Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2013 Mar 6.
Published in final edited form as: Future Microbiol. 2012 Mar;7(3):383–394. doi: 10.2217/fmb.12.1

Defense at the border: the blood–brain barrier versus bacterial foreigners

Nina M van Sorge 1,2, Kelly S Doran 2,3,*
PMCID: PMC3589978  NIHMSID: NIHMS446494  PMID: 22393891

Abstract

Bacterial meningitis is among the top ten causes of infectious disease-related deaths worldwide, with up to half of the survivors left with permanent neurological sequelae. The blood–brain barrier (BBB), composed mainly of specialized brain microvascular endothelial cells, maintains biochemical homeostasis in the CNS by regulating the passage of nutrients, molecules and cells from the blood to the brain. Despite its highly restrictive nature, certain bacterial pathogens are able to gain entry into the CNS resulting in serious disease. In recent years, important advances have been made in understanding the molecular and cellular events that are involved in the development of bacterial meningitis. In this review, we summarize the progress made in elucidating the molecular mechanisms of bacterial BBB-crossing, highlighting common themes of host–pathogen interaction, and the potential role of the BBB in innate defense during infection.

Keywords: blood–brain barrier, Escherichia coli K1, group B Streptococcus, Haemophilus influenzae type B, meningitis, Neisseria meningitidis, neutrophil, Streptococcus pneumoniae, tight/adherens junction

Dynamic epidemiology of bacterial meningitis

Despite our enormous progress in the treatment and prevention of infectious diseases, there are still 1.2 million cases of bacterial meningitis per year, 170,000 of which are fatal [201]. In addition, permanent neurological sequelae occur in up to 50% of survivors [1,2]. Although many bacteria can cause disease in humans, only a limited number of pathogens are isolated from the CNS of patients. The most prevalent cause of meningitis varies depending on geographic location, socioeconomic status, age, vaccination availability and overall health status of the individual.

Globally, Streptococcus pneumoniae, Neisseria meningitidis (meningococcus) and Haemophilus influenzae type B (HiB) are the most common causes of meningitis in infants and adults. In industrialized countries, both the incidence and epidemiology of meningitis have been dramatically affected by the introduction of vaccines against these organisms. Before the introduction of the conjugate HiB vaccine in the 1980s, HiB alone accounted for approximately half of the cases of meningitis in infants, compared with a 78% reduction in disease incidence following routine vaccination [3,4]. Globally, however, HiB is estimated to still cause 173,000 cases of meningitis each year [5] as HiB vaccination programs are not yet available in developing countries [4]. Strategies are in place to introduce the HiB vaccine in resource-poor countries over the next several years [202]. Currently, S. pneumoniae is the worldwide leading cause of bacterial invasive disease in children younger than 5 years of age [6] and of bacterial meningitis in all age groups except young infants [7]. In addition, in sub-Saharan Africa, also called the ‘meningitis belt’, N. meningitidis is a leading cause of large epidemics of meningococcal meningitis. Vaccination strategies against S. pneumonia and N. meningitidis have been limited to several serotypes and are overall hampered by concurrent increases in nonvaccine serotypes [8-10]. Therefore, a complete elimination or prevention of pneumococcal and meningococcal meningitis will only be possible when vaccines either include all serotypes, or a conserved antigen present in all disease isolates.

Although not classically thought of as a meningeal pathogen, non-typhoidal Salmonella (NTS) species are a frequent cause of meningitis in certain parts of Africa [11-13]. In northern Uganda, it has even been reported as the second most common cause of pediatric meningitis after the introduction of the HiB vaccine in 2002 [14]. The incidence of NTS meningitis is likely to increase given that NTS has become the most common cause of bacteremia in tropical Africa [15]. NTS meningitis has a high case fatality rate (up to 60%) and a high incidence of postinfectious sequelae in survivors as treatment is often complicated by multidrug resistance [11-13,15,16].

A different pathogen spectrum is associated with meningitis in the newborn. In many industrialized countries, Streptococcus agalactiae, also known as group B Streptococcus (GBS), Listeria monocytogenes and Escherichia coli K1 are the most common causes of meningitis in the neonatal period [7,17-19]. By contrast, in developing countries, additional pathogens such as Klebsiella, Staphylococcus aureus, and NTS species, are frequently isolated from neonates, and GBS is usually less common compared with many industrialized countries [20,21]. No vaccination strategies are currently in place to prevent these types of infections but they are anticipated to reduce the number of meningitis cases [7]. Fortunately, increased screening and intrapartum antibiotic prophylaxis has resulted in a decline in early-onset GBS invasive disease in the USA [22,23]. However, this treatment has not eliminated the incidence of GBS meningitis and concern has been raised about coinciding increases in non-GBS early-onset invasive disease, especially in preterm infants as a result of increased antibiotic use [24,25].

Besides these frequent causes of meningitis in different age groups, additional pathogens are known to cause meningitis in vulnerable groups, such as immunocompromised patients, which includes infants and the elderly adults (>65 years of age), or specific population groups. Meningitis due to Mycobacterium tuberculosis and NTS species occurs relatively frequently in HIV-infected and other immunocompromised patients [26-28], whereas Streptococcus suis is the most common cause of acute bacterial meningitis in southeast Asia, but is rare in other countries [29,30]. Although still considered a rare complication, increasing numbers of S. aureus meningitis cases are observed in the hospital setting [31], usually as a result of postoperative complications or hematogenous spread due to underlying diseases such as HIV, immune deficiencies, diabetes or cardiovascular disease [31,32]. The mortality rate seems especially high when disease was contracted in the community [31-34]. Finally, Bacillus anthracis, an exotic cause of infection and meningitis overall, has gained research interest as a result of its use in bioterrorism attacks in 2001 [35]. In addition to its potential use as a bioweapon, B. anthracis is still a relevant natural cause of infection to people living in endemic regions in agricultural settings or exposure due to occupation [36]. Just recently, an outbreak of systemic anthrax occurred in Scotland among intravenous drug users due to injection of contaminated heroin [37]. Anthrax meningitis is the main neurological complication of systemic infection, approaching 100% mortality despite intensive antibiotic therapy [38].

The pathogenesis & pathophysiology of bacterial meningitis

Many of the meningeal pathogens, including HiB, N. meningitidis, S. pneumoniae, GBS and S. aureus, are able to colonize the skin, upper respiratory, nasopharyngeal, GI or vaginal tract of healthy individuals. Carriage can be stable, transient or intermittent [39] and often remains asymptomatic. However, in certain cases, bacteria penetrate host cellular barriers to initiate a local infection that can result in systemic spread. An association between high-level bacteremia and development of meningitis has been suggested for E. coli, S. pneumoniae, GBS and HiB from experimental models of hematogeneous meningitis [40-45]. This implies that bloodstream survival is an important virulence trait of meningeal pathogens to avoid immune clearance by complement- and antibody-mediated phagocytic killing by host immune cells. Indeed, once bacteria reach the bloodstream, their transcriptional profile changes dramatically and includes survival strategies such as an altered cell membrane or cell wall composition [46,47] and increased expression of complement regulatory proteins [48,49] or iron-uptake systems [48]. Another strategy to prevent bloodstream clearance is adopted by E. coli K1 through expression of OmpA. This bacterium initiates its specific uptake in macrophages and dendritic cells through FcγRIa (CD64), and in neutrophils through gp96, to replicate intra-cellularly [50]. In this niche, the bacterium is protected from serum bactericidal activity and from phagocyte-mediated clearance by suppressing oxidative burst [51-53]. Increasing bacterial clearance from the blood would prevent or limit bacterial survival and multiplication, thereby limiting the possibility to reach the CNS. Vaccination poses a successful strategy to limit the risk for developing meningitis as it significantly decreases carriage, and reduces the levels of bacteremia due to enhanced phagocytic clearance by neutrophils and macrophages for vaccine-included serotypes. Once disease is established, early and aggressive antibiotic therapy can help reduce the bacterial load associated with disease progression.

Following bloodstream survival, bacteria will ultimately leave the bloodstream and invade the CNS, resulting in inflammation of the meninges, increased BBB permeability and pleocytosis. The molecular mechanisms involved in bacterial penetration of the BBB are complex, and common themes are described in more detail below. Subsequent CNS tissue injury results from cerebral ischemia, edema, hydrocephalus and increased intracranial pressure [54] and is caused by both toxic bacterial products and host inflammatory pathways initiated to clear the infection. In particular, the excessive inflammatory response of neutrophils has been associated with increased CNS injury [55,56]. Consequently, several large clinical trials have studied the potential benefit of corticosteroid therapy to improve disease outcome by reducing unwanted inflammatory responses [57-60]. Overall, adjuvant corticosteroid therapy does not decrease mortality [61], although it does reduce hearing loss and neurological sequelae, but only in high income countries [61].

BBB composition

The BBB is a structural and functional barrier that maintains the homeostasis of the neutral microenvironment by impeding the passage of virtually all molecules except those that are small and lipophilic [62,63]. The BBB is composed of brain microvascular endothelial cells (BMECs) that line cerebral microvessels along with periendothelial structures, which include pericytes, astrocytes and a basal membrane [64]. Together, these cells function in controlling the infiltration of blood proteins and cells through the vessel wall and into underlying tissues and in general provide functional barrier properties [65]. Brain endothelial cells are distinguished from the other cells of the BBB by possessing fewer cytoplasmic vesicles, more mitochondria and a large number of intercellular junctions that promote high transendothelial electrical resistance and retard paracellular flux [66]. Similar to epithelial cells, endothelial cells possess adherens junctions (AJs) and tight junctions (TJs) at their intercellular contacts [67,68]. Although TJs and AJs are formed by different molecules, cell–cell adhesion is accomplished similarly by a trans-membrane protein that interacts with the actin cytoskeleton and/or signaling proteins through an adaptor protein [65]. AJs in BMECs are dependent on the interaction between the cytoplasmic tail of cadherins with catenins, which in turn are linked to the actin cytoskeleton and/or signaling components [69]. TJs in BMECs are composed of four integral membrane proteins (occludin, claudins, junctional adhesion molecules and cell-selective adhesion molecules) that are linked through cytoplasmic proteins (ZO-1, -2 and -3 and cingulin) to the actin cytoskeleton [68,70,71]. Numerous endothelial functions are mediated by these junctional structures including maintenance of cell polarity, signaling, modulation of transcription and endothelial stabilization [72-74]. Another critically important function of AJs and TJs is regulation of BBB permeability. Until recently, AJs were considered to be important for basic cell–cell adhesion but not critically involved in regulating BBB permeability. However, a recent study demonstrated cross-talk between the TJ and AJ. Homophilic interactions between vascular endothelial cadherin expressed in AJs were demonstrated to regulate expression of TJ claudin-5 [75], providing a molecular mechanism for increased permeability when interfering with AJ formation.

