Skip to main content
Erschienen in: Journal of Neuroinflammation 1/2018

Open Access 01.12.2018 | Research

Characterization of a Listeria monocytogenes meningitis mouse model

verfasst von: Merel M. Koopmans, JooYeon Engelen-Lee, Matthijs C. Brouwer, Valery Jaspers, Wing Kit Man, Mercedes Vall Seron, Diederik van de Beek

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2018

Abstract

Background

Listeria monocytogenes is a common cause of bacterial meningitis. We developed an animal model of listerial meningitis.

Methods

In survival studies, C57BL/6 mice received intracisternal injections with different L. monocytogenes sequence type 1 (ST1) colony forming units per milliliter (CFU; n = 48, 105, 106, 107, 108, and 109 CFU/ml). Second, mice were inoculated with 108 CFU/ml ST1 and sacrificed at 6 h and 24 h (n = 12/group). Outcome parameters were clinical score, CFUs, cyto- and chemokine levels, and brain histopathology. Third, 84 mice were inoculated (109 CFU/ml ST1) to determine optimal antibiotic treatment with different doses of amoxicillin and gentamicin. Fourth, mice were inoculated with 109 CFU/ml ST1, treated with amoxicillin, and sacrificed at 16 h and 24 h (n = 12/group) for outcome assessment. Finally, time point experiments were repeated with ST6 (n = 24/group).

Results

Median survival time for inoculation with 108 and 109 CFU/ml ST1 was 46 h and 40 h; lower doses of bacteria led to minimal clinical signs of disease. Brain levels of IL-6, IL-17A, and IFN-γ were elevated at 24 h, and IL-1β, IL-6, IL-10, IFN-γ, and TNF-α were elevated in blood at 6 h and 24 h. Histopathology showed increased meningeal infiltration, vascular inflammation of meningeal vessels, hemorrhages, and ventriculitis. In the treatment model, brain levels of IL-6 and IL-17A and blood levels of IL-6 and IFN-γ were elevated. Compared to ST6, infection with ST1 led initially to higher levels of IL-1β and TNF-α in blood and more profound neuropathological damage. At 16 h post inoculation, IL-1β, IL-10, and TNF-α in blood and IL-6, IL17A, TNF-α, and IFN-γ levels in brain were higher in ST1 compared to ST6 without differences in CFUs between STs. At 24 h, neuropathology score was higher in ST1 compared to ST6 (p = 0.002) infected mice.

Conclusions

We developed and validated a murine model of listerial meningitis. ST1-infected mice had a more severe inflammatory response and brain damage as compared to ST6-infected mice.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12974-018-1293-3) contains supplementary material, which is available to authorized users.
Abkürzungen
ARRIVE
Animal Research: Reporting of In Vivo Experiments
CC
Clonal complex
CFU
Colony forming units
CSF
Cerebrospinal fluid
H and E
Hematoxylin and eosin staining
HEP
Humane endpoint
i.p.
Intraperitoneal
IQR
Interquartile range
LD50
Lethal dose for 50% of mice
LLOD
Lower limit of detection
NRLBM
Netherlands Reference Laboratory for Bacterial Meningitis
OD
Optical density
ST1
Sequence type 1

Background

Bacterial meningitis is a life-threatening infectious disease of the central nervous system, which is most commonly caused by Streptococcus pneumoniae and Neisseria meningitidis [1, 2]. Listeria monocytogenes is the third most common pathogen causing bacterial meningitis in adults, and is found in 5–10% of cases [24]. Listeria distributes easily in the environment and can be found in soil, ground water, and feces of animals [5, 6]. Main source for human infection is food, and it primarily affects elderly and immunocompromised persons, [7] in whom it can cause up to 40% of community-acquired bacterial meningitis cases [8, 9]. A nation-wide prospective cohort study on L. monocytogenes meningitis described an increasing mortality rate over time, from 17 to 36% over the past decade [10]. This trend was also reported by a French cohort study including 252 patients with listerial meningitis [7].
In the immune response neutrophils, monocytes and macrophages are activated by pro-inflammatory cytokines such as IL-1α, IL-1, IL-6, IL-12, TNF-α, and IFN-γ [1114]. Anti-inflammatory cytokine IL-10 plays an important role in limiting immune-mediated damage and at the same time antagonizes IFN-γ activity which makes the host more vulnerable for an invasive listerial infection [14]. Several mouse and rat listeria models have been developed to study invasive L. monocytogenes diseases including cerebral and meningeal infection, using oral [1517], intravenous [18], intracerebral [1923], or intracisternal inoculation methods [2426]. Problems with reproducibility, limited disease progression, or iatrogenic structural damage, combined with a need for a single model in which most pathological features seen in human listerial meningitis can be measured, have created the need for development of a new animal model. We developed a listerial meningitis mouse model to counter these problems and compared infection with listerial ST1 with ST6.

Methods

Bacterial strain L. monocytogenes sequence type 1 (ST1) was used for the experiments, time point studies were repeated with a ST6 strain. Both strains were obtained from human positive cerebrospinal fluid (CSF) isolates stored at the Netherlands Reference Laboratory for Bacterial Meningitis (NRLBM). The isolates were grown to mid-log phase in 1–1.5 h at 37 °C in BHI to an optical density (OD600) of 0.45–0.55, then, centrifuged at 2000 rpm for 20 min at 4 °C. Supernatant was removed, and sterile 0.9% NaCl was added to yield the needed concentration. Before and after inoculation, the dose was determined by serial dilution method and plated on blood agar plates overnight at 37 °C.
Experiments were performed with eight- to ten-week-old C57BL/6 mice (Charles River Laboratories, Germany). In the non-treatment survival experiments, both sexes were used; in the non-treatment time point experiments and all treatment experiments, male C57BL/6 mice were used. The mice were kept to a controlled 12-h light/dark cycle, and food and water were provided ad libitum. All experiments were approved by the Institutional Animal Care and Use Committee of the Academic Medical Center, Amsterdam and performed according to the institution and Animal Research: Reporting of In Vivo Experiments (ARRIVE) guidelines [27].

Mouse model of listerial meningitis: non-treatment

Survival experiments were performed to determine clinical course of disease with the aim to achieve a median lethal dose for 50% of mice (LD50) after 36–48 h. Mice were inoculated with 1 μl bacterial suspension L. monocytogenes ST1 into the cisterna magna using a 32-gauge needle and syringe to dispense 1–10 μl. During inoculation, mice received short-term anesthesia using 2% isoflurane (Baxter). Five inoculum sizes were tested between 105 and 109 CFU/ml (n = 6 in 105 and 106 CFU/ml, n = 12 in 107 and 109 CFU/ml groups). After inoculation, mice were checked according to a clinical scoring list for direct neurological deficits which could indicate puncture failure (such as occipital bleeding). If direct neurological deficits were found, mice were euthanized and excluded from the experiment. Mice were monitored every 4–6 h (starting from 12 h after inoculation), and the clinical score (Table 1) and the portion of surviving mice in each group was determined up to 90% mortality [28]. Mice were sacrificed by intraperitoneal injection (i.p.) with dexmedetomidine (0.3 mg/kg) in combination with ketamine (190 mg/kg) when a clinical score of ≥ 15 was reached. Subsequently, time point experiments were performed (n = 12 per time point) with 108 CFU/ml L. monocytogenes ST1 or ST6 which were compared to a control group (n = 6) receiving 1 μl 0.9% NaCl intracisternally. At 6 and 24 h post-inoculation, mice were sacrificed, and blood, CSF, and organs were collected, processed, and stored [28, 29].
Table 1
Clinical scoring list for bacterial meningitis mouse model
Parameter
Value
Weighted score
Max. score
Weight loss
Normal 5%
0
 
