Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 75
Filter
1.
BMC Med Genomics ; 17(1): 50, 2024 Feb 12.
Article in English | MEDLINE | ID: mdl-38347610

ABSTRACT

BACKGROUND: We aimed to investigate the involvement of long non-coding RNA (lncRNA) in bacterial and viral meningitis in children. METHODS: The peripheral blood of five bacterial meningitis patients, five viral meningitis samples, and five healthy individuals were collected for RNA sequencing. Then, the differentially expressed lncRNA and mRNA were detected in bacterial meningitis vs. controls, viral meningitis vs. healthy samples, and bacterial vs. viral meningitis patients. Besides, co-expression and the competing endogenous RNA (ceRNA) networks were constructed. Receiver operating characteristic curve (ROC) analysis was performed. RESULTS: Compared with the control group, 2 lncRNAs and 32 mRNAs were identified in bacterial meningitis patients, and 115 lncRNAs and 54 mRNAs were detected in viral meningitis. Compared with bacterial meningitis, 165 lncRNAs and 765 mRNAs were identified in viral meningitis. 2 lncRNAs and 31 mRNAs were specific to bacterial meningitis, and 115 lncRNAs and 53 mRNAs were specific to viral meningitis. The function enrichment results indicated that these mRNAs were involved in innate immune response, inflammatory response, and immune system process. A total of 8 and 1401 co-expression relationships were respectively found in bacterial and viral meningitis groups. The ceRNA networks contained 1 lncRNA-mRNA pair and 4 miRNA-mRNA pairs in viral meningitis group. GPR68 and KIF5C, identified in bacterial meningitis co-expression analysis, had an area under the curve (AUC) of 1.00, while the AUC of OR52K2 and CCR5 is 0.883 and 0.698, respectively. CONCLUSIONS: Our research is the first to profile the lncRNAs in bacterial and viral meningitis in children and may provide new insight into understanding meningitis regulatory mechanisms.


Subject(s)
Meningitis, Bacterial , Meningitis, Viral , MicroRNAs , RNA, Long Noncoding , Child , Humans , RNA, Long Noncoding/metabolism , Gene Regulatory Networks , MicroRNAs/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , RNA, Competitive Endogenous , Sequence Analysis, RNA , Meningitis, Bacterial/genetics , Meningitis, Viral/genetics , Receptors, G-Protein-Coupled/genetics , Kinesins/genetics
2.
Infect Genet Evol ; 118: 105553, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38228216

ABSTRACT

Interleukin (IL)-17 A plays a crucial role in protecting hosts from invading bacterial pathogens. In this study, we investigated if single nucleotide polymorphisms (SNPs) in IL-17A are associated with susceptibility and outcome of bacterial meningitis (BM) in Angolan children. The study sample comprised 241 confirmed BM patients and 265 controls, which were matched for age and ethnicity. Three IL-17A SNPs - rs2275913 (-197G > A), rs8193036 (-737C > T) and rs4711998 (-877 A > G) - were determined by high-resolution melting analysis (HRMA). The frequency of variant genotype rs4711998 was significantly higher in patients with BM caused by Haemophilus influenzae (odds ratio [OR] 3.5; 95% confidence interval [CI] 1.49-8.23; P = 0.0025) than in controls. Also, patients with BM caused by Gram-negative bacteria and who carried the variant genotype rs2275913 had a lower glucose level (P = 0.0051) in cerebrospinal fluid (CSF). Patients with BM caused by Streptococcus pneumoniae who carried the variant type rs8193036 had a reduced risk for severe neurological sequelae (OR: 0.14; 95% CI: 0.029-0.68; P = 0.0079), blindness (OR: 0.012; 95% CI: 0.012-0.87; P = 0.017) and ataxia (OR: 0.28; 95% CI: 0.091-0.83; P = 0.023). This study suggests an association of IL-17A genetic variations with susceptibility and outcome of bacterial meningitis in Angolan children.


