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

Open Access 01.12.2018 | Research

New insights into meningitic Escherichia coli infection of brain microvascular endothelial cells from quantitative proteomics analysis

verfasst von: Wen-Tong Liu, Yu-Jin Lv, Rui-Cheng Yang, Ji-Yang Fu, Lu Liu, Huan Wang, Qi Cao, Chen Tan, Huan-Chun Chen, Xiang-Ru Wang

Erschienen in: Journal of Neuroinflammation | Ausgabe 1/2018

Abstract

Background

Bacterial meningitis remains a big threat to the integrity of the central nervous system (CNS), despite the advancements in antimicrobial reagents. Escherichia coli is a bacterial pathogen that can disrupt the CNS function, especially in neonates. E. coli meningitis occurs after bacteria invade the brain microvascular endothelial cells (BMECs) that form a direct and essential barrier restricting the entry of circulating microbes and toxins to the brain. Previous studies have reported on several cellular proteins that function during meningitic E. coli infections; however, more comprehensive investigations to elucidate the potential targets involved in E. coli meningitis are essential to better understand this disease and discover new treatments for it.

Methods

The isobaric tags for relative and absolute quantification (iTRAQ) approach coupled with LC-MS/MS were applied to compare and characterize the different proteomic profiles of BMECs in response to meningitic or non-meningitic E. coli strains. KEGG and gene ontology annotations, ingenuity pathways analysis, and functional experiments were combined to identify the key host molecules involved in the meningitic E. coli-induced tight junction breakdown and neuroinflammatory responses.

Results

A total of 13 cellular proteins were found to be differentially expressed by meningitic E. coli strains PCN033 and RS218, including one that was also affected by HB101, a non-meningitic E. coli strain. Through bioinformatics analysis, we identified the macrophage migration inhibitory factor (MIF), granzyme A, NF-κB signaling, and mitogen-activated protein kinase (MAPK) pathways as being biologically involved in the meningitic E. coli-induced tight junction breakdown and neuroinflammation. Functionally, we showed that MIF facilitated meningitic E. coli-induced production of cytokines and chemokines and also helped to disrupt the blood-brain barrier by decreasing the expression of tight junction proteins like ZO-1, occludin. Moreover, we demonstrated the significant activation of NF-κB and MAPK signaling in BMECs in response to meningitic E. coli strains, which dominantly determined the generation of the proinflammatory cytokines including IL-6, IL-8, TNF-α, and IL-1β.

Conclusions

Our work identified 12 host cellular targets that are affected by meningitic E. coli strains and revealed MIF to be an important contributor to meningitic E. coli-induced cytokine production and tight junction disruption, and also the NF-κB and MAPK signaling pathways that are mainly involved in the infection-induced cytokines production. Characterization of these distinct proteins and pathways in BMECs will facilitate further elucidation of meningitis-causing mechanisms in humans and animals, thereby enabling the development of novel preventative and therapeutic strategies against infection with meningitic E. coli.
Hinweise

Electronic supplementary material

The online version of this article (https://​doi.​org/​10.​1186/​s12974-018-1325-z) contains supplementary material, which is available to authorized users.
Wen-Tong Liu and Yu-Jin Lv contributed equally to this work.
Abkürzungen
BBB
Blood-brain barrier
BMECs
Brain microvascular endothelial cells
CNS
Central nervous system
CSF
Cerebrospinal fluid
DEPs
Differentially expressed proteins
DMD
Dystrophin
E. coli
Escherichia coli
ECIS
Electric cell-substrate impedance sensing
EGFR
Epidermal growth factor receptor
ERK1/2
Extracellular signal-regulated kinases 1 and 2
ExPEC
Extraintestinal pathogenic Escherichia coli
GO
Gene Ontology
ICAM-1
Intercellular adhesion molecule-1
IL-1β
Interleukin 1 beta
IL-8
Interleukin-8
IPA
Ingenuity Pathways Analysis
ISO-1
(S, R)-3-(4-Hydroxyphenyl)-4, 5-dihydro-5-isoxazole acetic acid methyl ester DAPI
4′-6-Diamidino-2-phenylindole
iTRAQ
Isobaric tags for relative and absolute quantification
JNK
c-Jun N-terminal kinase
KEGG
Kyoto encyclopedia of genes and genomes
LC-MS/MS
Liquid chromatography tandem mass spectrometry
LGMN
Legumain
MAPK
Mitogen-activated protein kinase
MIF
Macrophage migration inhibitory factor
NF-κB
Nuclear factor-κB
NOS
Nitric oxide synthase
PI3K
Phosphatidylinositol 3-kinase
S1P
Sphingosine-1-phosphate
SCX
Strong cation exchange chromatography
TBPL1
TATA box-binding protein-like protein 1
TEAB
Tetraethyl-ammonium bromide
TEER
Trans-endothelial electric resistance
TNF-α
Tumor necrosis factor-alpha
VEGFA
Vascular endothelial growth factor A
ZO-1
Zonula occludens-1, IL-6, interleukin-6

Background

Bacterial meningitis is a severe, life-threatening infection of the central nervous system (CNS) with high morbidity and mortality. It is currently recognized as one of the top ten killers in infection-related deaths worldwide, with almost half of the survivors suffering from diverse neurological sequelae (e.g., mental retardation, hearing impairment and blindness), despite the advancements made in the field of antimicrobial treatment [13]. Most bacterial meningitis cases are initiated by hematogenous spread and develop when the circulating bacteria penetrate the blood-brain barrier (BBB), destroy brain parenchyma, and finally cause CNS disorders [1]. Among the meningitis-causing microbes, extraintestinal pathogenic Escherichia coli (ExPEC) has recently emerged as an important zoonotic bacterial pathogen with the potential to colonize multiple tissues outside the intestine and cause severe infections, with one typical outcome being meningitis. The evidence from recent in vivo and in vitro studies indicates that meningitic E. coli strains possess the ability to invade the brain, and the infection-induced BBB disruption that occurs is the hallmark event in the development of E. coli meningitis [4, 5].
The availability of in vitro and in vivo BBB infection models has made the study of meningitic E. coli penetration of the brain possible [69]. The in vitro BBB model uses brain microvascular endothelial cells (BMECs) that form distinctive tight junctions and exhibit high trans-endothelial electrical resistance, thereby mimicking the features of the natural in vivo barrier that protects the brain from circulating microorganisms and toxins [1013]. The in vivo model is established by inducing experimental hematogenous meningitis in newborn rats and mice [9, 14, 15]. With these models, it is now well-established that successful traversal of the BBB by circulating E. coli strains requires the following prerequisites: a high bacteremia, binding to and invasion of BMECs, rearrangement of actin cytoskeleton, and crossing the BBB as live bacteria [1, 2]. These require a series of complicated interactions between meningitic E. coli and the host. So far, several host targets have been found to be associated with this invasion process, including certain intracellular signaling molecules like focal adhesion kinase, phosphatidylinositol 3-kinase (PI3K), Rho GTPases, cytosolic phospholipase A2, nuclear factor-κB (NF-κB), inducible nitric oxide synthase (NOS), and several cellular surface molecules/receptors such as caveolin-1, Toll-like receptors, the intercellular adhesion molecule (ICAM-1), and some actin-binding molecules like ERM family proteins (ezrin, radixin, and moesin), most likely through their influences on the aforementioned prerequisites [8, 1619]. We have previously identified and characterized two essential cellular targets, S1P and EGFR, which are exploited by meningitic E. coli for successful invasion of the BBB [20]. In other work, we have also found that vascular endothelial growth factor A (VEGFA) and Snail-1, which are inducible by meningitic E. coli, can mediate the BBB disruption [5]. Despite these advances, the mechanisms involved in CNS infection by meningitic E. coli are still poorly understood, and a more comprehensive investigation to elucidate the cellular targets in infected BMECs is now required.
In the current study, we compared the different proteomic profiles of BMECs in response to meningitic and non-meningitic E. coli strains via the isobaric tags for relative and absolute quantification (iTRAQ) approach and investigated the potential host factors and mechanisms that were hijacked by meningitic E. coli to penetrate the BBB. Characterization of these potential host targets will expand our current knowledge on meningitic E. coli-induced CNS infections and provide new strategies to prevent this infection and develop novel therapeutic reagents against it.

Methods

Bacterial strains, cell culture, and infection

The E. coli K1 strain RS218 (O18:K1:H7) [GenBank: CP007149.1], whose genomic sequencing has been finalized and annotated, is a well-characterized cerebrospinal fluid (CSF) isolate from a neonatal meningitis case [21]. The porcine-originated ExPEC strain PCN033 (O11: K2) [GenBank: CP006632.1], which was isolated from swine CSF in China [22, 23], is evidenced to be highly virulent and capable of invading and disrupting the BBB, thereby causing CNS dysfunction [5, 24]. E. coli K12 strain HB101 is an avirulent and non-meningitic strain normally used as a negative control strain [25, 26]. All E. coli strains were grown aerobically at 37 °C in Luria–Bertani medium unless otherwise specified.
The immortalized human BMECs (hereafter called hBMECs) were kindly provided by Prof. Kwang Sik Kim in Johns Hopkins University School of Medicine and routinely cultured in RPMI 1640 supplemented with 10% fetal bovine serum, 2 mM L-glutamine, 1 mM sodium pyruvate, essential amino acids, nonessential amino acids, vitamins, and penicillin and streptomycin (100 U/mL) in a 37 °C incubator under 5% CO2 until monolayer confluence was reached [20, 27]. Confluent cells were washed with Hank’s balanced salt solution (Corning Cellgro, Manassas, VA, USA) and starved in serum-free medium for 16–18 h before further treatment. For bacterial challenge, the cells were infected with E. coli PCN033, RS218, or HB101 strains each at a multiplicity of infection of 10 for 2 h. In some assays, the cells were pretreated with specific inhibitors prior to bacterial challenge.

