B, Frentzen A, Normark S, Henriques-Normark B, Plant L Quail MA, Rajandream MA, Rutherford KM, Simmonds M, Skelton J,
(2009) Streptococcus pneumoniae evades human dendritic cell surveillance by Whitehead S, Spratt BG, Barrell BG (2000) Complete DNA sequence of
pneumolysin expression. EMBO Mol Med 1:211–222 a serogroup A strain of Neisseria meningitidis Z2491. Nature 404:502–506.
Loscher W, Potschka H (2005) Drug resistance in brain diseases and the role of doi:10.1038/35006655
drug efflux transporters. Nat Rev Neurosci 6:591–602. doi:10.1038/nrn1728, Paterson GK, Mitchell TJ (2006) Innate immunity and the pneumococcus.
nrn1728 [pii] Microbiology 152:285–293
Lu JJ, Perng CL, Lee SY, Wan CC (2000) Use of PCR with universal primers and Peltola H (2000) Worldwide Haemophilus influenzae type b disease at the begin-
restriction endonuclease digestions for detection and identification of com- ning of the 21st century: global analysis of the disease burden 25 years after
mon bacterial pathogens in cerebrospinal fluid. J Clin Microbiol 38:2076–2080 the use of the polysaccharide vaccine and a decade after the advent of
MacLennan J, Kafatos G, Neal K, Andrews N, Cameron JC, Roberts R, Evans MR, conjugates. Clin Microbiol Rev 13:302–317
Cann K, Baxter DN, Maiden MC, Stuart JM (2006) Social behavior and Perkins-Balding D, Ratliff-Griffin M, Stojiljkovic I (2004) Iron transport systems
meningococcal carriage in British teenagers. Emerg Infect Dis 12:950–957 in Neisseria meningitidis. Microbiol Mol Biol Rev 68:154–171
Mahdi LK, Wang H, Van der Hoek MB, Paton JC, Ogunniyi AD (2012) Identifi- Plant L, Sundqvist J, Zughaier S, Lovkvist L, Stephens DS, Jonsson AB
cation of a novel pneumococcal vaccine antigen preferentially expressed (2006) Lipooligosaccharide structure contributes to multiple steps in the
during meningitis in mice. J Clin Invest 122:2208–2220 virulence of Neisseria meningitidis. Infect Immun 74:1360–1367
Maiden MC, Bygraves JA, Feil E, Morelli G, Russell JE, Urwin R, Zhang Q, Zhou J, Plummer FA, Chubb H, Simonsen JN, Bosire M, Slaney L, Maclean I, Ndinya-
Zurth K, Caugant DA, Feavers IM, Achtman M, Spratt BG (1998) Multilocus Achola JO, Waiyaki P, Brunham RC (1993) Antibody to Rmp (outer mem-
sequence typing: a portable approach to the identification of clones within brane protein 3) increases susceptibility to gonococcal infection. J Clin Invest
populations of pathogenic microorganisms. Proc Natl Acad Sci USA 91:339–343
95:3140–3145 Power PM, Jennings MP (2003) The genetics of glycosylation in Gram-negative
Major M, Moss S, Gold R (2011) From genes to vaccine: a breakthrough in the bacteria. FEMS Microbiol Lett 218:211–222
prevention of meningococcal group B disease. Paediatr Child Health 16: Prymula R, Chlibek R, Ivaskeviciene I, Mangarov A, Meszner Z, Perenovska P,