Subject(s)
Interleukin-17 , Meningitis, Bacterial , Child , Humans , Case-Control Studies , Genetic Predisposition to Disease , Interleukin-17/genetics , Meningitis, Bacterial/genetics , Polymorphism, Single Nucleotide
4.
Front Immunol ; 13: 872832, 2022.
Article in English | MEDLINE | ID: mdl-36119025

ABSTRACT

Bacterial meningitis (BM) is a common life-threatening infection in children that occurs in the central nervous system (CNS). The cytologic examination of cerebrospinal fluid (CSF) is a key parameter in the diagnosis of BM, but the heterogeneity of cells in the CSF has not been elucidated, which limits the current understanding of BM neuroinflammation. In this study, CSF samples were collected from a number of BM patients who were in different stages of disease progression. Single-cell RNA-sequencing (scRNA-seq), with additional bulk transcriptome sequencing, was conducted to decipher the characteristics of CSF cells in BM progression. A total of 18 immune cell clusters in CSF were identified, including two neutrophils, two monocytes, one macrophage, four myeloid dendritic cells, five T cells, one natural killer cell, one B cell, one plasmacytoid dendritic cell, and one plasma cell subtype. Their population profiles and dynamics in the initial onset, remission, and recovery stages during BM progression were also characterized, which showed decreased proportions of myeloid cells and increased proportions of lymphoid cells with disease progression. One novel neutrophil subtype, FFAR2+TNFAIP6+ neutrophils, and one novel monocyte subtype, THBS1+IL1B+ monocytes, were discovered, and their quantity changes positively correlated with the intensity of the inflammatory response in the CSF during BM. In addition, the CSF of BM patients with unsatisfactory therapeutic responses presented with different cell heterogeneity compared to the CSF of BM patients with satisfactory therapeutic responses, and their CSF featured altered intercellular communications and increased proportions of type II myeloid dendritic cells and plasmacytoid dendritic cells. Moreover, the bulk transcriptome profiles of autologous CSF cells and peripheral blood leukocytes of BM patients showed that the immune cells in these two physiological compartments exhibited distinct immune responses under different onset conditions. In particular, the CSF cells showed a high expression of macrophage characteristic genes and a low expression of platelet characteristic genes compared with peripheral blood leukocytes. Our study conducted an in-depth exploration of the characteristics of CSF cells in BM progression, which provided novel insights into immune cell engagement in acute CNS infection.


Subject(s)
Meningitis, Bacterial , Child , Disease Progression , High-Throughput Nucleotide Sequencing , Humans , Meningitis, Bacterial/genetics , Monocytes , RNA
5.
Inflamm Res ; 71(4): 513-520, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35301550

ABSTRACT

OBJECTIVE/DESIGN: Pediatric meningitis is characterized by a colossal inflammatory response to the pathogen in the central nervous system (CNS). This unabated inflammatory response persists even after the removal of the pathogen by antibiotics/steroids causing collateral damage to CNS tissue. Toll-like receptors (TLRs) are the key players in the recognition and elicitation of innate-immune response against bacterial/viral components in cerebrospinal fluid (CSF). Till date, the precise understanding of TLR-triggered inflammatory response in pediatric meningitis is lacking. The present study was designed to delineate the role of TLR transcriptome and downstream signaling pathways in CSF of pediatric meningitis. METHODS: Children in the age group of > 3 months to 12 years with pediatric meningitis were included. A total of 249 cases of pediatric meningitis (bacterial = 89, viral = 160) were included. In addition, 71 children who tested negative to the pathogen in CSF tap and did not have signs of infection clinically constituted the controls. RNA was extracted from the CSF samples of both cases and controls. The relative gene expression profile of 42 TLR signaling pathway genes was performed. For the analysis of secretory cytokines and chemokines in CSF, Luminex assay was performed. RESULTS: We report global upregulation of TLR genes in patients with acute bacterial meningitis (ABM). The downstream signaling molecules were upregulated as well. The CSF of pediatric ABM patients revealed a predominant pro-inflammatory milieu marked by increased levels of pro-inflammatory cytokines. A significant correlation between poor clinical outcomes of patients and an increased expression of TLR/pro-inflammatory cytokine genes was observed. CONCLUSION: Our findings provide support for future studies exploring TLR-based adjunct therapy to limit the neurological sequelae, owing to persistent inflammation in pediatric ABM patients.