5–10%
1
 
10–15%
2
 
15–20%
3
 
20–25%
4
4
Activity
Normal
0
 
Increased/aggressive
1
 
Mildly diminished
1
 
Diminished
2
 
Severely diminished
3
3
Condition
Normal, does not lay on back
0
 
Upright within 5 s
2
 
Upright within 30 s
4
 
Does not turn upright
6
6
Coat
Normal
0
 
Diminished grooming
1
 
Soiled
1
 
Piloerection
1
3
Posture
Normal
0
 
Slightly hunched back
1
2
Severe hunched back
2
 
Eyes
Normal
0
 
Protruding
1
 
Closed eyelids
1
 
Discharge
1
4
Respiration rate (per min)
> 150
0
 
100–150
1
 
75–100
2
 
50–75
3
 
< 50
4
4
Breathing
Irregular
2
 
Labored
2
4
Neurological examination
Normal
0
 
Coordination problem
2
 
Paresis/paralysis
2
 
Epileptic seizure
2
 
Status epilepticus
6
10
Humane endpoints
Total score ≥ 15
  
Status epilepticus
  
≥ 2 seizures in 15 min
  
Hemiparalysis
  
≥ 25% weight loss
  

Mouse model of listerial meningitis: treatment

Three survival experiments with 84 male mice (12 mice per subgroup) were performed to test dosage and frequency of intraperitoneal amoxicillin treatment, and the potentially beneficial effect of adding intraperitoneal gentamicin, as is the preferred antibiotic treatment used in human listerial meningitis [30]. After each experiment, brains of one or two surviving mice per subgroup were harvested to measure bacterial outgrowth. In the first experiment, mice were inoculated with 109 CFU/ml L. monocytogenes ST1 and treated 16 h post-inoculation with 50 or 100 mg/kg amoxicillin every 24 h. In the second experiment, higher doses (100 vs. 200 mg/kg/24 h amoxicillin) and shorter treatment interval (100 mg/kg amoxicillin every 12 h vs. every 24 h) was tested, starting 16 h post-inoculation. To determine the effect of additional gentamicin, 20 mg/kg/24 h of gentamicin was administered concomitant to 100 mg/kg/24 h amoxicillin 16 h post-inoculation compared to 100 mg/kg/24 h amoxicillin only in mice who were inoculated with 108 CFU/ml. In the time point treatment experiments, mice were inoculated with 109 CFU/ml L. monocytogenes ST1 or ST6 per strain. Twelve mice were sacrificed 16 h after infection, and 12 mice were treated with 100 mg/kg amoxicillin i.p. 16 h after infection and sacrificed after 24 h.

Scoring, harvesting, and cytokine analyses

Clinical scoring was performed by two observers according to a previously developed scoring list for a pneumococcal meningitis mouse model (Table 1) [28]. Each scoring parameter ranges from zero, corresponding to no abnormalities, to a variable maximum score. Animals reaching humane endpoint (HEP) criteria were humanely killed. After anesthetizing the mice, cardiac puncture was performed for blood collection, and intracisternal puncture for CSF collection. Brain, spleen, liver, and lungs were harvested and processed as described previously [28]. Supernatant, plasma, and CSF were stored at − 80 °C until further use. Cytokine analyses were performed using Luminex technology Bio-plex Pro Mouse Cytokine 6-plex Assay (Bio-Rad Laboratories, Veenendaal, the Netherlands). We analyzed levels of IL-1β, IL-6, IL-10, IL-17A, TNF-α, and IFN-γ to include early and late pro- and anti-inflammatory cytokines. For cytokine values below the lower limit of detection (LLOD), the LLOD was used in the calculation of median and interquartile range. Histopathology was performed on the left hemisphere of the brain. Brain was fixed in 4% paraformaldehyde and paraffin embedded in seven coronal plaques. In all mice, a hematoxylin and eosin (H and E) staining and Gram staining were performed. Histopathology was scored (blinded) in six categories by a neuropathologist as previously described. An additional table shows this in more detail (Additional file 1) [29].
Comparison of survival curves between groups within each model was calculated using the log-rank test. Clinical scores were compared using a linear mixed model, assuming an exponential and group-specific time effect. Comparisons of cytokine levels between groups were calculated using the Mann–Whitney U test, and for dichotomous variables in histopathology scoring, the Fisher’s exact test was used. All statistical tests were two-tailed, and a p value of < 0.05 was considered to be significant.

Results

Non-treatment model

In the survival study, mice inoculated with 105, 106 and 107 CFU/ml L. monocytogenes ST1 showed minimal clinical signs of disease limited to diminished grooming and/or a slightly hunched back. They had to be sacrificed based on weight loss, ≥ 25% according to the predefined endpoint in the clinical scoring (Fig. 1a), and therefore considered not representable for a listerial meningitis model. Mice inoculated with 108 CFU/ml L. monocytogenes ST1 showed signs of illness 12 h after inoculation, consisting of discharge in eyes, a slightly hunched back, mildly diminished activity and/or piloerection (median clinical score of 2; interquartile range [IQR 2–2]), and had a median survival time of 46 h (IQR 34–52 h; Fig. 1b). Mice inoculated with 109 CFU/ml L. monocytogenes ST1 had signs of illness 12 h after inoculation (median clinical score of 3 [IQR 3–4]), and a median survival time of 40 h (IQR 23–46 h). The concentration of 108 CFU/ml was determined to be the optimal inoculum size for the non-treatment model. There was no difference in male or female mice in survival. An additional figure shows this in more detail (Additional file 2).
After intracisternal injection of 108 CFU/ml L. monocytogenes ST1, CFUs increased significantly over time in CSF, blood, and collected organs (Fig. 1c). Median L. monocytogenes concentration in the CSF was 6.8 × 104 CFU after 6 h and 1.2 × 106 CFU/ml after 24 h. In the brain homogenates, median titres were 2.0 × 105 after 6 h and 1.0 × 106 CFU/mg after 24 h. In the spleen, lungs, and liver homogenates, median bacterial titres were 2.1 × 103, 2.0 × 102, and 2.0 × 102 CFU/mg after 6 h and 4.9 × 105, 2.0 × 103, and 1.5 × 105 CFU/mg after 24 h respectively. Median titre in blood at 6 h was under the LLOD (< 1.0 × 102 CFU/ml) and 1.5 × 102 CFU/ml after 24 h.
Luminex analyses showed similar cytokine levels in the brain at the 6-h time point compared to negative controls, except for IL-1β in which levels were lower compared to the controls. Levels of IL-6, IL-17A, and IFN-γ were elevated in brain homogenates at the 24-h time point in infected mice compared to controls (Fig. 2), and all increased after 24 h compared to the 6-h time point. Plasma levels of all measured cytokines with the exception of IL-17A were elevated at both time points compared to the controls, and IL-6, IL-10, and IFN-γ increased between 6 h and 24 h (Fig. 2).
Histopathology showed increased brain damage between t = 6 and t = 24 in the non-treatment model. Median pathology score increased from 2 [IQR 1–4] to 7 [IQR 6–9] (p = 0.001, an additional table shows this in more detail Additional file 3) due to increase from mild focal meningeal infiltration (Fig. 3a) to severe meningeal infiltration (Fig. 3b), and meningeal vascular inflammation from one mouse (8%) 6 h post-inoculation to 12 mice (100%) 24 h post-inoculation. Focal small parenchymal and subarachnoid bleedings were present in 6 of 12 mice at 6 h (50%), and developed into multiple parenchymal and subarachnoid hemorrhages at 24 h (in 58%, Fig. 3c). Ventriculitis was present in 17% of mice at 6 h to 42% at 24 h.

Treatment model

Treatment time point was chosen at 16 h post-infection based on clinical course of disease in the survival experiments in mice inoculated with 109 CFU/ml bacteria. Treatment with 100 mg/kg/24 h of amoxicillin was superior to 50 mg/kg/24 h of amoxicillin in survival (median survival 70 h vs. 46 h, p < 0.001; an additional figure shows this in more detail Additional file 4a). Further experiments showed survival did not improve by treating mice with 200 mg/kg/24 h or 100 mg/kg/12 h of amoxicillin or adjuvant 20 mg/kg/24 h of gentamicin compared to 100 mg/kg/24 h of amoxicillin, and no significant differences were found in clinical scoring (Additional files 2 and 4). In all survival experiments, brain homogenates from mice that survived till the end of the experiment (70 h post-inoculation) showed bacterial outgrowth. Decreasing the inoculum size to 108 CFU/ml did not improve clearance of bacteria. An additional table shows this in more detail (Additional file 5).
In the time point study, bacterial titres in all collected fluids and organs decreased between 16 h and 24 h after treatment with 100 mg/kg/24 h of amoxicillin. Sixteen hours post-inoculation median bacterial titre in CSF was 4.3 × 106 CFU/ml, and decreased after treatment with amoxicillin to 5.2 × 105 CFU/ml at 24 h. An additional figure shows this in more detail (Additional file 4). Median bacterial concentration in the brain homogenates was 6.4 × 107 after 16 h and 1.0 × 106 CFU/mg after 24 h, and in blood, spleen, lungs, and liver homogenates, median bacterial titres were 2.0 × 103, 3.3 × 106, 8.2 × 103, and 4.1 × 104 CFU/mg after 16 h and 1.6 × 102, 1.3 × 105, 2.3 × 103, and 2.2 × 104 CFU/mg after 24 h respectively.
Cytokine measurements showed elevated IL-1β, IL-6, and IL-17A concentration in ST1-infected mice compared to negative controls in both time points (Fig. 4). Luminex of plasma in the treatment model showed elevated levels of IL-6, IL-10, and IFN-γ and decreased levels of IL-17A at both time points. IL-1β was elevated at the 16-h time point in ST1-infected mice compared to controls (Fig. 4).
Overall, histopathological score was similar between 16- and 24-h time points (median score 7 [IQR 5–8] vs. 9 [IQR 8–12]). An additional table shows this in more detail (Additional file 3). Meningeal infiltration and meningeal vascular inflammation were present in all mice. As seen in the non-treatment model, bleeding and ventriculitis were frequently present. Hemorrhages were found parenchymal or subarachnoidal, and were categorized as small and focal, 16 h post-inoculation in 73% of mice and as large hemorrhages at multiple locations in 91% after 24 h.