e61–e64 Richter D, Salman N, Simurka P, Tamm E, Tesovic G, Urbancikova I, Usonis V
Manchester M, Eto DS, Valsamakis A, Liton PB, Fernandez-Munoz R, Rota PA, (2011) Paediatric pneumococcal disease in Central Europe. Eur J Clin
Bellini WJ, Forthal DN, Oldstone MB (2000) Clinical isolates of measles Microbiol Infect Dis 30:1311–1320
virus use CD46 as a cellular receptor. J Virol 74:3967–3974 Quagliarello VJ, Long WJ, Scheld WM (1986) Morphologic alterations of the
Marchiafava E, Celli A (1884) Spra i micrococchi della meningite cerebrospinale blood–brain barrier with experimental meningitis in the rat. Temporal
epidemica. Gazz degli Ospedali 5:59 sequence and role of encapsulation. J Clin Invest 77:1084–1095
McGee L (2007) The coming of age of niche vaccines? Effect of vaccines on Radin JN, Orihuela CJ, Murti G, Guglielmo C, Murray PJ, Tuomanen EI (2005) beta-
resistance profiles in Streptococcus pneumoniae. Curr Opin Microbiol Arrestin 1 participates in platelet-activating factor receptor-mediated endocy-
10:473–478 tosis of Streptococcus pneumoniae. Infect Immun 73:7827–7835
Merz AJ, So M (2000) Interactions of pathogenic Neisseriae with epithelial cell Rappuoli R (2000) Reverse vaccinology. Curr Opin Microbiol 3:445–450
membranes. Annu Rev Cell Dev Biol 16:423–457 Read RC, Pullin J, Gregory S, Borrow R, Kaczmarski EB, di Giovine FS, Dower SK,
Meyer TF, Pohlner J, van Putten JP (1994) Biology of the pathogenic Neisseriae. Cannings C, Wilson AG (2001) A functional polymorphism of toll-like
Curr Top Microbiol Immunol 192:283–317 receptor 4 is not associated with likelihood or severity of meningococcal
Miner JR, Heegaard W, Mapes A, Biros M (2001) Presentation, time to antibi- disease. J Infect Dis 184:640–642
otics, and mortality of patients with bacterial meningitis at an urban county Reiss A, Braun JS, Jager K, Freyer D, Laube G, Buhrer C, Felderhoff-Muser U,
medical center. J Emerg Med 21:387–392 Stadelmann C, Nizet V, Weber JR (2011) Bacterial pore-forming cytolysins
Molyneux E, Riordan FAI, Walsh A (2006) Acute bacterial meningitis in children induce neuronal damage in a rat model of neonatal meningitis. J Infect Dis
presenting to the Royal Liverpool Children’s Hospital, Liverpool, UK and the 203:393–400
Queen Elisabeth Central Hospital in Blantyre, Malawi: a world of difference. Ridda I, Macintyre CR, Lindley R, McIntyre PB, Brown M, Oftadeh S, Sullivan J,
Ann Trop Paediatr 26:29–37 Gilbert GL (2010) Lack of pneumococcal carriage in the hospitalised elderly.
Mustafa MM, Lebel MH, Ramilo O, Olsen KD, Reisch JS, Beutler B, McCracken Vaccine 28:3902–3904
GH Jr (1989) Correlation of interleukin-1 beta and cachectin concentrations Ring A, Weiser JN, Tuomanen EI (1998) Pneumococcal trafficking across the
in cerebrospinal fluid and outcome from bacterial meningitis. J Pediatr blood–brain barrier. Molecular analysis of a novel bidirectional pathway.
