Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

DOI: 10.1002/JLB.

MR0318-102R

REVIEW

The pathogenesis of tuberculous meningitis

Angharad Grace Davis1,2,3 Ursula Karin Rohlwink4 Alizé Proust1


Anthony A. Figaji4 Robert J. Wilkinson1,2,3,5,6

1 The Francis Crick Institute, Midland Road,

London, United Kingdom Abstract


2 Faculty of Life Sciences, University College Tuberculosis (TB) remains a leading cause of death globally. Dissemination of TB to the brain
London, United Kingdom results in the most severe form of extrapulmonary TB, tuberculous meningitis (TBM), which rep-
3 Department of Medicine, University of Cape
resents a medical emergency associated with high rates of mortality and disability. Via various
Town, Republic of South Africa mechanisms the Mycobacterium tuberculosis (M.tb) bacillus disseminates from the primary site of
4 Neuroscience Institute, Division of Neuro-
infection and overcomes protective barriers to enter the CNS. There it induces an inflammatory
surgery, University of Cape Town, Republic of
South Africa
response involving both the peripheral and resident immune cells, which initiates a cascade of
5 Wellcome Centre for Infectious Dis- pathologic mechanisms that may either contain the disease or result in significant brain injury.
eases Research in Africa, Institute Here we review the steps from primary infection to cerebral disease, factors that contribute to
of Infectious Disease and Molecular the virulence of the organism and the vulnerability of the host and discuss the immune response
Medicine, University of Cape Town,
and the clinical manifestations arising. Priorities for future research directions are suggested.
Republic of South Africa
6 Department of Medicine, Imperial College,

London, United Kingdom KEYWORDS


Correspondence mycobacterial, endothelial cells, granulocytes, microglia cells, monocytes/macrophages, myeloid
Robert J. Wilkinson, The Francis Crick Institute,
cells, neutrophils, T Lymphocytes, Th17 cells
Midland Road, London, NW1 1AT,
United Kingdom.
Email: Robert.Wilkinson@uct.ac.za

1 INTRODUCTION 2 TEXT FROM PRIMARY INFECTION


TO THE CNS
Tuberculous meningitis (TBM) is the most serious extrapulmonary
manifestation of tuberculosis (TB) with mortality rates up to 50% 2.1 The systemic immune response
observed in the context of HIV-1 co-infection.1 The pathogenesis
to tuberculous infection
of TBM is incompletely understood and further understanding is
required in order to develop effective vaccines, optimal antibiotic and TB infection occurs through the inhalation of infectious droplets of

host-directed therapies. We present an overview of the pathogenesis aerosolized Mycobacterium tuberculosis (M.tb), which cross the lung

of TBM following the pathogen from site of primary infection to epithelium and infect lung alveolar macrophages, neutrophils, and den-

dissemination to and within the brain. We have included observations dritic cells (DCs). This leads to the secretion of antimicrobial peptides,
from important studies that have contributed to understanding cytokines (including IL-1𝛼 and IL-1𝛽, TNF-𝛼, IL-6 and -12, chemokines,
pathogenic mechanisms in TBM. We also highlight areas important for lipoxins that may stimulate necrosis and contribute to immune pro-
future research. tection, and prostaglandins that may induce apoptosis.2 Under the
influence of IL-12 and chemokines CCL-19 and -21, infected DCs
migrate to the local draining lymph nodes to stimulate the differen-
tiation of T-helper I (Th1) cells. Th1 cell release of IFN-𝛾 and TNF-𝛼

Abbreviations: ART, Antiretroviral therapy; BBB, Blood brain barrier; BCSFB,


at the site of infection activates macrophages and DCs to produce
Blood-cerebrospinal fluid barrier; CSF, Cerebrospinal fluid; DC, Dendritic cell; IRIS, Immune cytokines and antimicrobial factors that contribute to containment of
reconstitution inflammatory syndrome; LEC, Lymphoid endothelial cell; M.tb, Mycobacterium
the TB bacillus.2 This inflammatory process results in the formation
tuberculosis; PGL, Phenolic glycolipid; RD, Region of difference; TB, Tuberculosis; TBM,
Tuberculous meningitis; TPH, Trypophan hydrolase; VDR, Vitamin D receptor; VEGF, Vascular of a granuloma, which encapsulates the infected cells and retards the
endothelial growth factor
replication of TB bacilli and can lead to latent infection. However, in

Received: 31 July 2018 Revised: 17 November 2018 Accepted: 5 December 2018


J Leukoc Biol. 2019;105:267–280. www.jleukbio.org 2019
c Society for Leukocyte Biology 267
268 DAVIS ET AL .

the elderly, immune compromised or very young, the ongoing immune Another potential route of entry is the “Trojan horse” mechanism by
response may progress to active primary TB disease associated with which M.tb are trafficked in infected macrophages and neutrophils
tissue destruction in the lung and dissemination of the TB bacillus to across the BBB.7
other organ systems.3,4 Once the TB bacilli gain access to the brain, limited local innate
immunity allows their survival and replication and the development
of silent tuberculous lesions. Based on postmortem studies, Rich
2.2 Dissemination to the brain
and McCordock suggested that TBM is initiated by the rupture of
Dissemination of TB involves seeding of M.tb to other sites including one of these lesions, the Rich focus, located under the cortical pia
the CNS. Various mechanisms by which the bacilli migrate into the or adjacent to the meninges or ventricles, which releases M.tb bacilli
lymphatic system or blood stream have been suggested.5–7 Bacterial into the subarachnoid space causing a granulomatous infection of
proteins early secretory antigenic target 6 kDa (ESAT-6) and culture the meninges15–18 and the subsequent induction of inflammation.
filtrate protein 10 kDA (CFP-10) are involved in cell lysis, whereas Recently the relationship between the Rich focus and the onset of
heparin binding hemagglutinin adhesion (HBHA) aids M.tb transloca- miliary TB has been reviewed. Rich and McCordock did not acknowl-
tion across the epithelium without lysis.5 M.tb may also invade and edge a role of miliary TB in the pathogenesis of TBM; however, several
traverse vascular endothelial cells,6 replicate in lymphatic endothelial studies since have suggested that the bacteraemia seen in these
cells (LEC),8 and be trafficked to distant locations in phagocytes.5 cases increased the likelihood that a meningeal of subcortical focus is
Furthermore, mycobacteria are able to survive and replicate in established with subsequent rupture giving rise to TBM (reviewed in
infected macrophages and LEC surrounding granulomas in the lymph reference 16).
nodes. Research on LECs demonstrates that although M.tb bacilli are
initially phagocytosed upon infecting the cell, through their genetic
locus termed the region of difference 1 (RD1), the bacilli are able to
escape from the phagosomes into the cytosol, where they more readily
3 PATHOGENIC AND PATHOPHYSIOLOGIC
replicate. For those bacteria remaining in the phagosome they are able
MECHANISMS WITHIN THE BRAIN (FIG. 1)
to prevent fusion of lysosomes with the phagosome, also in an RD1
dependent manner, thereby allowing bacterial replication in the phago-
3.1 The host immune response to TB in the brain
some and contributing to lymphatic TB.8 The activation of LEC by
IFN-𝛾 is key to restricting these RD1 mechanisms of replication.8 Microglia within the cerebral parenchyma are the principal CNS cells
Additionally, host immunity and M.tb strain variation may play a infected by M.tb19,20 and are involved in immune regulation. Other
role; polymorphisms in the genes encoding for antigen recognition CNS cells that have potential roles in this process are astrocytes and
and macrophage activation9 or impaired proinflammatory cytokine neurons20,21 (Fig. 1). Randall et al. have demonstrated direct infection
release may influence the ability of the initial innate response to of neurons with M.tb; however, the effect on neuronal function and
control infection.5 TB strains assigned greater virulence, like the implications for intercellular interactions is not clear.21 Although not
lineage 2 strain, which is postulated to subvert the innate immune as prominent as microglia in their role, astrocytes have also been
response, promoting its survival and replication and thereby more implicated with a study by Rock and colleagues demonstrating 15% of
severe disease.10–12 astrocytes having cell-associated bacilli (average of 1.3 bacilli per cell)
The CNS is protected from influx of potentially harmful blood-borne compared to 76% of microglia (average of 4.2 bacilli/cell) under the
bacteria by 2 vascular barriers; the blood-brain barrier (BBB) and the same conditions.20
blood-cerebrospinal fluid barrier (BCSFB)13 (Fig. 1). The BBB is mainly M.tb is recognized by microglial cells via innate immune and neuro-
formed by brain microvascular endothelial cells that are characterized specific receptors, including pattern recognition receptors. The TLRs, a
by intercellular tight junctions and a paucity of endocytotic vesicles family of 10 pattern recognition molecules, play a crucial role in innate
and fenestrae, and exhibit dedicated transport mechanisms to regu- immunity. Internalization of M.tb by human microglia is dependent on
late influx and efflux across the CNS and blood compartments.13,14 CD14, a monocyte differentiation antigen, which binds to lipopolysac-
Pericytes, embedded within a basement membrane, and astrocytes’ charide with TLR-4.22 This was demonstrated in a study in which
end-feet support the endothelial cells and also “make an indispens- uptake of nonopsonized tubercle bacilli by microglia was reduced by
able contribution to BBB integrity.” In contrast, the BCSFB is com- 64% and 62% in the presence of anti-CD14 monoclonal antibodies
posed of choroid plexus epithelial cells joined together by tight junc- and soluble CD14 ligand respectively. This is in contrast to peripheral
tions and the arachnoid membrane. However, despite these protec- mononuclear phagocytes where CD14 neutralizing anti-CD14 anti-
tive mechanisms, M.tb bacilli migrate across these barriers. In vitro bodies did not affect bacillary uptake,23 but interestingly the presence
and animal models demonstrate that M.tb invades and traverses brain of CD14 led to up-regulation of CD14 expression on these cells per-
endothelial cells in the microvasculature through rearrangement of haps facilitating pathogenic immune responses. This receptor, along
their actin.6,14 Further, the M.tb gene Rv0931c (pknD) has been identi- with the 𝛽2-integrin CD18 and TNF-𝛼, is also involved in the forma-
fied as a potential virulence factor promoting CNS infection in certain tion of histologically characteristic multinucleated giant cells seen at
TB strains as it enables the bacilli to interact with extracellular factors autopsy and experimentally identified in porcine microglia infected
on the brain endothelium facilitating bacillary endothelial adhesion.14 with Mycobacterium bovis.24
DAVIS ET AL .