Reagents, antibodies, and inhibitors

The p38 inhibitor SB202190, extracellular signal-regulated kinases 1 and 2 (ERK1/2) inhibitor U0126, c-Jun N-terminal kinase (JNK) inhibitor SP600125, NF-κB inhibitor BAY11-7082, and (S, R)-3-(4-hydroxyphenyl)-4, 5-dihydro-5-isoxazole acetic acid methyl ester (ISO-1), an inhibitor of macrophage migration inhibitory factor (MIF), were purchased from MedChem Express (Monmouth, NJ, USA). Recombinant MIF protein was purchased from Novoprotein (Summit, NJ, USA). The nucleic acid dye, 4′-6-diamidino-2-phenylindole (DAPI), was obtained from Solarbio (Beijing, China). Anti-ZO-1, anti-MIF, anti-TATA box-binding protein-like protein 1 (TBPL1), anti-legumain (LGMN), anti-ERK1/2, and anti-phospho-ERK1/2 antibodies (all rabbit) were purchased from ABclonal (Wuhan, Hubei, China). Anti-occludin, anti-dystrophin (DMD), anti-HISTIHIC, anti-JNK, and anti-p38 mitogen-activated protein kinase (MAPK) antibodies (all rabbit) were purchased from Proteintech (Chicago, IL, USA). Anti-phospho-JNK (rabbit) antibody was from R&D Systems (Minneapolis, MO, USA). Anti-phospho-p38, anti-p65, anti-phospho-p65, and anti-IκBα antibodies (all rabbit) were purchased from Cell Signaling Technology (Danvers, MA, USA). Cy3-labeled goat anti-rabbit antibody was purchased from Beyotime Institute of Biotechnology (Shanghai, China). Anti-GAPDH (mouse) antibody was purchased from Beijing Biodragon Immunotechnologies Co., Ltd. (Beijing, China).

Protein isolation, digestion, and labeling with iTRAQ reagents

Bacterial-infected and non-infected cells in 10 cm dishes were collected 2-h post-infection and gently washed with pre-chilled PBS buffer. The cells were lysed in 1 mL lysis buffer, and the soluble protein fraction was harvested by 5 min of ultrasonication treatment (pulse on 2 s, pulse off 3 s, power 180 W) followed by centrifugation at 20000×g for 30 min at 4 °C, and the protein concentration was determined via the Bradford protein assay method with BSA as the standard substance. The proteins were reduced with 10 mM iodoacetamide at room temperature for 45 min in the dark and then precipitated in acetone at − 20 °C for 3 h. After centrifugation at 20000×g for 20 min, the protein pellet was resuspended and ultrasonicated in pre-chilled 50% (w/v) tetraethyl-ammonium bromide (TEAB) buffer supplemented with 0.1% SDS. The proteins were obtained after centrifugation at 20000×g and their concentrations were measured by Bradford assays.
Subsequently, protein (100 μg) in TEAB buffer was incubated with 3.3 μL of trypsin (1 μg/μL) (Promega, Madison, WI, USA) at 37 °C for 24 h in a sealed tube. The tryptic peptides were lyophilized and dissolved in 50% TEAB buffer, and iTRAQ labeling was performed according to the manufacturer’s instructions (AB Sciex, Foster City, CA, USA). Briefly, one unit of iTRAQ reagent was thawed and reconstituted in 24 μL isopropanol and the peptides were incubated at room temperature for 2 h. The peptides from the control, HB101, PCN033, and RS218 groups were designated 114, 115, 116, and 117, respectively. The labeled samples were then mixed and dried with a rotary vacuum concentrator. The labeling efficiency was examined by mass spectrometry (MS).

Strong cation exchange chromatography (SCX) fractionation and liquid chromatography (LC)–MS/MS analysis

The labeled samples were pooled and purified using an SCX column (Phenomenex, USA), and separated by LC using an LC-20AB HPLC pump system (Shimadzu, Japan). The peptides were then mixed with nine times their volume in buffer A (25% ACN, 10 mM KH2PO4, pH = 3) and loaded onto a 4.6 × 250 mm Ultremex SCX column containing 5-μm particles (Phenomenex). The peptides were eluted at a flow rate of 1 ml/min in a buffer B (25% ACN, 2 M KCL, 10 mM KH2PO4, pH = 3) gradient as follows: 0–5% buffer B for 30 min, 5–30% buffer B for 20 min, 30–50% buffer B for 5 min, 50% buffer B for 5 min, 50–100% buffer B for 5 min, and 100% buffer B for 1 min before equilibrating with buffer A for 10 min prior to the next injection. Next, the eluted peptides were desalted with a Strata X C18 column (100 mm × 75 mm, 5-um particles, 300A aperture) (Phenomenex, Torrance, CA, USA) and vacuum dried. The fractions were then dissolved in aqueous solution containing 0.1% formic acid (FA) and 2% ACN and centrifuged at 12000g for 10 min at 4 °C. Five micrograms supernatant was loaded on an LC-20AD nano HPLC (Shimadzu, Kyoto, Japan) by the autosampler onto a 2 cm C18 trap column (inner diameter 200 μm, Waters), and the peptides were eluted onto a resolving 10 cm analytical C18 column (inner diameter 75 μm, Waters). The mobile phases used were composed of solvent A (0.1% FA and 5% ACN) and solvent B (0.1% FA and 95% ACN). The gradient was run at 400 nL/min for 48 min at 5–80% solvent B, followed by running a linear gradient to 80% for 7 min, maintained at 80% B for 3 min, and finally returned to 5% in 7 min.
The peptides were subjected to nano-electrospray ionization followed by tandem mass spectrometry (MS/MS) in a Q EXACTIVE (Thermo Fisher Scientific, San Jose, CA, USA) coupled to the HPLC. Intact peptides were detected in the Orbitrap at a resolution of 70,000 and a mass range of 350–2000 m/z. Peptides were selected for MS/MS using high-energy collision dissociation (HCD), and ion fragments were detected in the Orbitrap at a resolution of 17,500. The electrospray voltage applied was 1.8 kV. MS/MS analysis was required for the 15 most abundant precursor ions, which were above a threshold ion count of 20,000 in the MS survey scan, including a following dynamic exclusion duration of 15 s.

iTRAQ data analysis

The raw data files acquired from the mass spectrometers were converted into MGF files using 5600 MS Converter. Protein identification and quantification were performed using the Mascot Server (http://​www.​matrixscience.​com/​search_​form_​select.​html) against the Uniprot_2015_human database (Matrix Science, London, UK; version 2.3.0) and Proteome Discoverer 1.3 (Thermo Fisher Scientific Inc.). To reduce the probability of false peptide identification, only peptides with significance scores at the 95% confidence interval as determined by a Mascot probability analysis were included. The quantitative protein ratios were weighted and normalized by the median ratio in Mascot. Statistical significance analyses were evaluated using two-way ANOVA. The proteins were considered to be differentially expressed if the ratio of mean fold change > 1.2 (or < 0.83) with an Exp pr > 0.05 and a Group pr < 0.05 (Exp pr, three-experiment p value; Group pr, group p value; fold change = experiment + group + error).
The Gene Ontology (GO) annotation of the identified proteins was performed via the online GO program (http://​geneontology.​org/). The biological functions, networks, and signaling pathways of the differentially expressed proteins (DEPs) were analyzed with Ingenuity Pathways Analysis (IPA) software (version 7.5, http://​www.​ingenuity.​com) (Additional files 8, 9 and 10).

RNA extraction and quantitative real-time PCR

Total RNA from the uninfected or infected cells was extracted with RNAiso Plus reagent according to the manufacturer’s instructions (TakaRa, Japan). Any genomic DNA contamination was eliminated by DNase I treatment, and the RNA was reverse-transcribed into cDNA using the PrimeScript™ RT reagent kit with gDNA Eraser, following the manufacturer’s instructions (Takara, Japan). Quantitative real-time PCR was performed in triplicate using the Power SYBR Green PCR Master Mix (Applied BioSystems, Foster City, CA, USA). The PCR primers for these experiments are listed in Table 1. The expression levels of the target genes were normalized to GAPDH by the 2−ΔΔCT method.
Table 1
Primers used for real-time PCR in this study
Primers
Nucleotide sequence(5′-3′)
Gene symbol(s)
P1
ACGAATCTCCGACCACT
IL-1β
P2
CCATGGCCACAACAACTGAC
P3
CTCAGCCTCTTCTCCTTC
TNF-α
P4
GGGTTTGCTACAACATGG
P5
CCACTCACCTCTTCAGAA
IL-6
P6
GGCAAGTCTCCTCATTGA
P7
GACATACTCCAAACCTTTCC
IL-8
P8
ATTCTCAGCCCTCTTCAAA
P9
TGCCTCCTGCACCACCAACT
GAPDH
P10
CGCCTGCTTCACCACCTTC

Western blotting

Uninfected and infected hBMECs were collected and lysed in RIPA buffer supplemented with a protease inhibitor cocktail (Sigma-Aldrich, St. Louis, MO, USA) and then sonicated and centrifuged at 10,000×g for 10 min at 4 °C. The soluble protein concentration in the supernatants was measured using the BCA protein assay kit (Beyotime, China). Aliquots from each sample were separated by 12% SDS-PAGE, and then transferred to polyvinylidene difluoride membranes (Bio-Rad, CA, USA). The blots were blocked with 5% BSA in Tris-buffered saline with Tween 20 at room temperature for 1 h and then incubated overnight at 4 °C with primary antibodies against GAPDH, DMD, MIF, HIST1H1C, TBPL1 or LGMN. The blots were subsequently washed and incubated with horseradish peroxidase-conjugated anti-rabbit or anti-mouse IgG at 37 °C for 1 h, and visualized with ECL reagents (Bio-Rad, USA). The blots were densitometrically quantified and analyzed with Image Lab software (Bio-Rad).

Immunofluorescence microscopy

Uninfected and infected hBMECs were fixed with 4% paraformaldehyde and permeabilized with 0.2% Triton X-100. After 2 h of blocking in PBS buffer with 5% BSA, the cells were incubated with the primary antibody (1:100) overnight at 4 °C, washed thrice with PBS, and then incubated with fluorescently labeled anti-mouse or anti-rabbit IgG (1500) for 1 h. Nuclei were stained with DAPI (0.5 μg/mL) for 30 min. Finally, the cells were mounted and then visualized with fluorescence microscopy.

Electric cell substrate impedance sensing (ECIS)

To explore the influence of recombinant MIF on the permeability of the BBB, hBMECs were seeded at 7 × 104 cells on collagen-coated, gold-plated electrodes in 96-well chamber slides (96W1E+) linked to ECIS Zθ equipment (Applied BioPhysics, Troy, NY, USA) and continuously cultured until confluence, and the trans-endothelial electric resistance (TEER) was monitored to reflect the formation of the barrier [28]. After stable maximal TEER was reached, the recombinant human MIF protein was added into the cells at multiple dosages (10, 100, and 200 ng/mL), and the possible TEER alteration of the monolayer cells was automatically recorded by the ECIS system.

Statistical analysis

Data were expressed as the mean ± standard deviation (mean ± SD) from three replicates. Statistical significance of the differences between each group was analyzed by a one-way analysis of variance (ANOVA) or two-way ANOVA embedded in GraphPad Prism, version 6.0 (GraphPad Software Inc., La Jolla, CA, USA). P < 0.05 (*) was considered statistically significant, and p < 0.01 (**), as well as p < 0.001 (***) were all considered extremely significant.