Transcellular penetration of the BBB

The initial attachment of blood-borne bacteria to brain endothelium and subsequent invasion may represent the initial step in penetration and/or disruption of the BBB. This interaction involves a complex interplay between host receptors and bacterial components. In recent years, significant progress has been made in understanding the molecular interaction between the BBB and meningeal pathogens because of the availability of in vitro tissue culture models of human BMECs (hBMECs) [76-78] and in vivo animal models of hematogeneous meningitis [79-83]. Many meningeal bacteria cross the BBB transcellularly as live organisms, requiring host actin cytoskeletal rearrangements that promote the initial bacterial uptake into the cell [78,84-91]. The signal transduction pathways involved display some common themes shared by different organisms, as will be discussed in more detail below. Most in vitro work has focused on identifying the bacterial molecules and host receptors involved in bacterial adherence to, and invasion of, the BBB, as recently reviewed by Kim [92,93]. Identification of critical bacterial adhesion/invasion molecules can be achieved using different approaches. An unbiased and comprehensive approach to identify new bacterial components involved in BBB interaction uses random mutant libraries and screens for loss of adherence and/or invasion [94,95]. This approach identified lipoteichoic acid as a critical mediator in brain endothelial cell invasion for GBS, S. aureus and S. pneumoniae both in vitro and in vivo [83,95]. Complementarily, one can determine critical adhesins and invasins by analyzing bacterial transcriptional profiles [87,96,97] or differential fluorescence gene induction [98] during BBB interaction. Subsequent ana lysis of hBMEC adherence/invasion using defined isogenic mutants of candidate genes is needed to confirm protein involvement. For example, this approach identified a strong contribution for type I fimbriae in E. coli K1 in hBMEC adherence [97]. Bioinformatic approaches comparing bacterial whole genome sequences can also identify key molecules as recently exemplified for the HvgA protein found in hypervirulent GBS strains [99]. In addition to expression of specific adhesion molecules, brain tropism seems to be critically determined by low blood flow in brain capillaries, as was recently demonstrated for N. meningitidis [100]. Importantly, bacterial adherence to endothelial cells was similar under low flow versus static conditions [100].

From the body of work studying molecular interactions between the BBB and bacteria, some commonalities in the way different Gram-positive and Gram-negative bacteria interact with brain endothelial cells can be discerned. First, many meningeal bacteria, including E. coli K1 [97], GBS [101,102] and N. meningitidis [103] use pili (also called fimbriae) or the shorter version, fibrils [104], to initiate binding to hBMECs. A similar mechanism is probably involved in BMEC interaction with S. pneumoniae [105,106] and HiB [107,108] as their pili or fibrils have been shown to contribute to interaction with other epithelial and endothelial cell types. Second, many meningeal pathogens possess a capsular polysaccharide [80,84,109-111], which promotes bloodstream survival and high-level bacteremia prior to BBB penetration. Studies performed using in vitro assays demonstrate that the capsule actually inhibits bacterial invasion, probably due to electrostatic repulsion or masking of bacterial surface structures that could function as adhesins. Thus, it is likely that capsule expression is highly regulated and may be induced during bloodstream replication and repressed while on mucosal or endothelial cell surfaces [86,112]. Finally, the expression of bacterial toxins can increase BBB penetration through different mechanisms. In the case of E. coli K1, cytotoxic necrotizing factor-1 activates RhoA, resulting in increased invasion of brain endothelial cells in vitro and increased penetration of the BBB in vivo [113]. By contrast, GBS β hemolysin (β-h/c) [84], S. pneumoniae pneumolysin [114] and HiB lipopolysaccharide [115] damage brain endothelial cells, resulting in increased BBB permeability in vivo [80,116].

A broad range of host receptors has been described to mediate interaction with meningeal pathogens (reviewed by [92]); also here, common themes can be recognized. First, bacterial binding to host receptors on brain endothelium seems to be important for initial attachment. The laminin receptor (LR) and platelet activating factor receptor (PAFr) have been identified as common portals of CNS entry for the leading meningeal pathogens N. meningitidis, S. pneumoniae and H. influenzae [86,117-119]. In addition, LR was identified in previous studies as an important internalization receptor for E. coli K1 on hBMECs [120,121]. Different bacterial adhesins, N. meningitidis PilQ and PorA, S. pneumoniae CbpA and H. influenzae OmpP2, all target a common carboxy-terminal domain of LR to establish initial contact with brain endothelium [117]. Sequential binding to PAFr through bacterial surface phosphorylcholine results in β-arrestin-mediated pneumococcal invasion of brain endothelial cells [86,122], and interfering with PAFr-mediated uptake protects the host from pneumococcal meningitis in vivo [122,123]. It is likely that both N. meningitidis and Haemophilus species similarly interact with PAFr on brain endothelial cells as both express phosphorylcholine on their pili and lipopolysaccharide, respectively [119,124-126]; however, further experimentation is needed to definitively demonstrate this. Second, the BBB interaction is not always direct but may involve bridging molecules such as components of the extracellular matrix. For example, human collagen bridges GBS pili adhesin, PilA, and α2β1 integrin on BMECs resulting in bacterial attachment, immune activation and ultimately penetration of the CNS [56]. Similarly, fibronectin bridges N. meningitidis and the α5β1 integrin on BMECs, promoting bacterial internalization [109]. Also, glucosaminoglycans on brain endothelium, which are also known to interact with integrins [127], have recently been shown to bind the αC protein of GBS [128]. Therefore, the LR, PAFr and integrin signaling pathways may represent attractive targets for therapeutic intervention, as they would prevent a broad range of bacterial meningitis. Third, inflammatory activation of brain endothelial cells by cytokines, which are typically elevated in meningitis patients [129-134], can increase host receptor expression resulting in enhanced bacterial invasion. For example, stimulation of cells with TNF-α, IL-1 or TGF-β resulted in increased bacterial uptake [86,135-137]. Some bacteria take advantage of this mechanism to promote their own uptake; the surface-expressed neuraminidase, NanA on S. pneumoniae promotes penetration of the BBB by inducing chemokine release from brain endothelial cells [82,136]. Ultimately, a more detailed understanding of host–bacteria interactions at the molecular level may result in therapeutic treatment strategies to block bacterial entry at an early stage of infection.

Breakdown of BBB integrity

Besides evidence of transcellular migration, many in vitro studies have demonstrated that bacteria can affect endothelial barrier integrity, allowing for direct entry or paracellular translocation. As discussed above, BMECs are tightly interconnected by AJs and TJs, which limit paracellular passage of foreign particles including bacteria. Disruption of the BBB is a hallmark event in the pathophysiology of bacterial meningitis. Many meningeal pathogens affect endothelium barrier integrity by direct toxic effects and/or by interfering specifically with AJ/TJ formation. For example, GBS and S. pneumoniae directly affect barrier function by secreting a pore-forming toxin [84,114,138]. For GBS, higher toxin production has been associated with an increased capacity to cause meningitis [80]. In addition to pathogen-derived toxins, increased expression of inflammatory cytokines/chemokines/molecules by the host in response to infection can negatively impact BBB function and disease outcome. Increased systemic expression of TNF-α is linked to enhanced permeability of the BBB [139-141]. In addition, E. coli K1 (through OmpA), S. pneumoniae and GBS increase nitric oxide production from brain endothelial cells by inducing expression of inducible nitric oxide synthase (iNOS) [142-145]. Consequently, this attenuates BBB integrity [142,143] and promotes bacterial invasion in the case of E. coli K1 [142]. However, inhibition of iNOS function does not provide unambiguous answers; in the case of GBS meningitis, pharmacological inhibition of iNOS results in increased pathology [144], whereas genetic deletion of iNOS confers complete protection in models of E. coli K1 meningitis [146].

Besides general BBB insult, some pathogens use sophisticated strategies to target endothelial cell junctions to promote barrier permeability resulting in increased bacterial BBB traversal. The critical AJ protein, vascular endothelial cadherin, is a common target for meningeal pathogens. E. coli K1 and N. meningitidis use their surface adhesion molecules to exploit brain endothelial cell signaling to increase paracellular translocation [147-150]. OmpA-expressing E. coli K1 causes permeability changes by dissociating β-catenins from cadherins and activating protein kinase Cα [150]. For meningococcal meningitis initial observations suggested that attachment of meningococci to brain endothelial cells through type IV pili signals the formation of mislocalized AJs, opening up the paracellular route for N. meningitidis translocation into the CNS [148]. In a follow-up study, the β2-adrenoceptor/β-arrestin signaling pathway was found to be involved in the induced junctional protein rearrangements and bacterial crossing of the BBB [147]. In addition to targeting AJs, N. meningitidis induces specific cleavage of the TJ component occludin through the release of host matrix metalloproteinase 8, resulting in endothelial cell detachment and increased paracellular permeability [149]. Pharmacological intervention of OmpA–hBMEC and type IV pili–β2-adrenoceptor interaction would therefore not only block the initial interaction of bacteria with the BBB, but would also prevent exploitation of host signaling machinery by the pathogen.