Blood-brain Host Cell Recognition Pro-inflammatory Permeable


barrier and Internalisation cytokines BBB
Rearrange actin
cytoskeleton ICAM-1
Release of endotoxins
TLR 2, 4, 9 TNF alpha
and reactive oxygen
free radicals
VEGF-A
Bacterial virulence CD14 IFN gamma Impaired neuro-signalling
factors (Rv0931c)
enable bacillary VCAM-1 Metabolic perturbations Cell damage
IL-6 Cytotoxic oedema
endothelial
Microglia Perturbations in
adhesion MMPs cerebral perfusion Mortality
Raised ICP
characteristics

Extracellular IL-1B Alteration of neuronal Infarcts


microenvironment
bacteria invade Decreased cerebral
endothelial cells and CCL-2 blood flow Morbidity
Infiltration cerebral Vascular stenosis,
may contribute to vessel wall occlusion, spasm
vasculopathy Neurons
CCL-5 T cells Exudate Hydrocephalus

CXCL-10 NK Leaky BBB Vasogenic oedema


cells
macrophage
Macrophage Tuberculomas
IL-1 alpha Containment of bacilli
Tuberculous abscesses
Bacteria use Disease
Astrocytes Monocyte resolution
infected phagocytes IL-12p40 Extension of disease Spinal arachnoiditis,
as a Trojan horse to into spinal canal tuberculomas, exudate
migrate into the
CNS dendritic cell Cellular
Genetic polymorphisms in the following
genes affect host immune response: Influx
TIRAP, TLR2, TLR9, CD43, MDL, PKP3-
Blood-CSF SIGIRR-TMEM16J, VDR, LTA4H

barrier

F I G U R E 1 Summary of the pathogenesis of TBM. A: Mycobacterium tuberculosis bacilli (M.tb) disseminate from the primary site of infection in the lung to seed the brain. The bacilli are able to traverse
the blood brain barrier (BBB) and blood cerebrospinal fluid barrier (BCSFB) through various virulence factors that enable the invasion of and migration through cerebral vascular endothelial cells, or are
carried into the CNS by infected peripheral innate immune cells. B: In the CNS antigen recognition and internalization by microglia, neurons and astrocytes occurs, mediated by numerous host genetic
factors. C: The resulting immune response stimulates the release of proinflammatory cytokines and chemokines and other immune mediators that contribute to the breakdown of the BBB and the influx
of innate and adaptive immune cells from the periphery. D: A prolific inflammatory response ensues. The inflammatory exudate in the basal cisterns contributes to cerebral vascular pathology and the
development of hydrocephalus and raised intracranial pressure. Vasogenic edema due to an influx of proteins through the leaky BBB, and cytotoxic edema as a result of cellular damage contribute to the
raised pressure. The overall decrease in cerebral blood flow puts the brain at risk of ischemia, infarction and poor patient outcomes. In some cases the infection is controlled in discrete tuberculomas or
abscesses, which resolve with treatment and time. Extension of the disease into the spinal canal manifests as spinal arachnoiditis, tuberculomas, or collection of exudate
269
270 DAVIS ET AL .

Activation of microglia leads to secretion of a number of cytokines cells, and S100B, a protein synthesized by astrocytes, oligodendro-
(Fig. 1). In murine microglia the intracellular signaling pathways leading cytes and Schwann cells known to be involved in cell-to-cell commu-
to cytokine release are M.tb inducible leading to a proinflammatory nication, cell growth, and intracellular signal transduction, as well as
response through NAPDH oxidase-dependent reactive oxygen species the development and maintenance of the CNS.45 These biomarkers of
(ROS) generation.25 Although cytokines play a critical role in the cerebral injury are known to increase in TBM,42 and therefore sug-
host defence to infection with M.tb, they can also mediate inflam- gest that PARP1 may be a potential new target in the development of
mation. TNF is central to the pathogenesis of CNS TB.26–28 It has a host-directed therapies.46 S100A8/9, also from the S100 family has
protective role in the immune response to mycobacteria29 ; but is also a role in neutrophil chemoattraction and stimulation47 and is impli-
associated with pathology in vivo via induction of fever, activation of cated in the pathogenesis of TB in pulmonary disease. In HIV-1 unin-
the hypothalamo-adrenal axis and by triggering the release of other fected patients raised S100A8/9 in serum correlated with increased
cytokines.30–32 Local TNF-𝛼 production in the CNS also increases radiographic disease severity.48,49 In murine models IL-17 induced
permeability of the BBB and thus influx of other immune mediators S100A8/9 was a key factor in neutrophil accumulation and led to exac-
to the CNS.31 In a murine CNS TB model, Tsenova et al. demonstrated erbated lung inflammation due to increased levels of proinflamma-
a correlation between levels of TNF-𝛼 and the extent of cerebral tory cytokines.50 In TBM Marais et al. demonstrated that in patients
pathology as measured by CSF leukocytosis, protein accumulation, with TBM and HIV infection, levels of S100A8/9 were significantly ele-
meningeal inflammation, persistence of bacillary load, and clinical vated 2 wk after the initiation of antiretroviral therapy (ART) in those
deterioration.28 TNF-𝛼 antagonists such as thalidomide33 and ana- who developed immune reconstitution inflammatory syndrome (IRIS)
logues thereof,34 used in rabbit models of TBM, down-regulated the defined as a paradoxical worsening of infection despite adequate treat-
production of TNF-𝛼 and subsequently improved survival. This finding ment following the initiation of antiretroviral drugs, compared to those
was not replicated in human studies where a clinical trial of thalido- who did not. This observation may explain in part the ongoing para-
mide used in conjunction with standard antituberculous therapy and doxical inflammation observed in IRIS discussed in more detail later in
corticosteroids in children with TBM was stopped early due to adverse this review.51
events associated with thalidomide use.35 Since then there has been In addition to the inflammatory milieu described above, there are
some suggestion of benefit with the use of thalidomide in the context several other factors implicated in the pathogenesis of TBM, in partic-
of tuberculous mass lesions.36 ular the subsequent increasing permeability of the BBB and influx of
CSF concentrations of IL-6 independently associate with more inflammatory mediators and cells (Fig. 1). Vascular endothelial growth
severe presentation of TBM.37 In this context it is unclear whether this factor (VEGF) is a potent endothelial growth factor playing diverse
is due to its proinflammatory or anti-inflammatory effects. In murine roles in vasculogenesis and angiogenesis. In TB it is now considered
models of TB IL-6 has been implicated in the stimulation of IFN-𝛾 a useful biomarker of disease, where it may be used as an indicator
production but not necessarily essential for the protective immunity of active versus latent disease activity or a marker of extrapul-
to M.tb.38 It may also have an anti-inflammatory role by suppressing monary versus primary lung disease. In several types of cancer, VEGF
gene expression of proinflammatory cytokines.39 In a study by Rock inhibitors such as Bevacizumab are well established as an effective
et al., proinflammatory cytokines other than TNF-𝛼, IFN-𝛾, and IL-6 therapeutic approach. In ischemic conditions of the brain, VEGF has a
found to be secreted by microglia in response to TBM included pathologic and protective role depending on pathogenic stage due to
IL-1𝛽, CCL2, CCL5, and CXCL-10. In contrast to microglia, astro- either effect on permeability of the microvasculature or subsequent
cytes produced only moderate amounts of CXCL10.20 Other reparative angiogenesis. In age-related neurologic conditions such as
cytokines, confirmed experimentally, to be secreted by microglia Alzheimer’s disease, Parkinson’s disease, and motor neuron disease;
following M.tb stimulation include: IL-1𝛼, IL-10, IL-12p40, G-CSF, however, VEGF is thought to be pathogenic due to its effect on BBB
and GM-CSF.25,40 dysfunction.52 In these conditions the effect of VEGF on increasing
Recent focus has turned to the pathogenic role of inflamma- endothelial permeability is clear; however, the mechanism by which
tory mediators such as DAMPs (damage-associated molecular pat- this happens remains less understood with possibilities including its
terns) and PAMPs (pathogen-associated molecular patterns),41 their effect on cell-cell junctions including tight junctions and adherens
validity as biomarkers of cerebral injury,42 and as potential targets junctions as well as on transcytosis.53 In TBM VEGF disrupts the per-
for novel host-directed therapies in TBM.43 PAMPs are by-products meability of the BBB,54,55 which has been proposed as a mechanism
released from pathogens that are recognized by host cell receptors by which dexamethasone exerts efficacy as a host-directed therapy
subsequently leading to activation of the innate immune response. in TBM. Also interesting is its neuroprotective effects, which have
DAMPs, which are released by damaged host immune cells, interact been more thoroughly explored in the context of amyotrophic lateral
with PAMPs leading to an accelerated cycle of cell death and injury. sclerosis where low VEGF has been reported with BBB dysfunction
Hostpoly(ADP-ribose) polymerase 1 (PARP1; also known as ARTD1) and the therapeutic use of a VEGF analogue (VEGF-A165 ) is currently
is an ADP-ribosylating enzyme essential for initiating various forms of under investigation in a clinical trial (NCT02269436). The release of
DNA repair.44 Recent studies have suggested a role for PARP1 in the intercellular and vascular adhesion molecules (ICAMs and VCAMs)
pathogenesis of TBM via its potential to modulate the release and acti- as well as matric metalloproteinases (MMPs) from inflammatory cells
vation of DAMPs. This includes high mobility group box-1 (HMGB1), a within the CNS have also been shown to increase the permeability of
nonhistone nuclear DNA binding protein expressed in all mammalian the BBB.56
DAVIS ET AL . 271

