Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

29 Pathogenesis and 

Pathophysiology
of Bacterial Infections of the CNS
Loic Le Guennec1,2 and Sandrine Bourdoulous1
1
Inserm, U1016, Institut Cochin, CNRS, UMR8104, Paris University, Paris, France
2
La Pitie‐Salpetriere Hospital, APHP, Paris, France

PCR polymerase chain reaction


Abbreviations
Pcho Phosphorylcholine
pIgR polymeric immunoglobulin
ACP alpha C protein
receptor
β2AR beta‐2‐adrenergic receptor
PMNL polymorphonuclear leukocyte
BBB blood‐brain barrier
SCID severe combined
CNS central nervous system
immunodeficient
CSF cerebrospinal fluid
SOL space‐occupying lesion
E. coli Escherichia coli
Sfb streptococcal
EMPRINN extracellular matrix metallo­
fibronectin‐binding
proteinase inducer for
S. pneumoniae Streptococcus pneumoniae
N. meningitidis
Srr serine‐rich repeat
EoD early‐onset disease
T4P type IV pili
fHBP factor H‐binding protein
TNF‐α tumor necrosis factor alpha
GBS group B Streptococcus
TRL toll‐like receptor
H. influenzae Haemophilus influenzae
ZO1 zona occludens 1
HvgA hypervirulent GBS adhesin
IL interleukin
ICP intracranial pressure Introduction
L. monocytogenes Listeria monocytogenes
LoD late‐onset disease Bacterial infections of the CNS pose a unique
LTA lipoteichoic acid ­challenge to physicians because of their potential
N. meningitidis Neisseria meningitidis morbidity and mortality and inherent difficulties
MMP matrix metalloproteinase involved in their treatment. Rapid diagnosis and
MRI magnetic resonance image prompt treatment are mandatory, but despite
M. tuberculosis Mycobacterium tuberculosis advances in  antibacterial therapy and vaccines,
PAF platelet‐activating factor complications frequently occur, resulting in an

Infections of the Central Nervous System: Pathology and Genetics, First Edition. Edited by Fabrice Chrétien,
Kum Thong Wong, Leroy R. Sharer, Catherine (Katy) Keohane and Françoise Gray.
© 2020 John Wiley & Sons Ltd. Published 2020 by John Wiley & Sons Ltd.

279
Infections of the Central Nervous System

unfavorable outcome in a high proportion of Acute meningitis


patients and neurological sequelae in survivors [1]. Acute meningitis is the most frequent and life‐
The most important infections are meningitis, threatening bacterial infection of the CNS, charac­
encephalitis, and brain abscesses. They differ con­ terized by an acute purulent infection of the
siderably from systemic infections, due to the par­ meninges [5]. It is usually caused by Streptococcus
ticular structure and function of the CNS. pneumoniae, Neisseria meningitidis, or Haemophilus
Structurally, the CNS is covered by three protective influenzae in children and adults and by Escherichia
membranes: the skull, the spine, and the meninges. coli and group B Streptococcus (GBS) during the
Additionally the blood‐brain barrier (BBB) consti­ neonatal period. Listeria monocytogenes can affect
tutes a selective filter that controls the entry of toxic neonates and patients who are immunocompro­
compounds or pathogens [2]. The presence of a mised. The main route of infection is hematoge­
lymphatic vasculature within the brain is still con­ nous, but the responsible bacteria can also spread
troversial [3]. Consequently, leukocytes (i.e. granu­ contiguously from adjacent tissues or gain entry as
locytes and T and B cells) stay within blood vessels a complication of trauma, surgery, or developmen­
and usually do not enter healthy brain tissue. tal malformations. Usually, patients present with
Within the CNS, host defenses rely on innate fever associated with acute or subacute meningeal
immune cells including parenchymal (microglia) syndromes (i.e. headache, stiff neck, Kernig and
and nonparenchymal macrophages and perivascu­ Brudzinski signs, nausea, vomiting, and phono‐ or
lar blood‐derived monocytes [4]; these defenses photophobia) [5].
are not sufficient to control bacterial proliferation Assuming clinical guidelines for safe lumbar punc­
and pathogenicity. During CNS infection or dis­ ture are followed, in a patient with suspected menin­
ease, peripheral blood immune cells cross the BBB gitis, rapid examination of the cerebrospinal fluid
and reach the nervous system parenchyma. The (CSF) is the cornerstone of diagnosis. Typical CSF
host inflammatory response then plays a central examination for bacterial meningitis shows a cloudy
role in contributing to local complications. and turbid fluid with pleocytosis (white blood cell
Depending on the pathogen involved, systemic count >10 cells/mm3) with marked predominance of
complications such as septic shock, pneumonia, polymorphonuclear leukocytes (PMNL) (>50%), low
and disseminated intravascular coagulation also or absent glucose (<40% of serum glucose), and an
contribute to a poor outcome. elevated protein levels (>50 mg/dl). Direct micros­
In this chapter, we will briefly review the most copy with appropriate stains for organisms, followed
common clinical and pathological features of bac­ by culture and antibiotic sensitivity tests, allow
terial CNS infections. identification of the responsible bacteria. Blood
­
­culture is also useful, especially when CSF sampling is
precluded by significant raised ICP.
Clinical presentation of bacterial
infections of the CNS Chronic meningitis
Chronic meningitis is defined as an inflammatory
The clinical features of CNS infections depend on CSF profile and meningitis symptoms that persist
the bacterial agent, the site of infection, and host for at least one month. Worldwide, the main caus­
characteristics, such as underlying risk factors and ative pathogen is Mycobacterium tuberculosis, the
immune status. In a patient who is febrile, the com­ agent responsible for tuberculosis, which contin­
mon neurologic manifestations suggesting CNS ues to cause high morbidity and mortality. It is fre­
infection are disturbance of consciousness, behav­ quently associated with spinal tuberculosis and
ioral changes, headache, symptoms of raised CSF analysis reveals elevation of PMNL early in
intracranial pressure (ICP), seizures, and focal the infection, later followed by lymphocytic men­
neurologic deficits. Overall, fever associated with ingitis with high CSF protein. Early detection of
neurological impairment should suggest one of the M. tuberculosis in the CSF of patients with
following medical emergencies. ­tuberculous meningitis still remains a challenge