In contrast to E. coli K1 and N. meningitidis, which use surface-associated molecules to affect BBB permeability, B. anthracis disrupts both AJs and TJs by secreted non-pore-forming factors including proteases and edema and lethal toxin complexes. Whereas edema toxin and secreted protease InhA affect the distribution of the critical TJ component ZO-1 [151-153], AJ formation is blocked by a synergistic effect of lethal toxin and edema toxin on endosomal transport of cadherins [154]. Disruption of endosome recycling by anthrax toxins has more widespread effects on vascular function as important cell-cell communication pathways, such as the Notch signaling pathway, also depend on proper endosome transport [154].

Finally, host inflammatory factors may also contribute to break down of the BBB. It was recently demonstrated that host neutrophils contribute to BBB permeability during the pathogenesis of GBS meningitis [56]. Although neutrophils were found to be critically important to prevent bacterial sepsis, depletion of peripheral neutrophils in a murine hematogenous model of GBS meningitis prolonged survival, and decreased bacterial brain load and BBB permeability in GBS-infected mice [56]. Correspondingly, prevention of leukocyte infiltration into the CNS using anti-CD18 antibody was previously shown to improve pneumococcal and HiB meningitis outcome [155,156].

The BBB innate defense response

Many studies have shown that the host’s inflammatory response contributes to many adverse events during bacterial meningitis. However, little is known about the role of the BBB as a functional, rather than just a physical, barrier against the initial threat of an invading pathogen. A better understanding of the host BBB responses to pathogen infection can aid in the development of preventive therapies for CNS infection.

The first comprehensive microarray ana lysis of the BBB transcriptional response to a pathogen was examined during GBS infection [80]. Interestingly, in this and subsequent studies, infection triggered a specific gene expression program for neutrophil recruitment (i.e., IL-8, CXCL1, CXCL2, ICAM-1) and activation (IL-6, IL-8), which was strongly dependent on GBS β-h/c toxin [80], the transcriptional regulator of virulence, CovR [138] and PilA expression [56]. Strong proinflammatory cytokines such as TNF-α or IL-1 were absent [80], precluding the development of an unchecked pattern of activation that is detrimental to CNS structures. Subsequent comparative microarray experiments with S. pneumoniae, HiB and Salmonella enterica Typhimurium reveal the presence of a core hBMEC transcriptional response in addition to pathogen-specific gene expression profiles [van Sorge NM, Doran KS, Unpublished Data] [89,136]. A group of core response genes function to orchestrate neutrophil recruitment (IL8, CXCL1, CXCL2), extravasation (ICAM-1), activation (IL6, IL8) and survival (GM-CSF). Similarly, other meningeal bacteria including E. coli K1 [157], S. suis [158], L. monocytogenes [159] and N. meningitidis [160,161] have been reported to induce the upregulation and secretion of neutrophil-specific factors upon hBMEC infection. Again TNF-α and IL-1 are typically absent from this response, except during infection with HiB [van Sorge NM, Doran KS, Unpublished Data] or N. meningitidis [161]. Whether this broader spectrum of inflammation in response to HiB or N. meningitidis contributes to the high prevalence of these two pathogens in bacterial meningitis or the often fulminant disease course during infection, remains to be determined. Overall, these data suggest that the BBB response serves a sentinel function by recognizing the threat of a bacterial pathogen, resulting in effective clearance of the bacteria before it can enter the CNS. However, the timing and magnitude of the neutrophil recruitment response is critical for the outcome of infection. Continued exposure and invasion of the pathogen may result in overactivation of BBB endothelium leading to increased inflammation that may compromise BBB integrity or cause neuronal damage. Indeed, experimental delay of neutrophil apoptosis results in prolonged inflammatory activity and more severe disease in a mouse pneumococcal meningitis model [55]. A similar observation was recently published for GBS meningitis. Pilus adhesion protein PilA, located at the tip of the pilus structure [162], induces hBMEC IL-8 expression through α2β1 integrin/focal adhesion kinase signaling [56]. In an in vivo model of GBS meningitis, PilA-induced host signaling resulted in a disproportionate neutrophil inflammatory response that increased BBB permeability, bacterial CNS penetration and mortality [56]. In the case of anthrax meningitis, the opposite has been observed. Infection of hBMEC with avirulent B. anthracis initiates the common neutrophil recruitment response as observed with other meningeal pathogens. However, the presence of the toxin-encoding pXO1 plasmid results in active downregulation of this innate defense pathway [163]. The resulting suppression of neutrophil chemotaxis allows for unrestricted proliferation and dissemination of the bacteria into the CNS [163].

Analysis of changes in transcriptional regulation upon BBB infection is just a starting point for future studies. It will be important to use this information to determine the signal transduction pathways and initiating receptors involved in this response, and whether their activation contributes to host resistance or neuronal damage. Based on published studies, some common downstream signaling pathways for bacterial internalization into hBMECs can be identified. Both E. coli K1 and GBS activate focal adhesion kinase signaling to trigger internalization in hBMECs [56,164,165]. Similarly, S. pneumoniae and N. meningitidis both trigger β-arrestin signaling downstream of PAFr and β2-adrenoceptor, respectively [147,148], which ultimately results in bacterial translocation across the BBB. Interestingly, the effects downstream of β-arrestin are different in both cases: S. pneunoniae β-arrestin signaling triggers vacuolar trafficking across brain endothelial cells [122], whereas N. meningitidis β-arrestin signaling depletes intercellular junctions, allowing paracellular translocation across the BBB [147].

In addition to signaling involved in bacterial translocation, unraveling signaling pathways that trigger protective or unwanted cytokine release could aid the development of pharmacological intervention strategies. For example, N. meningitidis, GBS and S. pneumoniae trigger IL-6 and IL-8 production in hBMECs by activating MAPK pathways [56,136,160]. The upstream receptors that induce this MAPK signaling have not been identified but probably involve immune pattern recognition receptors, such as integrins, Toll-like receptors and intracellular NOD-like receptors. Interestingly, in all these studies it was found that activation of cytokine responses did not require bacterial invasion of brain endothelial cells [56,136,160], suggesting that signaling is initiated at the host cell surface. Also, for other bacteria, this dissociation between bacteria-host cell interaction and cytokine production has been observed [157,166,167]. This suggests that interference with bacterial attachment to brain endothelium could prevent many of the downstream effects of meningitis pathogenesis.

Future perspective

Significant progress has been made in identifying molecular mechanisms that contribute to bacterial-BBB interaction and signaling during the progression of CNS disease. Identification of common pathways employed by bacterial pathogens to cross and penetrate BBB endothelium will assist in the identification of important bacterial and host cell targets for the development of effective therapies. However, a multi-disciplinary and systems biology approach is necessary to incorporate all this knowledge into new testable hypotheses that will provide insight into the pathogenesis and pathophysiology of bacterial meningitis and the discovery of novel therapeutic strategies.

Executive summary.

Dynamic epidemiology of bacterial meningitis

  • There are still 1.2 million cases of bacterial meningitis per year, 170,000 of which are fatal and survivors are left with permanent neurological sequelae.

  • Only a limited number of bacterial pathogens are capable of causing meningitis, the severity of which varies depending on geographic location, socioeconomic status, age, vaccination availability and overall health status of the individual.

The pathogenesis & pathophysiology of bacterial meningitis

  • In order to cause meningitis, bacterial pathogens must survive in the bloodstream and penetrate or transmigrate across the blood–brain barrier (BBB), which is primarily comprised of a single layer of specialized endothelial cells.

  • Once inside the CNS, the bacteria multiply and induce inflammation of the subarachnoid and ventrical spaces with associated pathophysiologic alterations such as increased BBB permeability and pleocytosis.

BBB composition

  • The BBB is composed of a single cell layer of brain microvascular endothelial cells that line cerebral microvessels.

  • Brain endothelial cells contain adherens and tight junctions that act to impede the passage of virtually all molecules, thereby maintaining the microenvironment of the CNS.

Transcellular penetration of the BBB

  • The attachment of blood-borne bacteria to brain endothelium and subsequent invasion may represent the initial step in penetration and/or disruption of the BBB; this interaction involves a complex interplay between host receptors and bacterial components.

Breakdown of BBB integrity

  • Many meningeal pathogens are capable of disrupting endothelium junction complexes by direct toxic effects, interfering specifically with junctional formation, and/or induction of an inflammatory response, which itself may compromise BBB integrity.

BBB innate defense response

  • Studies suggest that the BBB responds to bacterial encounter with a core gene activation program orchestrated to promote the targeted recruitment and activation of neutrophils.

  • Continued exposure and invasion of the pathogen may result in overactivation of BBB endothelium, leading to increased inflammation and BBB breakdown.

Acknowledgements

The authors appreciate the contribution of the anonymous peer reviewers and apologize to researchers whose work has not been discussed in detail or reviewed here due to space constraints.

Work on the blood–brain barrier and bacterial meningitis in KS Doran’s laboratory is supported by funding from the NIH/NINDS (grant no. RO1NS051247).