(a)

Third ventricle

Cerebral aqueduct

Quadrigeminal cistern

Midbrain
Interpenduncular cistern
Pons
Fourth ventricle
Prepontine cistern

(c)

(b) Third ventricle

Hypothalamus Ambient cistern

Pituitary stalk Subarachnoid space

Anterior Pituitary
Posterior Pituitary

F I G U R E 2 Basal cisterns and pituitary anatomy. A: Basal cisterns affected in TBM are represented here in a sagittal view of the brain. Note
the quadrigeminal cistern, which extends laterally to become a thin sheet like cistern surrounding the midbrain and posterior thalamus, named the
ambient cistern shown in C. B: Anatomy of the pituitary gland and surrounding structures

3.2 Metabolic factors in the host Glucose and its metabolic effects are also of interest. In a pivotal
study where adjunctive dexamethasone was found to decrease short
Neuroendocrine-associated metabolic abnormalities are common in
term mortality in HIV-1 uninfected patients with TBM, low glucose
TBM. In an observational study of patients newly diagnosed with TBM,
at presentation predicted poor outcome.62 In more recent studies,
neuroendocrine dysfunction occurred in half.57 This is likely due to the
including those in children, CSF glucose, lactate, and protein levels
tendency for TBM to affect the basal structures such as the interpe-
have been linked to poor outcome.37,63 If these metabolic markers are
duncular fossa, cisterna ambiens and cisterna pontis, which surround
simply markers of disease activity as in most forms of neurologic infec-
the pituitary gland, pituitary stalk, and hypothalamus (Fig. 2). Exu-
tion, then this finding is unsurprising. Mason et al. demonstrated that
dates here lead to edema, perivascular infiltration, and subsequent
the increase in lactate levels commonly observed in the CSF of patients
microglial reactions known collectively as “borderzone encephalitis.”
who go on to develop poor outcomes is of the L-form and therefore
Subsequent metabolic abnormalities include gonadotropin deficiency,
solely a response from the host to the infection, rather than being of
hyperprolactinemia, thyrotropin deficiency, corticotropin deficiency,
microbial origin.64 This finding contributes to this groups’ hypothesis
and somatotropic hormone deficiency.57 Hyponatremia is also com-
that in the context of neuroinflammatory-inducing infection, energy
mon, although the mechanism by which is occurs is debated. Cerebral
flow in brain metabolism is shifted away from the neurons and shunted
salt wasting (CSW) and a syndrome of inappropriate antidiuretic hor-
toward the microglia. They theorize that this leads to lactic acid pro-
mone (SIADH) are possible explanations. In a prospective study of 76
duction by glycolysis in astrocytes, which subsequently participates in
TBM patients conducted in India, 34 patients had hyponatremia, which
the activated immune response by contributing to oxidative phospho-
was due to CSW in 17, SIADH in 3 and miscellaneous causes in 14.58
rylation and hence production of high levels of ATP and forms of ROS
Although the mechanism of hyponatremia is unclear, its role in the
required for degradation of the invading pathogen.
pathophysiology TBM is important as it independently associates with
The metabolism of tryptophan is also of interest in TBM. Tryp-
poor outcome,59,60 and also more recently as a possible predictor for
tophan is a key amino acid required for protein biosynthesis, and a
stroke in the context of CSW.61 Corticotropin deficiency may be mod-
precursor for various metabolites, including serotonin and melatonin
ulated by treatment with dexamethasone, which at present remains
(serotonin pathway) and kynurenine and quinolinic acid (kynure-
the only host-directed therapy of proven benefit in TBM. Pituitary dys-
nine pathway)65 (Fig. 3). Proinflammatory cytokines such as IL-6,
function and its role in the pathogenesis of TBM remains an area of
TNF-𝛼, and IFN-𝛾 are known to trigger the kynurenine pathway by
interest, particularly given the observations of worsening outcomes
stimulating indoleamine 2, 3-dioxygenase (IDO).66,67 Once IDO is
and possible benefit of cortisol replacement.
272 DAVIS ET AL .

⊝ IL-6 F I G U R E 3 Mycobacterium tuberculosis and vitamin D


Vitamin D impact on the tryptophan metabolism. M.tb infection
⊕ Tryptophan ⊕ Mycobacterium tuberculosis activates the inflammatory response resulting in proinflam-
TPH IDO matory cytokines secretion. Along with M.tb, these cytokines
KAT Kynurenic acid can trigger the kynurenine pathway by stimulating IDO.
Serotonin Kynurenine (astrocytes) Once the pathway is activated, astrocytes and microglia
KMO respectively produce kynurenic acid and quinolinic acid, the
latter is neurotoxic. Thus, an imbalance in the synthesis of
Neurotransmission 3-HKyn
these two products may result in neurotoxicity. Vitamin D,
3-HAO
⊝ cofactor of TPH, promotes serotonin production and neuro-
Quinolinic acid transmission. Alone or when bound with one of its receptor,
(microglia) VDR, vitamin D attenuates the inflammatory response.
M.tb: Mycobacterium tuberculosis; IDO: indoleamine 2,

Oxydative stress NMDA 3-dioxygenase; TPH: tryptophan hydrolase; KAT: kynurenine
aminotransferase; KMO: kynurenine-3-monooxygenase;
3-HAO: 3-hydroxyanthranilic acid dioxygenase; NMDA:
Neurotoxicity N-methyl-d-aspartate; VDR: vitamin D receptor. Blue: neuro-
SEROTONIN PATHWAY KYNURENINE PATHWAY protection; red: neurotoxicity; green: influential factors

activated, the kynurenine pathway is promoted at the expense of the polymorphisms within the genes involved in the TLR pathway have
serotonin pathway. Microglia and astrocytes then secrete quinolinic been associated with susceptibility to infection in TB. A recent study
acid and kynurenic acid, respectively. Both products have opposite of patients with TBM in Vietnam was the first to demonstrate an
roles; whereas kynurenic acid acts as an antagonist of the glutamate association between single nucleotide polymorphisms (SNPs) in the
N-methyl-D-aspartate receptor (NMDAr), quinolinic acid acts as its TLR9 gene region and TBM.72 Further, a Vietnamese study found an
agonist and leads to neurotoxicity.66 Activation of the kynurenine association between TLR2 SNP T597C and the development of TBM
pathway is also known to exacerbate progression of neurodegenera- and miliary TB, suggesting that TLR2 influences the dissemination of
tive diseases and has been described in HIV-associated dementia.68 M.tb.73 Toll IL-1 receptor domain containing adaptor protein (TIRAP)
Recently, Van Laarhoven et al. conducted a metabolomic study mediates signals from TLR1, -2, -4, and -6 to activate macrophages
examining serum and CSF of TBM patients. Their results showed that and DCs. Hawn et al. investigated the association of the TIRAP
whereas most metabolites demonstrated elevated concentrations in SNP C558T with (i) susceptibility to TB (odds ratio, 2.25, P < 0.001)
TBM patients compared to controls, concentrations of tryptophan and (ii) susceptibility to meningeal TB (OR, 3.02; P < 0.001) versus
were low in TBM and further reduced in patients who survived. They pulmonary TB (OR, 1.55; P = 0.22). They also demonstrated that
further demonstrated up-regulation of the gene IDO1 and identified compared to the to the 558CC genotype, the 558TT genotype was
11 trait loci that correlated with tryptophan concentrations and were associated with decreased whole-blood IL-6 production, suggest-
prognostic of survival when combined with sex and age. The prognos- ing that TIRAP influences disease susceptibility by modulating the
tic potential of these genetic correlates was demonstrated in a valida- inflammatory response.9
tion cohort. These data suggest that tryptophan metabolism may play Another gene of interest is leukotriene A4 hydrolase (LTA4H).
an important role in TBM outcome, and that further investigation into LTA4H influences the balance of pro- and anti- inflammatory
this metabolic pathway is warranted.69 eicosanoids and subsequent TNF-𝛼 regulation through either reduced
Whereas M.tb may directly stimulate of IDO at the site of inflammation due to excess lipoxins or augmented inflammation due to
infection,70 vitamin D on the other hand is an essential cofactor of excess leukotriene B4. In a study of zebrafish and humans, mutations
the tryptophan hydrolase (TPH) and promotes serotonin production in the gene encoding LTA4H led to immunosuppressive Lipoxin A4
and thus neurotransmission. Furthermore, Vitamin D alone or bound (LXA4) accumulation and increased susceptibility to mycobacteria.27
to the vitamin D receptor (VDR) expressed on various cell types, In this study, heterozygosity at several LTA4H SNPs was associ-
including astrocytes and microglia, leads to a decrease in inflammatory ated with protection against TBM.27 These findings supported a
response.71 Thus tryptophan metabolism could be a modality by which hypothesis that excess LTA4H activity leads to a “hyperinflammatory
the vitamin D status of an individual modulates both susceptibility to state,” whereas lack of LTA4H activity leads to an inadequate host
M.tb infection and TBM pathogenesis. By restoring a noninflammatory response.74 In a later study by the same group the LTA4H promoter
environment and restricting M.tb replication, vitamin D could promote region SNP rs17525495, defining 3 genotypes—TT, CT, and CC—was
the serotonin pathway at the expense of the kynurenine one and pro- identified as a likely molecular determinant of genetic susceptibility. In
tect the brain from neurotoxicity. this study lymphoblastoid cell lines with SNP genotype CC conferred
a hypoinflammatory and TT a hyperinflammatory phenotype.27 It was
also demonstrated that (i) genotype correlated with pretreatment CSF
3.3 Host genetic factors
leukocytosis and survival and (ii) those benefiting from adjunctive dex-
A number of immune response genes encoding the pathways amethasone were carriers of the “hyperinflammatory” TT genotype.27
described are under genetic influence. For instance, mutations and Several follow-up studies to further investigate the relevance of this
DAVIS ET AL . 273