Bacterial strain ST1 vs. ST6

In the non-treatment time point experiments, clinical score and bacterial outgrowth of ST6 in CSF, blood, and collected organs were comparable to ST1. Cytokine and chemokine levels in brain homogenates measured at similar time points did not differ between ST1 and ST6. In plasma, IL-1β and TNF-α of both STs were significantly elevated 6 h post-inoculation, and ST1 24 h post-inoculation compared to the negative controls. At t = 24, there was a significant difference between strains in median IL-1β levels, ST1 432 pg/ml [IQR 282–533 pg/ml] and ST6 192 pg/ml [IQR 137–307 pg/ml] (p = 0.04), and at both time points, there was a significant difference in TNF-α levels between ST1 and ST6 (TNF-α t = 6; ST1 900 pg/ml [IQR 766–1034] vs. ST6 746 pg/ml [IQR 639–812] (p = 0.03) and at t = 24; ST1 1022 pg/ml [IQR 674–1137] vs. 470 pg/ml [IQR 370–746], p = 0.045). Histopathology scores in ST1- and ST6-infected mice were similar in terms of meningeal infiltration, severity of ventriculitis, thrombosis, and increase of meningeal vascular inflammation over time. Six hours post-inoculation, focal small parenchymal and meningeal bleeding was present in 6 of 12 ST1 mice (50%), while it was only seen in one of the 12 ST6-infected mice (8%, p = 0.03), but overall pathology scores were similar between ST1- and ST6-infected mice at 24 h.
There were no significant differences in clinical score and bacterial outgrowth in collected fluids or organs between ST1 and ST6 in both non-treatment and treatment model. An additional figure shows this in more detail (Additional file 6). In the treatment model, five ST6 mice had to be euthanized (three because of puncture failure and two because of wounds after fighting). However, brain levels of IL-6, IL17A, TNF-α, and IFN-γ in ST1-inoculated mice compared to ST6 16 h. An additional table shows this in more detail (Additional file 7). In plasma, IL-1β levels were higher in ST1-inoculated mice at both time points compared to ST6-infected mice, and IL-10 and TNF-α levels were higher at the 24-h time point in ST1-infected mice. Overall pathology score was higher at 24 h in ST1-infected mice (median score 9; [811, 31]) compared to ST6-inoculated mice (median score 7; [6, 7], p = 0.002). This was mainly driven by increased meningeal infiltration, ventriculitis, and hemorrhages in ST1-infected mice.

Discussion

We developed and validated a murine model of listerial meningitis. We used Listeria sequence types [18, 32] that commonly cause invasive disease [18, 31]. Previously described intracisternally inoculation mice and rat studies used a serotype 4b strain with unknown sequence type [2426]; intracerebral inoculation mice studies [22, 23] used a less virulent laboratory EGD strain compared to the ST1 and ST6 strains [18, 33]. We used intracisternal inoculation aiming for a reproducible meningitis model. Previous studies using oral and intravenous inoculation reported difficulties with respect to neuro-invasion reproducibility [1517, 3436], while intracerebral injection primarily causes cerebritis. Intracerebral inoculation methods have been used successfully previously to study the role of macrophage inflammatory protein and TNF-α in listerial meningitis [22, 23]. Intracisternal inoculation has been used to study heat production [24], compare effectiveness of antibiotics [26], and the role of reactive oxygen and nitric oxide in listeria growth [25]. Our model allows evaluation of multiple features including bacterial growth, host immune response, clinical severity, and histopathological damage.
Main histopathological characteristics of listerial meningitis were meningeal inflammation, ventriculitis, and abscesses. This is in line with previous studies [37, 38]. We also observed a high rate of cerebral hemorrhages, an uncommon feature in human listerial meningitis (2% of cases) [10]. This is consistent with the observed difference in pneumococcal meningitis in a human and mouse model [28, 39]. In human bacterial meningitis, it has been suggested that dysregulation of coagulation and fibrinolytic pathways, vascular endothelial cell swelling, and vasculitis plays a role in the pathophysiology of hemorrhages [4044].
IL-1β, IL-6, IL-10, IL-17A, TNF-α, and IFN-γ mediated the hosts immune response against L. monocytogenes. Previous listerial mouse models showed that monocyte recruitment to the brain is triggered by pro-inflammatory cytokines in particular IFN-γ- , TNF- , and IL-6-related immune response [45, 46]. These cytokines and IL-1β and IL-17A are able to mobilize phagocytes and activate other cytokines [1114, 4654], whereas IL-10 limits the immune-mediated injury; nonetheless can increase severity of L. monocytogenes disease by reducing the immune response [5557]. Since IL-6 and IFN-γ were elevated in the brain and blood of both our treatment and non-treatment mouse models, these cytokines are relevant outcome measures in our model to study changes in the inflammatory response. An interesting aspect of IFN-γ is its ambiguous role in listerial infections. It is known for its protective and controlling role in the early immune response, though it seems to promote susceptibility for L. monocytogenes later on. Study models with interferon-deficient mice showed protective effects during systemic listerial infections [5860]. In CSF of patients with listerial meningitis, elevation of IFN-γ, IFN-α2, and interferon-related cytokines IL-18, CX3CL1, and CCL20 were associated with an unfavorable outcome [61]. The use of amoxicillin, a bacteriolytic antibiotic, in our model did not lead to a significant increase in cytokine levels after therapy, as observed in other experimental models of bacterial meningitis also using bacteriolytic antibiotic.
Infection with Listeria strain of the ST1 type led to a more rigorous inflammatory response and more brain damage as compared to infection with ST6. Both STs have been marked as hypervirulent strains with a tropism for neuro-invasion [18]. ST1 has been among the most common genotypes causing listerial meningitis in the Netherlands over the last 25 years [31]. ST6 has been emerging over the last years and has been associated with an increasing rate of unfavorable outcome among adults with listerial meningitis, from 27 to 61% over a 14-year period [10]. The increased incidence of ST6 listerial meningitis in the Netherlands has been associated with the introduction of a novel plasmid, carrying the efflux transporter emrC [62]. Although speculative, differences in virulence between ST1 and ST6 found in our model could be explained by (i) degree of cell-to-cell spread from infected phagocytes to endothelial cells [63, 64]; (ii) the interaction with macrophages, neutrophils, and subsequently the cytokine signaling [65]; (iii) the proportion of Listeria bacteria residing in the brain parenchyma rather than extracellularly in the CSF, and thereby causing different degrees of histopathological damage [66]; (iv) degree of expression of the specific neuro-invasive internalin InlF and its binding to the filament protein vimentin [67, 68]; (v) presence of certain genetic elements in the bacteria such as LIPI-3 (in both ST1 and ST6) [69], LGI 2 (found in ST1) [70], or pLMST6 (found in ST6) [62]; (vi) other yet unknown factors influencing and differentiating the virulence of L. monocytogenes strains. Since ST1 and ST6 are clinically relevant strains, these unknown factors should be investigated and might help to unravel the pathophysiology of L. monocytogenes.
Our model has several limitations, of which some are inherent to the use of modeling of human disease in animals. First, we infected mice by inoculating directly into the cisterna magna, while the route of infection in humans mainly is through the digestive system. However, meningitis is difficult to evoke unless bacteria are injected directly intracranial, partially because animals tend to die due to systemic illness before meningitis develops [71]. Furthermore, the amount of bacteria reaching the brain cannot be controlled using digestive tract or intravenous inoculation. Second, in the treatment survival experiments, we observed that Listeria could be cultured from the murine brains despite high doses antibiotic treatment. This can be explained as the pathogen is intracellular and has a relatively slow growth rate. Patients with listerial meningitis are therefore treated for at least 3 weeks. To make sure we did not use insufficient dosage or type of antibiotics, we increased the dose and frequency of the amoxicillin and added gentamycin, but these changes did not influence outcome or bacterial outgrowth at the end of the experiment. Therefore, we feel that we achieved an optimal amoxicillin dose to perform the experiments with. We did observe that bacterial counts decreased in all treated mice. It could be argued that other antibiotics with a previously suggested effect and/or had a synergism in treatment of listerial meningitis should have been tested [30]. However, amoxicillin with or without gentamicin is the most commonly used treatment in human listerial meningitis, and therefore testing other antibiotics is beyond the scope of this article.