Footnotes

Financial & competing interests disclosure

The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

Papers of special note have been highlighted as:

■ of interest

■■ of considerable interest

  • 1.Baraff LJ, Lee SI, Schriger DL. Outcomes of bacterial meningitis in children: a meta-analysis. Pediatr. Infect. Dis. J. 1993;12(5):389–394. doi: 10.1097/00006454-199305000-00008. [DOI] [PubMed] [Google Scholar]
  • 2.Grimwood K, Anderson P, Anderson V, Tan L, Nolan T. Twelve year outcomes following bacterial meningitis: further evidence for persisting effects. Arch. Dis. Child. 2000;83(2):111–116. doi: 10.1136/adc.83.2.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Eskola J, Peltola H, Takala AK, et al. Efficacy of Haemophilus influenzae type b polysaccharide-diphtheria toxoid conjugate vaccine in infancy. N. Engl. J. Med. 1987;317(12):717–722. doi: 10.1056/NEJM198709173171201. [DOI] [PubMed] [Google Scholar]
  • 4.Peltola H. Worldwide Haemophilus influenzae type b disease at the beginning of the 21st century: global ana lysis of the disease burden 25 years after the use of the polysaccharide vaccine and a decade after the advent of conjugates. Clin. Microbiol. Rev. 2000;13(2):302–317. doi: 10.1128/cmr.13.2.302-317.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Watt JP, Wolfson LJ, O’Brien KL, et al. Burden of disease caused by Haemophilus influenzae type b in children younger than 5 years: global estimates. Lancet. 2009;374(9693):903–911. doi: 10.1016/S0140-6736(09)61203-4. [DOI] [PubMed] [Google Scholar]
  • 6.O’Brien KL, Wolfson LJ, Watt JP, et al. Burden of disease caused by Streptococcus pneumoniae in children younger than 5 years: global estimates. Lancet. 2009;374(9693):893–902. doi: 10.1016/S0140-6736(09)61204-6. [DOI] [PubMed] [Google Scholar]
  • 7.Thigpen MC, Whitney CG, Messonnier NE, et al. Bacterial meningitis in the United States, 1998-2007. N. Engl. J. Med. 2011;364(21):2016–2025. doi: 10.1056/NEJMoa1005384. [DOI] [PubMed] [Google Scholar]
  • 8.Kaplan SL, Mason EO, Jr, Wald ER, et al. Decrease of invasive pneumococcal infections in children among 8 children’s hospitals in the United States after the introduction of the 7-valent pneumococcal conjugate vaccine. Pediatrics. 2004;113(3 Pt 1):443–449. doi: 10.1542/peds.113.3.443. [DOI] [PubMed] [Google Scholar]
  • 9.Nakhla I, Frenck RW, Jr, Teleb NA, et al. The changing epidemiology of meningococcal meningitis after introduction of bivalent A/C polysaccharide vaccine into school-based vaccination programs in Egypt. Vaccine. 2005;23(25):3288–3293. doi: 10.1016/j.vaccine.2005.01.084. [DOI] [PubMed] [Google Scholar]
  • 10.Gray SJ, Trotter CL, Ramsay ME, et al. Epidemiology of meningococcal disease in England and Wales 1993/94 to 2003/04: contribution and experiences of the Meningococcal Reference Unit. J. Med. Microbiol. 2006;55(Pt 7):887–896. doi: 10.1099/jmm.0.46288-0. [DOI] [PubMed] [Google Scholar]
  • 11.Molyneux EM, Mankhambo LA, Phiri A, et al. The outcome of non-typhoidal Salmonella meningitis in Malawian children, 1997-2006. Ann. Trop. Paediatr. 2009;29(1):13–22. doi: 10.1179/146532809X401980. [DOI] [PubMed] [Google Scholar]
  • 12.Molyneux EM, Walsh AL, Malenga G, Rogerson S, Molyneux ME. Salmonella meningitis in children in Blantyre, Malawi, 1996-1999. Ann. Trop. Paediatr. 2000;20(1):41–44. doi: 10.1080/02724930092057. [DOI] [PubMed] [Google Scholar]
  • 13.Vaagland H, Blomberg B, Kruger C, Naman N, Jureen R, Langeland N. Nosocomial outbreak of neonatal Salmonella enterica serotype Enteritidis meningitis in a rural hospital in northern Tanzania. BMC Infect. Dis. 2004;4:35. doi: 10.1186/1471-2334-4-35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Iriso R, Ocakacon R, Acayo JA, Mawanda MA, Kisayke A. Bacterial meningitis following introduction of Hib conjugate vaccine in northern Uganda. Ann. Trop. Paediatr. 2008;28(3):211–216. doi: 10.1179/146532808X335660. [DOI] [PubMed] [Google Scholar]
  • 15.Gordon MA, Graham SM, Walsh AL, et al. Epidemics of invasive Salmonella enterica serovar enteritidis and S. enterica serovar Typhimurium infection associated with multidrug resistance among adults and children in Malawi. Clin. Infect. Dis. 2008;46(7):963–969. doi: 10.1086/529146. [DOI] [PubMed] [Google Scholar]
  • 16.Owusu-Ofori A, Scheld WM. Treatment of Salmonella meningitis: two case reports and a review of the literature. Int. J. Infect. Dis. 2003;7(1):53–60. doi: 10.1016/s1201-9712(03)90043-9. [DOI] [PubMed] [Google Scholar]
  • 17.Brouwer MC, Tunkel AR, Van De Beek D. Epidemiology, diagnosis, and antimicrobial treatment of acute bacterial meningitis. Clin. Microbiol. Rev. 2010;23(3):467–492. doi: 10.1128/CMR.00070-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Dawson KG, Emerson JC, Burns JL. Fifteen years of experience with bacterial meningitis. Pediatr. Infect. Dis. J. 1999;18(9):816–822. doi: 10.1097/00006454-199909000-00014. [DOI] [PubMed] [Google Scholar]
  • 19.Nigrovic LE, Kuppermann N, Malley R. Children with bacterial meningitis presenting to the emergency department during the pneumococcal conjugate vaccine era. Acad. Emerg. Med. 2008;15(6):522–528. doi: 10.1111/j.1553-2712.2008.00117.x. [DOI] [PubMed] [Google Scholar]
  • 20.Group TWYIS Bacterial etiology of serious infections in young infants in developing countries: results of a multicenter study. Pediatr. Infect. Dis. J. 1999;18(10 Suppl.):S17–S22. doi: 10.1097/00006454-199910001-00004. [DOI] [PubMed] [Google Scholar]
  • 21.Zaidi AK, Thaver D, Ali SA, Khan TA. Pathogens associated with sepsis in newborns and young infants in developing countries. Pediatr. Infect. Dis. J. 2009;28(1 Suppl):S10–S18. doi: 10.1097/INF.0b013e3181958769. [DOI] [PubMed] [Google Scholar]
  • 22.Phares CR, Lynfield R, Farley MM, et al. Epidemiology of invasive group B streptococcal disease in the United States, 1999-2005. JAMA. 2008;299(17):2056–2065. doi: 10.1001/jama.299.17.2056. [DOI] [PubMed] [Google Scholar]
  • 23.Van Dyke MK, Phares CR, Lynfield R, et al. Evaluation of universal antenatal screening for group B. Streptococcus. N. Engl. J. Med. 2009;360(25):2626–2636. doi: 10.1056/NEJMoa0806820. [DOI] [PubMed] [Google Scholar]
  • 24.Stoll BJ, Hansen N, Fanaroff AA, et al. Changes in pathogens causing early-onset sepsis in very-low-birth-weight infants. N. Engl. J. Med. 2002;347(4):240–247. doi: 10.1056/NEJMoa012657. [DOI] [PubMed] [Google Scholar]
  • 25.Stoll BJ, Hansen NI, Sanchez PJ, et al. Early onset neonatal sepsis: the burden of group B Streptococcal and E. coli disease continues. Pediatrics. 2011;127(5):817–826. doi: 10.1542/peds.2010-2217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Bekondi C, Bernede C, Passone N, et al. Primary and opportunistic pathogens associated with meningitis in adults in Bangui, Central African Republic, in relation to human immunodeficiency virus serostatus. Int. J. Infect. Dis. 2006;10(5):387–395. doi: 10.1016/j.ijid.2005.07.004. [DOI] [PubMed] [Google Scholar]
  • 27.Swe KS, Nagel G, Van Der Westhuizen M, Hoosen AA. Salmonella Typhimurium meningitis in an adult patient with AIDS. J. Clin. Pathol. 2008;61(1):138–139. doi: 10.1136/jcp.2006.043216. [DOI] [PubMed] [Google Scholar]
  • 28.Fernandez Guerrero ML, Ramos JM, Nunez A, De Gorgolas M. Focal infections due to non-typhi Salmonella in patients with AIDS: report of 10 cases and review. Clin. Infect. Dis. 1997;25(3):690–697. doi: 10.1086/513747. [DOI] [PubMed] [Google Scholar]
  • 29.Lun ZR, Wang QP, Chen XG, Li AX, Zhu XQ. Streptococcus suis: an emerging zoonotic pathogen. Lancet Infect. Dis. 2007;7(3):201–209. doi: 10.1016/S1473-3099(07)70001-4. [DOI] [PubMed] [Google Scholar]
  • 30.Gottschalk M, Xu J, Calzas C, Segura M. Streptococcus suis: a new emerging or an old neglected zoonotic pathogen? Future Microbiol. 2010;5(3):371–391. doi: 10.2217/fmb.10.2. [DOI] [PubMed] [Google Scholar]
  • 31.Aguilar J, Urday-Cornejo V, Donabedian S, Perri M, Tibbetts R, Zervos M. Staphylococcus aureus meningitis: case series and literature review. Medicine. 2010;89(2):117–125. doi: 10.1097/MD.0b013e3181d5453d. [DOI] [PubMed] [Google Scholar]
  • 32.Pintado V, Meseguer MA, Fortun J, et al. Clinical study of 44 cases of Staphylococcus aureus meningitis. Eur. J. Clin. Microbiol. Infect. Dis. 2002;21(12):864–868. doi: 10.1007/s10096-002-0814-1. [DOI] [PubMed] [Google Scholar]
  • 33.Lerche A, Rasmussen N, Wandall JH, Bohr VA. Staphylococcus aureus meningitis: a review of 28 consecutive community-acquired cases. Scand. J. Infect. Dis. 1995;27(6):569–573. doi: 10.3109/00365549509047069. [DOI] [PubMed] [Google Scholar]
  • 34.Brouwer MC, Keizerweerd GD, De Gans J, Spanjaard L, Van De Beek D. Community acquired Staphylococcus aureus meningitis in adults. Scand. J. Infect. Dis. 2009;41(5):375–377. doi: 10.1080/00365540902744766. [DOI] [PubMed] [Google Scholar]