280
Pathogenesis and Pathophysiology of Bacterial Infections of the CNS  Chapter 29

for clinicians, mainly because of the lack of rapid,


efficient and practical detection methods. Isolation
of M. tuberculosis from the CSF is complicated as
culture is unacceptably slow and requires special­
ized laboratory safety procedures (see Chapter 35).
Polymerase chain reaction (PCR) assays were
found to be insensitive at detecting M. tuberculosis
in CSF samples, and interferon gamma (IFN‐γ)
release assays are not recommended for diagnosis
of active tuberculous disease [6].
Less commonly, other intracellular or extracel­
lular bacteria can cause chronic meningitis. These
include Brucella spp., which causes brucellosis (see
Chapter  38); Treponema pallidum, the causative
agent of syphilis (see Chapter  37); and Borrelia
burgdorferi, which causes Lyme disease (see
Chapter  38). For these pathogens, CSF analysis
usually shows a lymphocytic meningitis, and direct
microscopy and culture are negative because they
cannot be cultivated in culture media. Diagnosis is Figure 29.1  Axial computed tomography (CT) brain scan
showing right frontal sinusitis in an adult with past medical
mostly based on the patient’s history, positive serol­
history of open right frontal bone fracture in childhood,
ogy, and extraneurological symptoms.
complicated by right frontal lobe brain abscess. Bacterial culture
In all cases, repeat CSF examination is required of the abscess was positive for Streptococcus pneumoniae.
over time to survey bacterial infection and inflam­
mation within the meninges.
The abscess location depends on the site of the
Brain abscess initial infective focus: frontotemporal regions for
Abscess is the second‐most frequent infection of the sinuses, cerebellar and temporal regions for the
the CNS and the most frequent infective cause of middle ear, and frontoparietal regions for hema­
intracranial space‐occupying lesion (SOL). When togenous spread. Post‐traumatic abscesses occur at
formed, it is a focal brain parenchymal suppura­ the sites of craniocerebral injury. Multiple abscesses
tive infection, generally enclosed by a capsule of tend to be found at the gray and white matter junc­
granulation tissue. At the early stage, following tions and should suggest an embolic cause, such as
local vascular injury after inoculation of bacteria, an infective endocarditis (Figure 29.2).
brain abscess is characterized by cerebritis with Neurological examination shows clinical signs of
PMNL infiltration, edema, and a perivascular raised ICP associated with focal neurologic signs
fibrinous exudate causing raised ICP. The sequen­ (i.e. seizures, alteration of behavior or conscious­
tial changes in the following days are described in ness, sensorimotor deficit). Forty percent of
detail in Chapter 31. patients have fever and only 25% have a meningeal
In 75% of cases, the abscess is single. Micro­ syndrome. Magnetic resonance imaging (MRI)
organisms reach the brain (i) from an adjacent cranial shows a ring‐enhancing SOL associated with
or extracranial focus of infection, (ii) from distant foci edema and mass effect. Imaging can also demon­
via the bloodstream, or (iii) when introduced by strate complications such as subdural empyema or
trauma (Figure  29.1) or via a congenital defect (see epidural abscess. In the presence of significant
Chapter 31 for more details). Infections of paranasal raised ICP, lumbar puncture is contraindicated.
sinuses, ears, lungs, and odontogenic foci were Neurosurgical drainage and antibiotic therapy fol­
regarded as the sources of infection in approximately lowing microbiological analysis are usually neces­
70% in a series of 45 cases [7]. sary for treatment.

281
Infections of the Central Nervous System

Figure 29.2  Brain magnetic resonance image (MRI) (axial plane, T2 fluid attenuated inversion recovery [FLAIR] sequence) showing
multiple embolic abscesses within the right frontal lobe, the basal ganglia, and the brainstem in a patient with Streptococcus pneumoniae
endocarditis.

The causative agent depends on the primary site surgery) or by bacteremia (Figure 29.3) because of
of infection and on host factors, particularly the distant‐site infections (endocarditis, furuncle, or
immune status. Immunosuppression is a well‐ soft‐tissue abscess) [8].
established risk factor and is reported in 80% of The clinical picture is similar to spinal cord com­
patients. The most common pathogens are anaero­ pression with intense spinal pain, radicular signs,
bic streptococci (e.g. Streptococcus milleri) and and paraparesis associated with sensory, bladder,
anaerobic Gram‐negative bacilli (e.g. Bacteroides and bowel sphincter disturbance. MRI shows a
fragilis). In patients who are immunocompetent, biconvex extra‐axial lesion outlined by the bony
when the primary source is a dental infection, a skull or vertebra. On T1‐weighted sequences the
mixed growth of bacteria is commonly found in
the brain abscess. In patients who are immuno­
compromised, other species such as Actinomyces
israelii and Nocardia spp. can be responsible for
brain abscesses. L. monocyogenes can also cause
abscesses within the brainstem with or without
meningitis. Finally, intracranial tuberculous
abscesses (tuberculomas) are an uncommon mani­
festation of CNS tuberculosis. The initial empirical
antibiotic treatment depends on the immunologi­
cal status of the host, adjusted following bacterial
identification and results of sensitivity.

Epidural abscess
Infection of the epidural space is rare. It is located
in the virtual space between the skull (or the spine)
and the outer layer of the dura mater. These infec­
tions mainly affect immunocompromised adults Figure 29.3  Cervicomedullary magnetic resonance image (MRI)
older than 50 years of age. The abscess is caused by (sagittal plane, T1 sequence) showing anterior epidural abscess
an adjacent infection (i.e. osteomyelitis, or follow­ contiguous with cervical spondylodiscitis resulting from embolism
ing cranial or spinal trauma, epidural anesthesia, or in a patient with Staphylococcus aureus endocarditis.