Results

Differential protein profiling of hBMECs in response to E. coli infection

The protein extracts prepared from the hBMECs with or without meningitic E. coli challenge were subjected to the iTRAQ proteomics analysis. The whole work flow was shown in Fig. 1. Approximately 3000 different proteins were identified and quantified by iTRAQ-coupled LC-MS/MS analysis of the hBMECs infected with E. coli HB101, PCN033, or RS218 strains (Additional file 1: Table S1, Additional file 2: Table S2, Additional file 3: Table S3). As shown in Fig. 2a–d, four proteins were identified as being significantly upregulated and two were significantly downregulated upon HB101 infection, six were significantly upregulated, and 72 were significantly downregulated upon PCN033 infection, while 16 significantly upregulated and 27 significantly downregulated proteins were identified in cells challenged with RS218. The details of these differentially expressed proteins (DEPs) are listed in Tables 2, 3, and 4. The meningitic E. coli PCN033 group displayed 65 unique proteins, while the RS218 group displayed 27 unique proteins. They both shared 13 DEPs with 12 of them being distinct proteins in the hBMECs in response to meningitic strains PCN033 and RS218 (Fig. 2e, Table 5). Only one protein, EXOSC4, was shared by the three groups, and it showed a 0.74-, 0.759-, and 0.8-fold decrease in HB101, PCN033 and RS218 groups, respectively (Fig. 2e, Table 5). In contrast, infection with the non-meningitic HB101 strain induced only two unique, differentially altered proteins. Four proteins were shared between HB101 and RS218 groups, and the three of them altered in response to HB101 and RS218 were specific host proteins in both of these human isolates (Fig. 2e).
Table 2
Significantly changed proteins in HB101-infected hBMECs
Accession
Description
MW [kDa]
Fold change
P valuea
P02656
Apolipoprotein C-III
10.8
1.544
0.000144***
Q07020
60S ribosomal protein L18
21.6
1.422
0.000429***
Q96HP4
Oxidoreductase NAD-binding domain-containing protein 1
34.8
2.715
0.002873**
Q9NPD3
Exosome complex component RRP41
26.4
0.737
0.008415**
Q9Y2Q5
Ragulator complex protein LAMTOR2
13.5
0.769
0.038574*
O14556
Glyceraldehyde-3-phosphate dehydrogenase, testis-specific
44.5
2.134
0.000347***
aP < 0.05 (*) was considered significant, and P < 0.01 (**), as well as < 0.001 (***) were all considered extremely significant
Table 3
Significantly changed proteins in PCN033-infected hBMECs
Accession
Description
MW [kDa]
Fold change
P valuea
A6ZKI3
Protein FAM127A
13.2
0.756
0.028947*
O00625
Pirin
32.1
1.228
0.037445*
O43633
Charged multivesicular body protein 2a
25.1
0.758
0.002376**
O43752
Syntaxin-6
29.2
0.79
0.025619*
O60524
Nuclear export mediator factor NEMF
122.9
0.83
0.004194**
O75190
DnaJ homolog subfamily B member 6
36.1
0.799
0.020038*
O75251
NADH dehydrogenase [ubiquinone] iron-sulfur protein 7, mitochondrial
23.5
0.711
0.004822**
O75817
Ribonuclease P protein subunit p20
15.6
0.815
0.017814*
O95229
ZW10 interactor
31.3
0.668
0.003727**
P04004
Vitronectin
54.3
0.808
0.003548**
P07305
Histone H1.0
20.9
0.776
0.008176**
P11532
Dystrophin
426.5
0.691
0.003088**
P14174
Macrophage migration inhibitory factor
12.5
1.486
0.000377***
P16401
Histone H1.5
22.6
0.631
0.000673***
P16402
Histone H1.3
22.3
0.6
0.000896***
P16403
Histone H1.2
21.4
0.572
7.57E−05***
P35251
Replication factor C subunit 1
128.2
0.786
0.013936*
P35527
Keratin, type I cytoskeletal 9
62
0.72
0.001431**
P39060
Collagen alpha-1(XVIII) chain
178.1
0.792
0.031694*
P46013
Antigen KI-67
358.5
0.793
0.005934**
P48651
Phosphatidylserine synthase 1
55.5
0.83
0.000234***
P49585
Choline-phosphate cytidylyltransferase A
41.7
0.608
0.002306**
P50914
60S ribosomal protein L14
23.4
0.71
0.005946**
P52756
RNA-binding protein 5
92.1
0.758
0.005815**
P56377
AP-1 complex subunit sigma-2
18.6
0.765
2.4E−06***
P61966
AP-1 complex subunit sigma-1A
18.7
0.814
4.81E−08***
P62277
40S ribosomal protein S13
17.2
0.792
0.002441**
P62380
TATA box-binding protein-like protein 1
20.9
0.621
0.000503***
Q13625
Apoptosis-stimulating of p53 protein 2
125.5
0.724
0.006481**
Q14241
Transcription elongation factor B polypeptide 3
89.9
0.647
0.006556**
Q14686
Nuclear receptor coactivator 6
219
0.792
0.005234**
Q15388
Mitochondrial import receptor subunit TOM20 homolog
16.3
0.823
0.007395**
Q15629
Translocating chain-associated membrane protein 1
43
0.809
0.023084*
Q17RN3
Protein FAM98C
37.3
0.821
0.010317*
Q4V339
COBW domain-containing protein 6
43.9
0.747
1.2E−05***
Q567U6
Coiled-coil domain-containing protein 93
73.2
0.814
0.001351**
Q5SSJ5
Heterochromatin protein 1-binding protein 3
61.2
0.828
0.000364***
Q6N069
N-alpha-acetyltransferase 16, NatA auxiliary subunit
101.4
0.775
0.001734**
Q709C8
Vacuolar protein sorting-associated protein 13C
422.1
0.799
0.000576***
Q7Z422
SUZ domain-containing protein 1
17
0.808
0.002542**
Q8IXJ9
Putative Polycomb group protein ASXL1
165.3
0.807
0.007037**
Q8N2K0
Monoacylglycerol lipase ABHD12
45.1
0.786
0.0034**
Q8N884
Cyclic GMP-AMP synthase
58.8
0.82
0.013058*
Q8NC44
Protein FAM134A
57.8
0.78
0.010814*
Q8NC60
Nitric oxide-associated protein 1
78.4
0.81
0.013637*
Q8NEY1
Neuron navigator 1
202.3
0.797
0.020924*
Q8TEM1
Nuclear pore membrane glycoprotein 210
205
0.833
0.032212*
Q8WUP2
Filamin-binding LIM protein 1
40.6
0.809
0.002633**
Q8WVV9
Heterogeneous nuclear ribonucleoprotein L-like
60
0.804
0.013638*
Q8WXA3
RUN and FYVE domain-containing protein 2
75
0.744
0.041934*
Q92604
Acyl-CoA:lysophosphatidylglycerol acyltransferase 1
43.1
0.789
0.033787 *
Q96A57
Transmembrane protein 230
13.2
0.786
0.000449***
Q96LB3
Intraflagellar transport protein 74 homolog
69.2
0.543
2.53E−05***
Q96RU3
Formin-binding protein 1
71.3
0.679
2.07E−05***
Q96T37
Putative RNA-binding protein 15
107.1
0.728
0.024086*
Q9GZP8
Immortalization upregulated protein
10.9
1.207
0.032624*
Q9H074
Polyadenylate-binding protein-interacting protein 1
53.5
1.266
0.004395**
Q9H5N1
Rab GTPase-binding effector protein 2
63.5
0.77
0.001156**
Q9H5X1
MIP18 family protein FAM96A
18.3
0.8
0.000118***
Q9HB40
Retinoid-inducible serine carboxypeptidase
50.8
1.215
0.000733***
Q9HC52
Chromobox protein homolog 8
43.4
1.201
0.023152*
Q9NPD3
Exosome complex component RRP41
26.4
0.759
0.000746***
Q9NRY4
Rho GTPase-activating protein 35
170.4
0.792
0.011172*
Q9NS87
Kinesin-like protein KIF15
160.1
0.785
0.010039*
Q9NSP4
Centromere protein M
19.7
0.802
0.021316*
Q9NTI5
Sister chromatid cohesion protein PDS5 homolog B
164.6
0.826
0.003399**
Q9NWU5
39S ribosomal protein L22, mitochondrial
23.6
0.812
0.016677*
Q9NZQ3
NCK-interacting protein with SH3 domain
78.9
0.661
0.00317**
Q9P0V3
SH3 domain-binding protein 4
107.4
0.797
0.001833**
Q9UBL6
Copine-7
70.2
0.823
2.73E−05***
Q9UJW0
Dynactin subunit 4
52.3
0.823
0.012604*
Q9UNP9
Peptidyl-prolyl cis-trans isomerase E
33.4
0.75
0.044207*
Q9Y2R0
Cytochrome c oxidase assembly protein 3 homolog, mitochondrial
11.7
0.792
0.003694**
Q9Y5Y2
Cytosolic Fe-S cluster assembly factor NUBP2
28.8
0.787
0.000891***
Q9Y6I9
Testis-expressed sequence 264 protein
34.2
0.814
0.047637*
Q9Y3Y2
Chromatin target of PRMT1 protein
26.4
0.828
0.008622**
Q9Y4R8
Telomere length regulation protein TEL2 homolog
91.7
0.735
0.013443*
P10412
Histone H1.4
21.9
0.655
0.001429**
aP < 0.05 (*) was considered significant, and P < 0.01 (**), as well as < 0.001 (***), were all considered extremely significant
Table 4
Significantly changed proteins in RS218-infected hBMECs
Accession
Description
MW [kDa]
Fold change
P valuea
O00592
Podocalyxin
58.6
1.214
0.001481**
O14556
Glyceraldehyde-3-phosphate dehydrogenase, testis-specific
44.5
2.514
0.000183***
O43598
2′-Deoxynucleoside 5′-phosphate N-hydrolase 1
19.1
0.8
0.020803*
O76024
Wolframin
100.2
0.732
0.000347***
O76095
Protein JTB
16.3
0.815
0.026287*
O95989
Diphosphoinositol polyphosphate phosphohydrolase 1
19.5
0.821
0.003332**
P05067
Amyloid beta A4 protein
86.9
0.813
0.004913**
P10412
Histone H1.4
21.9
1.271
0.001736**
P11532
Dystrophin
426.5
0.799
0.014535*
P14174
Macrophage migration inhibitory factor
12.5
1.276
0.008267**
P16401
Histone H1.5
22.6
1.221
0.025445*
P16402
Histone H1.3
22.3
1.306
0.001514**
P16403
Histone H1.2
21.4
1.332
0.021727*
P30154
Serine/threonine-protein phosphatase 2A 65 kDa regulatory subunit A beta isoform
66.2
0.809
0.005474**
P35527
Keratin, type I cytoskeletal 9
62
0.822
0.038701*
P42167
Lamina-associated polypeptide 2, isoforms beta/gamma
50.6
0.826
0.0494*
P46781
40S ribosomal protein S9
22.6
1.207
0.013518*
P50402
Emerin
29
0.8
0.000916***
P52756
RNA-binding protein 5
92.1
0.74
4.23E−06***
P55789
FAD-linked sulfhydryl oxidase ALR
23.4
1.537
0.014932*
P61313
60S ribosomal protein L15
24.1
1.286
0.000236***
P62380
TATA box-binding protein-like protein 1
20.9
0.66
0.008696**
Q07020
60S ribosomal protein L18
21.6
1.367
0.002799**
Q4V339
COBW domain-containing protein 6
43.9
0.756
0.00457**
Q8N4H5
Mitochondrial import receptor subunit TOM5 homolog
6
1.223
0.000783***
Q8ND56
Protein LSM14 homolog A
50.5
0.793
0.017061*
Q96BZ8
Leukocyte receptor cluster member 1
30.5
0.693
0.005542**
Q96HP4
Oxidoreductase NAD-binding domain-containing protein 1
34.8
3.845
0.008111**
Q96KR1
Zinc finger RNA-binding protein
116.9
0.811
0.01368*
Q96LB3
Intraflagellar transport protein 74 homolog
69.2
0.71
0.012006*
Q96P47
Arf-GAP with GTPase, ANK repeat and PH domain-containing protein 3
95
0.783
0.004713**
Q99538
Legumain
49.4
0.692
0.001173**
Q9BTA9
WW domain-containing adapter protein with coiled-coil
70.7
0.743
0.01886*
Q9BZF9
Uveal autoantigen with coiled-coil domains and ankyrin repeats
162.4
0.824
0.031539*
Q9H7B2
Ribosome production factor 2 homolog
35.6
1.367
0.020862*
Q9HCD5
Nuclear receptor coactivator 5
65.5
0.771
0.001324**
Q9NPD3
Exosome complex component RRP41
26.4
0.784
0.015931*
Q9NZR1
Tropomodulin-2
39.6
1.216
0.030742*
Q9UI10
Translation initiation factor eIF-2B subunit delta
57.5
0.828
0.001214**
Q9UIC8
Leucine carboxyl methyltransferase 1
38.4
0.811
0.0312*
Q9UK41
Vacuolar protein sorting-associated protein 28 homolog
25.4
0.715
0.032545*
Q9Y4R8
Telomere length regulation protein TEL2 homolog
91.7
0.807
0.019858*
Q9Y5V3
Melanoma-associated antigen D1
86.1
1.238
0.007536**
aP < 0.05 (*) was considered significant, and P < 0.01 (**), as well as < 0.001 (***), were all considered extremely significant
Table 5
The distinct differential proteins in hBMECs in response to meningitic E. coli strains PCN033 and RS218
ID
Name
Protein
Fold Change
RS218
PCN033
HB101
Q9NPD3
EXOSC4
Exosome complex component RRP41
0.8
0.759
0.74
Q96LB3
IFT74
Intraflagellar transport protein 74 homolog
0.7
0.543
/
P11532
DMD
Dystrophin
0.8
0.691
/
P52756
RBM5
RNA-binding protein 5
0.7
0.758
/
Q4V339
CBWD6
COBW domain-containing protein 6
0.8
0.747
/
Q9Y4R8
TELO2
Telomere length regulation protein TEL2 homolog
0.8
0.735
/
P35527
KRT9
Keratin, type I cytoskeletal 9
0.8
0.72
/
P62380
TBOL1
TATA box-binding protein-like protein 1
0.7
0.621
/
P16403
HIST1H1C
Histone H1.2
1.3
0.572
/
P16402
HIST1H1D
Histone H1.3
1.3
0.6
/
P10412
HIST1H1E
Histone H1.4
1.3
0.655
/
P16401
HIST1H1B
Histone H1.5
1.2
0.631
/
P14174
MIF
Macrophage migration inhibitory factor
1.3
1.486
/