Conclusions

The listerial meningitis mouse model provides an experimental setting of listerial meningitis with multiple outcome parameters. Similar model set up in pneumococcal meningitis has proven to be useful in exploring inflammatory hypotheses in pneumococcal meningitis [7274]. Integration of these pathological features in a single model is a valuable tool in the further investigation of both pathophysiological and therapeutic intervention studies in listerial meningitis.

Funding

This study was supported by the Netherlands Organization for Health Research and Development (ZonMw; NWO-Veni-Grant [916.13.078] to MB, NWO-Vidi-Grant [016.116.358] to DB), the Academic Medical Center (AMC Fellowship to DB), and the European Research Council (ERC Starting Grant to DB).

Availability of data and materials

Data of the MeninGene study is available for all researchers at www.​MeninGene.​eu.

Ethics approval

The study was approved by the Medical Ethical Committee of the Academic Medical Centre, Amsterdam, the Netherlands. Experiments were approved by the Institutional Animal Care and Use Committee of the Academic Medical Center, Amsterdam, the Netherlands.
Not applicable.

Competing interests

The authors declare that they have no competing interests.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://​creativecommons.​org/​licenses/​by/​4.​0/​), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://​creativecommons.​org/​publicdomain/​zero/​1.​0/​) applies to the data made available in this article, unless otherwise stated.
Anhänge