115:208–213 J Clin Invest 102:347–360
426 18 Meningitis

Roberts MC (1989) Plasmids of Neisseria gonorrhoeae and other Neisseria species. Sjoholm AG, Braconier JH, Soderstrom C (1982) Properdin deficiency in a family
Clin Microbiol Rev 2(Suppl):S18–S23 with fulminant meningococcal infections. Clin Exp Immunol 50:291–297
Roine I, Peltola H, Fernandez J, Zavala I, Gonzalez Mata A, Gonzalez Ayala S, Sjostrom K, Spindler C, Ortqvist A, Kalin M, Sandgren A, Kuhlmann-Berenzon S,
Arbo A, Bologna R, Mino G, Goyo J, Lopez E, Dourado de Andrade S, Henriques-Normark B (2006) Clonal and capsular types decide whether
Sarna S (2008) Influence of admission findings on death and neurological pneumococci will act as a primary or opportunistic pathogen. Clin Infect
outcome from childhood bacterial meningitis. Clin Infect Dis 46: Dis 42:451–459
1248–1252 Smirnova I, Mann N, Dols A, Derkx HH, Hibberd ML, Levin M, Beutler B (2003)
Rosenstein NE, Perkins BA, Stephens DS, Popovic T, Hughes JM (2001) Menin- Assay of locus-specific genetic load implicates rare Toll-like receptor 4
gococcal disease. N Engl J Med 344:1378–1388 mutations in meningococcal susceptibility. Proc Natl Acad Sci USA
Roy S, Knox K, Segal S, Griffiths D, Moore CE, Welsh KI, Smarason A, Day NP, 100:6075–6080
McPheat WL, Crook DW, Hill AV (2002) MBL genotype and risk of invasive Snape MD, Pollard AJ (2005) Meningococcal polysaccharide-protein conjugate
pneumococcal disease: a case–control study. Lancet 359:1569–1573 vaccines. Lancet Infect Dis 5:21–30
Rubin LL, Staddon JM (1999) The cell biology of the blood–brain barrier. Annu Snyder LA, Saunders NJ (2006) The majority of genes in the pathogenic Neisseria
Rev Neurosci 22:11–28 species are present in non-pathogenic Neisseria lactamica, including those
Rudel T, Facius D, Barten R, Scheuerpflug I, Nonnenmacher E, Meyer TF designated as ‘virulence genes’. BMC Genomics 7:128
(1995) Role of pili and the phase-variable PilC protein in natural competence Snyder LA, Jarvis SA, Saunders NJ (2005) Complete and variant forms of the
for transformation of Neisseria gonorrhoeae. Proc Natl Acad Sci USA ‘gonococcal genetic island’ in Neisseria meningitidis. Microbiology
92:7986–7990 151:4005–4013
Rupprecht TA, Angele B, Klein M, Heesemann J, Pfister HW, Botto M, Koedel U Sparling PF (1966) Genetic transformation of Neisseria gonorrhoeae to strepto-
(2007) Complement C1q and C3 are critical for the innate immune response mycin resistance. J Bacteriol 92:1364–1371
to Streptococcus pneumoniae in the central nervous system. J Immunol Spratt BG, Zhang QY, Jones DM, Hutchison A, Brannigan JA, Dowson CG
178:1861–1869 (1989) Recruitment of a penicillin-binding protein gene from Neisseria
Saha SK, Darmstadt GL, Yamanaka N, Billal DS, Nasreen T, Islam M, Hamer DH flavescens during the emergence of penicillin resistance in Neisseria
(2005) Rapid diagnosis of pneumococcal meningitis: implications for treat- meningitidis. Proc Natl Acad Sci USA 86:8988–8992
ment and measuring disease burden. Pediatr Infect Dis J 24:1093–1098 Stephens DS (2007) Conquering the meningococcus. FEMS Microbiol Rev
Saha SK, Darmstadt GL, Baqui AH, Hossain B, Islam M, Foster D, Al-Emran H, 31:3–14
Naheed A, Arifeen SE, Luby SP, Santosham M, Crook D (2008) Identification Stephens DS, McGee ZA (1981) Attachment of Neisseria meningitidis to human
of serotype in culture negative pneumococcal meningitis using sequential mucosal surfaces: influence of pili and type of receptor cell. J Infect Dis
multiplex PCR: implication for surveillance and vaccine design. PLoS One 3: 143:525–532
e3576 Stephens DS, Hajjeh RA, Baughman WS, Harvey RC, Wenger JD, Farley MM
Sanders MS, van Well GT, Ouburg S, Morre SA, van Furth AM (2011) Genetic (1995) Sporadic meningococcal disease in adults: results of a 5-year popu-
variation of innate immune response genes in invasive pneumococcal and lation-based study. Ann Intern Med 123:937–940
meningococcal disease applied to the pathogenesis of meningitis. Genes Stephens DS, Greenwood B, Brandtzaeg P (2007) Epidemic meningitis,
Immun 12:321–334 meningococcaemia, and Neisseria meningitidis. Lancet 369:2196–2210
Sandgren A, Sjostrom K, Olsson-Liljequist B, Christensson B, Samuelsson A, Strom MS, Nunn DN, Lory S (1993) A single bifunctional enzyme, PilD, catalyzes
Kronvall G, Henriques Normark B (2004) Effect of clonal and serotype- cleavage and N-methylation of proteins belonging to the type IV pilin family.