282
Pathogenesis and Pathophysiology of Bacterial Infections of the CNS  Chapter 29

lesion is iso‐ or hypodense and hyperdense on T2‐ crossing the BBB transcellularly, paracellularly or
weighted sequences with peripheral or diffuse con­ in infected phagocytes (the so‐called Trojan horse
trast enhancement. Bacterial identification can be mechanism); (ii) by retrograde axonal transport
from blood cultures, from the primary site of infec­ to  the brain via cranial or peripheral nerves; and
tion, from CSF, or by ultrasound‐guided or neuro­ (iii) by direct invasion.
surgically acquired biopsy from the epidural abscess. The most common route of entry to the CNS for
Treatment consists of urgent surgical drainage extracellular bacteria is via the bloodstream. Most
together with broad spectrum antibiotics against pathogens responsible for CNS infections can asymp­
Staphylococcus aureus, Streptococcus spp., and tomatically colonize skin and mucosa of healthy indi­
Gram‐negative rod‐shaped bacteria and anaer­ viduals. These pathogens can reach the bloodstream,
obes. M. tuberculosis can also be responsible for survive within the blood, and ultimately cross the
epidural abscess, as a complication of tuberculosis BBB. Extracellular pathogens that can cause menin­
of the spine (Pott’s disease). gitis are well‐adapted to survive in the extracellular
environment: they can all resist killing by PMNLs or
Subdural empyema complement and can obtain the ferric iron required
The mode of development and bacteriology of for their growth and survival. Sustained bacteremia
subdural empyema is similar to that of epidural seems to be required to cause meningeal invasion [9].
abscess. Subdural empyema is a collection of pus The duration of bacteremia is also important, for
between the dura and the subarachnoid mem­ example in H. influenzae meningitis, both the density
brane. The subdural space is traversed by veins and duration of bacteremia correlate with meningeal
and divided into several large compartments. invasion [10]. Furthermore, autopsies have shown
Subdural empyema is usually caused by aerobic that most extracellular bacteria can interact directly
and anaerobic bacteria, which reach the subdural with brain endothelial cells [11]. A dense bacteremia
space from veins draining the skull. The primary probably increases the likelihood of bacterial interac­
sites of infection are mainly in the sinuses, fol­ tion with the components of CNS barriers, which is a
lowed by mastoid and middle ear infections. prerequisite for meningeal invasion.
Usually, the empyema is located above the tento­ Numerous in vitro studies using brain‐derived
rium or adjacent to the falx cerebri and rarely in endothelial cell lines expressing tight junctions have
the spinal canal. Treatment includes antibiotics investigated how circulating bacteria responsible
and surgical intervention. for meningitis interact with the BBB. These studies
have identified bacterial adhesins and their specific
Ventriculitis endothelial cell receptors such as platelet activating
Ventriculitis generally occurs as a complication of a factor receptor for S. pneumoniae [12] or CD147
previous CNS bacterial infection such as meningi­ (also known as Basigin or extracellular matrix met­
tis or a brain abscess that ruptures into the cerebral alloproteinase inducer [EMPRINN]) for N. menin-
ventricles. The main complication is hydrocepha­ gitidis [13]. Relevant in vivo experimental models
lus. Clinical symptoms are due to raised ICP have been set up to study how pathogens such as E.
because of hydrocephalus. coli, GBS, S. pneumoniae, or N. meningitidis can
reach the CNS, but the precise site or method of
entry by which they cross into the CSF is still
Bacterial invasion of the CNS unknown. The choroid plexus is suggested as one
site where bacteria could cross. If this were the case,
In the past few decades, our knowledge of the bacterial meningitis might be expected to be associ­
mechanisms of bacterial invasion of the CNS has ated with ventriculitis, which is not always present.
greatly improved because of basic research and Another theory suggests bacteria cross the BBB
experimental animal models. through postcapillary perforating venules because
The routes of entry by which bacteria access the of their proximity to the subarachnoid space.
CNS are: (i) hematogenous dissemination, by Bacteria crossing the endothelial monolayer at this

283
Infections of the Central Nervous System

site would be close to the Virchow Robin space. nasopharyngeal mucosa in asymptomatic individu­
Also, the low blood flow in these venules could als but is responsible for otitis media, sinusitis, and
facilitate bacterial adhesion to endothelial cells [11]. for severe invasive infections, such as pneumonia,
There are several hypotheses as to how bacteria bacteremia, and acute meningitis (Figure  29.4).
cross the endothelial cell barrier. One possibility is a Findings regarding the pathophysiology of pneu­
facilitated transport occurring during diapedesis of mococcal meningitis are either derived from brain
infected leukocytes, but extracellular bacteria autopsies or from animal models.
responsible for meningitis probably do not use this To reach the meninges, pneumococci have to
route to cross the BBB because several studies have escape ear‐nose‐throat or respiratory mucosal
shown a specific interaction between bacteria and defenses and either cause sinusitis or mastoiditis,
endothelial cells [13]. Many studies hypothesized with local spread through the skull, or survive
that bacterial adhesion to endothelial cells induces within the bloodstream and be carried into the
intracellular signaling that could cause disruption CNS (Figure 29.5). The pneumococcal capsule pro­
of intercellular tight junctions, allowing bacteria to vides protection against phagocytosis, comple­
cross between or across cells (transcytosis) of the ment‐mediated immunity, and opsonophagocytic
endothelial monolayer. Transcytosis across this killing. The capsule also reduces pneumococcal
monolayer has been demonstrated for S. pneumo- capture by neutrophil extracellular traps and inter­
niae [14], E. coli [15], GBS [16], and through human feres with innate immune responses by masking
umbilical vein endothelial cells for H. influenzae several toll‐like receptor (TLR) ligands associated
[17]. In vitro, although N. meningitidis internalized with the pneumococcal cell wall. These include
in human brain microvascular endothelial cells lipoteichoic acid and lipopeptides. Furthermore, its
[18], the pathway used by meningococci to cross ability to form biofilms is important in naso­
endothelial cells is still debated. Another hypothesis pharyngeal colonization, pneumonia, and otitis
is that bacteria exert a direct cytotoxic effect result­
ing in disruption of the endothelial barrier, but BBB
leakage because of bacterial cytotoxicity might be
expected to cause subarachnoid hemorrhage, which
is a rare event in bacterial meningitis. These patho­
gens seem to respect the architecture of the BBB
[19]. Inflammation may also play a role in bacterial
entry into the CNS. The innate immune response
induced by bacteremia, with or without septic
shock, may increase BBB permeability, thereby pro­
moting bacterial entry into the CNS.
The following section describes the most fre­
quent bacteria responsible for acute meningitis and
their respective pathophysiological mechanisms
leading to mucosal colonization, dissemination,
and crossing of the BBB (Table 29.1).