Additional files

Literatur
1.
Zurück zum Zitat van de Beek D, de Gans J, Spanjaard L, Weisfelt M, Reitsma JB, Vermeulen M. Clinical features and prognostic factors in adults with bacterial meningitis. N Engl J Med. 2004;351:1849–59.CrossRefPubMed van de Beek D, de Gans J, Spanjaard L, Weisfelt M, Reitsma JB, Vermeulen M. Clinical features and prognostic factors in adults with bacterial meningitis. N Engl J Med. 2004;351:1849–59.CrossRefPubMed
2.
Zurück zum Zitat Bijlsma MW, Brouwer MC, Kasanmoentalib ES, Kloek AT, Lucas MJ, Tanck MW, van der Ende A, van de Beek D. Community-acquired bacterial meningitis in adults in the Netherlands, 2006-14: a prospective cohort study. Lancet Infect Dis. 2016;16:339–47.CrossRefPubMed Bijlsma MW, Brouwer MC, Kasanmoentalib ES, Kloek AT, Lucas MJ, Tanck MW, van der Ende A, van de Beek D. Community-acquired bacterial meningitis in adults in the Netherlands, 2006-14: a prospective cohort study. Lancet Infect Dis. 2016;16:339–47.CrossRefPubMed
3.
Zurück zum Zitat Moon SY, Chung DR, Kim SW, Chang HH, Lee H, Jung DS, Kim YS, Jung SI, Ryu SY, Heo ST, et al. Changing etiology of community-acquired bacterial meningitis in adults: a nationwide multicenter study in Korea. Eur J Clin Microbiol Infect Dis. 2010;29:793–800.CrossRefPubMed Moon SY, Chung DR, Kim SW, Chang HH, Lee H, Jung DS, Kim YS, Jung SI, Ryu SY, Heo ST, et al. Changing etiology of community-acquired bacterial meningitis in adults: a nationwide multicenter study in Korea. Eur J Clin Microbiol Infect Dis. 2010;29:793–800.CrossRefPubMed
4.
Zurück zum Zitat Thigpen MC, Whitney CG, Messonnier NE, Zell ER, Lynfield R, Hadler JL, Harrison LH, Farley MM, Reingold A, Bennett NM, et al. Bacterial meningitis in the United States, 1998-2007. N Engl J Med. 2011;364:2016–25.CrossRefPubMed Thigpen MC, Whitney CG, Messonnier NE, Zell ER, Lynfield R, Hadler JL, Harrison LH, Farley MM, Reingold A, Bennett NM, et al. Bacterial meningitis in the United States, 1998-2007. N Engl J Med. 2011;364:2016–25.CrossRefPubMed
6.
Zurück zum Zitat Stea EC, Purdue LM, Jamieson RC, Yost CK, Truelstrup Hansen L. Comparison of the prevalences and diversities of listeria species and listeria monocytogenes in an urban and a rural agricultural watershed. Appl Environ Microbiol. 2015;81:3812–22.CrossRefPubMedPubMedCentral Stea EC, Purdue LM, Jamieson RC, Yost CK, Truelstrup Hansen L. Comparison of the prevalences and diversities of listeria species and listeria monocytogenes in an urban and a rural agricultural watershed. Appl Environ Microbiol. 2015;81:3812–22.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Charlier C, Perrodeau E, Leclercq A, Cazenave B, Pilmis B, Henry B, Lopes A, Maury MM, Moura A, Goffinet F, et al. Clinical features and prognostic factors of listeriosis: the MONALISA national prospective cohort study. Lancet Infect Dis. 2017;15:150–9. Charlier C, Perrodeau E, Leclercq A, Cazenave B, Pilmis B, Henry B, Lopes A, Maury MM, Moura A, Goffinet F, et al. Clinical features and prognostic factors of listeriosis: the MONALISA national prospective cohort study. Lancet Infect Dis. 2017;15:150–9.
8.
Zurück zum Zitat van Ettekoven CN, van de Beek D, Brouwer MC. Update on community-acquired bacterial meningitis: guidance and challenges. Clin Microbiol Infect. 2017;23:601–6.CrossRefPubMed van Ettekoven CN, van de Beek D, Brouwer MC. Update on community-acquired bacterial meningitis: guidance and challenges. Clin Microbiol Infect. 2017;23:601–6.CrossRefPubMed
9.
Zurück zum Zitat van Veen KEB, Brouwer MC, van der Ende A, van de Beek D. Bacterial meningitis in patients using immunosuppressive medication: a population-based prospective nationwide study. J NeuroImmune Pharmacol. 2017;12:213–8.CrossRefPubMed van Veen KEB, Brouwer MC, van der Ende A, van de Beek D. Bacterial meningitis in patients using immunosuppressive medication: a population-based prospective nationwide study. J NeuroImmune Pharmacol. 2017;12:213–8.CrossRefPubMed
10.
Zurück zum Zitat Koopmans MM, Brouwer MC, Bijlsma MW, Bovenkerk S, Keijzers W, van der Ende A, van de Beek D. Listeria monocytogenes sequence type 6 and increased rate of unfavorable outcome in meningitis: epidemiologic cohort study. Clin Infect Dis. 2013;57:247–53.CrossRefPubMed Koopmans MM, Brouwer MC, Bijlsma MW, Bovenkerk S, Keijzers W, van der Ende A, van de Beek D. Listeria monocytogenes sequence type 6 and increased rate of unfavorable outcome in meningitis: epidemiologic cohort study. Clin Infect Dis. 2013;57:247–53.CrossRefPubMed
11.
Zurück zum Zitat Labow M, Shuster D, Zetterstrom M, Nunes P, Terry R, Cullinan EB, Bartfai T, Solorzano C, Moldawer LL, Chizzonite R, McIntyre KW. Absence of IL-1 signaling and reduced inflammatory response in IL-1 type I receptor-deficient mice. J Immunol. 1997;159:2452–61.PubMed Labow M, Shuster D, Zetterstrom M, Nunes P, Terry R, Cullinan EB, Bartfai T, Solorzano C, Moldawer LL, Chizzonite R, McIntyre KW. Absence of IL-1 signaling and reduced inflammatory response in IL-1 type I receptor-deficient mice. J Immunol. 1997;159:2452–61.PubMed
12.
Zurück zum Zitat Dalrymple SA, Lucian LA, Slattery R, McNeil T, Aud DM, Fuchino S, Lee F, Murray R. Interleukin-6-deficient mice are highly susceptible to listeria monocytogenes infection: correlation with inefficient neutrophilia. Infect Immun. 1995;63:2262–8.PubMedPubMedCentral Dalrymple SA, Lucian LA, Slattery R, McNeil T, Aud DM, Fuchino S, Lee F, Murray R. Interleukin-6-deficient mice are highly susceptible to listeria monocytogenes infection: correlation with inefficient neutrophilia. Infect Immun. 1995;63:2262–8.PubMedPubMedCentral
13.
Zurück zum Zitat Tripp CS, Gately MK, Hakimi J, Ling P, Unanue ER. Neutralization of IL-12 decreases resistance to listeria in SCID and C.B-17 mice. Reversal by IFN-gamma. J Immunol. 1994;152:1883–7.PubMed Tripp CS, Gately MK, Hakimi J, Ling P, Unanue ER. Neutralization of IL-12 decreases resistance to listeria in SCID and C.B-17 mice. Reversal by IFN-gamma. J Immunol. 1994;152:1883–7.PubMed
14.
Zurück zum Zitat Tripp CS, Wolf SF, Unanue ER. Interleukin 12 and tumor necrosis factor alpha are costimulators of interferon gamma production by natural killer cells in severe combined immunodeficiency mice with listeriosis, and interleukin 10 is a physiologic antagonist. Proc Natl Acad Sci U S A. 1993;90:3725–9.CrossRefPubMedPubMedCentral Tripp CS, Wolf SF, Unanue ER. Interleukin 12 and tumor necrosis factor alpha are costimulators of interferon gamma production by natural killer cells in severe combined immunodeficiency mice with listeriosis, and interleukin 10 is a physiologic antagonist. Proc Natl Acad Sci U S A. 1993;90:3725–9.CrossRefPubMedPubMedCentral
15.
Zurück zum Zitat Czuprynski CJ, Faith NG, Steinberg H. A/J mice are susceptible and C57BL/6 mice are resistant to listeria monocytogenes infection by intragastric inoculation. Infect Immun. 2003;71:682–9.CrossRefPubMedPubMedCentral Czuprynski CJ, Faith NG, Steinberg H. A/J mice are susceptible and C57BL/6 mice are resistant to listeria monocytogenes infection by intragastric inoculation. Infect Immun. 2003;71:682–9.CrossRefPubMedPubMedCentral
16.
Zurück zum Zitat Bou Ghanem EN, Myers-Morales T, D'Orazio SE. A mouse model of foodborne listeria monocytogenes infection. Curr Protoc Microbiol. 2013;31:9B 3 1–9B 3 16. Bou Ghanem EN, Myers-Morales T, D'Orazio SE. A mouse model of foodborne listeria monocytogenes infection. Curr Protoc Microbiol. 2013;31:9B 3 1–9B 3 16.
17.
Zurück zum Zitat Bergmann S, Beard PM, Pasche B, Lienenklaus S, Weiss S, Gahan CG, Schughart K, Lengeling A. Influence of internalin A murinisation on host resistance to orally acquired listeriosis in mice. BMC Microbiol. 2013;13:90.CrossRefPubMedPubMedCentral Bergmann S, Beard PM, Pasche B, Lienenklaus S, Weiss S, Gahan CG, Schughart K, Lengeling A. Influence of internalin A murinisation on host resistance to orally acquired listeriosis in mice. BMC Microbiol. 2013;13:90.CrossRefPubMedPubMedCentral
18.
Zurück zum Zitat Maury MM, Tsai YH, Charlier C, Touchon M, Chenal-Francisque V, Leclercq A, Criscuolo A, Gaultier C, Roussel S, Brisabois A, et al. Uncovering listeria monocytogenes hypervirulence by harnessing its biodiversity. Nat Genet. 2016;48:308–13. Maury MM, Tsai YH, Charlier C, Touchon M, Chenal-Francisque V, Leclercq A, Criscuolo A, Gaultier C, Roussel S, Brisabois A, et al. Uncovering listeria monocytogenes hypervirulence by harnessing its biodiversity. Nat Genet. 2016;48:308–13.
19.
Zurück zum Zitat Virna S, Deckert M, Lutjen S, Soltek S, Foulds KE, Shen H, Korner H, Sedgwick JD, Schluter D. TNF is important for pathogen control and limits brain damage in murine cerebral listeriosis. J Immunol. 2006;177:3972–82.CrossRefPubMed Virna S, Deckert M, Lutjen S, Soltek S, Foulds KE, Shen H, Korner H, Sedgwick JD, Schluter D. TNF is important for pathogen control and limits brain damage in murine cerebral listeriosis. J Immunol. 2006;177:3972–82.CrossRefPubMed
20.
Zurück zum Zitat Deckert M, Soltek S, Geginat G, Lutjen S, Montesinos-Rongen M, Hof H, Schluter D. Endogenous interleukin-10 is required for prevention of a hyperinflammatory intracerebral immune response in listeria monocytogenes meningoencephalitis. Infect Immun. 2001;69:4561–71.CrossRefPubMedPubMedCentral Deckert M, Soltek S, Geginat G, Lutjen S, Montesinos-Rongen M, Hof H, Schluter D. Endogenous interleukin-10 is required for prevention of a hyperinflammatory intracerebral immune response in listeria monocytogenes meningoencephalitis. Infect Immun. 2001;69:4561–71.CrossRefPubMedPubMedCentral
21.
Zurück zum Zitat Schluter D, Chahoud S, Lassmann H, Schumann A, Hof H, Deckert-Schluter M. Intracerebral targets and immunomodulation of murine listeria monocytogenes meningoencephalitis. J Neuropathol Exp Neurol. 1996;55:14–24.CrossRefPubMed Schluter D, Chahoud S, Lassmann H, Schumann A, Hof H, Deckert-Schluter M. Intracerebral targets and immunomodulation of murine listeria monocytogenes meningoencephalitis. J Neuropathol Exp Neurol. 1996;55:14–24.CrossRefPubMed
22.
Zurück zum Zitat Seebach J, Bartholdi D, Frei K, Spanaus KS, Ferrero E, Widmer U, Isenmann S, Strieter RM, Schwab M, Pfister H, Fontana A. Experimental listeria meningoencephalitis. Macrophage inflammatory protein-1 alpha and -2 are produced intrathecally and mediate chemotactic activity in cerebrospinal fluid of infected mice. J Immunol. 1995;155:4367–75.PubMed Seebach J, Bartholdi D, Frei K, Spanaus KS, Ferrero E, Widmer U, Isenmann S, Strieter RM, Schwab M, Pfister H, Fontana A. Experimental listeria meningoencephalitis. Macrophage inflammatory protein-1 alpha and -2 are produced intrathecally and mediate chemotactic activity in cerebrospinal fluid of infected mice. J Immunol. 1995;155:4367–75.PubMed
23.
Zurück zum Zitat 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;167:1743–8.CrossRefPubMed 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;167:1743–8.CrossRefPubMed