  • 35.Jernigan JA, Stephens DS, Ashford DA, et al. Bioterrorism-related inhalational anthrax: the first 10 cases reported in the United States. Emerg. Infect. Dis. 2001;7(6):933–944. doi: 10.3201/eid0706.010604. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bales ME, Dannenberg AL, Brachman PS, Kaufmann AF, Klatsky PC, Ashford DA. Epidemiologic response to anthrax outbreaks: field investigations, 1950-2001. Emerg. Infect. Dis. 2002;8(10):1163–1174. doi: 10.3201/eid0810.020223. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ramsay CN, Stirling A, Smith J, et al. An outbreak of infection with Bacillus anthracis in injecting drug users in Scotland. EuroSurveill. 2010;15(2) doi: 10.2807/ese.15.02.19465-en. [DOI] [PubMed] [Google Scholar]
  • 38.Lanska DJ. Anthrax meningoencephalitis. Neurology. 2002;59(3):327–334. doi: 10.1212/wnl.59.3.327. [DOI] [PubMed] [Google Scholar]
  • 39.Stephens DS. Uncloaking the meningococcus: dynamics of carriage and disease. Lancet. 1999;353(9157):941–942. doi: 10.1016/S0140-6736(98)00279-7. [DOI] [PubMed] [Google Scholar]
  • 40.Kim KS, Itabashi H, Gemski P, Sadoff J, Warren RL, Cross AS. The K1 capsule is the critical determinant in the development of Escherichia coli meningitis in the rat. J. Clin. Invest. 1992;90(3):897–905. doi: 10.1172/JCI115965. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Moxon ER, Ostrow PT. Haemophilus influenzae meningitis in infant rats: role of bacteremia in pathogenesis of age-dependent inflammatory responses in cerebrospinal fluid. J. Infect. Dis. 1977;135(2):303–307. doi: 10.1093/infdis/135.2.303. [DOI] [PubMed] [Google Scholar]
  • 42.La Scolea LJ, Jr, Dryja D. Quantitation of bacteria in cerebrospinal fluid and blood of children with meningitis and its diagnostic significance. J. Clin. Microbiol. 1984;19(2):187–190. doi: 10.1128/jcm.19.2.187-190.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Petersdorf RG, Swarner DR, Garcia M. Studies on the pathogenesis of meningitis. II. Development of meningitis during pneumococcal bacteremia. J. Clin. Invest. 1962;41:320–327. doi: 10.1172/JCI104485. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ferrieri P, Burke B, Nelson J. Production of bacteremia and meningitis in infant rats with group B streptococcal serotypes. Infect. Immun. 1980;27(3):1023–1032. doi: 10.1128/iai.27.3.1023-1032.1980. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Bell LM, Alpert G, Campos JM, Plotkin SA. Routine quantitative blood cultures in children with Haemophilus influenzae or Streptococcus pneumoniae bacteremia. Pediatrics. 1985;76(6):901–904. [PubMed] [Google Scholar]
  • 46.Nakamura S, Shchepetov M, Dalia AB, et al. Molecular basis of increased serum resistance among pulmonary isolates of non-typeable Haemophilus influenzae. PLoS Pathog. 2011;7(1):E1001247. doi: 10.1371/journal.ppat.1001247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Mereghetti L, Sitkiewicz I, Green NM, Musser JM. Extensive adaptive changes occur in the transcriptome of Streptococcus agalactiae (group B Streptococcus) in response to incubation with human blood. PLoS One. 2008;3(9):E3143. doi: 10.1371/journal.pone.0003143. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Echenique-Rivera H, Muzzi A, Del Tordello E, et al. Transcriptome ana lysis of Neisseria meningitidis in human whole blood and mutagenesis studies identify virulence factors involved in blood survival. PLoS Pathog. 2011;7(5):E1002027. doi: 10.1371/journal.ppat.1002027. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Santi I, Scarselli M, Mariani M, et al. BibA: a novel immunogenic bacterial adhesin contributing to group B Streptococcus survival in human blood. Mol. Microbiol. 2007;63(3):754–767. doi: 10.1111/j.1365-2958.2006.05555.x. [DOI] [PubMed] [Google Scholar]
  • 50.Sukumaran SK, Shimada H, Prasadarao NV. Entry and intracellular replication of Escherichia coli K1 in macrophages require expression of outer membrane protein A. Infect. Immun. 2003;71(10):5951–5961. doi: 10.1128/IAI.71.10.5951-5961.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Mittal R, Prasadarao NV. gp96 expression in neutrophils is critical for the onset of Escherichia coli K1 (RS218) meningitis. Nat. Commun. 2011;2:552. doi: 10.1038/ncomms1554. ■ Demonstrates mechanisms that promote bacterial bloodstream survival and subsequent blood–brain barrier (BBB) penetration.
  • 52.Mittal R, Sukumaran SK, Selvaraj SK, et al. Fcgamma receptor I alpha chain (CD64) expression in macrophages is critical for the onset of meningitis by Escherichia coli K1. PLoS Pathog. 2010;6(11):E1001203. doi: 10.1371/journal.ppat.1001203. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Mittal R, Krishnan S, Gonzalez-Gomez I, Prasadarao NV. Deciphering the roles of outer membrane protein A extracellular loops in the pathogenesis of Escherichia coli K1 meningitis. J. Biol. Chem. 2011;286(3):2183–2193. doi: 10.1074/jbc.M110.178236. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Pfister HW, Borasio GD, Dirnagl U, Bauer M, Einhaupl KM. Cerebrovascular complications of bacterial meningitis in adults. Neurology. 1992;42(8):1497–1504. doi: 10.1212/wnl.42.8.1497. [DOI] [PubMed] [Google Scholar]
  • 55.Koedel U, Frankenberg T, Kirschnek S, et al. Apoptosis is essential for neutrophil functional shutdown and determines tissue damage in experimental pneumococcal meningitis. PLoS Pathog. 2009;5(5):E1000461. doi: 10.1371/journal.ppat.1000461. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Banerjee A, Kim BJ, Carmona EM, et al. Bacterial Pili exploit integrin machinery to promote immune activation and efficient blood-brain barrier penetration. Nat. Commun. 2011;2:462. doi: 10.1038/ncomms1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Nguyen TH, Tran TH, Thwaites G, et al. Dexamethasone in Vietnamese adolescents and adults with bacterial meningitis. N. Engl. J. Med. 2007;357(24):2431–2440. doi: 10.1056/NEJMoa070852. [DOI] [PubMed] [Google Scholar]
  • 58.Peltola H, Roine I, Fernandez J, et al. Adjuvant glycerol and/or dexamethasone to improve the outcomes of childhood bacterial meningitis: a prospective, randomized, double-blind, placebo-controlled trial. Clin. Infect. Dis. 2007;45(10):1277–1286. doi: 10.1086/522534. [DOI] [PubMed] [Google Scholar]
  • 59.Van De Beek D, Farrar JJ, De Gans J, et al. Adjunctive dexamethasone in bacterial meningitis: a meta-ana lysis of individual patient data. Lancet Neurol. 2010;9(3):254–263. doi: 10.1016/S1474-4422(10)70023-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Scarborough M, Gordon SB, Whitty CJ, et al. Corticosteroids for bacterial meningitis in adults in sub-Saharan Africa. N. Engl. J. Med. 2007;357(24):2441–2450. doi: 10.1056/NEJMoa065711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Brouwer MC, Mcintyre P, De Gans J, Prasad K, Van De Beek D. Corticosteroids for acute bacterial meningitis. Cochrane Database Syst. Rev. 2010;(9):CD004405. doi: 10.1002/14651858.CD004405.pub3. [DOI] [PubMed] [Google Scholar]
  • 62.Abbott NJ, Ronnback L, Hansson E. Astrocyte–endothelial interactions at the blood–brain barrier. Nat. Rev. Neurosci. 2006;7(1):41–53. doi: 10.1038/nrn1824. [DOI] [PubMed] [Google Scholar]
  • 63.Rubin LL, Staddon JM. The cell biology of the blood–brain barrier. Annu. Rev. Neurosci. 1999;22:11–28. doi: 10.1146/annurev.neuro.22.1.11. [DOI] [PubMed] [Google Scholar]
  • 64.Pulzova L, Bhide MR, Andrej K. Pathogen translocation across the blood–brain barrier. FEMS Immunol. Med. Microbiol. 2009;57(3):203–213. doi: 10.1111/j.1574-695X.2009.00594.x. [DOI] [PubMed] [Google Scholar]
  • 65.Dejana E, Corada M, Lampugnani MG. Endothelial cell-to-cell junctions. FASEB. 1995;9(10):910–918. [PubMed] [Google Scholar]
  • 66.Hawkins RA, O’Kane RL, Simpson IA, Vina JR. Structure of the blood–brain barrier and its role in the transport of amino acids. J. Nutr. 2006;136(1 Suppl.):218S–226S. doi: 10.1093/jn/136.1.218S. [DOI] [PubMed] [Google Scholar]
  • 67.Schulze C, Firth JA. Immunohistochemical localization of adherens junction components in blood–brain barrier microvessels of the rat. Cell Sci. 1993;104(Pt 3):773–782. doi: 10.1242/jcs.104.3.773. [DOI] [PubMed] [Google Scholar]
  • 68.Wolburg H, Lippoldt A. Tight junctions of the blood–brain barrier: development, composition and regulation. Vascul. Pharmacol. 2002;38(6):323–337. doi: 10.1016/s1537-1891(02)00200-8. [DOI] [PubMed] [Google Scholar]
  • 69.Gumbiner BM. Cell adhesion: the molecular basis of tissue architecture and morphogenesis. Cell. 1996;84(3):345–357. doi: 10.1016/s0092-8674(00)81279-9. [DOI] [PubMed] [Google Scholar]
  • 70.Furuse M, Itoh M, Hirase T, Nagafuchi A, Yonemura S, Tsukita S. Direct association of occludin with ZO-1 and its possible involvement in the localization of occludin at tight junctions. J. Cell. Biol. 1994;127(6 Pt 1):1617–1626. doi: 10.1083/jcb.127.6.1617. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Martin-Padura I, Lostaglio S, Schneemann M, et al. Junctional adhesion molecule, a novel member of the immunoglobulin superfamily that distributes at intercellular junctions and modulates monocyte transmigration. J. Cell. Biol. 1998;142(1):117–127. doi: 10.1083/jcb.142.1.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Braga VM. Cell-cell adhesion and signalling. Curr. Opin. Cell Biol. 2002;14(5):546–556. doi: 10.1016/s0955-0674(02)00373-3. [DOI] [PubMed] [Google Scholar]