S. pneumoniae
Figure 29.4  Bronchioalveolar lavage (BAL), stained with
S. pneumoniae (pneumococcus) is an alpha‐hemolytic
May‐Grünwald Giemsa, showing diplococci within altered
Gram‐positive extracellular coccus. It frequently
polymorphonuclear neutrophils in a patient with acute
resides in the upper respiratory tract of humans and community‐acquired pneumonia and clinical meningitis. Blood
other related mammalian species. It is the most cultures were positive for Gram‐positive cocci. The patient also
frequent organism responsible for community‐ had purpura fulminans with coagulopathy, so lumbar puncture
acquired bacterial meningitis worldwide, affecting was contraindicated. BAL and blood cultures were positive for
young children and the elderly. It can colonize the Streptococcus pneumoniae.

284
Table 29.1  Main bacterial pathogens responsible for acute meningitis.

Streptococcus Neisseria Haemophilus


pneumoniae meningitidis Influenzae type b Group B Streptococcus Escherichia coli K1 Listeria monocytogenes

Type of bacteria Gram‐positive Gram‐negative Gram‐negative rod Gram‐positive diplococcus Gram‐negative rod Gram‐positive rod shaped
diplococcus diplococcus shaped shaped
Population Between 2 months Between 2 months and Children <5 years <3 months <3 months <3 months, and adults
affected and 5 years, and 5 years, and young
>50 years adults
Risk factors Splenectomy or Alternative and terminal Insufficient access to Maternal carriage and Maternal carriage and Ingestion of contaminated
functional complement vaccines prematurity prematurity food in pregnant women
hyposplenia pathway deficiencies or patients with
immunocompromise
Carriage site/ Nasopharynx, lung Nasopharynx Nasopharynx Rectal or vaginal Rectal Digestive tract
Site of entry
Virulence factors Capsule, CbpA, Capsule, LOS, Type IV Capsule, gA1 protease Capsule, lipotechoic acid, Capsule, O‐LPS, OmpA, Capsule, InIA, InIB,
PCho (PAF), pili LOS, Hib Type I pili, ScpB, type 1 fimbriae, Listeriolysin O
pneumolysin Omp FbsA, b‐hemolysin CNF1
Host cell pIgR, laminin CD147, β2‐adrenergic Mucin, Lactoferrin Fibronectin, fibrinogen Laminin receptor E‐cadherin, Met
receptors receptor, PAF receptor and laminin
receptor

CbpA, Choline‐binding protein A; CNF1, cytotoxic necrotizing factor 1; LOS, lipooligosaccharide; LPS, lipopolysaccharide; Omp, outer member protein; PAF, platelet‐activating factor;
PCho, phosphorylcholine (mimics the chemokine PAF); plgR, polymeric immunoglobulin receptor.

0004466174.INDD 285 18­12­2019 13:29:05


Infections of the Central Nervous System

Autolysin LytA
Streptococcus and Pneumolysin release
pneumoniae

PCho
CbpA
PAFr
pIgR Laminin
Blood

Endothelial cell

Cell wall degradation


Brain

Transcellular pathway
Paracellular pathway

Legend:

Streptococcus pneumoniae Platelet-activating factor receptor (PAFr) Junction disruption

Autolysin LytA and Pneumolysin Polymeric immunoglobulin receptor (pIgR)

Phosphorylcholine (PCho) Laminin

Figure 29.5  Mechanism of CNS invasion by Streptococcus pneumoniae. S. pneumoniae adheres to endothelial cells by the bacterial
choline‐binding protein CbpA (also called PspC, SpsA or Hic), which binds to human cell laminin and polymeric immunoglobulin receptor
(pIgR), and bacterial phosphorylcholine (PCho), which mimics the chemokine platelet‐activating factor (PAF) and binds to the cell receptor
PAF receptor (PAFr), enabling transcytosis across the endothelium. Additional release of pneumococcus autolysin LytA and pneumolysin
promote cell wall degradation and endothelial monolayer disruption, allowing paracellular bacterial invasion.

media. Pneumolysin, a key virulence factor a polymeric immunoglobulin receptor (pIgR) at


secreted by S. pneumoniae, binds to cholesterol in the cell surface of nasopharyngeal or respiratory
cell membranes. This pore‐forming toxin, pro­ epithelial cells. This interaction promotes bacte­
duced by virtually all clinical isolates of S. pneumo- rial transcytosis across the epithelial barrier [20].
niae, contributes significantly to its virulence by Once in the bloodstream, bacterial CbpA binds
inducing tissue damage and plays a role in biofilm brain endothelial cells through laminin receptor.
formation [20]. Pneumoccocci can also anchor to human epithe­
The first step in pneumococcal invasion is the lial and endothelial cells through an interaction
interaction between the bacterial choline‐binding between bacterial phosphorylcholine (PCho),
protein CbpA (also called PspC, SpsA or Hic) and which mimics the chemokine platelet‐activating