finding have tended to reproduce an association with susceptibility to proinflammatory cytokines.91 A later study by Caws et al. comparing
disease, but not necessarily outcome.75–78 The two largest of these bacterial and host genotype across two groups of Vietnamese adults
both published in 2017 investigated the association in Vietnamese78 with pulmonary or meningeal TB found that disease caused by the
and Indonesian77 populations. The first showed that individuals with Euro-American lineage was significantly more likely in pulmonary dis-
the LTA4H CC genotype had a higher risk of early death, whereas the ease; however, by contrast found no association between the lineage 2
second did not find an association between genotype and mortality. and disease phenotype.10
Possible explanations may include differences in linkage disequilibrium Epidemiologic studies have reported several differences in disease
as well as the observed overall differences in patient characteristics phenotype between M.tb lineages in terms of pathogen virulence,92,93
such as mortality (40.7% in Indonesia vs. 18.9% in Vietnam), fre- transmission of disease,87,94,95 progression from latent to active
quency of culture confirmed diagnosis (55.3% vs. 42.8% in Vietnam), disease,96 and in response to treatment.97,98 In vitro studies have
severity of disease (BMC grade I severity in Vietnam 37% vs. 11% in explored whether the host immune response is specific to genotype.
Indonesia), and median age of patients (41 yr vs. 29 yr in Vietnam) A study in South Africa found differential mycobacterial growth and
(reviewed in reference 79 ). Nonetheless this remains an area of great levels of early proinflammatory cytokines including TNF and IL-12p40
interest and a clinical trial is under way to determine whether LTA4H between three modern lineages.99 Others have found difference in
genotype, defined at randomization, influences dexamethasone’s human macrophage responses between lineages and have hypoth-
clinical efficacy when added to the first 6–8 wk of anti-TB treatment of esized that the lack of early proinflammatory response observed
TBM (NCT03100786). with modern lineages may contribute to more rapid disease pro-
There are a number of other polymorphisms documented in the gression and transmission and therefore confers survival advantage
literature to relate to the pathogenesis of TBM. Polymorphisms in for these strains of M.tb.100 In Madagascar differences between
CD43 encoding a surface glycoprotein involved in M.tb adhesion ancient and modern lineages were characterized by contrasting
and proinflammatory cytokine induction has been associated with IFN-𝛾 responses.101 Specifically, comparison of the IFN-𝛾 responses
decreased survival from TBM.80 The mannose binding protein (MBP) with the spoligotype of the infecting clinical strains showed that mod-
binds mycobacteria and acts as an opsonin in vitro. Although MBP ern M.tb strains tended to induce lower IFN-𝛼 responses than ancient
plays a role in the first line of defence against M.tb, it may also facil- strains in index cases and their household contacts.101 The aforemen-
itate the spread of the intracellular pathogen.81 A hypothesis, was tioned study by Caws et al. was the first to demonstrate an interac-
therefore advanced that phenotypes in which levels of MBP are low tion between pathogen and host genetic factors as a predictor of dis-
may result in protection from TBM. In a study to test this, a mutation ease phenotype by showing that individuals with the C allele of the
in the MBP B allele (G54D), which leads to disruption of the collagen TLR02-T597C genotype were more likely to have TB caused by the Bei-
region of the MBP protein, was found to be associated with reduced jing genotype (OR 1.57; 95% CI 1.15–2.15).10 The most recent study
dissemination to the brain and suggested protection against TBM.82 in this field performed whole genome sequencing of M.tb strains from
Polymorphism in the PKP3-SIGIRR-TMEM16J gene region encoding 322 HIV-1 uninfected patients with TBM (n = 106) and PTB (n = 216).
a negative regulator of TLR/IL-1R signaling has been associated Unlike the previous studies1090 there was no association with dis-
with reduced survival in both pulmonary TB (PTB) and TBM.83 Last, ease phenotype and lineage; however, using a homoplasy based asso-
VDR gene polymorphisms with heterozygous (TC) and mutant (CC) ciation analysis they identified three M.tb genes associated with dis-
genotypes of Taq1 VDR SNP associate with TBM,84 although further ease phenotype. This included Rv0218, a secretome gene encoding a
research is required to investigate whether this is universal across protein that influences pathogen recognition and host-pathogen inter-
all populations.85 action. They hypothesize that a SNP in the region of Rv0218 would
alter the appearance of the surface of M.tb therefore allowing it to
evade recognition and the immune response, enabling dissemination to
3.4 Pathogen virulence factors and their
extrapulmonary sites.
effect on pathogenesis
M.tb was formerly regarded as an organism exhibiting little genetic
variation. More recent studies using genetic typing techniques to ana- 4 DIFFERENCES IN THE INTRACEREBRAL
lyze M.tb isolates from diverse geographic populations have revealed IMMUNE RESPONSE IN THE HIV-1
a cladal phylogeographic distribution with variation between different INFECTED HOST
lineages.86,87 Seven lineages are now identified classified as “ancient”
(lineages 1, 5, 6, and 7) or “modern” (2, 3, and 4).88 One lineage of There is a relative paucity of data directly comparing the intracere-
particular interest has been lineage 2, which is highly prevalent in bral immune response to TBM in HIV-1 infected and uninfected hosts.
East Asia (and therefore known as “Beijing”) and has been ascribed The impact of HIV-1 infection on the clinical presentation of TBM
hypervirulence in a rabbit model89 as well as some human studies is debated, some studies report that HIV-1 infection does not alter
demonstrating increased risk of disseminated disease.90 This has been the cerebral features of TBM,102,103 whereas others suggest a higher
attributed to an intact polyketide synthase (pks 15/1) gene and the pro- rate of extrameningeal and extrapulmonary disease associated with
duction of a phenolic glycoprotein (PGL), which is thought to attenu- HIV-1 infection.104–106 These studies consistently report that HIV-1
ate the early host immune response leading to a reduced production of infection associates with death102 and decreased efficacy of treatment
274 DAVIS ET AL .

against M.tb.105 A better characterization of the effect of HIV-1 in the TA B L E 1 Cell types within the CNS
pathogenesis of TBM is important. Cell Types Description
A study conducted in Vietnam demonstrated a small reduction in Neuron Primary cell type within the CNS and
IFN-𝛾 yet more significant reduction in the anti-inflammatory cytokine peripheral nervous system (PNS) distinct
from other cell types due to their electrical
IL-10 in HIV-1 infected patients, the latter being 6-fold decreased in
excitability. Responsible for receiving,
the context of HIV. This skewed the CSF IFN-𝛾: IL-10 ratio toward processing and transmitting electrical and
excess IFN-𝛾 with subsequent analysis that was independently asso- chemical signals. A typical neuron consists
of a cell body (soma), dendrites and an axon.
ciated with HIV co-infection (P = −0.009, odds ratio [95% CI], 2.47
Neuroglia Non-neuronal cells within the peripheral and
(1.25–4.88)). central nervous system.
Torok et al.107 described a higher percentage of CSF neutrophils Macroglia Cells derived from the ectoderm.
and suggested this may be due to prior HIV-1 infection. Indeed, this ele-
CNS
vated percentage of CSF neutrophils in HIV-1 infected patients may be
Astrocytes Most abundant macroglia in the CNS. Provide
caused by the microglial response to HIV-1 infection and HIV-1 pro- a vascular supply to neurons and in doing so
teins gp120, Tat (trans-activator of transcription), Nef (negative reg- form an important part of the blood brain
barrier. They affect potassium influx and
ulatory factor), and Vpr (viral protein R). Atluri et al. demonstrated efflux and influence neurotransmitters to
that these four proteins disrupt the BBB integrity by triggering the regulate the external environment of
neurons.
decline of tight proteins expression on brain microvascular endothe-
Oligodendrocytes These cells create the “myelin sheath”; a
lial cells, MMP expression, and apoptosis, and leading to infiltration of
protective coating around the axon, which
leukocytes.108 Moreover, it has been reported in vitro that microglia enables effective propagation of electrical
secrete numerous proinflammatory cytokines and chemokines when impulses along the neuron. In addition they
provide trophic support by the production
infected by HIV-1, including the proinflammatory neutrophil chemo- of glial cell line-derived neurotrophic factor
tactic factor IL-8.20 Those cells also secrete TNF, a cytokine implicated (GDNF), brain-derived neurotrophic factor
(BDNF), or insulin-like growth factor-1
in BBB permeability and whose role in pathogenesis of M.tb is critical, (IGF-1).
indicating the influence of HIV-1 in the promotion of TBM and M.tb Ependymal cells These cells comprise the blood-CSF barrier
spread in the CNS (Table 1). line the ventricular system and spinal cord
and producing cerebrospinal fluid and
Further insight into the mechanisms underlying immunopathogen-
enabling its flow throughout the CNS.
esis of TBM have been observed through the study of paradoxical
PNS
immune reactions. A paradoxical reaction in TB is characterized by
Schwann cells Responsible for the production of the myelin
worsening of preexisting tuberculous lesions, or the appearance of sheath in peripheral nerve axons are known
new tuberculous lesions despite effective anti-TB treatment in patients to have phagocytic properties, which allows
for ongoing production of PNS neurons.
who have demonstrated initial improvement to therapy. In HIV-1 co-
Satellite cells These cells comprise sensory,
infection, paradoxical worsening often occurs following the introduc-
parasympathetic and sympathetic ganglia
tion of ART: a phenomenon known as IRIS. Although initiation of ART and are responsible for regulation of the
during treatment of TBM is associated with a reduced 6 mo mortal- external chemical environment.