  • 73.Matter K, Balda MS. Signalling to and from tight junctions. Nat. Rev. Mol. Cell Biol. 2003;4(3):225–236. doi: 10.1038/nrm1055. [DOI] [PubMed] [Google Scholar]
  • 74.Wheelock MJ, Johnson KR. Cadherin-mediated cellular signaling. Curr. Opin. Cell Biol. 2003;15(5):509–514. doi: 10.1016/s0955-0674(03)00101-7. [DOI] [PubMed] [Google Scholar]
  • 75.Taddei A, Giampietro C, Conti A, et al. Endothelial adherens junctions control tight junctions by VE-cadherin-mediated upregulation of claudin-5. Nat. Cell Biol. 2008;10(8):923–934. doi: 10.1038/ncb1752. [DOI] [PubMed] [Google Scholar]
  • 76.Stins MF, Prasadarao NV, Ibric L, Wass CA, Luckett P, Kim KS. Binding characteristics of S fimbriated Escherichia coli to isolated brain microvascular endothelial cells. Am. J. Pathol. 1994;145(5):1228–1236. ■ Pioneering study describing the development of the human brain microvascular cell line.
  • 77.Weksler BB, Subileau EA, Perriere N, et al. Blood–brain barrier-specific properties of a human adult brain endothelial cell line. FASEB. 2005;19(13):1872–1874. doi: 10.1096/fj.04-3458fje. [DOI] [PubMed] [Google Scholar]
  • 78.Greiffenberg L, Goebel W, Kim KS, et al. Interaction of Listeria monocytogenes with human brain microvascular endothelial cells: InlB-dependent invasion, long-term intracellular growth, and spread from macrophages to endothelial cells. Infect. Immun. 1998;66(11):5260–5267. doi: 10.1128/iai.66.11.5260-5267.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Wickham ME, Brown NF, Provias J, Finlay BB, Coombes BK. Oral infection of mice with Salmonella enterica serovar Typhimurium causes meningitis and infection of the brain. BMC Infect. Dis. 2007;7:65. doi: 10.1186/1471-2334-7-65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Doran KS, Liu GY, Nizet V. Group B streptococcal beta-hemolysin/cytolysin activates neutrophil signaling pathways in brain endothelium and contributes to development of meningitis. J. Clin. Invest. 2003;112(5):736–744. doi: 10.1172/JCI17335. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Smith AL, Smith DH, Averill DR, Jr, Marino J, Moxon ER. Production of Haemophilus influenzae b meningitis in infant rats by intraperitoneal inoculation. Infect. Immun. 1973;8(2):278–290. doi: 10.1128/iai.8.2.278-290.1973. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Uchiyama S, Carlin AF, Khosravi A, et al. The surface-anchored NanA protein promotes pneumococcal brain endothelial cell invasion. J. Exp. Med. 2009;206(9):1845–1852. doi: 10.1084/jem.20090386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Sheen TR, Ebrahimi CM, Hiemstra IH, Barlow SB, Peschel A, Doran KS. Penetration of the blood–brain barrier by Staphylococcus aureus: contribution of membrane-anchored lipoteichoic acid. J. Mol. Med. 2010;88(6):633–639. doi: 10.1007/s00109-010-0630-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Nizet V, Kim KS, Stins M, et al. Invasion of brain microvascular endothelial cells by group B streptococci. Infect. Immun. 1997;65(12):5074–5081. doi: 10.1128/iai.65.12.5074-5081.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Stins MF, Badger J, Sik Kim K. Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Microb. Pathog. 2001;30(1):19–28. doi: 10.1006/mpat.2000.0406. [DOI] [PubMed] [Google Scholar]
  • 86.Ring A, Weiser JN, Tuomanen EI. Pneumococcal trafficking across the blood–brain barrier. Molecular ana lysis of a novel bidirectional pathway. J. Clin. Invest. 1998;102(2):347–360. doi: 10.1172/JCI2406. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Jain SK, Paul-Satyaseela M, Lamichhane G, Kim KS, Bishai WR. Mycobacterium tuberculosis invasion and traversal across an in vitro human blood–brain barrier as a pathogenic mechanism for central nervous system tuberculosis. J. Infect. Dis. 2006;193(9):1287–1295. doi: 10.1086/502631. [DOI] [PubMed] [Google Scholar]
  • 88.Nikulin J, Panzner U, Frosch M, Schubert-Unkmeir A. Intracellular survival and replication of Neisseria meningitidis in human brain microvascular endothelial cells. Int. J. Med. Microbiol. 2006;296(8):553–558. doi: 10.1016/j.ijmm.2006.06.006. [DOI] [PubMed] [Google Scholar]
  • 89.Van Sorge NM, Zialcita PA, Browne SH, Quach D, Guiney DG, Doran KS. Penetration and activation of brain endothelium by Salmonella enterica serovar Typhimurium. J. Infect. Dis. 2011;203(3):401–405. doi: 10.1093/infdis/jiq048. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Prasadarao NV, Wass CA, Stins MF, Shimada H, Kim KS. Outer membrane protein A-promoted actin condensation of brain microvascular endothelial cells is required for Escherichia coli invasion. Infect. Immun. 1999;67(11):5775–5783. doi: 10.1128/iai.67.11.5775-5783.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 91.Das A, Asatryan L, Reddy MA, et al. Differential role of cytosolic phospholipase A2 in the invasion of brain microvascular endothelial cells by Escherichia coli and Listeria monocytogenes. J. Infect. Dis. 2001;184(6):732–737. doi: 10.1086/322986. [DOI] [PubMed] [Google Scholar]
  • 92.Kim KS. Mechanisms of microbial traversal of the blood–brain barrier. Nat. Rev. Microbiol. 2008;6(8):625–634. doi: 10.1038/nrmicro1952. ■■ Excellent review of bacterial and host factors involved in BBB penetration and pathogenesis of meningitis.
  • 93.Kim KS. Acute bacterial meningitis in infants and children. Lancet Infect. Dis. 2010;10(1):32–42. doi: 10.1016/S1473-3099(09)70306-8. ■■ Excellent review on the pathogenesis of bacterial meningitis.
  • 94.Badger JL, Wass CA, Weissman SJ, Kim KS. Application of signature-tagged mutagenesis for identification of Escherichia coli K1 genes that contribute to invasion of human brain microvascular endothelial cells. Infect. Immun. 2000;68(9):5056–5061. doi: 10.1128/iai.68.9.5056-5061.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Doran KS, Engelson EJ, Khosravi A, et al. Blood–brain barrier invasion by group B Streptococcus depends upon proper cell-surface anchoring of lipoteichoic acid. J. Clin. Invest. 2005;115(9):2499–2507. doi: 10.1172/JCI23829. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Dietrich G, Kurz S, Hubner C, et al. Transcriptome ana lysis of Neisseria meningitidis during infection. J. Bacteriol. 2003;185(1):155–164. doi: 10.1128/JB.185.1.155-164.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97.Teng CH, Cai M, Shin S, et al. Escherichia coli K1 RS218 interacts with human brain microvascular endothelial cells via type 1 fimbria bacteria in the fimbriated state. Infect. Immun. 2005;73(5):2923–2931. doi: 10.1128/IAI.73.5.2923-2931.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Badger JL, Wass CA, Kim KS. Identification of Escherichia coli K1 genes contributing to human brain microvascular endothelial cell invasion by differential fluorescence induction. Mol. Microbiol. 2000;36(1):174–182. doi: 10.1046/j.1365-2958.2000.01840.x. [DOI] [PubMed] [Google Scholar]
  • 99.Tazi A, Disson O, Bellais S, et al. The surface protein HvgA mediates group B Streptococcus hypervirulence and meningeal tropism in neonates. J. Exp. Med. 2010;207(11):2313–2322. doi: 10.1084/jem.20092594. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Mairey E, Genovesio A, Donnadieu E, et al. Cerebral microcirculation shear stress levels determine Neisseria meningitidis attachment sites along the blood–brain barrier. J. Exp. Med. 2006;203(8):1939–1950. doi: 10.1084/jem.20060482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Maisey HC, Hensler M, Nizet V, Doran KS. Group B streptococcal pilus proteins contribute to adherence to and invasion of brain microvascular endothelial cells. J. Bacteriol. 2007;189(4):1464–1467. doi: 10.1128/JB.01153-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 102.Van Sorge NM, Quach D, Gurney MA, Sullam PM, Nizet V, Doran KS. The group B streptococcal serine-rich repeat 1 glycoprotein mediates penetration of the blood–brain barrier. J. Infect. Dis. 2009;199(10):1479–1487. doi: 10.1086/598217. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 103.Kirchner M, Meyer TF. The PilC adhesin of the Neisseria type IV pilus-binding specificities and new insights into the nature of the host cell receptor. Mol. Microbiol. 2005;56(4):945–957. doi: 10.1111/j.1365-2958.2005.04600.x. [DOI] [PubMed] [Google Scholar]
  • 104.Telford JL, Barocchi MA, Margarit I, Rappuoli R, Grandi G. Pili in Gram-positive pathogens. Nat. Rev. Microbiol. 2006;4(7):509–519. doi: 10.1038/nrmicro1443. [DOI] [PubMed] [Google Scholar]