Western blot verification of the DEPs

We next used western blotting to further test the DEPs identified by iTRAQ. We selected several proteins from the iTRAQ results from both PCN033 and RS218 groups. The test proteins were HIST1H1C, TBPL1, and MIF for the PCN033 group (Fig. 3a), and DMD, LGMN, and HIST1H1C for the RS218 group (Fig. 3c). The western blot and densitometry analyses produced the similar expression alteration to those of the iTRAQ results following either PCN033 or RS218 infection (Fig. 3b, d).

Bioinformatic analysis of the DEPs in hBMECs

We next investigated and characterized the DEPs by searching the GO and UniProt databases. The DEPs were assigned to the categories of different “biological processes,” “cellular components,” and “molecular functions.” Within the biological processes class, the DEPs from the three groups (RS218, PCN033, and HB101) were mainly divided into metabolic processes, localization, cellular process, and cellular component organization or biogenesis. The immune system process and developmental process classes were found in both RS218 and PCN033 infection groups, but not in the HB101 group. Within the cellular component class, the DEPs were mainly divided into organelle, macromolecular complex, and cell parts, and the membrane-associated ones were only identified in the meningitic strains RS218 and PCN033, not in HB101. As for molecular function, the DEPs were mainly associated with structural molecule activity, catalytic activity, and binding (Fig. 4a, Additional file 4: Table S4).
We next performed canonical pathway prediction through IPA on the DEPs. The top ranked canonical pathways in each group are shown in Fig. 4b. We found that protein kinase A signaling, eumelanin biosynthesis, EIF2 signaling, and granzyme A signaling were simultaneously enriched in both RS218 and PCN033 infection groups, but not in the HB101 group (Fig. 4b). Noticeably, granzyme A signaling was much more significantly enriched in the DEPs from both meningitic groups, suggesting a potential role for granzyme A in meningitic E. coli invasion of the BBB. Additionally, phosphatidylcholine biosynthesis I, choline biosynthesis III, and glioma invasiveness signaling were only enriched in the PCN033 group, while neuronal NOS signaling and regulation of eIF4 and p70S6K signaling were only identified in the RS218 group, which exhibited distinct signaling pathways that might have strain specificity (Fig. 4b).
The IPA tool was used to further analyze the potential networks based on the DEPs from the E. coli infections. Two networks were drawn for these differential cellular proteins in response to HB101 infection (Fig. 5a, b, Additional file 5: Table S5). In addition, four networks were generated based on the DEPs from the PCN033 infection (Fig. 5c–f, Additional file 6: Table S6), while two networks were generated from the DEPs upon RS218 infection (Fig. 5g, h, Additional file 7: Table S7). It should be noted that the NF-κB complex, as well as ERK, were included in the networks of both PCN033 and RS218 groups, while they were not observed in the cells in response to the non-meningitic HB101 strain, suggesting that these two essential signaling molecules exert regulatory effects during meningitic E. coli penetration of the BBB.

MIF contributes to meningitic E. coli-induced cytokine production and tight junction disruption

Based on the aforementioned network analysis, we noticed the presence of MIF in the meningitic PCN033 and RS218 strain groups, suggesting potential roles for it in meningitic E. coli invasion of the BBB. Here, by pretreating the hBMECs with 20 μM ISO-1 (a MIF inhibitor), we found that the multiple cytokines [e.g. interleukin (IL)-6, IL-8, tumor necrosis factor (TNF)-α, IL-1β] significantly induced by meningitic E. coli PCN033 or RS218 infection had decreased levels (Fig. 6a, b). Moreover, the ECIS system was applied to evaluate the potential effects of recombinant MIF protein on the barrier function of hBMECs. The results showed that recombinant MIF obviously decreased the resistance formed by the cells in a dose-dependent manner (Fig. 6c). We also observed that treatment with recombinant MIF (200 ng/ml) for 12 and 24 h led to decreased expression of tight junction proteins like ZO-1 and occludin (Fig. 6d); moreover, use of the MIF inhibitor ISO-1 could partially recover the PCN033 or RS218 infection-caused downregulation of tight junction proteins like ZO-1 and Occludin (Fig. 6e, f). Together, these observations support the conclusion that MIF contributes to the induction of proinflammatory cytokines and the decrease in tight junction proteins during meningitic E. coli invasion of the BBB.

Meningitic E. coli activation of NF-κB signaling mediates the production of cytokines

As mentioned above in the network analysis, involvement of the NF-κB complex was observed in cells following the challenge with meningitic E. coli strains PCN033 and RS218, but not with non-meningitic HB101. Therefore, we investigated NF-κB signaling activation in hBMECs in response to infection. Phosphorylation of the NF-κB p65 subunit increased significantly in response to PCN033 and RS218 infection, and this was much higher than that observed during the response to HB101 infection. Also, degradation of IκBα upon PCN033 or RS218 infection was much greater than that upon HB101 infection (Fig. 7a, b). Using immunofluorescence microscopy, we also observed p65 translocation to the nucleus upon PCN033 and RS218 infection (Fig. 7c), while this nuclear translocation was barely observed in response to HB101 infection (Fig. 7c). These results indicate that the NF-κB signaling pathway is activated during meningitic E. coli interaction with hBMECs. Moreover, by using the NF-κB inhibitor BAY11-7082, we observed that the meningitic E. coli PCN033- or RS218-induced cytokines production (including IL-6, IL-8, TNF-α, and IL-1β) was significantly decreased when compared with DMSO treatment (Fig. 7d, e). Together, these data firmly support our network analysis that the NF-κB signaling pathway is involved in both PCN033 and RS218 infection of hBMECs, and their activation of NF-κB signaling in hBMECs mediates the induction of proinflammatory cytokines.

MAPK signaling pathways are involved in proinflammatory cytokine induction by meningitic E. coli strains

Because ERK was assumed to be involved in infections with PCN033 and RS218 based on our network prediction, we next investigated the activation of MAPK pathways in hBMECs in response to meningitic E. coli. The results showed that the phosphorylation of p38, JNK, and ERK1/2 significantly increased in response to meningitic strains PCN033 or RS218 (Fig. 8a, b), indicating the activation of all three MAPK pathways in hBMECs upon meningitic E. coli challenge. After demonstrating the significant induction of several proinflammatory cytokines above, we next investigated whether the MAPK pathways were involved in these cytokines production. Following pretreatment with U0126 (a specific ERK1/2 inhibitor), SB202190 (a selective inhibitor of p38), and SP600125 (a JNK-specific inhibitor), the proinflammatory cytokines (IL-6, IL-8, TNF-α, IL-1β) induced in hBMECs upon PCN033 or RS218 infection were significantly reduced (to different extents), compared with that in each DMSO control group (Fig. 8c). These results indicate that the MAPK signaling pathways, including MAPK-p38, MAPK-ERK1/2, and MAPK-JNK, were all activated and at least participated in meningitic E. coli-induced neuroinflammatory responses.