ity (HR 0.3, 95% CI = 0.08–0.82)1 IRIS in this context carries a poor Enteric glial cells Within the intrinsic ganglia of the digestive
system these cells contribute to regulation
prognosis with one study reporting a mortality rate of 25% with severe of the enteric system.
disability at 9 mo observed in 23% of those who survived.109 The role Microglia Located within the brain and spinal cord these
of inflammasomes, immune complexes of receptors, and sensors that cells are the resident macrophages within
the CNS.
mediate innate immune responses and lead to inflammation, have been
described in this setting.110–112
In TBM specifically, IRIS is characterized by high CSF neutrophil together with the finding that inflammasome activation can contribute
counts and M.tb positivity at presentation.109 In those who develop to pyroptosis114 suggest that tissue injury observed in TBM may be
IRIS CSF TNF was found to be high whereas CNF IFN-𝛼 was low.109 partly induced by inflammasome-mediated pyroptosis. It is unclear as
Further analysis of predictive factors has demonstrated the role of to whether similar mechanisms exist in the absence of HIV.
neutrophil mediators in particular S100A8/9, which 2 wk after the ini- HIV-1 infiltrates the CNS soon after systemic infection and around
tiation of ART was found to be significantly higher in the CSF of those half of HIV-1 infected people develop HIV-associated neurocognitive
who develop TBM-IRIS compared to those who do not.51 Using longi- disorders.115 It has been reported that IRIS may worsen established
tudinal microarray analysis of blood from patients with HIV and TBM HIV-related cognitive impairment.116 Little is known of the pathologic
Marais et al. have also demonstrated that neutrophil abundant tran- mechanisms that lead to worsening cognition in the context of IRIS
scripts were significantly higher in those who develop IRIS from before but some have postulated a role of the HIV-1 trans-activator of
ART initiation until the onset of TBM-IRIS.113 After initiation of ART a transcription (Tat) protein.117 They described the presence of Tat in
significantly higher number of transcripts associated with canonic and the CSF of the virologically controlled ART-treated population and
noncanonic inflammasome activation were found in patients who went proposed that Tat would contribute to neuroinflammation and IL-17
on to develop IRIS than in those who did not.113 These observations production by infiltrating Th17 cells. Given that an increase of IL-17
DAVIS ET AL . 275

F I G U R E 4 Radiological features of TBM. Top left: Exudate. Contrast-enhanced T1-weighted MRI scan images: A: Normal scan showing cere-
brospinal fluid in the cisterns (interpeduncular cistern in front of the midbrain, black arrow) and vessels at the base of the brain in normal cisterns
(posterior cerebral artery, white arrow); B: Scan of a patient with TBM showing exudate in the basal cisterns of the brain (interpeduncular cistern,
anterior to the brainstem and beneath the hypothalamus, black arrow) and vessels coursing through the exudate in the cisterns (posterior cerebral
artery, white arrow). Middle left: Hydrocephalus. A: Initial head CT scan images of a patient with TBM showing acute hydrocephalus with dilated
ventricles and a compressed brain; B: Head CT scan of the same patient after 3 wk of medical therapy showing resolution of the hydrocephalus.
Lower left: Tuberculomas. MRI scans demonstrating different patterns and imaging characteristics of brain tuberculomas: A: Contrast-enhanced
T1-weighted MRI scan showing multiple ring-enhancing small tuberculomas (arrowed); B: T2-weighted MRI scan showing a large tuberculoma
in the cerebellum compressing the brainstem, surrounding edema, and hydrocephalus from obstruction of the cerebral aqueduct. Right: Infarcts.
Demonstration of infarction patterns in TBM, A: T2-weighted MRI scan showing normal ventricular size and no infarcts; B: T2-weighted axial MRI
scan of a patient with TBM showing discrete unilateral small perforator vessel infarcts (arrowed); C: T2-weighted MRI scan showing more extensive
infarcts in the thalami and basal ganglia (arrowed); D: Head CT scan of a patient with TBM showing global infarction with hypodense hemispheres
bilaterally and a swollen brain

secretion predisposes to IRIS in HIV-1 infected individuals in the important implications; first, the major cerebral vessels originating
context of cryptococcal meningitis,118 it is possible that Tat is a key from the base of the brain become encased with exudate; second,
player in the pathogenesis of HIV-related cognitive impairment in the exudate blocks the circulation of CSF; and third, it envelopes and
context of IRIS. compresses the local cranial nerves resulting in cranial nerve palsies.18

5.1 Vasculitis
5 MACROSCOPIC MANIFESTATIONS
OF THE DISEASE IN RELATION As the exudate spreads along the cisternal extensions from the base
TO THE IMMUNE RESPONSE of the brain it coats the large arteries and their small perforators,
although it has a predilection for extension into the Sylvian fissures.
The characteristic feature of TBM in post-mortem studies is the Therefore, the middle cerebral artery (MCA) and its perforators
presence of a thick, gelatinous inflammatory exudate in the basal cis- around the floor of the 3rd ventricle are most commonly involved,
terns and subarachnoid spaces of the brain (Fig. 4), which may extend resulting in ischemic damage to the subthalamic nuclei and lower
into the spinal canal.18,119,120 The predominantly basal location has internal capsule18 (Fig. 5). Involvement of small vessels supplying the
276 DAVIS ET AL .

Lateral ventricle
Caudate nucleus
F I G U R E 5 Lenticulostriate arteries. Lenticulostriate
Internal capsule arteries branching from the M1 segment of the mid-
dle cerebral artery supply the basal ganglia and internal
capsule
Lenticulostriate arteries

Middle cerebral artery

Internal carotid artery

mid-brain may lead to infarction119 and exudate in the interpeduncular associated with peripheral edema and vascular proliferation.15,18 With
cistern can compromise the vessels supplying the hypothalamic region time the tubercles become discretely organized and bound by a rim
and precipitate hypothalamic symptoms in these patients.119 Vascular of connective tissue of reticulin fibers, which enhances on contrasted
pathology includes infiltration of inflammation from the adventitia brain imaging. They may occur throughout the brain in the cerebrum,
of arteries and veins inward, resulting in peri- or pan-arteritis that cerebellum, and brainstem.18,122
involves segments or the full thickness of the vascular wall tissue.121 TB brain abscesses may follow TBM or develop independen-
Evolution of vascular inflammation may involve thickening of the vessel tly125,126 ; however, they occur infrequently.127 Abscesses manifest
intima resulting in vessel stenosis or occlusion.121 Further, vasospasm as encapsulated collections of pus that contain tubercular bacilli in
is also an important contributor to brain ischemia.21 Both cerebral and absence of typical TB granuloma features including epithelioid and
spinal vessels may be affected by any or all of these processes and this giant cells and mononuclear cell infiltrates.127 These abscesses have
may reflect the duration of illness.121 Infarcts (Fig. 4) and changes on been reported in the cerebrum125,127 and posterior fossa,125,128,129
angiography are reported in the majority of TBM patients122 and are occur infrequently in the brain stem,126 and may be singular
associated with post-mortem cerebral and vascular pathology.18 or multiple.15

5.4 Spinal TB
5.2 Hydrocephalus (Fig. 4)
Spinal TB may develop as a primary tuberculous lesion, from the
Extension of exudative material into the basal cisterns results in a col-
downward extension of intracranial TBM, or secondary to vertebral
lar of exudate around the midbrain and a blockage in CSF flow around
TB.18,130,131 It involves the cord, meninges and nerve roots and mani-
the upper brain stem. Exudate may also collect around the cerebral
fests as spinal arachnoiditis, intradural (extramedullary) tuberculomas,
aqueduct of the third and fourth ventricles hindering the flow of CSF
or rarely as intramedullary tuberculomas.122,132–134 Spinal nerve roots
through the ventricular system (Fig. 2). Consequently, hydrocephalus is
may become entrapped and matted in exudate filling the subarachnoid
common, occurring in 75–80% of TBM patients.122–124 The pressure of
space, and complete obliteration of the thecal space can occur in severe
expanding ventricles and the opposing pressure of brain edema due to
cases.122 Exudate involvement of the lower lumbar segments is asso-
pathologic processes can negatively impact the grey and white matter,
ciated with difficulty in performing lumbar punctures and a high CSF
leading to pallor and diffuse loss of myelin,18 and the raised intracranial
protein.18,122 The microscopic appearance of spinal exudate is identi-
pressure can severely compromise cerebral blood flow.
cal to intracranial exudate, consisting of giant cells and caseous areas,
and causing vasculitis of the spinal veins and arteries, which may lead

5.3 Tuberculomas and tuberculous brain to cord ischemia and infarction.18,135

abscess (Fig. 4)
Tuberculomas may coexist with TBM or occur independently and are 6 RESEARCH GAPS AND THE
thought to originate from expanding tubercles in the parenchyma. PATH FORWARD
These lesions are typically granulomatous consisting of a necrotic cen-
ter surrounded by epithelioid cells (which may merge to form Lang- Intracerebral and spinal pathology in TBM is mediated by a dys-
hans giant cells), lymphocytes, and a border of astrocytes, and are regulated inflammatory response that contributes to meningitis,
DAVIS ET AL . 277

tuberculoma formation, arteritis, obstruction of cerebrospinal fluid 7. Nguyen L, Pieters J. The Trojan horse: survival tactics of pathogenic
(CSF) flow, and vascular complications including stroke. The develop- mycobacteria in macrophages. Trends Cell Biol. 2005;15:269–276.