24.
Zurück zum Zitat Trampuz A, Steinhuber A, Wittwer M, Leib SL. Rapid diagnosis of experimental meningitis by bacterial heat production in cerebrospinal fluid. BMC Infect Dis. 2007;7:116.CrossRefPubMedPubMedCentral Trampuz A, Steinhuber A, Wittwer M, Leib SL. Rapid diagnosis of experimental meningitis by bacterial heat production in cerebrospinal fluid. BMC Infect Dis. 2007;7:116.CrossRefPubMedPubMedCentral
25.
Zurück zum Zitat Remer KA, Jungi TW, Fatzer R, Tauber MG, Leib SL. Nitric oxide is protective in listeric meningoencephalitis of rats. Infect Immun. 2001;69:4086–93.CrossRefPubMedPubMedCentral Remer KA, Jungi TW, Fatzer R, Tauber MG, Leib SL. Nitric oxide is protective in listeric meningoencephalitis of rats. Infect Immun. 2001;69:4086–93.CrossRefPubMedPubMedCentral
26.
Zurück zum Zitat Michelet C, Leib SL, Bentue-Ferrer D, Tauber MG. Comparative efficacies of antibiotics in a rat model of meningoencephalitis due to listeria monocytogenes. Antimicrob Agents Chemother. 1999;43:1651–6.PubMedPubMedCentralCrossRef Michelet C, Leib SL, Bentue-Ferrer D, Tauber MG. Comparative efficacies of antibiotics in a rat model of meningoencephalitis due to listeria monocytogenes. Antimicrob Agents Chemother. 1999;43:1651–6.PubMedPubMedCentralCrossRef
27.
Zurück zum Zitat Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010;8:e1000412.CrossRefPubMedPubMedCentral Kilkenny C, Browne WJ, Cuthill IC, Emerson M, Altman DG. Improving bioscience research reporting: the ARRIVE guidelines for reporting animal research. PLoS Biol. 2010;8:e1000412.CrossRefPubMedPubMedCentral
28.
Zurück zum Zitat Mook-Kanamori B, Geldhoff M, Troost D, van der Poll T, van de Beek D. Characterization of a pneumococcal meningitis mouse model. BMC Infect Dis. 2012;12:71.CrossRefPubMedPubMedCentral Mook-Kanamori B, Geldhoff M, Troost D, van der Poll T, van de Beek D. Characterization of a pneumococcal meningitis mouse model. BMC Infect Dis. 2012;12:71.CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Engelen-Lee JY, Brouwer MC, Aronica E, van de Beek D. Pneumococcal meningitis: clinical-pathological correlations (meningene-path). Acta Neuropathol Commun. 2016;4:26.CrossRefPubMedPubMedCentral Engelen-Lee JY, Brouwer MC, Aronica E, van de Beek D. Pneumococcal meningitis: clinical-pathological correlations (meningene-path). Acta Neuropathol Commun. 2016;4:26.CrossRefPubMedPubMedCentral
30.
Zurück zum Zitat van de Beek D, Cabellos C, Dzupova O, Esposito S, Klein M, Kloek AT, Leib SL, Mourvillier B, Ostergaard C, Pagliano P, et al. ESCMID guideline: diagnosis and treatment of acute bacterial meningitis. Clin Microbiol Infect. 2016;22(Suppl 3):S37–62.CrossRefPubMed van de Beek D, Cabellos C, Dzupova O, Esposito S, Klein M, Kloek AT, Leib SL, Mourvillier B, Ostergaard C, Pagliano P, et al. ESCMID guideline: diagnosis and treatment of acute bacterial meningitis. Clin Microbiol Infect. 2016;22(Suppl 3):S37–62.CrossRefPubMed
31.
Zurück zum Zitat Koopmans MM, Bijlsma MW, Brouwer MC, van de Beek D, van der Ende A. Listeria monocytogenes meningitis in the Netherlands, 1985-2014: a nationwide surveillance study. J Inf. 2017;75:12–9. Koopmans MM, Bijlsma MW, Brouwer MC, van de Beek D, van der Ende A. Listeria monocytogenes meningitis in the Netherlands, 1985-2014: a nationwide surveillance study. J Inf. 2017;75:12–9.
32.
Zurück zum Zitat Ragon M, Wirth T, Hollandt F, Lavenir R, Lecuit M, Le Monnier A, Brisse S. A new perspective on listeria monocytogenes evolution. PLoS Pathog. 2008;4:e1000146.CrossRefPubMedPubMedCentral Ragon M, Wirth T, Hollandt F, Lavenir R, Lecuit M, Le Monnier A, Brisse S. A new perspective on listeria monocytogenes evolution. PLoS Pathog. 2008;4:e1000146.CrossRefPubMedPubMedCentral
33.
Zurück zum Zitat Becavin C, Bouchier C, Lechat P, Archambaud C, Creno S, Gouin E, Wu Z, Kuhbacher A, Brisse S, Pucciarelli MG, et al. Comparison of widely used listeria monocytogenes strains EGD, 10403S, and EGD-e highlights genomic variations underlying differences in pathogenicity. MBio. 2014;5:e00969–14.CrossRefPubMedPubMedCentral Becavin C, Bouchier C, Lechat P, Archambaud C, Creno S, Gouin E, Wu Z, Kuhbacher A, Brisse S, Pucciarelli MG, et al. Comparison of widely used listeria monocytogenes strains EGD, 10403S, and EGD-e highlights genomic variations underlying differences in pathogenicity. MBio. 2014;5:e00969–14.CrossRefPubMedPubMedCentral
34.
Zurück zum Zitat Barbour AH, Rampling A, Hormaeche CE. Comparison of the infectivity of isolates of listeria monocytogenes following intragastric and intravenous inoculation in mice. Microb Pathog. 1996;20:247–53.CrossRefPubMed Barbour AH, Rampling A, Hormaeche CE. Comparison of the infectivity of isolates of listeria monocytogenes following intragastric and intravenous inoculation in mice. Microb Pathog. 1996;20:247–53.CrossRefPubMed
35.
Zurück zum Zitat Lecuit M, Cossart P. Genetically-modified-animal models for human infections: the listeria paradigm. Trends Mol Med. 2002;8:537–42.CrossRefPubMed Lecuit M, Cossart P. Genetically-modified-animal models for human infections: the listeria paradigm. Trends Mol Med. 2002;8:537–42.CrossRefPubMed
36.
37.
Zurück zum Zitat Uldry PA, Kuntzer T, Bogousslavsky J, Regli F, Miklossy J, Bille J, Francioli P, Janzer R. Early symptoms and outcome of listeria monocytogenes rhombencephalitis: 14 adult cases. J Neurol. 1993;240:235–42.CrossRefPubMed Uldry PA, Kuntzer T, Bogousslavsky J, Regli F, Miklossy J, Bille J, Francioli P, Janzer R. Early symptoms and outcome of listeria monocytogenes rhombencephalitis: 14 adult cases. J Neurol. 1993;240:235–42.CrossRefPubMed
38.
Zurück zum Zitat Prats N, Briones V, Blanco MM, Altimira J, Ramos JA, Dominguez L, Marco A. Choroiditis and meningitis in experimental murine infection with listeria monocytogenes. Eur J Clin Microbiol Infect Dis. 1992;11:744–7.CrossRefPubMed Prats N, Briones V, Blanco MM, Altimira J, Ramos JA, Dominguez L, Marco A. Choroiditis and meningitis in experimental murine infection with listeria monocytogenes. Eur J Clin Microbiol Infect Dis. 1992;11:744–7.CrossRefPubMed
39.
Zurück zum Zitat Mook-Kanamori BB, Fritz D, Brouwer MC, van der Ende A, van de Beek D. Intracerebral hemorrhages in adults with community associated bacterial meningitis in adults: should we reconsider anticoagulant therapy? PLoS One. 2012;7:e45271.CrossRefPubMedPubMedCentral Mook-Kanamori BB, Fritz D, Brouwer MC, van der Ende A, van de Beek D. Intracerebral hemorrhages in adults with community associated bacterial meningitis in adults: should we reconsider anticoagulant therapy? PLoS One. 2012;7:e45271.CrossRefPubMedPubMedCentral
40.
Zurück zum Zitat Winkler F, Kastenbauer S, Koedel U, Pfister HW. Role of the urokinase plasminogen activator system in patients with bacterial meningitis. Neurology. 2002;59:1350–5.CrossRefPubMed Winkler F, Kastenbauer S, Koedel U, Pfister HW. Role of the urokinase plasminogen activator system in patients with bacterial meningitis. Neurology. 2002;59:1350–5.CrossRefPubMed
41.
Zurück zum Zitat Mook-Kanamori BB, Geldhoff M, van der Poll T, van de Beek D. Pathogenesis and pathophysiology of pneumococcal meningitis. Clin Microbiol Rev. 2011;24:557–91.CrossRefPubMedPubMedCentral Mook-Kanamori BB, Geldhoff M, van der Poll T, van de Beek D. Pathogenesis and pathophysiology of pneumococcal meningitis. Clin Microbiol Rev. 2011;24:557–91.CrossRefPubMedPubMedCentral
42.
Zurück zum Zitat Vergouwen MD, Schut ES, Troost D, van de Beek D. Diffuse cerebral intravascular coagulation and cerebral infarction in pneumococcal meningitis. Neurocrit Care. 2010;13:217–27.CrossRefPubMed Vergouwen MD, Schut ES, Troost D, van de Beek D. Diffuse cerebral intravascular coagulation and cerebral infarction in pneumococcal meningitis. Neurocrit Care. 2010;13:217–27.CrossRefPubMed
43.
Zurück zum Zitat Kowalik MM, Smiatacz T, Hlebowicz M, Pajuro R, Trocha H. Coagulation, coma, and outcome in bacterial meningitis--an observational study of 38 adult cases. J Inf Secur. 2007;55:141–8. Kowalik MM, Smiatacz T, Hlebowicz M, Pajuro R, Trocha H. Coagulation, coma, and outcome in bacterial meningitis--an observational study of 38 adult cases. J Inf Secur. 2007;55:141–8.
44.
Zurück zum Zitat Weisfelt M, Determann RM, de Gans J, van der Ende A, Levi M, van de Beek D, Schultz MJ. Procoagulant and fibrinolytic activity in cerebrospinal fluid from adults with bacterial meningitis. J Inf Secur. 2007;54:545–50. Weisfelt M, Determann RM, de Gans J, van der Ende A, Levi M, van de Beek D, Schultz MJ. Procoagulant and fibrinolytic activity in cerebrospinal fluid from adults with bacterial meningitis. J Inf Secur. 2007;54:545–50.
45.
Zurück zum Zitat Drevets DA, Bronze MS. Listeria monocytogenes: epidemiology, human disease, and mechanisms of brain invasion. FEMS Immunol Med Microbiol. 2008;53:151–65.CrossRefPubMed Drevets DA, Bronze MS. Listeria monocytogenes: epidemiology, human disease, and mechanisms of brain invasion. FEMS Immunol Med Microbiol. 2008;53:151–65.CrossRefPubMed
46.
Zurück zum Zitat Havell EA, Sehgal PB. Tumor necrosis factor-independent IL-6 production during murine listeriosis. J Immunol. 1991;146:756–61.PubMed Havell EA, Sehgal PB. Tumor necrosis factor-independent IL-6 production during murine listeriosis. J Immunol. 1991;146:756–61.PubMed
47.
Zurück zum Zitat Huang S, Hendriks W, Althage A, Hemmi S, Bluethmann H, Kamijo R, Vilcek J, Zinkernagel RM, Aguet M. Immune response in mice that lack the interferon-gamma receptor. Science. 1993;259:1742–5.CrossRefPubMed Huang S, Hendriks W, Althage A, Hemmi S, Bluethmann H, Kamijo R, Vilcek J, Zinkernagel RM, Aguet M. Immune response in mice that lack the interferon-gamma receptor. Science. 1993;259:1742–5.CrossRefPubMed
48.
Zurück zum Zitat Rothe J, Lesslauer W, Lotscher H, Lang Y, Koebel P, Kontgen F, Althage A, Zinkernagel R, Steinmetz M, Bluethmann H. Mice lacking the tumour necrosis factor receptor 1 are resistant to TNF-mediated toxicity but highly susceptible to infection by listeria monocytogenes. Nature. 1993;364:798–802.CrossRefPubMed Rothe J, Lesslauer W, Lotscher H, Lang Y, Koebel P, Kontgen F, Althage A, Zinkernagel R, Steinmetz M, Bluethmann H. Mice lacking the tumour necrosis factor receptor 1 are resistant to TNF-mediated toxicity but highly susceptible to infection by listeria monocytogenes. Nature. 1993;364:798–802.CrossRefPubMed
49.
Zurück zum Zitat Pfeffer K, Matsuyama T, Kundig TM, Wakeham A, Kishihara K, Shahinian A, Wiegmann K, Ohashi PS, Kronke M, Mak TW. Mice deficient for the 55 kd tumor necrosis factor receptor are resistant to endotoxic shock, yet succumb to L. monocytogenes infection. Cell. 1993;73:457–67. Pfeffer K, Matsuyama T, Kundig TM, Wakeham A, Kishihara K, Shahinian A, Wiegmann K, Ohashi PS, Kronke M, Mak TW. Mice deficient for the 55 kd tumor necrosis factor receptor are resistant to endotoxic shock, yet succumb to L. monocytogenes infection. Cell. 1993;73:457–67.