specific properties on the invasive capacity of Streptococcus pneumoniae. Proc Natl Acad Sci USA 90:2404–2408
J Infect Dis 189:785–796 Swanson J (1973) Studies on gonococcus infection. IV. Pili: their role in attach-
Sandgren A, Albiger B, Orihuela CJ, Tuomanen E, Normark S, Henriques- ment of gonococci to tissue culture cells. J Exp Med 137:571–589
Normark B (2005) Virulence in mice of pneumococcal clonal types with Taha MK, Fox A (2007) Quality assessed nonculture techniques for detection and
known invasive disease potential in humans. J Infect Dis 192:791–800 typing of meningococci. FEMS Microbiol Rev 31:37–42
Santoro F, Greenstone HL, Insinga A, Liszewski MK, Atkinson JP, Lusso P, Berger EA Tettelin H, Saunders NJ, Heidelberg J, Jeffries AC, Nelson KE, Eisen JA, Ketchum KA,
(2003) Interaction of glycoprotein H of human herpesvirus 6 with the cellular Hood DW, Peden JF, Dodson RJ, Nelson WC, Gwinn ML, DeBoy R,
receptor CD46. J Biol Chem 278:25964–25969 Peterson JD, Hickey EK, Haft DH, Salzberg SL, White O, Fleischmann RD,
Schneider MC, Exley RM, Ram S, Sim RB, Tang CM (2007) Interactions between Dougherty BA, Mason T, Ciecko A, Parksey DS, Blair E, Cittone H, Clark EB,
Neisseria meningitidis and the complement system. Trends Microbiol Cotton MD, Utterback TR, Khouri H, Qin H, Vamathevan J, Gill J, Scarlato V,
15:233–240 Masignani V, Pizza M, Grandi G, Sun L, Smith HO, Fraser CM, Moxon ER,
Schoen C, Blom J, Claus H, Schramm-Gluck A, Brandt P, Muller T, Goesmann A, Rappuoli R, Venter JC (2000) Complete genome sequence of Neisseria
Joseph B, Konietzny S, Kurzai O, Schmitt C, Friedrich T, Linke B, Vogel U, meningitidis serogroup B strain MC58. Science 287:1809–1815, 8338 [pii]
Frosch M (2008) Whole-genome comparison of disease and carriage strains Thigpen MC, Whitney CG, Messonnier NE, Zell ER, Lynfield R, Hadler JL,
provides insights into virulence evolution in Neisseria meningitidis. Proc Natl Harrison LH, Farley MM, Reingold A, Bennett NM, Craig AS, Schaffner W,
Acad Sci USA 105:3473–3478 Thomas A, Lewis MM, Scallan E, Schuchat A (2011) Bacterial meningitis in
Schut ES, Lucas MJ, Brouwer MC, Vergouwen MD, van der Ende A, van de Beek D the United States, 1998–2007. N Engl J Med 364:2016–2025
(2012) Cerebral infarction in adults with bacterial meningitis. Neurocrit Tocheva AS, Jefferies JM, Rubery H, Bennett J, Afimeke G, Garland J,
Care 16:421–427 Christodoulides M, Faust SN, Clarke SC (2011) Declining serotype coverage
Schuurman T, de Boer RF, Kooistra-Smid AM, van Zwet AA (2004) Prospective of new pneumococcal conjugate vaccines relating to the carriage of Strepto-
study of use of PCR amplification and sequencing of 16S ribosomal DNA coccus pneumoniae in young children. Vaccine 29:4400–4404
from cerebrospinal fluid for diagnosis of bacterial meningitis in a clinical Tonjum T (2005) Family Neisseriaceae and genus Neisseria. In: Garrity G (ed)
setting. J Clin Microbiol 42:734–740 Bergey’s manual of systematic bacteriology: the proteobacteria. Springer,
Seki M, Yamashita Y, Torigoe H, Tsuda H, Sato S, Maeno M (2005) Loop- New York, pp 775–798