ment of animal or in vitro cellular models could aid understanding of 8. Lerner TR, de Souza Carvalho-Wodarz C, Repnik U, et al. Lymphatic
endothelial cells are a replicative niche for Mycobacterium tubercu-
the underlying immune mechanisms and point the way to adjunctive
losis. J Clin Invest. 2016;126:1093–1108.
anti-inflammatory and improved antibiotic therapies. In human stud-
9. Hawn TR, Dunstan SJ, Thwaites GE, et al. A polymorphism in Toll-
ies high-resolution MRI or CT imaging to assess TBM disease activ-
interleukin 1 receptor domain containing adaptor protein is asso-
ity could provide a more detailed clinical phenotype for TBM than ciated with susceptibility to meningeal tuberculosis. J Infect Dis.
is currently available. Unbiased application of “omics” technologies 2006;194:1127–1134.
(particularly transcriptomics) in particular analysis of cells and fluid 10. Caws M, Thwaites G, Dunstan S, et al. The influence of host and bac-
arising from the site of disease is likely to be particularly valuable. terial genotype on the development of disseminated disease with
Mycobacterium tuberculosis. PLoS Pathog. 2008;4:e1000034.
Genes encoding proteins involved in metabolism, cell growth, trans-
port, immune response, cell communication, and signal transduction 11. Fernando SL, Saunders BM, Sluyter R, et al. A polymorphism in the
P2X7 gene increases susceptibility to extrapulmonary tuberculosis.
are particularly of interest. Longitudinal sampling of blood or CSF
Am J Respir Crit Care Med. 2007;175:360–366.
during observational studies or randomized controlled trials is highly
12. Caws M, Thwaites G, Stepniewska K, et al. Beijing genotype of
informative regarding the dynamics of the disease, such as disease Mycobacterium tuberculosis is significantly associated with human
progression and drug response. immunodeficiency virus infection and multidrug resistance in cases
of tuberculous meningitis. J Clin Microbiol. 2006;44:3934–3939.
AUTHORSHIP 13. Pulzova L, Bhide MR, Andrej K. Pathogen translocation across the
blood-brain barrier. FEMS Immunol Med Microbiol. 2009;57:203–213.
R.J.W. conceived the idea for the review. A.G.D., U.K.R., A.P., and R.J.W.
14. Be N, Kim K, Bishai W, Jain S. Pathogenesis of central nervous system
wrote the text and compiled figures. A.A.F. provided images and crit-
tuberculosis. Curr Mol Med. 2009;9:94–99.
ically reviewed the text. All authors agreed to the submission of
15. Rock RB, Olin M, Baker CA, Molitor TW, Peterson PK. Central ner-
the article.
vous system tuberculosis: pathogenesis and clinical aspects. Clin
Microbiol Rev. 2008;21:243–261.
ACKNOWLEDGMENTS 16. Donald P, Schaaf H, Schoeman J. Tuberculous meningitis and miliary
R.J.W. receives support from the Francis Crick Institute, which is tuberculosis: the Rich focus revisited. J Infect. 2005;50:193–195.
supported by Cancer Research UK (10218), Wellcome (10218), and 17. Rich A, MH. The pathogenesis of tuberculous meningitis. Bull John
UK Research and Innovation (10218). He also receives support from Hopkins Hosp. 1933;52:5–37.

Wellcome (104803, 203135) and the National Institutes of Health 18. Dastur DK, Manghani DK, Udani PM. Pathology and patho-
genetic mechanisms in neurotuberculosis. Radiol Clin North Am.
(U01AI115940). A.G.D. is supported by a Wellcome Clinicians Ph.D.
1995;33:733–752.
grant to University College London. A.A.F. is supported by the National
19. Peterson PK, et al. CD14 receptor-mediated uptake of nonopsonized
Research Foundation of South Africa through the SARChI Chair of
Mycobacterium tuberculosis by human microglia. Infect Immun.
Clinical Neurosciences. The funders had no role in the writing of the 1995;63:1598–1602.
review or decision to publish it. 20. Rock RB, Hu S, Gekker G, et al. Mycobacterium tuberculosis-induced
cytokine and chemokine expression by human microglia and astro-
DISCLOSURES cytes: effects of dexamethasone. J Infect Dis. 2005;192:2054–2058.

The authors declare no conflicts of interest. 21. Randall PJ, Hsu N-J, Lang D, et al. Neurons are host cells for Mycobac-
terium tuberculosis. Infect Immun. 2014;82:1880–1890.
22. Wright S, Ramos R, Tobias P, Ulevitch R, Mathison J. CD14, a receptor
REFERENCES for complexes of lipopolysaccharide (LPS) and LPS binding protein.
1. Marais S, Pepper DJ, Schutz C, Wilkinson RJ, Meintjes G. Presenta- Science. 1990;249:1431–1433.
tion and outcome of tuberculous meningitis in a high HIV prevalence
23. Shams H, Wizel B, Lakey DL, et al. The CD14 receptor does not medi-
setting. PLoS One. 2011;6:e20077.
ate entry of Mycobacterium tuberculosis into human mononuclear
2. O’Garra A, Redford PS, McNab FW, Bloom CI, Wilkinson RJ, Berry phagocytes. FEMS Immunol Med Microbiol. 2003;36:63–69.
MPR. The immune response in tuberculosis. Annu Rev Immunol.
24. Peterson PK, Gekker G, Hu S, et al. Multinucleated giant cell forma-
2013;31:475–527.
tion of swine microglia induced by Mycobacterium bovis. J Infect Dis.
3. Kumar V, Kumar V. Robbins and Cotran’s Pathologic Basis of Disease. 1996;173:1194–1201.
Philadelphia, PA; London: Saunders; 2010. xiv, 1450: ill.; 28 cm.
25. Yang C-S, Lee H-M, Lee J-Y, et al. Reactive oxygen species
4. Coico R, Sunshine G. Immunology: A Short Course. Hoboken, NJ: Wiley- and p47phox activation are essential for the Mycobacterium
Blackwell; 2009. xx, 391: col. ill.; 28 cm. tuberculosis-induced pro-inflammatory response in murine
5. Krishnan N, Robertson BD, Thwaites G. The mechanisms and con- microglia. J Neuroinflammation. 2007;4:27.
sequences of the extra-pulmonary dissemination of Mycobacterium 26. Mastroianni CM, Paoletti F, Lichtner M, D’Agostino C, Vullo V, Delia
tuberculosis. Tuberculosis. 2010;90:361–366. S. Cerebrospinal fluid cytokines in patients with tuberculous menin-
6. Jain SK, Paul-Satyaseela M, Lamichhane G, Kim KS, Bishai WR. gitis. Clin Immunol Immunopathol. 1997;84:171–176.
Mycobacterium tuberculosis invasion and traversal across an in vitro 27. Tobin DM, Roca FJ, Oh SF, et al. Host genotype-specific therapies can
human blood-brain barrier as a pathogenic mechanism for central optimize the inflammatory response to mycobacterial infections. Cell.
nervous system tuberculosis. J Infect Dis. 2006;193:1287–1295. 2012;148:434–446.
278 DAVIS ET AL .

28. Tsenova L, Bergtold A, Freedman VH, Young RA, Kaplan G. Tumor 48. Pechkovsky DV, Zalutskaya OM, Ivanov GI, Misuno NI. Calprotectin
necrosis factor alpha is a determinant of pathogenesis and disease (MRP8/14 protein complex) release during mycobacterial infection in
progression in mycobacterial infection in the central nervous system. vitro and in vivo. FEMS Immunol Med Microbiol. 2000;29:27–33.
Proc Natl Acad Sci. 1999;96:5657–5662. 49. Gopal R, Monin L, Torres D, et al. S100A8/A9 proteins mediate neu-
29. Kaplan G, Freedman VH. The role of cytokines in the immune trophilic inflammation and lung pathology during tuberculosis. Am J
response to tuberculosis. Res Immunol. 1996;147:565–572. Respir Crit Care Med. 2013;188:1137–1146.
30. Hashimoto M, Ishikawa Y, Yokota S, et al. Action site of circulating 50. Tadokera R, Meintjes G, Skolimowska KH, et al. Hypercytokinaemia
interleukin-1 on the rabbit brain. Brain Res. 1991;540:217–223. accompanies HIV-tuberculosis immune reconstitution inflammatory
31. de Vries HE, et al. The blood-brain barrier in neuroinflammatory dis- syndrome. Eur Respir J. 2011;37:1248–1259.
eases. Pharmacol Rev. 1997;49:143–155. 51. Marais S, Wilkinson KA, Lesosky M, et al. Neutrophil-associated
32. Ramilo O. Tumor necrosis factor alpha/cachectin and interleukin 1 central nervous system inflammation in tuberculous meningitis
beta initiate meningeal inflammation. J Exp Med. 1990;172:497–507. immune reconstitution inflammatory syndrome. Clin Infect Dis.
2014;59:1638–1647.
33. Tsenova L, Sokol K, Freedman VH, Kaplan G. A combination of
thalidomide plus antibiotics protects rabbits from mycobacterial 52. Lange C, Storkebaum E, de Almodóvar CR, Dewerchin M, Carmeliet
meningitis-associated death. J Infect Dis. 1998;177:1563–1572. P. Vascular endothelial growth factor: a neurovascular target in neu-
rological diseases. Nat Rev Neurol. 2016;12:439–454.
34. Tsenova L, Mangaliso B, Muller G, et al. Use of IMiD3, a thalidomide
analog, as an adjunct to therapy for experimental tuberculous menin- 53. Ayata C, Ropper AH. Ischaemic brain oedema. J Clin Neurosci.
gitis. Antimicrob Agents Chemother. 2002;46:1887–1895. 2002;9:113–124.

35. Schoeman JF, Springer P, van Rensburg AJ, et al. Adjunctive thalido- 54. van der Flier M, Hoppenreijs S, van Rensburg AJ, et al. Vascu-
mide therapy for childhood tuberculous meningitis: results of a ran- lar endothelial growth factor and blood-brain barrier disruption in
domized study. J Child Neurol. 2004;19:250–257. tuberculous meningitis. Pediatr Infect Dis J. 2004;23:608–613.

36. van Toorn R, du Plessis A-M, Schaaf HS, Buys H, Hewlett RH, Schoe- 55. Kim H, Lee JM, Park JS, et al. Dexamethasone coordinately regulates
man JF. Clinicoradiologic response of neurologic tuberculous mass angiopoietin-1 and VEGF: a mechanism of glucocorticoid-induced
lesions in children treated with thalidomide. Pediatr Infect Dis J. stabilization of blood-brain barrier. Biochem Biophys Res Commun.
2015;34:214–218. 2008;372:243–248.

37. Simmons CP, Thwaites GE, Quyen NTH, et al. Pretreatment intrac- 56. Garg R, Mahdi A, Jain A, et al. Cerebrospinal fluid cytokines and
erebral and peripheral blood immune responses in Vietnamese adults matrix metalloproteinases in human immunodeficiency seropositive
with tuberculous meningitis: diagnostic value and relationship to dis- and seronegative patients of tuberculous meningitis. Ann Indian Acad
ease severity and outcome. J Immunol. 2006;176:2007–2014. Neurol. 2014;17:171–178.