50.
Zurück zum Zitat Guo Y, Niesel DW, Ziegler HK, Klimpel GR. Listeria monocytogenes activation of human peripheral blood lymphocytes: induction of non-major histocompatibility complex-restricted cytotoxic activity and cytokine production. Infect Immun. 1992;60:1813–9.PubMedPubMedCentral Guo Y, Niesel DW, Ziegler HK, Klimpel GR. Listeria monocytogenes activation of human peripheral blood lymphocytes: induction of non-major histocompatibility complex-restricted cytotoxic activity and cytokine production. Infect Immun. 1992;60:1813–9.PubMedPubMedCentral
51.
Zurück zum Zitat Gregory SH, Jiang X, Wing EJ. Lymphokine-activated killer cells lyse listeria-infected hepatocytes and produce elevated quantities of interferon-gamma. J Infect Dis. 1996;174:1073–9.CrossRefPubMed Gregory SH, Jiang X, Wing EJ. Lymphokine-activated killer cells lyse listeria-infected hepatocytes and produce elevated quantities of interferon-gamma. J Infect Dis. 1996;174:1073–9.CrossRefPubMed
52.
Zurück zum Zitat Buchmeier NA, Schreiber RD. Requirement of endogenous interferon-gamma production for resolution of listeria monocytogenes infection. Proc Natl Acad Sci U S A. 1985;82:7404–8.CrossRefPubMedPubMedCentral Buchmeier NA, Schreiber RD. Requirement of endogenous interferon-gamma production for resolution of listeria monocytogenes infection. Proc Natl Acad Sci U S A. 1985;82:7404–8.CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Pappu R, Ramirez-Carrozzi V, Ota N, Ouyang W, Hu Y. The IL-17 family cytokines in immunity and disease. J Clin Immunol. 2010;30:185–95.CrossRefPubMed Pappu R, Ramirez-Carrozzi V, Ota N, Ouyang W, Hu Y. The IL-17 family cytokines in immunity and disease. J Clin Immunol. 2010;30:185–95.CrossRefPubMed
54.
55.
Zurück zum Zitat Couper KN, Blount DG, Riley EM. IL-10: the master regulator of immunity to infection. J Immunol. 2008;180:5771–7.CrossRefPubMed Couper KN, Blount DG, Riley EM. IL-10: the master regulator of immunity to infection. J Immunol. 2008;180:5771–7.CrossRefPubMed
56.
Zurück zum Zitat Dai WJ, Kohler G, Brombacher F. Both innate and acquired immunity to listeria monocytogenes infection are increased in IL-10-deficient mice. J Immunol. 1997;158:2259–67.PubMed Dai WJ, Kohler G, Brombacher F. Both innate and acquired immunity to listeria monocytogenes infection are increased in IL-10-deficient mice. J Immunol. 1997;158:2259–67.PubMed
57.
Zurück zum Zitat Wagner RD, Maroushek NM, Brown JF, Czuprynski CJ. Treatment with anti-interleukin-10 monoclonal antibody enhances early resistance to but impairs complete clearance of listeria monocytogenes infection in mice. Infect Immun. 1994;62:2345–53.PubMedPubMedCentral Wagner RD, Maroushek NM, Brown JF, Czuprynski CJ. Treatment with anti-interleukin-10 monoclonal antibody enhances early resistance to but impairs complete clearance of listeria monocytogenes infection in mice. Infect Immun. 1994;62:2345–53.PubMedPubMedCentral
58.
Zurück zum Zitat Auerbuch V, Brockstedt DG, Meyer-Morse N, O'Riordan M, Portnoy DA. Mice lacking the type I interferon receptor are resistant to listeria monocytogenes. J Exp Med. 2004;200:527–33.CrossRefPubMedPubMedCentral Auerbuch V, Brockstedt DG, Meyer-Morse N, O'Riordan M, Portnoy DA. Mice lacking the type I interferon receptor are resistant to listeria monocytogenes. J Exp Med. 2004;200:527–33.CrossRefPubMedPubMedCentral
59.
Zurück zum Zitat Carrero JA, Calderon B, Unanue ER. Type I interferon sensitizes lymphocytes to apoptosis and reduces resistance to listeria infection. J Exp Med. 2004;200:535–40.CrossRefPubMedPubMedCentral Carrero JA, Calderon B, Unanue ER. Type I interferon sensitizes lymphocytes to apoptosis and reduces resistance to listeria infection. J Exp Med. 2004;200:535–40.CrossRefPubMedPubMedCentral
60.
Zurück zum Zitat O'Connell RM, Saha SK, Vaidya SA, Bruhn KW, Miranda GA, Zarnegar B, Perry AK, Nguyen BO, Lane TF, Taniguchi T, et al. Type I interferon production enhances susceptibility to Listeria monocytogenes infection. J Exp Med. 2004;200:437–45.CrossRefPubMedPubMedCentral O'Connell RM, Saha SK, Vaidya SA, Bruhn KW, Miranda GA, Zarnegar B, Perry AK, Nguyen BO, Lane TF, Taniguchi T, et al. Type I interferon production enhances susceptibility to Listeria monocytogenes infection. J Exp Med. 2004;200:437–45.CrossRefPubMedPubMedCentral
61.
Zurück zum Zitat Koopmans MM, Brouwer MC, Geldhoff M, Seron MV, Houben J, van der Ende A, van de Beek D. Cerebrospinal fluid inflammatory markers in patients with listeria monocytogenes meningitis. BBA Clin. 2014;1:44–51.CrossRefPubMedPubMedCentral Koopmans MM, Brouwer MC, Geldhoff M, Seron MV, Houben J, van der Ende A, van de Beek D. Cerebrospinal fluid inflammatory markers in patients with listeria monocytogenes meningitis. BBA Clin. 2014;1:44–51.CrossRefPubMedPubMedCentral
62.
Zurück zum Zitat Kremer PH, Lees JA, Koopmans MM, Ferwerda B, Arends AW, Feller MM, Schipper K, Valls Seron M, van der Ende A, Brouwer MC, et al. Benzalkonium tolerance genes and outcome in listeria monocytogenes meningitis. Clin Microbiol Infect. 2017;23:265.e1-265.e7. Kremer PH, Lees JA, Koopmans MM, Ferwerda B, Arends AW, Feller MM, Schipper K, Valls Seron M, van der Ende A, Brouwer MC, et al. Benzalkonium tolerance genes and outcome in listeria monocytogenes meningitis. Clin Microbiol Infect. 2017;23:265.e1-265.e7.
63.
64.
Zurück zum Zitat Drevets DA, Sawyer RT, Potter TA, Campbell PA. Listeria monocytogenes infects human endothelial cells by two distinct mechanisms. Infect Immun. 1995;63:4268–76.PubMedPubMedCentral Drevets DA, Sawyer RT, Potter TA, Campbell PA. Listeria monocytogenes infects human endothelial cells by two distinct mechanisms. Infect Immun. 1995;63:4268–76.PubMedPubMedCentral
65.
Zurück zum Zitat Wilson SL, Drevets DA. Listeria monocytogenes infection and activation of human brain microvascular endothelial cells. J Infect Dis. 1998;178:1658–66.CrossRefPubMed Wilson SL, Drevets DA. Listeria monocytogenes infection and activation of human brain microvascular endothelial cells. J Infect Dis. 1998;178:1658–66.CrossRefPubMed
66.
Zurück zum Zitat Blanot S, Joly MM, Vilde F, Jaubert F, Clement O, Frija G, Berche P. A gerbil model for rhombencephalitis due to listeria monocytogenes. Microb Pathog. 1997;23:39–48.CrossRefPubMed Blanot S, Joly MM, Vilde F, Jaubert F, Clement O, Frija G, Berche P. A gerbil model for rhombencephalitis due to listeria monocytogenes. Microb Pathog. 1997;23:39–48.CrossRefPubMed
67.
Zurück zum Zitat Dramsi S, Dehoux P, Lebrun M, Goossens PL, Cossart P. Identification of four new members of the internalin multigene family of listeria monocytogenes EGD. Infect Immun. 1997;65:1615–25.PubMedPubMedCentral Dramsi S, Dehoux P, Lebrun M, Goossens PL, Cossart P. Identification of four new members of the internalin multigene family of listeria monocytogenes EGD. Infect Immun. 1997;65:1615–25.PubMedPubMedCentral
68.
Zurück zum Zitat Ghosh P, Halvorsen EM, Ammendolia DA, Mor-Vaknin N, O'Riordan MXD, Brumell JH, Markovitz DM, Higgins DE. Invasion of the brain by listeria monocytogenes is mediated by InlF and host cell Vimentin. MBio. 2018;9 Ghosh P, Halvorsen EM, Ammendolia DA, Mor-Vaknin N, O'Riordan MXD, Brumell JH, Markovitz DM, Higgins DE. Invasion of the brain by listeria monocytogenes is mediated by InlF and host cell Vimentin. MBio. 2018;9
69.
Zurück zum Zitat Moura A, Criscuolo A, Pouseele H, Maury MM, Leclercq A, Tarr C, Bjorkman JT, Dallman T, Reimer A, Enouf V, et al. Whole genome-based population biology and epidemiological surveillance of listeria monocytogenes. Nat Microbiol. 2016;2:16185.CrossRefPubMed Moura A, Criscuolo A, Pouseele H, Maury MM, Leclercq A, Tarr C, Bjorkman JT, Dallman T, Reimer A, Enouf V, et al. Whole genome-based population biology and epidemiological surveillance of listeria monocytogenes. Nat Microbiol. 2016;2:16185.CrossRefPubMed
70.
Zurück zum Zitat Lee S, Ward TJ, Jima DD, Parsons C, Kathariou S. The arsenic resistance-associated listeria genomic island LGI2 exhibits sequence and integration site diversity and a propensity for three listeria monocytogenes clones with enhanced virulence. Appl Environ Microbiol. 2017;83. Lee S, Ward TJ, Jima DD, Parsons C, Kathariou S. The arsenic resistance-associated listeria genomic island LGI2 exhibits sequence and integration site diversity and a propensity for three listeria monocytogenes clones with enhanced virulence. Appl Environ Microbiol. 2017;83.
71.
72.
Zurück zum Zitat Kasanmoentalib ES, Valls Seron M, Ferwerda B, Tanck MW, Zwinderman AH, Baas F, van der Ende A, Brouwer MC, van de Beek D. Mannose-binding lectin-associated serine protease 2 (MASP-2) contributes to poor disease outcome in humans and mice with pneumococcal meningitis. J Neuroinflammation. 2017;14:2.CrossRefPubMedPubMedCentral Kasanmoentalib ES, Valls Seron M, Ferwerda B, Tanck MW, Zwinderman AH, Baas F, van der Ende A, Brouwer MC, van de Beek D. Mannose-binding lectin-associated serine protease 2 (MASP-2) contributes to poor disease outcome in humans and mice with pneumococcal meningitis. J Neuroinflammation. 2017;14:2.CrossRefPubMedPubMedCentral
73.
Zurück zum Zitat Mook-Kanamori BB, Valls Seron M, Geldhoff M, Havik SR, van der Ende A, Baas F, van der Poll T, Meijers JC, B PM, Brouwer MC, van de Beek D. Thrombin-activatable fibrinolysis inhibitor influences disease severity in humans and mice with pneumococcal meningitis. J Thromb Haemost. 2015;13:2076–86.CrossRefPubMed Mook-Kanamori BB, Valls Seron M, Geldhoff M, Havik SR, van der Ende A, Baas F, van der Poll T, Meijers JC, B PM, Brouwer MC, van de Beek D. Thrombin-activatable fibrinolysis inhibitor influences disease severity in humans and mice with pneumococcal meningitis. J Thromb Haemost. 2015;13:2076–86.CrossRefPubMed
74.
Zurück zum Zitat Geldhoff M, Mook-Kanamori BB, Brouwer MC, Troost D, Leemans JC, Flavell RA, Van der Ende A, Van der Poll T, Van de Beek D. Inflammasome activation mediates inflammation and outcome in humans and mice with pneumococcal meningitis. BMC Infect Dis. 2013;13:358.CrossRefPubMedPubMedCentral Geldhoff M, Mook-Kanamori BB, Brouwer MC, Troost D, Leemans JC, Flavell RA, Van der Ende A, Van der Poll T, Van de Beek D. Inflammasome activation mediates inflammation and outcome in humans and mice with pneumococcal meningitis. BMC Infect Dis. 2013;13:358.CrossRefPubMedPubMedCentral
Metadaten
Titel
Characterization of a Listeria monocytogenes meningitis mouse model
verfasst von
Merel M. Koopmans
JooYeon Engelen-Lee
Matthijs C. Brouwer
Valery Jaspers
Wing Kit Man
Mercedes Vall Seron
Diederik van de Beek
Publikationsdatum
01.12.2018
Verlag
BioMed Central
Erschienen in
Journal of Neuroinflammation / Ausgabe 1/2018
Elektronische ISSN: 1742-2094
DOI
https://doi.org/10.1186/s12974-018-1293-3