286
Pathogenesis and Pathophysiology of Bacterial Infections of the CNS  Chapter 29

factor (PAF) and binds its cell receptor (PAF carriage in healthy individuals in 8‐ 20% of the popu­
receptor [PAFr]), the first known host cell recep­ lation and up to 25% in children [24]. Meningococci
tor for S. pneumoniae. This interaction promotes are transmitted by respiratory secretions, saliva, or by
bacterial transcytosis, through a clathrin‐medi­ inhalation of contaminated aerosol droplets.
ated endocytosis in brain endothelial cells [20]. Nasopharyngeal acquisition may be transient, lead to
Pneumococcus has an additional important prop­ colonization, or result in invasive disease. Blood‐
erty of undergoing autolysis, characterized by cell borne infections can rapidly progress to septic shock.
wall degradation by a peptidoglycan hydrolase As meningococci adhere to, and proliferate in, sys­
(autolysin Lyt A), leading to pneumolysin release temic microvessels, vascular colonization leads to
(and its associated virulence). In a microvascular abnormal inflammation and coagulation, endothe­
endothelial cell culture model, pneumococci lial dysfunction and, in severe cases, to vascular leak­
expressing pneumolysin could breach endothelial age associated with the typical extensive necrotic
cells, whereas mutant pneumococci deficient in purpuric rash. Interaction with brain microvessels
pneumolysin could not [21]. This effect on brain allows bacteria to cross the BBB, reach the meninges,
endothelial cells could potentially induce apopto­ and cause meningitis (Figure  29.6), while brain
sis and disruption of BBB, favoring bacterial entry endothelial cell junctions are relatively intact with no
into the CNS. Pneumolysin impairs PMN leuko­ signs of hemorrhage or thrombosis [13].
cyte activity and also activates different proin­ The prerequisite for meningococcal pathogenic­
flammatory signaling pathways, which may ity is its interaction with the nasopharyngeal epi­
accentuate brain inflammation and neuronal thelial surface and transfer across this mucosal
damage. Both CbpA and pneumolysin seem barrier to reach the circulation [25]. Mucosal colo­
essential for pneumococcal pathogenesis. In mice, nization and crossing mechanisms are poorly
immunization against CbpA and pneumolysin described. Once in the bloodstream, many viru­
can protect against pneumococcal infection and lence factors enable bacterial growth and immune
prevent nasopharyngeal carriage [22]. Finally, escape from host effectors and the complement
nasal cavity infection in a mouse model suggests system. They include the iron chelation systems,
that pneumococci may enter the brain directly by the polysaccharide capsule, the sialylated lipooligo­
axonal transport through olfactory nerves [23]. saccharide, and factor H‐binding protein (fHBP).
Teichoic acid in the pneumococcal cell wall can Pathogenesis is also linked to some bacterial
interact with gangliosides, so it has been sug­ genetic factors (e.g. expression of a gene encoding a
gested that pneumococcal carriage can induce hemoglobin receptor and the expression of a fila­
olfactory tissue infection and bacterial entry into mentous prophage were detected at a higher fre­
the brain in the absence of bacteremia. However, quency in pathogenic isolates than in carriage
these colonizing bacteria fail to cause extensive isolates) [26].
brain infection. Whether these events occur in However, meningococcal pathogenicity princi­
humans remains to be proven, so further studies pally relies on the capacity to interact with human
are required to determine the precise pathophysi­ microvessels. This was elegantly demonstrated in
ology of pneumococcal meningitis. vivo in a severe combined immunodeficient
(SCID) mouse model, grafted with human skin.
N. meningitidis The grafted human vessels connected with the
N. meningitidis (meningococcus) is an extracellular mouse vasculature and remained functional.
Gram‐negative diplococcus. It is one of the most Following infection with N. meningitidis, circulat­
virulent bacteria, responsible for 10% of meningitis ing meningococci specifically colonized human
deaths. Systemic complications, such as septic shock, endothelial cells within the grafts and promoted
purpura fulminans, and disseminated intravascular vascular lesions similar to purpuric lesions in
coagulation, contribute to the unfavorable outcome. humans [27]. Mouse vessels were not colonized
N. meningitidis is restricted to humans; it is a fre­ and remained intact. The model also demonstrated
quent nasopharynx commensal with asymptomatic that pathogenic capsulated meningococci required

287
Infections of the Central Nervous System

Neisseria
meningitidis

Type IV Pili Type IV Pili

CD147 β2AR
Blood

Endothelial cell
Intercellular junction
Brain disruption

Paracellular pathway
Legend:

Neisseria meningitidis CD147 Junction disruption

Type IV Pili B2-Adrenergic receptor (β2AR)

Figure 29.6  Mechanism of CNS invasion by Neisseria meningitidis. Type IV pili (T4P) of N. meningitidis interact with endothelial cell
receptor CD147, leading to the activation of both β2‐adrenergic receptor (β2AR) and downstream signaling cascades. In vitro, infection
promotes the disruption of intercellular junctions, which could favor paracellular bacterial entry into the CNS across the blood‐brain barrier.

type IV pili (T4P) to colonize and induce vascular At the molecular level, to colonize human
damage. T4P are long dynamic filamentous struc­ endothelium, meningococcal T4P interact with
tures extending from the bacterial surface through two key endothelial cell receptors: CD147 (also
the capsule. Nonpiliated mutants failed to target called EMMPRIN or Basigin), a member of the
human graft vessels or induce vascular damage immunoglobulin superfamily, and endothelial
[27]. This model also showed that the vascular cell beta‐2‐adrenergic receptor (β2AR) leading to its
wall provided an environment for meningococcal bias activation [25]. β2AR activation promotes
multiplication promoting a sustained bacteremia local recruitment and activation of cytoskeleton‐
and mouse lethality. These studies highlighted the associated and signaling proteins. This results in
importance of pilus‐mediated vascular coloniza­ (i) forming protrusions of the apical plasma
tion in the development and outcome of meningo­ membrane, which stabilize bacterial colonies at
coccal sepsis. the endothelial cell surface (Figure  29.7), and

288
Pathogenesis and Pathophysiology of Bacterial Infections of the CNS  Chapter 29

Scanning electron microscopy Nuclei CD147

5 µm

Ezrin Actin Merge

10 µm

Figure 29.7  Mechanism of CNS invasion by Neisseria meningitidis. Right panel: Upon adhesion to human brain endothelial cells
(hCMEC/D3 cells) meningococci proliferate at the endothelial cell surface forming compact microcolonies observed by scanning electron
microscopy (left panel). Subsequently, local membrane protrusions form which stabilize bacterial colonies at the endothelial cell surface
upon shear stress. The protrusions result from local changes in endothelial cytoskeletal architecture. Immunofluorescence microscopy
(right panels) shows surface microcolonies, with local accumulation of the endothelial cell receptor CD147, of the membrane‐cytoskeleton
linker Ezrin and of cortical actin polymerization, all involved in protrusion formation.

(ii) ­opening of intercellular ­junctions that con­ crosses the BBB by disrupting cell‐cell junctions.
tribute to vascular changes [25]. Colonization In vitro studies indicate that meningococcal
also favors endothelial cell survival, the expres­ ­adhesion to human brain endothelium induces
sion of adhesion molecules, and the secretion of signaling events leading to altered endothelial
cytokines [28]. permeability. N. meningitidis actively disrupts
Although the affinity of T4P for CD147 and cell‐cell junctions and promotes sequestration of
β2AR is weak, these receptors are assembled into cell‐cell junction molecules, such as VE‐cadherin,
multimers [29], and CD147 expression on brain zona occludens 1 (ZO1), or claudin 5, to the sites
endothelial cells is strong, thus enhancing of bacterial adhesion [25]. Meningococcus also
­avidity [13]. The low levels of shear stress in the activates matrix metalloproteinase 8 (MMP8)
cerebral microcirculation also favor bacterial production, which promotes proteolytic cleavage
adhesion [11]. Other meningococcal factors can of the tight junction protein occludin, favoring
reinforce or modulate adhesion (detailed cell detachment. Because N. meningitidis infec­
in [30]). tion is specific to humans, there is no relevant in
Colonization of brain capillaries in the suba­ vivo animal model of CNS invasion to test
rachnoid space, the parenchyma and the choroid whether the same process occurs in vivo at the
plexus suggested that N. meningitidis actively human BBB.