Discussion

The iTRAQ-based proteomics, a powerful approach for obtaining comprehensive and quantitative protein expression profiling data, has been used widely to identify and characterize potential cellular targets. In current study, we used iTRAQ to explore the proteomic differences in hBMECs in response to meningitic or non-meningitic E. coli infections. The E. coli strains PCN033 and RS218 were selected for this study because they are representative meningitis-causing strains capable of penetrating the BBB as well as inducing severe neuroinflammation [5, 20], while the E. coli strain HB101 is avirulent and non-meningitic and was therefore used as the negative control.
Based on our data, 13 significantly differentiated proteins in total were found to be shared by PCN033 and RS218 (Fig. 1). They are TELO2, IFT74, CBWD6, EXOSC4, TBOL1, RBM5, KRT9, HIST1H1C, HIST1H1D, HIST1H1B, HIST1H1E, MIF, and DMD (Table 5). Among these, EXOSC4 was the only protein that was also significantly changed in response to non-meningitic E. coli HB101 (Fig. 2, Table 5). EXOSC4, a non-catalytic component of the RNA exosome machinery, has 3′-5′ exoribonuclease activity and participates in a multitude of cellular RNA processing and degradation events [29]. It was reported that EXOSC4 was a potential factor involved in the maintenance of genome stability, by eliminating the RNA processing by-products and non-coding “pervasive” transcripts thereby limiting or excluding their export to the cytoplasm, or by preventing translation of aberrant mRNAs [3032]. In lung adenocarcinoma, EXOSC4 has been reported to be extremely highly expressed and closely associated with cancer cell proliferation and was, therefore, recognized as a new prognostic marker [30]. Similarly, in patients with liver cancer, the EXOSC4 gene was found to be highly expressed, and its knock-down commonly inhibited cancer cell growth and invasion [33]. Here, we found that EXOSC4 was commonly targeted by the meningitic and the non-meningitic E. coli strains, indicating that this cellular protein is a non-specific infection-related protein. Other than EXOSC4, the remaining 12 proteins were shared by the meningitic strains (PCN033 and RS218) alone, suggesting that these proteins might represent the potential targets hijacked by these meningitic E. coli strains.
Among these 12 meningitic E. coli-specific “cellular responders,” we firstly focused on MIF, which was the only one to exhibit common upregulation in response to both meningitic E. coli PCN033 and RS218 (Table 5). MIF is a proinflammatory cytokine, which has been highlighted as a key player in infection and septic shock [34, 35]. It is reported to be involved in the cytokine storm, which facilitates the uncontrolled release of cytokines into the circulation during pathogen infection or sepsis [36]. As previously evidenced in E. coli-induced meningitis, cytokines and chemokines potentially contribute to BBB damage [5]. The burst of proinflammatory cytokines during infection may lead directly to dysfunction of the endothelial barrier and an increase in vascular permeability in the brain, thus finally leading to severe CNS injury. Moreover, MIF may be secreted by a wide variety of cells upon stimulation, and once MIF binds to its receptors (e.g., CXCR2, CXCR4, and/or CD74 [37, 38]), several downstream signal molecules such as PI3K/Akt or MAPK/ERK become activated, thus mediating the inflammatory response [39, 40]. In the present study, the effects of MIF on meningitic E. coli-induced inflammation were also verified by the observation that the MIF inhibitor ISO-1 significantly decreased meningitic E. coli PCN033- or RS218-induced upregulation of IL-6, IL-8, IL-Iβ, and TNF-α (Fig. 5). Noticeably however, although the ISO-1 inhibitory effects were significant, there was still a significant induction of IL-6 and IL-8 in response to PCN033 and RS218 infection, suggesting that other “switches” for proinflammatory cytokine and chemokine generation commonly exist in response to infection. Except for its role in inflammation, we also observed the involvement of MIF in BBB damage, as evidenced by the fact that recombinant MIF was able to deconstruct the endothelial barrier by inducing a significant decrease in the junction-associated protein ZO-1 and occludin (Fig. 6). Furthermore, when MIF inhibitor ISO-1 was used, the PCN033- and/or RS218-induced downregulation of ZO-1 and occludin was largely restored (Fig. 6). Considering the potential roles of MIF in mediating the neuroinflammatory response as well as in inducing BBB disruption, it is possible that MIF may represent a novel and potential target for clinical prevention and therapy for E. coli meningitis.
Our IPA-based canonical pathways prediction suggested that protein kinase A signaling, eumelanin biosynthesis, EIF2 signaling, and granzyme A signaling were simultaneously enriched in hBMECs upon infection with RS218 and PCN033, but not with HB101. Among these processes, granzyme A signaling was much more significantly enriched. In the RS218 group, HIST1H1B, HIST1H1C, HIST1H1E, and HIST1H1D are included in granzyme A signaling, while in the PCN033 group, HIST1H1B, HIST1H1C, HIST1H1E, HIST1H1D, and H1F0 are involved (Additional file 6: Table S6). Granzyme A was identified as a cytotoxic T lymphocyte protease with multiple roles in infectious diseases. For example, several studies have shown that granzyme A is highly expressed in patients with tuberculosis and may represent a promising diagnostic marker distinct from IFN-γ to discriminate between patients with tuberculosis and other pulmonary diseases [4143]. Granzyme A is also considered to participate in the host defense response in multiple ways, such as by generating superoxide and inactivating the oxidative defense enzymes that kill intracellular parasites [44], by unfavorably impairing host defenses during Streptococcus pneumoniae pneumonia [45], by performing as a proinflammatory protease that cleaves IL-1β intracellularly into bioactive IL-1β [46, 47], or by causing detachment of alveolar epithelial A549 cells accompanied by promotion of IL-8 release [48]. Here, in the present study, granzyme A signaling was significantly enriched by cellular differentiated proteins in response to both meningitic E. coli strains, but not in non-meningitic E. coli HB101. This result probably indicates that granzyme A could be a potential indicator of E. coli meningitis, but further supportive evidences are needed.
Based on the IPA functional network analysis, we also noticed that the NF-κB complex and MAPK/ERK signaling were involved in both PCN033 and RS218 infection of hBMECs, but barely in the HB101 group. The NF-κB complex comprises a family of closely related transcription factors with important roles in regulating the gene expression involved in inflammation and the immune response [49]. The NF-κB activation process is induced by the phosphorylation of serine residues in IkB proteins, which are subjected to ubiquitination and proteasome degradation and, subsequently, phosphorylation and nuclear translocation of the p65 subunit. Early studies have shown that NF-κB is activated in bacteria-induced CNS infections [50], and NF-κB inhibitors have been found to reduce neuroinflammation [51] as well as protect rat brains from inflammatory injury following transient focal cerebral ischemia [52] and pneumococcal meningitis [53]. In E. coli, it has been evidenced that OmpA+E. coli can induce ICAM-1 expression in hBMECs by activating NF-κB signaling [54] and that the IbeA+E. coli K1 strain can also induce activation and nuclear translocation of NF-κB in hBMECs [55]. In the current study, by western blotting, we also showed that the NF-κB pathway was activated more in hBMECs infected by meningitic strains PCN033 and RS218 compared with that by HB101 infection, where the phosphorylation of p65 and degradation of IκBα were compared, as well as with the immunofluorescence experiments that showed the nuclear translocation of p65. Not unexpectedly, treating hBMECs with the NF-κB inhibitor BAY11-7082 significantly attenuated those cytokines induction during meningitic E. coli infection, suggesting that NF-κB signaling works potently in mediating the neuroinflammatory response.
Likewise, we found that the effects of MAPK signaling were similarly associated with both PCN033 and RS218 infection of hBMECs. MAPK signaling cascades actually involve three major pathways: JNK (which acts as mediator of extracellular stress responses), ERK1/2 (which mediates proliferative stimuli), and p38 (which is also involved in mediating extracellular stress responses, particularly by regulating cytokine expression) [56]. Our IPA network analysis indicated the involvement of ERK during infection with meningitic E. coli PCN033 and RS218, which is consistent with our previous finding that MAPK/ERK signaling is involved in infection and mediates the induction of VEGFA and Snail-1 by the meningitic strain PCN033 [5]; however, via western blotting we showed the activation of all these three signaling molecules in response to PCN033 and RS218 infection. Also, by using specific inhibitors against ERK1/2, p38, and JNK, we observed that inhibition of all three MAPK pathways significantly decreased the infection-induced upregulation of proinflammatory cytokines IL-6, IL-8, IL-Iβ, and TNF-α. Therefore, collectively these data largely support the viewpoint that all three major MAPK signaling pathways play potent roles in meningitic E. coli infection and induce neuroinflammatory responses.

Conclusions

In our study, using the iTRAQ proteomics approach, we compared and analyzed the DEPs in hBMECs infected with meningitic or non-meningitic E. coli strains. Twelve DEPs were identified as the commonly responding proteins in hBMECs upon infection with meningitic E. coli strains PCN033 and RS218, except for only one cellular protein shared by both meningitic and non-meningitic strains. Our data revealed MIF to be an important contributor to meningitic E. coli-induced cytokine production and tight junction disruption, while also showing that the NF-κB and MAPK signaling pathways are involved in the infection process. Comparing and profiling these differential cellular proteins in hBMECs in response to meningitic E. coli strains should open up further research on host responses against meningitic strains and help with the development of more targets for better prevention and therapeutic control of E. coli meningitis.

Acknowledgements

We would like to thank Prof. Kwang Sik Kim in Johns Hopkins University School of Medicine to kindly provide the hBMECs cells.

Funding

This work was supported by grants from the National Key R&D Program of China (2016YFD0500406), the National Natural Science Foundation of China (NSFC) (Nos. 31772736, 31502062), the Outstanding youth project of Natural Science Foundation in Hubei Province (2018CFA070), and the Fundamental Research Funds for the Central Universities (Program No. 2662018PY032).