Weitere Artikel der Ausgabe 1/2018

Journal of Neuroinflammation 1/2018 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Schützt Olivenöl vor dem Tod durch Demenz?

10.05.2024 Morbus Alzheimer Nachrichten

Konsumieren Menschen täglich 7 Gramm Olivenöl, ist ihr Risiko, an einer Demenz zu sterben, um mehr als ein Vierten reduziert – und dies weitgehend unabhängig von ihrer sonstigen Ernährung. Dafür sprechen Auswertungen zweier großer US-Studien.

Darf man die Behandlung eines Neonazis ablehnen?

08.05.2024 Gesellschaft Nachrichten

In einer Leseranfrage in der Zeitschrift Journal of the American Academy of Dermatology möchte ein anonymer Dermatologe bzw. eine anonyme Dermatologin wissen, ob er oder sie einen Patienten behandeln muss, der eine rassistische Tätowierung trägt.

Chirurginnen und Chirurgen sind stark suizidgefährdet

07.05.2024 Suizid Nachrichten

Der belastende Arbeitsalltag wirkt sich negativ auf die psychische Gesundheit der Angehörigen ärztlicher Berufsgruppen aus. Chirurginnen und Chirurgen bilden da keine Ausnahme, im Gegenteil.

Ein Drittel der jungen Ärztinnen und Ärzte erwägt abzuwandern

07.05.2024 Klinik aktuell Nachrichten

Extreme Arbeitsverdichtung und kaum Supervision: Dr. Andrea Martini, Sprecherin des Bündnisses Junge Ärztinnen und Ärzte (BJÄ) über den Frust des ärztlichen Nachwuchses und die Vorteile des Rucksack-Modells.