289
Infections of the Central Nervous System

GBS because of small numbers of alveolar macrophages


GBS is a Gram‐positive extracellular coccus and is and PMNL and low levels of immunoglobulin A/
the main cause of bacterial meningitis in babies immunoglobulin G (IgA/IgG). The most common
younger than two months of age. Contamination route of entry is inhalation or ingestion of amniotic
usually occurs by maternal‐fetal transmission from fluid contaminated by the maternal genital tract,
the birth canal at the time of delivery. Mothers are which itself can be contaminated by pelvic exami­
frequently asymptomatic. About 30–70% of neo­ nation during the peripartum period. Bacteria then
nates born to mothers with GBS‐colonization cross the fetal respiratory or gastrointestinal
become transiently colonized by their mother’s mucosa, proliferate in the blood, and enter the CNS
organisms. Neonates, particularly if premature, (Figure  29.8). GBS is responsible for early‐onset
have an increased risk for bacterial meningitis disease (EoD), typically occurring within the first

Group B
Streptococcus
Srr2
ST-17

Plasminogen

FbsA HvgA
PilA
Fibrinogen Glycosamino-
Sfb Collagen
Fibronectin glycan α2β1Integrin
Blood

Endothelial cell

β-hemolysin Cell wall degradation


Brain

Transcellular pathway
Paracellular pathway
Legend:

Group B Streptococcus ST-17 Srr2 Glycosaminoglycan Junction disruption

β-hemolysin Plasminogen Collagen

FbsA Fibrinogen PilA


Sfb
Fibronectin A2β1 Integrin
HvgA

Figure 29.8  Mechanism of CNS invasion in Group B Streptococcus (GBS) meningitis. GBS adheres to brain endothelial cells through
interaction of the adhesins Srr2, HvgA, and FbsA with components of the extracellular matrix (ECM). GBS adhesin pilA binds to collagen
and indirectly interacts with α2β1 integrins at the endothelial cell surface. Following interaction, GBS can invade brain endothelial cells,
allowing transcellular crossing of the blood‐brain barrier (BBB) by an unknown mechanism. Additional release of β‐hemolysin promotes
cell wall degradation and endothelial monolayer disruption, allowing bacterial invasion through a paracellular pathway.

290
Pathogenesis and Pathophysiology of Bacterial Infections of the CNS  Chapter 29

week of life, with pneumonia, bacteremia, and passage. In addition, GBS β‐hemolysin/cytolysin is
sometimes meningitis. Late‐onset disease (LoD) directly cytolytic for human brain endothelial cells
occurs in infants up to two months of age, with and may contribute to BBB disruption.
fewer symptoms related to bacteremia and a higher
incidence of meningitis. A single clone, GBS ST‐17, E. coli
is strongly associated with LoD [31]. E. coli is an extracellular Gram‐negative bacillus. It
GBS possesses multiples adhesins [31], includ­ is the second cause of meningitis in neonates and
ing lipoteichoic acid (LTA), pili, serine‐rich repeat the leading cause in premature infants [34]. Rectal
(Srr) proteins, streptococcal fibronectin‐binding carriage of E. coli in women of childbearing age is
protein (Sfb), the surface protein hypervirulent up to 50%. Maternal‐fetal colonization affects about
GBS adhesin (HvgA), found in hypervirulent 70% of their newborns, without necessarily causing
ST‐17 strains, and alpha C protein (ACP; encoded pathogenicity. About 80% of E. coli strains isolated
by bca), which binds to glycosaminoglycan. These from pregnant women and newborns possess the
components enable GBS to interact with various K1 capsular polysaccharide antigen that protects
human cell types, including vaginal epithelium, the bacterial cell from immune attack. E. coli K1
placental membranes, airways, and brain endothe­ meningitis starts with gastrointestinal mucosa colo­
lial cells. Adhesion to epithelial cells is mediated by nization, followed by invasion, survival, and prolif­
lipoteichoic acid associated with the bacterial wall, eration within the blood [35]. E. coli K1 can induce
and by GBS surface proteins. The pilus protein high‐level bacteremia [35]. At the brain level, E. coli
PilA, Srr1, Sfb, and ACP also mediate GBS interac­ K1 binds to cerebral capillaries via bacterial surface
tion with basement membrane components. GBS structures such as OmpA and type 1 fimbriae. The
binds to extracellular matrix components, includ­ interaction induces host cell actin cytoskeleton
ing fibronectin, fibrinogen, and laminin, which rearrangement and microvilli‐like protrusions that
facilitate mucosal colonization [31]. This interac­ facilitate bacterial internalization. E. coli K1 is then
tion is mediated by ScpB, a homolog of the Lra1 stored in cell vacuoles and reaches the subarach­
adhesin family, and by FbsA, a surface‐anchored noid space after its release at the basal membrane of
protein. ST‐17 strains specifically express Srr2, a microvascular endothelial cells.
cell wall‐anchored protein, which binds plasmino­
gen and plasmin, increases bacterial survival from L. monocytogenes
phagocytic killing and bacterial persistence in a L. monocytogenes is a facultative intracellular
murine model of meningitis. This suggests that Gram‐positive bacillus, which can survive at freez­
Srr2 hijacks ligands of the host coagulation system, ing temperatures and proliferates at very low
contributing to GBS dissemination, invasiveness, (refrigeration) temperatures. For this reason, it is
and ultimately to meningitis [32]. associated with foodborne illness. L. monocy-
GBS has also developed strategies to penetrate togenes crosses the intestinal and fetoplacental
and cross host cell barriers and to avoid immuno­ barriers via the interaction of bacterial proteins
logical clearance and inflammatory response InIA and InIB with cell surface proteins E‐cadherin
because its sialylated capsular polysaccharide limits and Met respectively [36]. L. monocytogenes
host complement activation and phagocytosis. spreads through the CNS by the diapedesis of
Animal models and in vitro culture systems have infected monocytes (Trojan horse mechanism)
shown that GBS can invade brain endothelial cells, across the BBB, and by direct invasion of endothe­
supporting the transcellular theory of BBB crossing. lial cells [36]. After entering endothelial cells, the
Recent data support a Snail1‐dependent mecha­ bacteria escape from phagosomes via a hemolysin
nism of BBB disruption and penetration [33]. GBS called listeriolysin O or LLO, a pore‐forming mol­
can promote the induction of host transcriptional ecule. Once in the cytoplasm, L. monocytogenes
repressor Snail1, which impedes expression of tight highjacks actin cytoskeleton of the infected cell for
junction genes, facilitating tight junction disruption its own propulsion (comet tails), and spreads in
and promoting BBB permeability to allow bacterial adjacent cells. Direct intra‐axonal spread along