  • 105.Barocchi MA, Ries J, Zogaj X, et al. A pneumococcal pilus influences virulence and host inflammatory responses. Proc. Natl Acad. Sci. USA. 2006;103(8):2857–2862. doi: 10.1073/pnas.0511017103. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Bagnoli F, Moschioni M, Donati C, et al. A second pilus type in Streptococcus pneumoniae is prevalent in emerging serotypes and mediates adhesion to host cells. J. Bacteriol. 2008;190(15):5480–5492. doi: 10.1128/JB.00384-08. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.St Geme JW, 3rd, Cutter D. Influence of pili, fibrils, and capsule on in vitro adherence by Haemophilus influenzae type b. Mol. Microbiol. 1996;21(1):21–31. doi: 10.1046/j.1365-2958.1996.6241331.x. [DOI] [PubMed] [Google Scholar]
  • 108.St. Geme JW, 3rd, Cutter D. Evidence that surface fibrils expressed by Haemophilus influenzae type b promote attachment to human epithelial cells. Mol. Microbiol. 1995;15(1):77–85. doi: 10.1111/j.1365-2958.1995.tb02222.x. [DOI] [PubMed] [Google Scholar]
  • 109.Unkmeir A, Latsch K, Dietrich G, et al. Fibronectin mediates Opc-dependent internalization of Neisseria meningitidis in human brain microvascular endothelial cells. Mol. Microbiol. 2002;46(4):933–946. doi: 10.1046/j.1365-2958.2002.03222.x. [DOI] [PubMed] [Google Scholar]
  • 110.Kim KJ, Elliott SJ, Di Cello F, Stins MF, Kim KS. The K1 capsule modulates trafficking of E. coli-containing vacuoles and enhances intracellular bacterial survival in human brain microvascular endothelial cells. Cell. Microbiol. 2003;5(4):245–252. doi: 10.1046/j.1462-5822.2003.t01-1-00271.x. [DOI] [PubMed] [Google Scholar]
  • 111.St Geme JW, 3rd, Falkow S. Loss of capsule expression by Haemophilus influenzae type b results in enhanced adherence to and invasion of human cells. Infect. Immun. 1991;59(4):1325–1333. doi: 10.1128/iai.59.4.1325-1333.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Hammerschmidt S, Muller A, Sillmann H, et al. Capsule phase variation in Neisseria meningitidis serogroup B by slipped-strand mispairing in the polysialyltransferase gene (siaD): correlation with bacterial invasion and the outbreak of meningococcal disease. Mol. Microbiol. 1996;20(6):1211–1220. doi: 10.1111/j.1365-2958.1996.tb02641.x. [DOI] [PubMed] [Google Scholar]
  • 113.Khan NA, Wang Y, Kim KJ, Chung JW, Wass CA, Kim KS. Cytotoxic necrotizing factor-1 contributes to Escherichia coli K1 invasion of the central nervous system. J. Biol. Chem. 2002;277(18):15607–15612. doi: 10.1074/jbc.M112224200. [DOI] [PubMed] [Google Scholar]
  • 114.Zysk G, Schneider-Wald BK, Hwang JH, et al. Pneumolysin is the main inducer of cytotoxicity to brain microvascular endothelial cells caused by Streptococcus pneumoniae. Infect. Immun. 2001;69(2):845–852. doi: 10.1128/IAI.69.2.845-852.2001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Patrick D, Betts J, Frey EA, Prameya R, Dorovini-Zis K, Finlay BB. Haemophilus influenzae lipopolysaccharide disrupts confluent monolayers of bovine brain endothelial cells via a serum-dependent cytotoxic pathway. J. Infect. Dis. 1992;165(5):865–872. doi: 10.1093/infdis/165.5.865. [DOI] [PubMed] [Google Scholar]
  • 116.Wellmer A, Zysk G, Gerber J, et al. Decreased virulence of a pneumolysin-deficient strain of Streptococcus pneumoniae in murine meningitis. Infect. Immun. 2002;70(11):6504–6508. doi: 10.1128/IAI.70.11.6504-6508.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Orihuela CJ, Mahdavi J, Thornton J, et al. Laminin receptor initiates bacterial contact with the blood brain barrier in experimental meningitis models. J. Clin. Invest. 2009;119(6):1638–1646. doi: 10.1172/JCI36759. ■ Demonstrates a host receptor that acts as a common portal of bacterial entry.
  • 118.Swords WE, Ketterer MR, Shao J, Campbell CA, Weiser JN, Apicella MA. Binding of the non-typeable Haemophilus influenzae lipooligosaccharide to the PAF receptor initiates host cell signalling. Cell. Microbiol. 2001;3(8):525–536. doi: 10.1046/j.1462-5822.2001.00132.x. [DOI] [PubMed] [Google Scholar]
  • 119.Virji M, Saunders JR, Sims G, Makepeace K, Maskell D, Ferguson DJ. Pilus-facilitated adherence of Neisseria meningitidis to human epithelial and endothelial cells: modulation of adherence phenotype occurs concurrently with changes in primary amino acid sequence and the glycosylation status of pilin. Mol. Microbiol. 1993;10(5):1013–1028. doi: 10.1111/j.1365-2958.1993.tb00972.x. [DOI] [PubMed] [Google Scholar]
  • 120.Kim KJ, Chung JW, Kim KS. 67-kDa laminin receptor promotes internalization of cytotoxic necrotizing factor 1-expressing Escherichia coli K1 into human brain microvascular endothelial cells. J. Biol. Chem. 2005;280(2):1360–1368. doi: 10.1074/jbc.M410176200. [DOI] [PubMed] [Google Scholar]
  • 121.Chung JW, Hong SJ, Kim KJ, et al. 37-kDa laminin receptor precursor modulates cytotoxic necrotizing factor 1-mediated RhoA activation and bacterial uptake. J. Biol. Chem. 2003;278(19):16857–16862. doi: 10.1074/jbc.M301028200. [DOI] [PubMed] [Google Scholar]
  • 122.Radin JN, Orihuela CJ, Murti G, Guglielmo C, Murray PJ, Tuomanen EI. Beta-Arrestin 1 participates in platelet-activating factor receptor-mediated endocytosis of Streptococcus pneumoniae. Infect. Immun. 2005;73(12):7827–7835. doi: 10.1128/IAI.73.12.7827-7835.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123.Rijneveld AW, Weijer S, Florquin S, et al. Improved host defense against pneumococcal pneumonia in platelet-activating factor receptor-deficient mice. J. Infect. Dis. 2004;189(4):711–716. doi: 10.1086/381392. [DOI] [PubMed] [Google Scholar]
  • 124.Kolberg J, Hoiby EA, Jantzen E. Detection of the phosphorylcholine epitope in Streptococci, Haemophilus and pathogenic Neisseriae by immunoblotting. Microb. Pathog. 1997;22(6):321–329. doi: 10.1006/mpat.1996.0114. [DOI] [PubMed] [Google Scholar]
  • 125.Weiser JN, Pan N, McGowan KL, Musher D, Martin A, Richards J. Phosphorylcholine on the lipopolysaccharide of Haemophilus influenzae contributes to persistence in the respiratory tract and sensitivity to serum killing mediated by C-reactive protein. J. Exp. Med. 1998;187(4):631–640. doi: 10.1084/jem.187.4.631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Swords WE, Buscher BA, Ver Steeg Ii K, et al. Non-typeable Haemophilus influenzae adhere to and invade human bronchial epithelial cells via an interaction of lipooligosaccharide with the PAF receptor. Mol. Microbiol. 2000;37(1):13–27. doi: 10.1046/j.1365-2958.2000.01952.x. [DOI] [PubMed] [Google Scholar]
  • 127.Kim SH, Turnbull J, Guimond S. Extracellular matrix and cell signalling: the dynamic cooperation of integrin, proteoglycan and growth factor receptor. J. Endocrinol. 2011;209(2):139–151. doi: 10.1530/JOE-10-0377. [DOI] [PubMed] [Google Scholar]
  • 128.Chang YC, Wang Z, Flax LA, et al. Glycosaminoglycan binding facilitates entry of a bacterial pathogen into central nervous systems. PLoS Pathog. 2011;7(6):e1002082. doi: 10.1371/journal.ppat.1002082. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Fida NM, Al-Mughales J, Farouq M. Interleukin-1alpha, interleukin-6 and tumor necrosis factor-alpha levels in children with sepsis and meningitis. Pediatr. Int. 2006;48(2):118–124. doi: 10.1111/j.1442-200X.2006.02152.x. [DOI] [PubMed] [Google Scholar]
  • 130.Tang RB, Lee BH, Chung RL, Chen SJ, Wong TT. Interleukin-1beta and tumor necrosis factor-alpha in cerebrospinal fluid of children with bacterial meningitis. Childs Nerv. Syst. 2001;17(8):453–456. doi: 10.1007/s003810000422. [DOI] [PubMed] [Google Scholar]
  • 131.Lehmann AK, Halstensen A, Sornes S, Rokke O, Waage A. High levels of interleukin 10 in serum are associated with fatality in meningococcal disease. Infect. Immun. 1995;63(6):2109–2112. doi: 10.1128/iai.63.6.2109-2112.1995. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 132.Leist TP, Frei K, Kam-Hansen S, Zinkernagel RM, Fontana A. Tumor necrosis factor alpha in cerebrospinal fluid during bacterial, but not viral, meningitis. Evaluation in murine model infections and in patients. J. Exp. Med. 1988;167(5):1743–1748. doi: 10.1084/jem.167.5.1743. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Waage A, Brandtzaeg P, Halstensen A, Kierulf P, Espevik T. The complex pattern of cytokines in serum from patients with meningococcal septic shock. Association between interleukin 6, interleukin 1, and fatal outcome. J. Exp. Med. 1989;169(1):333–338. doi: 10.1084/jem.169.1.333. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Nagesh Babu G, Kumar A, Kalita J, Misra UK. Proinflammatory cytokine levels in the serum and cerebrospinal fluid of tuberculous meningitis patients. Neurosci. Lett. 2008;436(1):48–51. doi: 10.1016/j.neulet.2008.02.060. [DOI] [PubMed] [Google Scholar]
  • 135.Cundell DR, Gerard NP, Gerard C, Idanpaan-Heikkila I, Tuomanen EI. Streptococcus pneumoniae anchor to activated human cells by the receptor for platelet-activating factor. Nature. 1995;377(6548):435–438. doi: 10.1038/377435a0. [DOI] [PubMed] [Google Scholar]