mediated isothermal amplification method targeting the lytA gene for detec- Tonjum T, Koomey M (1997) The pilus colonization factor of pathogenic
tion of Streptococcus pneumoniae. J Clin Microbiol 43:1581–1586 neisserial species: organelle biogenesis and structure/function relation-
Shanholtzer CJ, Schaper PJ, Peterson LR (1982) Concentrated gram stain smears ships—a review. Gene 192:155–163
prepared with a cytospin centrifuge. J Clin Microbiol 16:1052–1056 Tonjum T, Caugant DA, Dunham SA, Koomey M (1998) Structure and function
Sim RJ, Harrison MM, Moxon ER, Tang CM (2000) Underestimation of menin- of repetitive sequence elements associated with a highly polymorphic
gococci in tonsillar tissue by nasopharyngeal swabbing. Lancet 356: domain of the Neisseria meningitidis PilQ protein. Mol Microbiol
1653–1654 29:111–124
Meningitis 18 427

Tonjum T, Nilsson F, Bruun JN, Haneberg B (1983) The early phase of meningo- van Well GT, Sanders MS, Ouburg S, van Furth AM, Morre SA (2012) Poly-
coccal disease. NIPH Ann 6:175–181 morphisms in Toll-like receptors 2, 4, and 9 are highly associated with
Trindade MB, Job V, Contreras-Martel C, Pelicic V, Dessen A (2008) Structure of hearing loss in survivors of bacterial meningitis. PLoS One 7:e35837
a widely conserved type IV pilus biogenesis factor that affects the stability of Vieusseux M (1805) Mémoire su la maladie qui a regné a Genêve au printemps de
secretin multimers. J Mol Biol 378:1031–1039 1804. J Med Chir Pharmacol 11:163
Tsai CJ, Griffin MR, Nuorti JP, Grijalva CG (2008) Changing epidemiology of Vipond C, Mulloy B, Rigsby P, Burkin K, Bolgiano B (2012) Evaluation of
pneumococcal meningitis after the introduction of pneumococcal conjugate a candidate International Standard for Meningococcal Group
vaccine in the United States. Clin Infect Dis 46:1664–1672 C polysaccharide. Biologicals 40:353–363
Tsirpouchtsidis A, Hurwitz R, Brinkmann V, Meyer TF, Haas G (2002) Neisserial Viriyakosol S, Tobias PS, Kitchens RL, Kirkland TN (2001) MD-2 binds to
immunoglobulin A1 protease induces specific T-cell responses in humans. bacterial lipopolysaccharide. J Biol Chem 276:38044–38051
Infect Immun 70:335–344 Vogel U, Claus H, Frosch M (2000) Rapid serogroup switching in Neisseria
Tully J, Viner RM, Coen PG, Stuart JM, Zambon M, Peckham C, Booth C, Klein N, meningitidis. N Engl J Med 342:219–220
Kaczmarski E, Booy R (2006) Risk and protective factors for meningococcal Weichselbaum A (1887) Ueber die Aetiologie der akuten Meningitis cerebro-
disease in adolescents: matched cohort study. BMJ 332:445–450 spinalis. Fortschr Med 5:573–583
Tunkel AR, Hartman BJ, Kaplan SL, Kaufman BA, Roos KL, Scheld WM, Whitley RJ Weisfelt M, van de Beek D, Spanjaard L, Reitsma JB, de Gans J (2006) Clinical
(2004) Practice guidelines for the management of bacterial meningitis. Clin features, complications, and outcome in adults with pneumococcal menin-
Infect Dis 39:1267–1284 gitis: a prospective case series. Lancet Neurol 5:123–129
Turner PC, Southern KW, Spencer NJ, Pullen H (1990) Treatment failure in Whitney CG, Farley MM, Hadler J, Harrison LH, Bennett NM, Lynfield R, Reingold