38. Saunders BM, Frank AA, Orme IM, Cooper AM. Interleukin-6 induces 57. More A, Verma R, Garg RK, et al. A study of neuroendocrine
early gamma interferon production in the infected lung but is not dysfunction in patients of tuberculous meningitis. J Neurol Sci.
required for generation of specific immunity to Mycobacterium 2017;379:198–206.
tuberculosis infection. Infect Immun. 2000;68:3322–3326. 58. Misra UK, Kalita J, Bhoi SK, Singh RK. A study of hyponatremia in
39. Xing Z, Gauldie J, Cox G, et al. IL-6 is an antiinflammatory cyto- tuberculous meningitis. J Neurol Sci. 2016;367:152–157.
kine required for controlling local or systemic acute inflammatory 59. Cotton MF, Donald PR, Schoeman JF, Aalbers C, Van Zyl LE, Lom-
responses. J Clin Invest. 1998;101:311–320. bard C. Plasma arginine vasopressin and the syndrome of inappropri-
40. Curto M, Reali C, Palmieri G, et al. Inhibition of cytokines expression ate antidiuretic hormone secretion in tuberculous meningitis. Pediatr
in human microglia infected by virulent and non-virulent mycobacte- Infect Dis J. 1991;10:837–842.
ria. Neurochem Int. 2004;44:381–392. 60. Singh BS, Patwari AK, Deb M. Serum sodium and osmolal changes in
41. Chen Y, Zhang J, Wang X, et al. HMGB1 level in cerebrospinal fluid as tuberculous meningitis. Indian Pediatr. 1994;31:1345–1350.
a complimentary biomarker for the diagnosis of tuberculous menin- 61. Misra UK, Kalita J, Kumar M, Neyaz Z. Hypovolemia due to cerebral
gitis. SpringerPlus. 2016;5:1775. salt wasting may contribute to stroke in tuberculous meningitis. QJM.
42. Rohlwink UK, Mauff K, Wilkinson KA, et al. Biomarkers of cere- 2018;111:455–460.
bral injury and inflammation in pediatric tuberculous meningitis. Clin 62. Thwaites GE, Bang ND, Dung NH, et al. Dexamethasone for the treat-
Infect Dis. 2017;65:1298–1307. ment of tuberculous meningitis in adolescents and adults. N Engl J
43. Berger NA, et al. The pathogenesis of tuberculous meningitis. Bull Med. 2004;351:1741–1751.
John Hopkins Hosp. 1933;175:192–222. 63. Bang ND, Caws M, Truc TT, et al. Clinical presentations, diagnosis,
44. Ko HL, Ren EC. Functional aspects of PARP1 in DNA repair and tran- mortality and prognostic markers of tuberculous meningitis in Viet-
scription. Biomolecules. 2012;2:524–548. namese children: a prospective descriptive study. BMC Infect Dis.
2016;16:573.
45. Heizmann CW, Fritz G, Schafer BW. S100 proteins: structure, func-
tions and pathology. Front Biosci. 2002;7:d1356–68. 64. Mason S, Reinecke CJ, Kulik W, van Cruchten A, Solomons R, van
Furth AMT. Cerebrospinal fluid in tuberculous meningitis exhibits
46. Mahon RN, Hafner R. Immune cell regulatory pathways unexplored only the L-enantiomer of lactic acid. BMC Infect Dis. 2016;16:251.
as host-directed therapeutic targets for mycobacterium tuberculosis:
an opportunity to apply precision medicine innovations to infectious 65. Lesniak WG, Jyoti A, Mishra MK, et al. Concurrent quantification
diseases. Clin Infect Dis. 2015;61(Suppl 3):S200–S216. of tryptophan and its major metabolites. Anal Biochem. 2013;443:
222–231.
47. Ryckman C, Vandal K, Rouleau P, Talbot M, Tessier PA. Proin-
flammatory activities of S100: proteins S100A8, S100A9, and 66. Campbell BM, Charych E, Lee AW, Möller T. Kynurenines in
S100A8/A9 induce neutrophil chemotaxis and adhesion. J Immunol. CNS disease: regulation by inflammatory cytokines. Front Neurosci.
2003;170:3233–3242. 2014;8:12.
DAVIS ET AL . 279

67. O’Connor JC, Andre C, Wang Y, et al. Interferon-gamma and 85. Areeshi MY, et al. Vitamin D receptor ApaI (rs7975232) polymor-
tumor necrosis factor-alpha mediate the upregulation of indoleamine phism confers decreased risk of pulmonary tuberculosis in overall
2,3-dioxygenase and the induction of depressive-like behavior in and African population, but not in Asians: evidence from a meta-
mice in response to bacillus Calmette-Guerin. J Neurosci. 2009;29: analysis. Ann Clin Lab Sci. 2017;47:628–637.
4200–4209. 86. Gagneux S, Small PM. Global phylogeography of Mycobacterium
68. Majláth ZF, Tajti JN, Vécsei L. Kynurenines and other novel therapeu- tuberculosis and implications for tuberculosis product development.
tic strategies in the treatment of dementia. Ther Adv Neurol Disord. Lancet Infect Dis. 2007;7:328–337.
2013;6:386–397. 87. Gagneux S, DeRiemer K, Van T, et al. Variable host-pathogen
69. van Laarhoven A, Dian S, Aguirre-Gamboa RL, et al. Cerebral trypto- compatibility in Mycobacterium tuberculosis. Proc Natl Acad Sci.
phan metabolism and outcome of tuberculous meningitis: an obser- 2006;103:2869–2873.
vational cohort study. Lancet Infect Dis. 2018;18:526–535. 88. Comas IA, Coscolla M, Luo T, et al. Out-of-Africa migration and
70. Weiner J, Parida SK, Maertzdorf J, et al. Biomarkers of inflam- Neolithic coexpansion of Mycobacterium tuberculosis with modern
mation, immunosuppression and stress with active disease are humans. Nat Genet. 2013;45:1176–1182.
revealed by metabolomic profiling of tuberculosis patients. PLoS One. 89. Tsenova L, Ellison E, Harbacheuski R, et al. Virulence of selected
2012;7:e40221. Mycobacterium tuberculosis clinical isolates in the rabbit model
71. Wobke TK, Sorg BL, Steinhilber D. Vitamin D in inflammatory dis- of meningitis is dependent on phenolic glycolipid produced by the
eases. Front Physiol. 2014;5:244. bacilli. J Infect Dis. 2005;192:98–106.
72. Graustein AD, Horne DJ, Arentz M, et al. TLR9 gene region poly- 90. Kong Y, Cave MD, Zhang L, et al. Association between Mycobac-
morphisms and susceptibility to tuberculosis in Vietnam. Tuberculosis. terium tuberculosis Beijing/W lineage strain infection and
2015;95:190–196. extrathoracic tuberculosis: insights from epidemiologic and clinical
73. Thuong NTT, Hawn TR, Thwaites GE, et al. A polymorphism in characterization of the three principal genetic groups of M. tubercu-
human TLR2 is associated with increased susceptibility to tubercu- losis clinical isolates. J Clin Microbiol. 2007;45:409–414.
lous meningitis. Genes Immun. 2007;8:422–428. 91. DORMANS J, BURGER M, AGUILAR D, et al. Correlation of vir-
74. Tobin DM, Vary JC, Ray JP, et al. The lta4h locus modulates sus- ulence, lung pathology, bacterial load and delayed type hyper-
ceptibility to mycobacterial infection in zebrafish and humans. Cell. sensitivity responses after infection with different Mycobacterium
2010;140:717–730. tuberculosis genotypes in a BALB/c mouse model. Clin Exp Immunol.
2004;137:460–468.
75. Yang J, Chen J, Yue J, Liu L, Han M, Wang H. Relationship between
human LTA4H polymorphisms and extra-pulmonary tuberculosis in 92. Guerra-Assunção JA, Houben RMGJ, Crampin AC, et al. Recurrence
an ethnic Han Chinese population in Eastern China. Tuberculosis. due to relapse or reinfection with Mycobacterium tuberculosis: a
2014;94:657–663. whole-genome sequencing approach in a large, population-based
cohort with a high HIV infection prevalence and active follow-up.
76. Dunstan SJ, Tram TTB, Thwaites GE, et al. LTA4H genotype is asso- J Infect Dis. 2015;211:1154–1163.
ciated with susceptibility to bacterial meningitis but is not a critical
determinant of outcome. PLoS One. 2015;10:e0118789. 93. Reed MB, Domenech P, Manca C, et al. A glycolipid of hypervirulent
tuberculosis strains that inhibits the innate immune response. Nature.
77. van Laarhoven A, Dian S, Ruesen C, et al. Clinical parameters, routine 2004;431:84–87.
inflammatory markers, and LTA4H genotype as predictors of mor-
tality among 608 patients with tuberculous meningitis in Indonesia. 94. Coscolla M, Gagneux S. Consequences of genomic diver-
J Infect Dis. 2017;215:1029–1039. sity in Mycobacterium tuberculosis. Semin Immunol. 2014;26:
431–444.
78. Thuong NTT, Heemskerk D, Tram TTB, et al. Leukotriene A4 hydro-
lase genotype and HIV infection influence intracerebral inflam- 95. Guerra-Assunção JA, Crampin A, Houben R, et al. Large-scale whole
mation and survival from tuberculous meningitis. J Infect Dis. genome sequencing of M. tuberculosis provides insights into trans-
2017;215:1020–1028. mission in a high prevalence area. eLife. 2015;4.

79. Fava VM, Schurr E. Evaluating the impact of LTA4H genotype and 96. de Jong BC, Hill PC, Aiken A, et al. Progression to active tuberculosis,
immune status on survival from tuberculous meningitis. J Infect Dis. but not transmission, varies by Mycobacterium tuberculosis lineage
2017;215:1011–1013. in the Gambia. J Infect Dis. 2008;198:1037–1043.