  • 136.Banerjee A, Van Sorge NM, Sheen TR, Uchiyama S, Mitchell TJ, Doran KS. Activation of brain endothelium by pneumococcal neuraminidase NanA promotes bacterial internalization. Cell. Microbiol. 2010;12(11):1576–1588. doi: 10.1111/j.1462-5822.2010.01490.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Zhang WG, Khan AN, Kim KJ, Stins M, Kim KS. Transforming growth factor-beta increases Escherichia coli K1 adherence, invasion, and transcytosis in human brain microvascular endothelial cells. Cell. Tissue Res. 2002;309(2):281–286. doi: 10.1007/s00441-002-0549-4. [DOI] [PubMed] [Google Scholar]
  • 138.Lembo A, Gurney MA, Burnside K, et al. Regulation of CovR expression in group B Streptococcus impacts blood–brain barrier penetration. Mol. Microbiol. 2010;77(2):431–443. doi: 10.1111/j.1365-2958.2010.07215.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Sharief MK, Ciardi M, Thompson EJ. Blood–brain barrier damage in patients with bacterial meningitis: association with tumor necrosis factor-alpha but not interleukin-1 beta. J. Infect. Dis. 1992;166(2):350–358. doi: 10.1093/infdis/166.2.350. [DOI] [PubMed] [Google Scholar]
  • 140.Barichello T, Pereira JS, Savi GD, et al. A kinetic study of the cytokine/chemokines levels and disruption of blood–brain barrier in infant rats after pneumococcal meningitis. J. Neuroimmunol. 2011;233(1-2):12–17. doi: 10.1016/j.jneuroim.2010.10.035. [DOI] [PubMed] [Google Scholar]
  • 141.Kim KS, Wass CA, Cross AS. Blood–brain barrier permeability during the development of experimental bacterial meningitis in the rat. Exp. Neurol. 1997;145(1):253–257. doi: 10.1006/exnr.1997.6458. [DOI] [PubMed] [Google Scholar]
  • 142.Mittal R, Prasadarao NV. Nitric oxide/cGMP signalling induces Escherichia coli K1 receptor expression and modulates the permeability in human brain endothelial cell monolayers during invasion. Cell Microbiol. 2010;12(1):67–83. doi: 10.1111/j.1462-5822.2009.01379.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Winkler F, Koedel U, Kastenbauer S, Pfister HW. Differential expression of nitric oxide synthases in bacterial meningitis: role of the inducible isoform for blood–brain barrier breakdown. J. Infect. Dis. 2001;183(12):1749–1759. doi: 10.1086/320730. [DOI] [PubMed] [Google Scholar]
  • 144.Leib SL, Kim YS, Black SM, Tureen JH, Tauber MG. Inducible nitric oxide synthase and the effect of aminoguanidine in experimental neonatal meningitis. J. Infect. Dis. 1998;177(3):692–700. doi: 10.1086/514226. [DOI] [PubMed] [Google Scholar]
  • 145.Koedel U, Bernatowicz A, Paul R, Frei K, Fontana A, Pfister HW. Experimental pneumococcal meningitis: cerebrovascular alterations, brain edema, and meningeal inflammation are linked to the production of nitric oxide. Ann. Neurol. 1995;37(3):313–323. doi: 10.1002/ana.410370307. [DOI] [PubMed] [Google Scholar]
  • 146.Mittal R, Gonzalez-Gomez I, Goth KA, Prasadarao NV. Inhibition of inducible nitric oxide controls pathogen load and brain damage by enhancing phagocytosis of Escherichia coli K1 in neonatal meningitis. Am. J. Pathol. 2010;176(3):1292–1305. doi: 10.2353/ajpath.2010.090851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Coureuil M, Lecuyer H, Scott MG, et al. Meningococcus Hijacks a beta2-adrenoceptor/beta-Arrestin pathway to cross brain microvasculature endothelium. Cell. 2010;143(7):1149–1160. doi: 10.1016/j.cell.2010.11.035. [DOI] [PubMed] [Google Scholar]
  • 148.Coureuil M, Mikaty G, Miller F, et al. Meningococcal type IV pili recruit the polarity complex to cross the brain endothelium. Science. 2009;325(5936):83–87. doi: 10.1126/science.1173196. ■ Demonstrates paracellular transit at the BBB.
  • 149.Schubert-Unkmeir A, Konrad C, Slanina H, Czapek F, Hebling S, Frosch M. Neisseria meningitidis induces brain microvascular endothelial cell detachment from the matrix and cleavage of occludin: a role for MMP-8. PLoS Pathog. 2010;6(4):e1000874. doi: 10.1371/journal.ppat.1000874. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 150.Sukumaran SK, Prasadarao NV. Escherichia coli K1 invasion increases human brain microvascular endothelial cell monolayer permeability by disassembling vascular-endothelial cadherins at tight junctions. J. Infect. Dis. 2003;188(9):1295–1309. doi: 10.1086/379042. [DOI] [PubMed] [Google Scholar]
  • 151.Ebrahimi CM, Kern JW, Sheen TR, et al. Penetration of the blood–brain barrier by Bacillus anthracis requires the pXO1-encoded BslA protein. J. Bacteriol. 2009;191(23):7165–7173. doi: 10.1128/JB.00903-09. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152.Ebrahimi CM, Sheen TR, Renken CW, Gottlieb RA, Doran KS. Contribution of lethal toxin and edema toxin to the pathogenesis of anthrax meningitis. Infect. Immun. 2011;79(7):2510–2518. doi: 10.1128/IAI.00006-11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Mukherjee DV, Tonry JH, Kim KS, et al. Bacillus anthracis protease InhA increases blood–brain barrier permeability and contributes to cerebral hemorrhages. PLoS One. 2011;6(3):E17921. doi: 10.1371/journal.pone.0017921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 154.Guichard A, McGillivray SM, Cruz-Moreno B, Van Sorge NM, Nizet V, Bier E. Anthrax toxins cooperatively inhibit endocytic recycling by the Rab11/Sec15 exocyst. Nature. 2010;467(7317):854–858. doi: 10.1038/nature09446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 155.Saez-Llorens X, Jafari HS, Severien C, et al. Enhanced attenuation of meningeal inflammation and brain edema by concomitant administration of anti-CD18 monoclonal antibodies and dexamethasone in experimental Haemophilus meningitis. J. Clin. Invest. 1991;88(6):2003–2011. doi: 10.1172/JCI115527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 156.Tuomanen EI, Saukkonen K, Sande S, Cioffe C, Wright SD. Reduction of inflammation, tissue damage, and mortality in bacterial meningitis in rabbits treated with monoclonal antibodies against adhesion-promoting receptors of leukocytes. J. Exp. Med. 1989;170(3):959–969. doi: 10.1084/jem.170.3.959. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Galanakis E, Di Cello F, Paul-Satyaseela M, Kim KS. Escherichia coli K1 induces IL-8 expression in human brain microvascular endothelial cells. Eur. Cytokine Netw. 2006;17(4):260–265. [PubMed] [Google Scholar]
  • 158.Vadeboncoeur N, Segura M, Al-Numani D, Vanier G, Gottschalk M. Pro-inflammatory cytokine and chemokine release by human brain microvascular endothelial cells stimulated by Streptococcus suis serotype 2. FEMS Immunol. Med. Microbiol. 2003;35(1):49–58. doi: 10.1111/j.1574-695X.2003.tb00648.x. [DOI] [PubMed] [Google Scholar]
  • 159.Wilson SL, Drevets DA. Listeria monocytogenes infection and activation of human brain microvascular endothelial cells. J. Infect. Dis. 1998;178(6):1658–1666. doi: 10.1086/314490. [DOI] [PubMed] [Google Scholar]
  • 160.Sokolova O, Heppel N, Jagerhuber R, et al. Interaction of Neisseria meningitidis with human brain microvascular endothelial cells: role of MAP- and tyrosine kinases in invasion and inflammatory cytokine release. Cell. Microbiol. 2004;6(12):1153–1166. doi: 10.1111/j.1462-5822.2004.00422.x. [DOI] [PubMed] [Google Scholar]
  • 161.Schubert-Unkmeir A, Sokolova O, Panzner U, Eigenthaler M, Frosch M. Gene expression pattern in human brain endothelial cells in response to Neisseria meningitidis. Infect. Immun. 2007;75(2):899–914. doi: 10.1128/IAI.01508-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Krishnan V, Gaspar AH, Ye N, Mandlik A, Ton-That H, Narayana SV. An IgG-like domain in the minor pilin GBS52 of Streptococcus agalactiae mediates lung epithelial cell adhesion. Structure. 2007;15(8):893–903. doi: 10.1016/j.str.2007.06.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 163.Van Sorge NM, Ebrahimi CM, McGillivray SM, et al. Anthrax toxins inhibit neutrophil signaling pathways in brain endothelium and contribute to the pathogenesis of meningitis. PLoS One. 2008;3(8):e2964. doi: 10.1371/journal.pone.0002964. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Reddy MA, Wass CA, Kim KS, Schlaepfer DD, Prasadarao NV. Involvement of focal adhesion kinase in Escherichia coli invasion of human brain microvascular endothelial cells. Infect. Immun. 2000;68(11):6423–6430. doi: 10.1128/iai.68.11.6423-6430.2000. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Sukumaran SK, Prasadarao NV. Regulation of protein kinase C in Escherichia coli K1 invasion of human brain microvascular endothelial cells. J. Biol. Chem. 2002;277(14):12253–12262. doi: 10.1074/jbc.M110740200. [DOI] [PubMed] [Google Scholar]
  • 166.Tenenbaum T, Bloier C, Adam R, Reinscheid DJ, Schroten H. Adherence to and invasion of human brain microvascular endothelial cells are promoted by fibrinogen-binding protein FbsA of Streptococcus agalactiae. Infect. Immun. 2005;73(7):4404–4409. doi: 10.1128/IAI.73.7.4404-4409.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Tenenbaum T, Spellerberg B, Adam R, Vogel M, Kim KS, Schroten H. Streptococcus agalactiae invasion of human brain microvascular endothelial cells is promoted by the laminin-binding protein Lmb. Microbes Infect. 2007;9(6):714–720. doi: 10.1016/j.micinf.2007.02.015. [DOI] [PubMed] [Google Scholar]

Websites

RESOURCES