291
Infections of the Central Nervous System

lower cranial nerves and within the brain has been


demonstrated in animal models and may also be
Conclusions
important in human listeria brainstem and rhom­
Many clinical syndromes are encountered in CNS
boencephalitis [37].
infections. Accurate diagnosis and prompt treat­
ment are mandatory. The pathophysiology is com­
Inflammation plex, contributed to by both direct bacterial damage
and the host inflammatory response. A full under­
Inflammation plays a central role during bacterial standing of these infections may help the design of
CNS infection. Although mandatory for host pro­ new therapies to block bacterial invasion and
tection against pathogens, inflammation is also reduce inflammation response‐induced injuries.
responsible for several crucial steps of bacterial
invasion and inflammation‐induced adverse
events, such as BBB rupture, CSF cytokine produc­ References
tion, leukocyte invasion in the CSF, and neuronal
injury. 1. Leber, A.L., Everhart, K., Balada‐Llasat, J.M. et  al.
A wide range of innate sensors are expressed by (2016). Multicenter evaluation of biofire filmarray
all neuronal and nonneuronal cells within the CNS. meningitis/encephalitis panel for detection of bacteria,
viruses, and yeast in cerebrospinal fluid specimens.
Locally, in response to bacterial invasion, endothe­
J. Clin. Microbiol. 54: 2251–2261.
lial, glial, and resident immune cells produce pro­
2. Weiss, N., Miller, F., Cazaubon, S., and Couraud, P.O.
inflammatory factors, including the cytokines (2009). Biology of the blood‐brain barrier: Part I. Rev.
tumor necrosis factor alpha (TNF‐α), interleukin Neurol. (Paris) 165: 863–874.
(IL)‐1β, IL‐8/CXCL‐8, and IL‐6. These initiate an 3. Aspelund, A., Antila, S., Proulx, S.T. et al. (2015). A dural
inflammatory cascade and attract immune cells. lymphatic vascular system that drains brain interstitial
Matrix metalloproteinases (MMPs) are also pro­ fluid and macromolecules. J. Exp. Med. 212: 991–999.
duced, which degrade the extracellular matrix, 4. Prinz, M. and Priller, J. (2017). The role of peripheral
allow cell migration, and increase BBB permeabil­ immune cells in the CNS in steady state and disease.
ity [38]. In the inflammatory state, adhesion mole­ Nat. Neurosci. 20: 136–144.
cules such as VCAM‐1 and ICAM‐1 are expressed 5. van de Beek, D., de Gans, J., Tunkel, A.R., and
Wijdicks, E.F. (2006). Community‐acquired bacterial
at the endothelial cell surface, facilitating leukocyte
meningitis in adults. N. Engl. J. Med. 354: 44–53.
adhesion [39]. PMNL are locally recruited in the
6. Wang, Y.Y. and Xie, B.D. (2018). Progress on diagnosis
acute phase, and a mix of T and B cells, monocytes, of Tuberculous Meningitis. Methods Mol. Biol. 1754:
and dendritic cells in the chronic stage; all are 375–386.
required for microbial control. Several portals of 7. Chun, C.H., Johnson, J.D., Hofstetter, M., and Raff,
entry into the brain exist for inflammatory cells, M.J. (1986). Brain abscess. A study of 45 consecutive
including the fenestrated vasculature within the cases. Medicine (Baltimore) 65: 415–431.
subarachnoid space and the choroid plexus, across 8. Heusner, A.P. (1948). Nontuberculous spinal epidural
epithelial and ependymal cells of the choroid infections. N. Engl. J. Med. 239: 845–854.
plexus or astrocyte endfeet of BBB. 9. Brandt, C.T., Holm, D., Liptrot, M. et al. (2008). Impact
Inflammation affects cerebral blood flow during of bacteremia on the pathogenesis of experimental pneu-
mococcal meningitis. J. Infect. Dis. 197: 235–244.
bacterial infection of the CNS. In acute meningitis,
10. Smith, A.L., Scheifele, D., Syriopoulou, V. et  al.
endothelin, a strong vasoconstrictor, is secreted by
(1982). Haemophilus Influenzae, Epidemiology,
brain endothelial cells, and contributes to cerebral Immunology and Prevention of the Disease. In:
blood flow reduction and ischemia [40]. Reactive Pathogenesis of Haemophilus influenzae meningitis
oxygen species (ROS) and reactive nitrogen species (eds. S.H. Sell and P.F. Wright), 89–109. New‐York:
produced by granulocytes, endothelial cells, and Elsevier Sciences Publishing Co. Inc.
microglial cells are responsible for vascular damage 11. Mairey, E., Genovesio, A., Donnadieu, E. et al. (2006).
within the brain [41]. Cerebral microcirculation shear stress levels determine