Availability of data and materials

There is no data, software, databases, and application/tool available apart from the reported in the present study. All data is provided in manuscript.
Not applicable.
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.
Literatur
1.
Zurück zum Zitat Kim KS. Pathogenesis of bacterial meningitis: from bacteraemia to neuronal injury. Nat Rev Neurosci. 2003;4:376–85.CrossRef Kim KS. Pathogenesis of bacterial meningitis: from bacteraemia to neuronal injury. Nat Rev Neurosci. 2003;4:376–85.CrossRef
2.
Zurück zum Zitat Kim KS. Mechanisms of microbial traversal of the blood-brain barrier. Nat Rev Microbiol. 2008;6:625–34.CrossRef Kim KS. Mechanisms of microbial traversal of the blood-brain barrier. Nat Rev Microbiol. 2008;6:625–34.CrossRef
3.
Zurück zum Zitat Kim KS. Acute bacterial meningitis in infants and children. Lancet Infect Dis. 2010;10:32–42.CrossRef Kim KS. Acute bacterial meningitis in infants and children. Lancet Infect Dis. 2010;10:32–42.CrossRef
4.
Zurück zum Zitat Candelario-Jalil E, Yang Y, Rosenberg GA. Diverse roles of matrix metalloproteinases and tissue inhibitors of metalloproteinases in neuroinflammation and cerebral ischemia. Neuroscience. 2009;158:983–94.CrossRef Candelario-Jalil E, Yang Y, Rosenberg GA. Diverse roles of matrix metalloproteinases and tissue inhibitors of metalloproteinases in neuroinflammation and cerebral ischemia. Neuroscience. 2009;158:983–94.CrossRef
5.
Zurück zum Zitat Yang RC, Liu WT, Miao L, Yang XP, Fu JY, Dou BB, Cai AL, Zong X, Tan C, Chen HC, Wang XR. Induction of VEGFA and Snail-1 by meningitic Escherichia coli mediates disruption of the blood-brain barrier. Oncotarget. 2016;7:63839–55.PubMedPubMedCentral Yang RC, Liu WT, Miao L, Yang XP, Fu JY, Dou BB, Cai AL, Zong X, Tan C, Chen HC, Wang XR. Induction of VEGFA and Snail-1 by meningitic Escherichia coli mediates disruption of the blood-brain barrier. Oncotarget. 2016;7:63839–55.PubMedPubMedCentral
6.
Zurück zum Zitat Stins MF, Badger J, Sik Kim K. Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Microb Pathog. 2001;30:19–28.CrossRef Stins MF, Badger J, Sik Kim K. Bacterial invasion and transcytosis in transfected human brain microvascular endothelial cells. Microb Pathog. 2001;30:19–28.CrossRef
7.
Zurück zum Zitat Stins MF, Gilles F, Kim KS. Selective expression of adhesion molecules on human brain microvascular endothelial cells. J Neuroimmunol. 1997;76:81–90.CrossRef Stins MF, Gilles F, Kim KS. Selective expression of adhesion molecules on human brain microvascular endothelial cells. J Neuroimmunol. 1997;76:81–90.CrossRef
8.
Zurück zum Zitat Das A, Asatryan L, Reddy MA, Wass CA, Stins MF, Joshi S, Bonventre JV, Kim KS. Differential role of cytosolic phospholipase A2 in the invasion of brain microvascular endothelial cells by Escherichia coli and Listeria monocytogenes. J Infect Dis. 2001;184:732–7.CrossRef Das A, Asatryan L, Reddy MA, Wass CA, Stins MF, Joshi S, Bonventre JV, Kim KS. Differential role of cytosolic phospholipase A2 in the invasion of brain microvascular endothelial cells by Escherichia coli and Listeria monocytogenes. J Infect Dis. 2001;184:732–7.CrossRef
9.
Zurück zum Zitat 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;90:897–905.CrossRef 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;90:897–905.CrossRef
10.
Zurück zum Zitat Burkhart A, Thomsen LB, Thomsen MS, Lichota J, Fazakas C, Krizbai I, Moos T. Transfection of brain capillary endothelial cells in primary culture with defined blood-brain barrier properties. Fluids Barriers CNS. 2015;12:19.CrossRef Burkhart A, Thomsen LB, Thomsen MS, Lichota J, Fazakas C, Krizbai I, Moos T. Transfection of brain capillary endothelial cells in primary culture with defined blood-brain barrier properties. Fluids Barriers CNS. 2015;12:19.CrossRef
11.
Zurück zum Zitat Eigenmann DE, Xue G, Kim KS, Moses AV, Hamburger M, Oufir M. Comparative study of four immortalized human brain capillary endothelial cell lines, hCMEC/D3, hBMEC, TY10, and BB19, and optimization of culture conditions, for an in vitro blood-brain barrier model for drug permeability studies. Fluids Barriers CNS. 2013;10:33.CrossRef Eigenmann DE, Xue G, Kim KS, Moses AV, Hamburger M, Oufir M. Comparative study of four immortalized human brain capillary endothelial cell lines, hCMEC/D3, hBMEC, TY10, and BB19, and optimization of culture conditions, for an in vitro blood-brain barrier model for drug permeability studies. Fluids Barriers CNS. 2013;10:33.CrossRef
12.
Zurück zum Zitat Abbott NJ. Blood-brain barrier structure and function and the challenges for CNS drug delivery. J Inherit Metab Dis. 2013;36:437–49.CrossRef Abbott NJ. Blood-brain barrier structure and function and the challenges for CNS drug delivery. J Inherit Metab Dis. 2013;36:437–49.CrossRef
13.
Zurück zum Zitat Tajes M, Ramos-Fernandez E, Weng-Jiang X, Bosch-Morato M, Guivernau B, Eraso-Pichot A, Salvador B, Fernandez-Busquets X, Roquer J, Munoz FJ. The blood-brain barrier: structure, function and therapeutic approaches to cross it. Mol Membr Biol. 2014;31:152–67.CrossRef Tajes M, Ramos-Fernandez E, Weng-Jiang X, Bosch-Morato M, Guivernau B, Eraso-Pichot A, Salvador B, Fernandez-Busquets X, Roquer J, Munoz FJ. The blood-brain barrier: structure, function and therapeutic approaches to cross it. Mol Membr Biol. 2014;31:152–67.CrossRef
14.
Zurück zum Zitat Hoffman JA, Badger JL, Zhang Y, Huang SH, Kim KS. Escherichia coli K1 aslA contributes to invasion of brain microvascular endothelial cells in vitro and in vivo. Infect Immun. 2000;68:5062–7.CrossRef Hoffman JA, Badger JL, Zhang Y, Huang SH, Kim KS. Escherichia coli K1 aslA contributes to invasion of brain microvascular endothelial cells in vitro and in vivo. Infect Immun. 2000;68:5062–7.CrossRef
15.
Zurück zum Zitat Wang Y, Kim KS. Role of OmpA and IbeB in Escherichia coli K1 invasion of brain microvascular endothelial cells in vitro and in vivo. Pediatr Res. 2002;51:559–63.CrossRef Wang Y, Kim KS. Role of OmpA and IbeB in Escherichia coli K1 invasion of brain microvascular endothelial cells in vitro and in vivo. Pediatr Res. 2002;51:559–63.CrossRef
16.
Zurück zum Zitat Reddy MA, Prasadarao NV, Wass CA, Kim KS. Phosphatidylinositol 3-kinase activation and interaction with focal adhesion kinase in Escherichia coli K1 invasion of human brain microvascular endothelial cells. J Biol Chem. 2000;275:36769–74.CrossRef Reddy MA, Prasadarao NV, Wass CA, Kim KS. Phosphatidylinositol 3-kinase activation and interaction with focal adhesion kinase in Escherichia coli K1 invasion of human brain microvascular endothelial cells. J Biol Chem. 2000;275:36769–74.CrossRef
17.
Zurück zum Zitat 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;277:15607–12.CrossRef 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;277:15607–12.CrossRef
18.
Zurück zum Zitat 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;68:6423–30.CrossRef 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;68:6423–30.CrossRef
19.
Zurück zum Zitat Chung JW, Hong SJ, Kim KJ, Goti D, Stins MF, Shin S, Dawson VL, Dawson TM, Kim KS. 37-kDa laminin receptor precursor modulates cytotoxic necrotizing factor 1-mediated RhoA activation and bacterial uptake. J Biol Chem. 2003;278:16857–62.CrossRef Chung JW, Hong SJ, Kim KJ, Goti D, Stins MF, Shin S, Dawson VL, Dawson TM, Kim KS. 37-kDa laminin receptor precursor modulates cytotoxic necrotizing factor 1-mediated RhoA activation and bacterial uptake. J Biol Chem. 2003;278:16857–62.CrossRef
20.
Zurück zum Zitat Wang X, Maruvada R, Morris AJ, Liu JO, Wolfgang MJ, Baek DJ, Bittman R, Kim KS. Sphingosine 1-phosphate activation of EGFR as a novel target for meningitic Escherichia coli penetration of the blood-brain barrier. PLoS Pathog. 2016;12:e1005926.CrossRef Wang X, Maruvada R, Morris AJ, Liu JO, Wolfgang MJ, Baek DJ, Bittman R, Kim KS. Sphingosine 1-phosphate activation of EGFR as a novel target for meningitic Escherichia coli penetration of the blood-brain barrier. PLoS Pathog. 2016;12:e1005926.CrossRef
21.
Zurück zum Zitat Wijetunge DS, Katani R, Kapur V, Kariyawasam S. Complete genome sequence of Escherichia coli strain RS218 (O18:H7:K1), associated with neonatal meningitis. Genome Announc. 2015;3:e00804-15. Wijetunge DS, Katani R, Kapur V, Kariyawasam S. Complete genome sequence of Escherichia coli strain RS218 (O18:H7:K1), associated with neonatal meningitis. Genome Announc. 2015;3:e00804-15.
22.
Zurück zum Zitat Tan C, Xu Z, Zheng H, Liu W, Tang X, Shou J, Wu B, Wang S, Zhao GP, Chen H. Genome sequence of a porcine extraintestinal pathogenic Escherichia coli strain. J Bacteriol. 2011;193:5038.CrossRef Tan C, Xu Z, Zheng H, Liu W, Tang X, Shou J, Wu B, Wang S, Zhao GP, Chen H. Genome sequence of a porcine extraintestinal pathogenic Escherichia coli strain. J Bacteriol. 2011;193:5038.CrossRef
23.
Zurück zum Zitat Liu C, Zheng H, Yang M, Xu Z, Wang X, Wei L, Tang B, Liu F, Zhang Y, Ding Y, et al. Genome analysis and in vivo virulence of porcine extraintestinal pathogenic Escherichia coli strain PCN033. BMC Genomics. 2015;16:717.CrossRef Liu C, Zheng H, Yang M, Xu Z, Wang X, Wei L, Tang B, Liu F, Zhang Y, Ding Y, et al. Genome analysis and in vivo virulence of porcine extraintestinal pathogenic Escherichia coli strain PCN033. BMC Genomics. 2015;16:717.CrossRef
24.
Zurück zum Zitat Tan C, Tang X, Zhang X, Ding Y, Zhao Z, Wu B, Cai X, Liu Z, He Q, Chen H. Serotypes and virulence genes of extraintestinal pathogenic Escherichia coli isolates from diseased pigs in China. Vet J. 2012;192:483–8.CrossRef Tan C, Tang X, Zhang X, Ding Y, Zhao Z, Wu B, Cai X, Liu Z, He Q, Chen H. Serotypes and virulence genes of extraintestinal pathogenic Escherichia coli isolates from diseased pigs in China. Vet J. 2012;192:483–8.CrossRef
25.
Zurück zum Zitat Zhu L, Maruvada R, Sapirstein A, Malik KU, Peters-Golden M, Kim KS. Arachidonic acid metabolism regulates Escherichia coli penetration of the blood-brain barrier. Infect Immun. 2010;78:4302–10.CrossRef Zhu L, Maruvada R, Sapirstein A, Malik KU, Peters-Golden M, Kim KS. Arachidonic acid metabolism regulates Escherichia coli penetration of the blood-brain barrier. Infect Immun. 2010;78:4302–10.CrossRef
26.
Zurück zum Zitat Khan NA, Kim Y, Shin S, Kim KS. FimH-mediated Escherichia coli K1 invasion of human brain microvascular endothelial cells. Cell Microbiol. 2007;9:169–78.CrossRef Khan NA, Kim Y, Shin S, Kim KS. FimH-mediated Escherichia coli K1 invasion of human brain microvascular endothelial cells. Cell Microbiol. 2007;9:169–78.CrossRef
27.
Zurück zum Zitat Yang R, Huang F, Fu J, Dou B, Xu B, Miao L, Liu W, Yang X, Tan C, Chen H, Wang X. Differential transcription profiles of long non-coding RNAs in primary human brain microvascular endothelial cells in response to meningitic Escherichia coli. Sci Rep. 2016;6:38903.CrossRef Yang R, Huang F, Fu J, Dou B, Xu B, Miao L, Liu W, Yang X, Tan C, Chen H, Wang X. Differential transcription profiles of long non-coding RNAs in primary human brain microvascular endothelial cells in response to meningitic Escherichia coli. Sci Rep. 2016;6:38903.CrossRef
28.
Zurück zum Zitat Szulcek R, Bogaard HJ, Amerongen GPV. Electric cell-substrate impedance sensing for the quantification of endothelial proliferation, barrier function, and motility. J Vis Exp. 2014;85. https://doi.org/10.3791/51300. Szulcek R, Bogaard HJ, Amerongen GPV. Electric cell-substrate impedance sensing for the quantification of endothelial proliferation, barrier function, and motility. J Vis Exp. 2014;85. https://​doi.​org/​10.​3791/​51300.
29.
Zurück zum Zitat Allmang C, Petfalski E, Podtelejnikov A, Mann M, Tollervey D, Mitchell P. The yeast exosome and human PM-Scl are related complexes of 3′ --> 5′ exonucleases. Genes Dev. 1999;13:2148–58.CrossRef Allmang C, Petfalski E, Podtelejnikov A, Mann M, Tollervey D, Mitchell P. The yeast exosome and human PM-Scl are related complexes of 3′ --> 5′ exonucleases. Genes Dev. 1999;13:2148–58.CrossRef
30.
Zurück zum Zitat O'Byrne K, Paquet N, Box JK, Adams M, Richard D. 17P examination of EXOSC4 as a new prognostic marker and a novel therapeutic avenue in lung adenocarcinoma. J Thorac Oncol. 2016;11:S63.CrossRef O'Byrne K, Paquet N, Box JK, Adams M, Richard D. 17P examination of EXOSC4 as a new prognostic marker and a novel therapeutic avenue in lung adenocarcinoma. J Thorac Oncol. 2016;11:S63.CrossRef
31.