80. Campo M, Randhawa AK, Dunstan S, et al. Common polymorphisms 97. van Crevel R, Nelwan RHH, de Lenne W, et al. Mycobacterium tuber-
in the CD43 gene region are associated with tuberculosis disease and culosis Beijing genotype strains associated with febrile response to
mortality. Am J Respir Cell Mol Biol. 2015;52:342–348. treatment. Emerg Infect Dis. 2001;7:880–883.

81. Hoppe HC, et al. Identification of phosphatidylinositol mannoside as 98. Parwati I, Alisjahbana B, Apriani L, et al. Mycobacterium tuber-
a mycobacterial adhesin mediating both direct and opsonic binding to culosis Beijing genotype is an independent risk factor for tuber-
nonphagocytic mammalian cells. Infect Immun. 1997;65:3896–3905. culosis treatment failure in Indonesia. J Infect Dis. 2010;201:
553–557.
82. Hoal-Van Helden EG, Epstein J, Victor TC, et al. Mannose-binding
protein B allele confers protection against tuberculous meningitis. 99. Sarkar R, Lenders L, Wilkinson KA, Wilkinson RJ, Nicol MP. Modern
Pediatr Res. 1999;45:459–464. lineages of Mycobacterium tuberculosis exhibit lineage-specific pat-
terns of growth and cytokine induction in human monocyte-derived
83. Horne DJ, Randhawa AK, Chau TTH, et al. Common polymorphisms macrophages. PLoS One. 2012;7:e43170.
in the PKP3-SIGIRR-TMEM16J gene region are associated with sus-
ceptibility to tuberculosis. J Infect Dis. 2012;205:586–594. 100. Portevin D, Gagneux SB, Comas IA, Young D. Human macrophage
responses to clinical isolates from the Mycobacterium tuberculosis
84. Rizvi I, Garg RK, Jain A, et al. Vitamin D status, vitamin D receptor and complex discriminate between ancient and modern lineages. PLoS
toll like receptor-2 polymorphisms in tuberculous meningitis: a case- Pathog. 2011;7:e1001307.
control study. Infection. 2016;44:633–640.
280 DAVIS ET AL .

101. Rakotosamimanana N, Raharimanga V, Andriamandimby SF, et al. 119. Daniel PM. Gross morbid anatomy of the central nervous system of
Variation in gamma interferon responses to different infecting strains cases of tuberculous meningitis treated with streptomycin. Proc R Soc
of Mycobacterium tuberculosis in acid-fast bacillus smear-positive Med. 1949;42:169–174.
patients and household contacts in Antananarivo, Madagascar. Clin 120. Shinoyama M, Suzuki M, Nomura S. Fulminant tubercu-
Vaccine Immunol. 2010;17:1094–1103. lous meningitis–autopsy case report. Neurol Med Chir (Tokyo).
102. Katrak SM, Shembalkar PK, Bijwe SR, Bhandarkar LD. The clini- 2012;52:761–764.
cal, radiological and pathological profile of tuberculous meningitis in 121. Lammie GA, Hewlett RH, Schoeman JF, Donald PR. Tuberculous cere-
patients with and without human immunodeficiency virus infection. brovascular disease: a review. J Infect. 2009;59:156–166.
J Neurol Sci. 2000;181:118–126.
122. Rohlwink UK, Kilborn T, Wieselthaler N, Banderker E, Zwane E, Figaji
103. Karande S, Gupta V, Kulkarni M, Joshi A, Rele M. Tuberculous menin- AA. Imaging features of the brain, cerebral vessels and spine in pedi-
gitis and HIV. Ind J Pediatr. 2005;72:755–760. atric tuberculous meningitis with associated hydrocephalus. Pediatr
104. Thwaites GE, Duc Bang N, Huy Dung N, et al. The influence Infect Dis J. 2016;35:e301–e310.
of HIV infection on clinical presentation, response to treatment, 123. van Well GTJ, Paes BF, Terwee CB, et al. Twenty years of pediatric
and outcome in adults with Tuberculous meningitis. J Infect Dis. tuberculous meningitis: a retrospective cohort study in the western
2005;192:2134–2141. cape of South Africa. Pediatrics. 2009;123:e1–e8.
105. Karstaedt A. Tuberculous meningitis in South African urban adults. 124. Thwaites GE, Macmullen-Price J, Chau TTH, et al. Serial MRI to
QJM. 1998;91:743–747. determine the effect of dexamethasone on the cerebral pathology of
106. Azuaje C, Fernández Hidalgo N, Almirante B, et al. Tubercu- tuberculous meningitis: an observational study. The Lancet Neurology.
lous meningitis: a comparative study in relation to concurrent 2007;6:230–236.
human immunodeficiency virus infection. Enferm Infecc Microbiol Clín. 125. Schoeman JF, Fieggen G, Seller N, Mendelson M, Hartzenberg B.
2006;24:245–250. Intractable intracranial tuberculous infection responsive to thalido-
107. Torok ME, Chau TTH, Mai PP, et al. Clinical and microbiological fea- mide: report of four cases. J Child Neurol. 2006;21:301–308.
tures of HIV-associated tuberculous meningitis in Vietnamese adults. 126. Kumar R, SV. Tuberculous brain stem abscesses in children. J Pediatr
PLoS One. 2008;3:e1772. Neurol. 2004;2:101–106.
108. Atluri VSR, Hidalgo M, Samikkannu T, et al. Effect of human immun- 127. Kumar R, Pandey C, Bose N, Sahay S. Tuberculous brain abscess: clini-
odeficiency virus on blood-brain barrier integrity and function: an cal presentation, pathophysiology and treatment (in children). Child’s
update. Front Cell Neurosci. 2015;9:212. Nerv Syst. 2002;18:118–123.
109. Marais S, Meintjes G, Pepper DJ, et al. Frequency, severity, and pre- 128. Schoeman JF, Morkel A, Seifart HI, et al. Massive posterior fossa
diction of tuberculous meningitis immune reconstitution inflamma- tuberculous abscess developing in a young child treated for miliary
tory syndrome. Clin Infect Dis. 2013;56:450–460. tuberculosis. Possible role of very rapid acetylation of isoniazid. Pedi-
110. Lai RPJ, Meintjes G, Wilkinson KA, et al. HIV-tuberculosis-associated atr Neurosurg. 1998;29:64–68.
immune reconstitution inflammatory syndrome is characterized 129. Saini AG, Dogra S, Kumar R, Nada R, Singh M. Primary tuberculous
by Toll-like receptor and inflammasome signalling. Nat Commun. cerebellar abscess: case report. Ann Trop Paediatr. 2011;31:367–369.
2015;6:8451.
130. Moghtaderi A, Alavi Naini R. Tuberculous radiculomyelitis: review
111. Tan HY, Yong YK, Andrade BB, et al. Plasma interleukin-18 levels are and presentation of five patients. Int J Tuberc Lung Dis. 2003;7:
a biomarker of innate immune responses that predict and charac- 1186–1190.
terize tuberculosis-associated immune reconstitution inflammatory
syndrome. AIDS. 2015;29:421–431. 131. Hernandez Pando R, Aguilar D, Cohen I, et al. Specific bacterial
genotypes of Mycobacterium tuberculosis cause extensive dissem-
112. Tan HY, Yong YK, Shankar EM, et al. Aberrant inflammasome acti- ination and brain infection in an experimental model. Tuberculosis.
vation characterizes tuberculosis-associated immune reconstitution 2010;90:268–277.
inflammatory syndrome. J Immunol. 2016;196:4052–4063.
132. Srivastava T, Kochar DK. Asymptomatic spinal arachnoiditis in
113. Marais S, et al. Inflammasome activation underlying central ner- patients with tuberculous meningitis. Neuroradiology. 2003;45:
vous system deterioration in HIV-associated tuberculosis. J Infect Dis. 727–729.
2017;215:677–686.
133. Skendros P, Kamaria F, Kontopoulos V, Tsitouridis I, Sidiropoulos L.
114. Bergsbaken T, Fink SL, Cookson BT. Pyroptosis: host cell death and Intradural, eextramedullary tuberculoma of the spinal cord as a com-
inflammation. Nat Rev Microbiol. 2009;7:99–109. plication of tuberculous meningitis. Infection. 2003;31:115–117.
115. Tozzi V, Balestra P, Lorenzini P, et al. Prevalence and risk factors for 134. Lim YS, Kim SB, Kim MK, Lim YJ. Disseminated tuberculosis of central
human immunodeficiency virus-associated neurocognitive impair- nervous system: spinal intramedullary and intracranial tuberculomas.
ment, 1996 to 2002: results from an urban observational cohort. J Korean Neurosurg Soc. 2013;54:61–64.
J Neurovirol. 2005;11:265–273.
135. Kato M, Mochizuki T, Negaro K, Fukusako T, Nogaki H, Morimatsu
116. Isaacson PG, Hall-Craggs M, Lucas S, et al. Cerebral CD8+ lymphocy- M. [Magnetic resonance imaging of a case of central nervous system
tosis in HIV-1 infected patients with immune restoration induced by tuberculosis with tuberculous arachnoiditis and multiple tuberculo-
HAART. Acta Neuropathol (Berl). 2004;108:17–23. mas]. Nippon Ronen Igakkai Zasshi. 1997;34:818–824.
117. Johnson TP, Patel K, Johnson KR, et al. Induction of IL-17 and non-
classical T-cell activation by HIV-Tat protein. Proc Natl Acad Sci.
2013;110:13588–13593. How to cite this article: Davis AG, Rohlwink UK, Proust A, Figaji
118. Boulware DR, Meya DB, Bergemann TL, et al. Clinical features and AA, Wilkinson RJ. The pathogenesis of tuberculous meningi-
serum biomarkers in HIV immune reconstitution inflammatory syn- tis. J Leukoc Biol. 2019;105:267–280. https://doi.org/10.1002/
drome after cryptococcal meningitis: a prospective cohort study. JLB.MR0318-102R
PLoS Med. 2010;7:e1000384.

You might also like