292
Pathogenesis and Pathophysiology of Bacterial Infections of the CNS  Chapter 29

Neisseria meningitidis attachment sites along the 25. Coureuil, M., Bourdoulous, S., Marullo, S., and
blood‐brain barrier. J. Exp. Med. 203: 1939–1950. Nassif, X. (2014). Invasive meningococcal disease: a
12. Cundell, D.R., Gerard, C., Idanpaan‐Heikkila, I. et al. disease of the endothelial cells. Trends Mol. Med. 20:
(1996). PAf receptor anchors Streptococcus pneumo- 571–578.
niae to activated human endothelial cells. Adv. Exp. 26. Bille, E., Ure, R., Gray, S.J. et al. (2008). Association of
Med. Biol. 416: 89–94. a bacteriophage with meningococcal disease in young
13. Bernard, S.C., Simpson, N., Join‐Lambert, O. et  al. adults. PLoS One 3: e3885.
(2014). Pathogenic Neisseria utilizes CD147 for vascu- 27. Join‐Lambert, O., Lecuyer, H., Miller, F. et al. (2013).
lar colonization. Nat. Med. 20: 725–731. Meningococcal interaction to microvasculature trig-
14. Ring, A., Weiser, J.N., and Tuomanen, E.I. (1998). gers the tissular lesions of purpura fulminans. J. Infect.
Pneumococcal trafficking across the blood‐brain bar- Dis. 208: 1590–1597.
rier. Molecular analysis of a novel bidirectional path- 28. Schubert‐Unkmeir, A., Sokolova, O., Panzner, U.
way. J. Clin. Invest. 102: 347–360. et al. (2007). Gene expression pattern in human brain
15. Stins, M.F., Badger, J., and Sik Kim, K. (2001). endothelial cells in response to Neisseria meningitidis.
Bacterial invasion and transcytosis in transfected Infect. Immun. 75: 899–914.
human brain microvascular endothelial cells. Microb. 29. Maissa, N., Covarelli, V., Janel, S. et  al. (2017).
Pathog. 30: 19–28. Strength of Neisseria meningitidis binding to endothe-
16. Nizet, V., Kim, K.S., Stins, M. et al. (1997). Invasion of lial cells requires highly‐ordered CD147/beta2‐adreno-
brain microvascular endothelial cells by group B strep- ceptor clusters assembled by alpha‐actinin‐4. Nat.
tococci. Infect. Immun. 65: 5074–5081. Commun. 8: 15764.
17. Virji, M., Kayhty, H., Ferguson, D.J. et  al. (1992). 30. Coureuil, M., Lecuyer, H., Bourdoulous, S., and
Interactions of Haemophilus influenzae with human Nassif, X. (2017). A journey into the brain: insight into
endothelial cells in vitro. J. Infect. Dis. 165 (Suppl 1): how bacterial pathogens cross blood‐brain barriers.
S115–S116. Nat. Rev. Microbiol. 15: 149–159.
18. Nikulin, J., Panzner, U., Frosch, M., and Schubert‐ 31. Tazi, A., Bellais, S., Tardieux, I. et al. (2012). Group B
Unkmeir, A. (2006). Intracellular survival and repli- Streptococcus surface proteins as major determinants for
cation of Neisseria meningitidis in human brain meningeal tropism. Curr. Opin. Microbiol. 15: 44–49.
microvascular endothelial cells. Int. J. Med. Microbiol. 32. Six, A., Bellais, S., Bouaboud, A. et al. (2015). Srr2, a
296: 553–558. multifaceted adhesin expressed by ST‐17 hypervirulent
19. Koedel, U., Scheld, W.M., and Pfister, H.W. (2002). Group B Streptococcus involved in binding to both fibrin-
Pathogenesis and pathophysiology of pneumococcal ogen and plasminogen. Mol. Microbiol. 97: 1209–1222.
meningitis. Lancet Infect. Dis. 2: 721–736. 33. Kim, B.J., Hancock, B.M., Bermudez, A. et al. (2015).
20. Mook‐Kanamori, B.B., Geldhoff, M., van der Poll, T., Bacterial induction of Snail1 contributes to blood‐
and van de Beek, D. (2011). Pathogenesis and patho- brain barrier disruption. J. Clin. Invest. 125:
physiology of pneumococcal meningitis. Clin. 2473–2483.
Microbiol. Rev. 24: 557–591. 34. Ouchenir, L., Renaud, C., Khan, S. et al. (2017). The
21. Zysk, G., Schneider‐Wald, B.K., Hwang, J.H. et  al. epidemiology, management, and outcomes of bacterial
(2001). Pneumolysin is the main inducer of cytotox- meningitis in infants. Pediatrics 140 (pii): e20170476.
icity to brain microvascular endothelial cells caused 35. Xie, Y., Kim, K.J., and Kim, K.S. (2004). Current concepts
by Streptococcus pneumoniae. Infect. Immun. 69: on Escherichia coli K1 translocation of the blood‐brain
845–852. barrier. FEMS Immunol. Med. Microbiol. 42: 271–279.
22. Mann, B., Thornton, J., Heath, R. et al. (2014). Broadly 36. Disson, O. and Lecuit, M. (2013). In vitro and in vivo
protective protein‐based pneumococcal vaccine com- models to study human listeriosis: mind the gap.
posed of pneumolysin toxoid‐CbpA peptide recombi- Microbes Infect. 15: 971–980.
nant fusion protein. J. Infect. Dis. 209: 1116–1125. 37. Antal, E.A., Løberg, E.M., Bracht, P. et  al. (2001).
23. van Ginkel, F.W., McGhee, J.R., Watt, J.M. et  al. Evidence for intraaxonal spread of Listeria monocy-
(2003). Pneumococcal carriage results in ganglioside‐ togenes from the periphery to the central nervous sys-
mediated olfactory tissue infection. Proc. Natl. Acad. tem. Brain Pathol. 11: 432–438.
Sci. U. S. A. 100: 14363–14367. 38. Sellner, J. and Leib, S.L. (2006). In bacterial meningitis
24. Pace, D. and Pollard, A.J. (2012). Meningococcal dis- cortical brain damage is associated with changes in
ease: clinical presentation and sequelae. Vaccine 30 parenchymal MMP‐9/TIMP‐1 ratio and increased col-
(Suppl 2): B3–B9. lagen type IV degradation. Neurobiol. Dis. 21: 647–656.

293
Infections of the Central Nervous System

39. Greenwood, J., Etienne‐Manneville, S., Adamson, P., 41. Schaper, M., Gergely, S., Lykkesfeldt, J. et  al. (2002).
and Couraud, P.O. (2002). Lymphocyte migration into Cerebral vasculature is the major target of oxidative pro-
the central nervous system: implication of ICAM‐1 sig- tein alterations in bacterial meningitis. J. Neuropathol.
nalling at the blood‐brain barrier. Vasc. Pharmacol. Exp. Neurol. 61: 605–613.
38: 315–322.
40. Eisenhut, M. (2014). Vasospasm in cerebral inflam-
mation. Int. J. Inflamm. 2014: 509707.

294

You might also like