Zurück zum Zitat Basu U, Meng FL, Keim C, Grinstein V, Pefanis E, Eccleston J, Zhang T, Myers D, Wasserman CR, Wesemann DR, et al. The RNA exosome targets the AID cytidine deaminase to both strands of transcribed duplex DNA substrates. Cell. 2011;144:353–63.CrossRef Basu U, Meng FL, Keim C, Grinstein V, Pefanis E, Eccleston J, Zhang T, Myers D, Wasserman CR, Wesemann DR, et al. The RNA exosome targets the AID cytidine deaminase to both strands of transcribed duplex DNA substrates. Cell. 2011;144:353–63.CrossRef
32.
Zurück zum Zitat Van Dijk EL, Schilders G, Pruijn GJ. Human cell growth requires a functional cytoplasmic exosome, which is involved in various mRNA decay pathways. RNA. 2007;13:1027–35.CrossRef Van Dijk EL, Schilders G, Pruijn GJ. Human cell growth requires a functional cytoplasmic exosome, which is involved in various mRNA decay pathways. RNA. 2007;13:1027–35.CrossRef
33.
Zurück zum Zitat Stefanska B, Cheishvili D, Suderman M, Arakelian A, Huang J, Hallett M, Han ZG, Al-Mahtab M, Akbar SM, Khan WA, et al. Genome-wide study of hypomethylated and induced genes in patients with liver cancer unravels novel anticancer targets. Clin Cancer Res. 2014;20:3118–32.CrossRef Stefanska B, Cheishvili D, Suderman M, Arakelian A, Huang J, Hallett M, Han ZG, Al-Mahtab M, Akbar SM, Khan WA, et al. Genome-wide study of hypomethylated and induced genes in patients with liver cancer unravels novel anticancer targets. Clin Cancer Res. 2014;20:3118–32.CrossRef
34.
Zurück zum Zitat Rosado Jde D, Rodriguez-Sosa M. Macrophage migration inhibitory factor (MIF): a key player in protozoan infections. Int J Biol Sci. 2011;7:1239–56.CrossRef Rosado Jde D, Rodriguez-Sosa M. Macrophage migration inhibitory factor (MIF): a key player in protozoan infections. Int J Biol Sci. 2011;7:1239–56.CrossRef
35.
Zurück zum Zitat Delaloye J, De Bruin IJ, Darling KE, Reymond MK, Sweep FC, Roger T, Calandra T, Cavassini M. Increased macrophage migration inhibitory factor (MIF) plasma levels in acute HIV-1 infection. Cytokine. 2012;60:338–40.CrossRef Delaloye J, De Bruin IJ, Darling KE, Reymond MK, Sweep FC, Roger T, Calandra T, Cavassini M. Increased macrophage migration inhibitory factor (MIF) plasma levels in acute HIV-1 infection. Cytokine. 2012;60:338–40.CrossRef
36.
Zurück zum Zitat Pang T, Cardosa MJ, Guzman MG. Of cascades and perfect storms: the immunopathogenesis of dengue haemorrhagic fever-dengue shock syndrome (DHF/DSS). Immunol Cell Biol. 2007;85:43–5.CrossRef Pang T, Cardosa MJ, Guzman MG. Of cascades and perfect storms: the immunopathogenesis of dengue haemorrhagic fever-dengue shock syndrome (DHF/DSS). Immunol Cell Biol. 2007;85:43–5.CrossRef
37.
Zurück zum Zitat Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, Dewor M, Georgiev I, Schober A, Leng L, et al. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med. 2007;13:587–96.CrossRef Bernhagen J, Krohn R, Lue H, Gregory JL, Zernecke A, Koenen RR, Dewor M, Georgiev I, Schober A, Leng L, et al. MIF is a noncognate ligand of CXC chemokine receptors in inflammatory and atherogenic cell recruitment. Nat Med. 2007;13:587–96.CrossRef
38.
Zurück zum Zitat Schwartz V, Lue H, Kraemer S, Korbiel J, Krohn R, Ohl K, Bucala R, Weber C, Bernhagen J. A functional heteromeric MIF receptor formed by CD74 and CXCR4. FEBS Lett. 2009;583:2749–57.CrossRef Schwartz V, Lue H, Kraemer S, Korbiel J, Krohn R, Ohl K, Bucala R, Weber C, Bernhagen J. A functional heteromeric MIF receptor formed by CD74 and CXCR4. FEBS Lett. 2009;583:2749–57.CrossRef
39.
Zurück zum Zitat Lue H, Kapurniotu A, Fingerle-Rowson G, Roger T, Leng L, Thiele M, Calandra T, Bucala R, Bernhagen J. Rapid and transient activation of the ERK MAPK signalling pathway by macrophage migration inhibitory factor (MIF) and dependence on JAB1/CSN5 and Src kinase activity. Cell Signal. 2006;18:688–703.CrossRef Lue H, Kapurniotu A, Fingerle-Rowson G, Roger T, Leng L, Thiele M, Calandra T, Bucala R, Bernhagen J. Rapid and transient activation of the ERK MAPK signalling pathway by macrophage migration inhibitory factor (MIF) and dependence on JAB1/CSN5 and Src kinase activity. Cell Signal. 2006;18:688–703.CrossRef
40.
Zurück zum Zitat Lue H, Thiele M, Franz J, Dahl E, Speckgens S, Leng L, Fingerle-Rowson G, Bucala R, Luscher B, Bernhagen J. Macrophage migration inhibitory factor (MIF) promotes cell survival by activation of the Akt pathway and role for CSN5/JAB1 in the control of autocrine MIF activity. Oncogene. 2007;26:5046–59.CrossRef Lue H, Thiele M, Franz J, Dahl E, Speckgens S, Leng L, Fingerle-Rowson G, Bucala R, Luscher B, Bernhagen J. Macrophage migration inhibitory factor (MIF) promotes cell survival by activation of the Akt pathway and role for CSN5/JAB1 in the control of autocrine MIF activity. Oncogene. 2007;26:5046–59.CrossRef
41.
Zurück zum Zitat Garcia-Laorden MI, Blok DC, Kager LM, Hoogendijk AJ, van Mierlo GJ, Lede IO, Rahman W, Afroz R, Ghose A, Visser CE, et al. Increased intra- and extracellular granzyme expression in patients with tuberculosis. Tuberculosis (Edinb). 2015;95:575–80.CrossRef Garcia-Laorden MI, Blok DC, Kager LM, Hoogendijk AJ, van Mierlo GJ, Lede IO, Rahman W, Afroz R, Ghose A, Visser CE, et al. Increased intra- and extracellular granzyme expression in patients with tuberculosis. Tuberculosis (Edinb). 2015;95:575–80.CrossRef
42.
Zurück zum Zitat Guggino G, Orlando V, Cutrera S, La Manna MP, Di Liberto D, Vanini V, Petruccioli E, Dieli F, Goletti D, Caccamo N. Granzyme A as a potential biomarker of Mycobacterium tuberculosis infection and disease. Immunol Lett. 2015;166:87–91.CrossRef Guggino G, Orlando V, Cutrera S, La Manna MP, Di Liberto D, Vanini V, Petruccioli E, Dieli F, Goletti D, Caccamo N. Granzyme A as a potential biomarker of Mycobacterium tuberculosis infection and disease. Immunol Lett. 2015;166:87–91.CrossRef
43.
Zurück zum Zitat Laux da Costa L, Delcroix M, Dalla Costa ER, Prestes IV, Milano M, Francis SS, Unis G, Silva DR, Riley LW, Rossetti ML. A real-time PCR signature to discriminate between tuberculosis and other pulmonary diseases. Tuberculosis (Edinb). 2015;95:421–5.CrossRef Laux da Costa L, Delcroix M, Dalla Costa ER, Prestes IV, Milano M, Francis SS, Unis G, Silva DR, Riley LW, Rossetti ML. A real-time PCR signature to discriminate between tuberculosis and other pulmonary diseases. Tuberculosis (Edinb). 2015;95:421–5.CrossRef
44.
Zurück zum Zitat Dotiwala F, Mulik S, Polidoro RB, Ansara JA, Burleigh BA, Walch M, Gazzinelli RT, Lieberman J. Killer lymphocytes use granulysin, perforin and granzymes to kill intracellular parasites. Nat Med. 2016;22:210–6.CrossRef Dotiwala F, Mulik S, Polidoro RB, Ansara JA, Burleigh BA, Walch M, Gazzinelli RT, Lieberman J. Killer lymphocytes use granulysin, perforin and granzymes to kill intracellular parasites. Nat Med. 2016;22:210–6.CrossRef
45.
Zurück zum Zitat Van den Boogaard FE, van Gisbergen KP, Vernooy JH, Medema JP, Roelofs JJ, van Zoelen MA, Endeman H, Biesma DH, Boon L, Van't Veer C, et al. Granzyme A impairs host defense during Streptococcus pneumoniae pneumonia. Am J Physiol Lung Cell Mol Physiol. 2016;311:L507–16.CrossRef Van den Boogaard FE, van Gisbergen KP, Vernooy JH, Medema JP, Roelofs JJ, van Zoelen MA, Endeman H, Biesma DH, Boon L, Van't Veer C, et al. Granzyme A impairs host defense during Streptococcus pneumoniae pneumonia. Am J Physiol Lung Cell Mol Physiol. 2016;311:L507–16.CrossRef
46.
Zurück zum Zitat Pardo J, Simon MM, Froelich CJ. Granzyme A is a proinflammatory protease. Blood. 2009;114:3968 author reply 3969-3970.CrossRef Pardo J, Simon MM, Froelich CJ. Granzyme A is a proinflammatory protease. Blood. 2009;114:3968 author reply 3969-3970.CrossRef
47.
Zurück zum Zitat Hildebrand D, Bode KA, Riess D, Cerny D, Waldhuber A, Rommler F, Strack J, Korten S, Orth JH, Miethke T, et al. Granzyme A produces bioactive IL-1beta through a nonapoptotic inflammasome-independent pathway. Cell Rep. 2014;9:910–7.CrossRef Hildebrand D, Bode KA, Riess D, Cerny D, Waldhuber A, Rommler F, Strack J, Korten S, Orth JH, Miethke T, et al. Granzyme A produces bioactive IL-1beta through a nonapoptotic inflammasome-independent pathway. Cell Rep. 2014;9:910–7.CrossRef
48.
Zurück zum Zitat Yoshikawa Y, Hirayasu H, Tsuzuki S, Fushiki T. Granzyme A causes detachment of alveolar epithelial A549 cells accompanied by promotion of interleukin-8 release. Biosci Biotechnol Biochem. 2008;72:2481–4.CrossRef Yoshikawa Y, Hirayasu H, Tsuzuki S, Fushiki T. Granzyme A causes detachment of alveolar epithelial A549 cells accompanied by promotion of interleukin-8 release. Biosci Biotechnol Biochem. 2008;72:2481–4.CrossRef
49.
Zurück zum Zitat Rothwarf DM, Karin M. The NF-kappa B activation pathway: a paradigm in information transfer from membrane to nucleus. Sci STKE. 1999;1999:RE1.PubMed Rothwarf DM, Karin M. The NF-kappa B activation pathway: a paradigm in information transfer from membrane to nucleus. Sci STKE. 1999;1999:RE1.PubMed
50.
Zurück zum Zitat Ichiyama T, Isumi H, Yoshitomi T, Nishikawa M, Matsubara T, Furukawa S. NF-kappaB activation in cerebrospinal fluid cells from patients with meningitis. Neurol Res. 2002;24:709–12.CrossRef Ichiyama T, Isumi H, Yoshitomi T, Nishikawa M, Matsubara T, Furukawa S. NF-kappaB activation in cerebrospinal fluid cells from patients with meningitis. Neurol Res. 2002;24:709–12.CrossRef
51.
Zurück zum Zitat Yang XP, Fu JY, Yang RC, Liu WT, Zhang T, Yang B, Miao L, Dou BB, Tan C, Chen HC, Wang XR. EGFR transactivation contributes to neuroinflammation in Streptococcus suis meningitis. J Neuroinflammation. 2016;13:274.CrossRef Yang XP, Fu JY, Yang RC, Liu WT, Zhang T, Yang B, Miao L, Dou BB, Tan C, Chen HC, Wang XR. EGFR transactivation contributes to neuroinflammation in Streptococcus suis meningitis. J Neuroinflammation. 2016;13:274.CrossRef
52.
Zurück zum Zitat Khan M, Elango C, Ansari MA, Singh I, Singh AK. Caffeic acid phenethyl ester reduces neurovascular inflammation and protects rat brain following transient focal cerebral ischemia. J Neurochem. 2007;102:365–77.CrossRef Khan M, Elango C, Ansari MA, Singh I, Singh AK. Caffeic acid phenethyl ester reduces neurovascular inflammation and protects rat brain following transient focal cerebral ischemia. J Neurochem. 2007;102:365–77.CrossRef
53.
Zurück zum Zitat Koedel U, Bayerlein I, Paul R, Sporer B, Pfister HW. Pharmacologic interference with NF-kappaB activation attenuates central nervous system complications in experimental Pneumococcal meningitis. J Infect Dis. 2000;182:1437–45.CrossRef Koedel U, Bayerlein I, Paul R, Sporer B, Pfister HW. Pharmacologic interference with NF-kappaB activation attenuates central nervous system complications in experimental Pneumococcal meningitis. J Infect Dis. 2000;182:1437–45.CrossRef
54.
Zurück zum Zitat Selvaraj SK, Periandythevar P, Prasadarao NV. Outer membrane protein A of Escherichia coli K1 selectively enhances the expression of intercellular adhesion molecule-1 in brain microvascular endothelial cells. Microbes Infect. 2007;9:547–57.CrossRef Selvaraj SK, Periandythevar P, Prasadarao NV. Outer membrane protein A of Escherichia coli K1 selectively enhances the expression of intercellular adhesion molecule-1 in brain microvascular endothelial cells. Microbes Infect. 2007;9:547–57.CrossRef
55.
Zurück zum Zitat Chi F, Bo T, Wu CH, Jong A, Huang SH. Vimentin and PSF act in concert to regulate IbeA+ E. coli K1 induced activation and nuclear translocation of NF-kappaB in human brain endothelial cells. PLoS One. 2012;7:e35862.CrossRef Chi F, Bo T, Wu CH, Jong A, Huang SH. Vimentin and PSF act in concert to regulate IbeA+ E. coli K1 induced activation and nuclear translocation of NF-kappaB in human brain endothelial cells. PLoS One. 2012;7:e35862.CrossRef
56.
Zurück zum Zitat Ramsauer K, Sadzak I, Porras A, Pilz A, Nebreda AR, Decker T, Kovarik P. p38 MAPK enhances STAT1-dependent transcription independently of Ser-727 phosphorylation. Proc Natl Acad Sci U S A. 2002;99:12859–64.CrossRef Ramsauer K, Sadzak I, Porras A, Pilz A, Nebreda AR, Decker T, Kovarik P. p38 MAPK enhances STAT1-dependent transcription independently of Ser-727 phosphorylation. Proc Natl Acad Sci U S A. 2002;99:12859–64.CrossRef
Metadaten
Titel
New insights into meningitic Escherichia coli infection of brain microvascular endothelial cells from quantitative proteomics analysis
verfasst von
Wen-Tong Liu
Yu-Jin Lv
Rui-Cheng Yang
Ji-Yang Fu
Lu Liu
Huan Wang
Qi Cao
Chen Tan
Huan-Chun Chen
Xiang-Ru Wang
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/s12974-018-1325-z