meningococcal meningitis. Lancet 335:732–733 A, Cieslak PR, Pilishvili T, Jackson D, Facklam RR, Jorgensen JH,
Uchiyama S, Carlin AF, Khosravi A, Weiman S, Banerjee A, Quach D, Hightower Schuchat A (2003) Decline in invasive pneumococcal disease after the introduc-
G, Mitchell TJ, Doran KS, Nizet V (2009) The surface-anchored NanA tion of protein-polysaccharide conjugate vaccine. N Engl J Med 348:1737–1746
protein promotes pneumococcal brain endothelial cell invasion. J Exp Med WHO (2012) Global alert and responses. http://www.who.int/csr/en
206:1845–1852 Wippel C, Fortsch C, Hupp S, Maier E, Benz R, Ma J, Mitchell TJ, Iliev AI
Unkmeir A, Latsch K, Dietrich G, Wintermeyer E, Schinke B, Schwender S, Kim KS, (2011) Extracellular calcium reduction strongly increases the lytic capacity
Eigenthaler M, Frosch M (2002) Fibronectin mediates Opc-dependent inter- of pneumolysin from streptococcus pneumoniae in brain tissue. J Infect Dis
nalization of Neisseria meningitidis in human brain microvascular endothe- 204:930–936
lial cells. Mol Microbiol 46:933–946 Wolfgang M, Lauer P, Park HS, Brossay L, Hebert J, Koomey M (1998) PilT
van de Beek D, Farrar JJ, de Gans J, Mai NT, Molyneux EM, Peltola H, Peto TE, mutations lead to simultaneous defects in competence for natural transfor-
Roine I, Scarborough M, Schultsz C, Thwaites GE, Tuan PQ, Zwinderman AH mation and twitching motility in piliated Neisseria gonorrhoeae. Mol
(2010) Adjunctive dexamethasone in bacterial meningitis: a meta-analysis of Microbiol 29:321–330
individual patient data. Lancet Neurol 9:254–263 Wolfgang M, van Putten JP, Hayes SF, Koomey M (1999) The comP locus of
van Deuren M, Brandtzaeg P, van der Meer JW (2000) Update on meningococcal Neisseria gonorrhoeae encodes a type IV prepilin that is dispensable for pilus
disease with emphasis on pathogenesis and clinical management. Clin biogenesis but essential for natural transformation. Mol Microbiol
Microbiol Rev 13:144–166, table of contents 31:1345–1357
van Ginkel FW, McGhee JR, Watt JM, Campos-Torres A, Parish LA, Briles DE Wu YD, Chen LH, Wu XJ, Shang SQ, Lou JT, Du LZ, Zhao ZY (2008) Gram stain-
(2003) Pneumococcal carriage results in ganglioside-mediated olfactory specific-probe-based real-time PCR for diagnosis and discrimination of
tissue infection. Proc Natl Acad Sci USA 100:14363–14367 bacterial neonatal sepsis. J Clin Microbiol 46:2613–2619
van Hoek AJ, Andrews N, Waight PA, George R, Miller E (2012) Effect of serotype Yazdankhah SP, Caugant DA (2004) Neisseria meningitidis: an overview of the
on focus and mortality of invasive pneumococcal disease: coverage of dif- carriage state. J Med Microbiol 53:821–832
ferent vaccines and insight into non-vaccine serotypes. PLoS One 7:e39150 Zhang Q, Li Y, Tang CM (2010) The role of the exopolyphosphatase PPX in
van Ulsen P, Tommassen J (2006) Protein secretion and secreted proteins in avoidance by Neisseria meningitidis of complement-mediated killing. J Biol
pathogenic Neisseriaceae. FEMS Microbiol Rev 30:292–319 Chem 285(44):34259–34268, PMID 20736171

View publication stats

You might also like