Weitere Artikel der Ausgabe 1/2018

Journal of Neuroinflammation 1/2018 Zur Ausgabe

Neu in den Fachgebieten Neurologie und Psychiatrie

Niedriger diastolischer Blutdruck erhöht Risiko für schwere kardiovaskuläre Komplikationen

25.04.2024 Hypotonie Nachrichten

Wenn unter einer medikamentösen Hochdrucktherapie der diastolische Blutdruck in den Keller geht, steigt das Risiko für schwere kardiovaskuläre Ereignisse: Darauf deutet eine Sekundäranalyse der SPRINT-Studie hin.

Frühe Alzheimertherapie lohnt sich

25.04.2024 AAN-Jahrestagung 2024 Nachrichten

Ist die Tau-Last noch gering, scheint der Vorteil von Lecanemab besonders groß zu sein. Und beginnen Erkrankte verzögert mit der Behandlung, erreichen sie nicht mehr die kognitive Leistung wie bei einem früheren Start. Darauf deuten neue Analysen der Phase-3-Studie Clarity AD.

Viel Bewegung in der Parkinsonforschung

25.04.2024 Parkinson-Krankheit Nachrichten

Neue arznei- und zellbasierte Ansätze, Frühdiagnose mit Bewegungssensoren, Rückenmarkstimulation gegen Gehblockaden – in der Parkinsonforschung tut sich einiges. Auf dem Deutschen Parkinsonkongress ging es auch viel um technische Innovationen.

Demenzkranke durch Antipsychotika vielfach gefährdet

23.04.2024 Demenz Nachrichten

Wenn Demenzkranke aufgrund von Symptomen wie Agitation oder Aggressivität mit Antipsychotika behandelt werden, sind damit offenbar noch mehr Risiken verbunden als bislang angenommen.