Subject(s)
Meningitis, Bacterial , Toll-Like Receptors , Transcriptome , Child , Child, Preschool , Cytokines/genetics , Humans , Meningitis, Bacterial/cerebrospinal fluid , Meningitis, Bacterial/genetics , Signal Transduction , Toll-Like Receptors/genetics
6.
APMIS ; 130(4): 221-229, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35122704

ABSTRACT

This study examined whether gene polymorphisms for toll-like receptor 10 (TLR10) associated with the susceptibility to and outcomes of bacterial meningitis (BM) in Angolan children. The study cohort consisted of 190 BM patients and the determination of ten single-nucleotide polymorphisms (SNPs) by Sanger sequencing. Patients with BM caused by Streptococcus pneumoniae who carried the following variants of TLR10 SNPs exhibited an increased risk of coexisting pneumonia: rs10004195 (T > A) (p = 0.025), rs10856837 (G > A) (p = 0.018) or rs11096956 (G > T) (p = 0.010). Yet, TLR10 SNPs rs11466652 (A > G), rs10856837 (G > A) and rs11096956 (G > T) influenced the protein levels in the cerebrospinal fluid (CSF). Moreover, compared with the wild type, patients with pneumococcal meningitis carrying a variant genotype of TLR10 SNP rs11466648 (A > G) exhibited an increased risk of developing blindness (p = 0.025), whereas patients with TLR10 SNP rs10004195 (T > A) exhibited a lower risk of convulsions at admission (p = 0.039) and a lower risk of altered consciousness (p = 0.029). This study suggests a relationship exists between coexisting pneumonia, protein levels in CSF, blindness, convulsions and an altered consciousness with genetic variations of TLR10 in BM in Angolan children.


Subject(s)
Meningitis, Bacterial , Meningitis, Pneumococcal , Angola/epidemiology , Child , Genotype , Humans , Meningitis, Bacterial/genetics , Meningitis, Pneumococcal/genetics , Polymorphism, Single Nucleotide , Toll-Like Receptor 10/genetics
7.
Mol Brain ; 15(1): 4, 2022 01 03.
Article in English | MEDLINE | ID: mdl-34980188

ABSTRACT

Escherichia coli is the most common gram-negative pathogenic bacterium causing meningitis. It penetrates the blood-brain barrier (BBB) and activates nuclear factor kappa B (NF-κB) signaling, which are vital events leading to the development of meningitis. Long non-coding RNAs (lncRNAs) have been implicated in regulating neuroinflammatory signaling, and our previous study showed that E. coli can induce differential expression of lncRNAs, including lncC11orf54-1, in human brain microvascular endothelial cells (hBMECs). The hBMECs constitute the structural and functional basis for the BBB, however, it is unclear whether lncRNAs are involved in the regulation of inflammatory responses of hBMECs during meningitic E. coli infection. In this study, we characterized an abundantly expressed lncRNA, lncC11orf54-1, which was degraded by translocated coilin to produce mgU2-19 and mgU2-30 in hBMECs during E. coli infection. Functionally, lncC11orf54-1-originated non-coding RNA mgU2-30 interacted with interleukin-1 receptor-associated kinase 1 (IRAK1) to induce its oligomerization and autophosphorylation, thus promoting the activation of NF-κB signaling and facilitating the production of pro-inflammatory cytokines. In summary, our study uncovers the involvement of lncC11orf54-1 in IRAK1-NF-κB signaling, and it functions as a positive regulator of inflammatory responses in meningitic E. coli-induced neuroinflammation, which may be a valuable therapeutic and diagnostic target for bacterial meningitis.


Subject(s)
Escherichia coli Infections , Meningitis, Bacterial , RNA, Long Noncoding , Endothelial Cells/metabolism , Escherichia coli/metabolism , Escherichia coli Infections/metabolism , Humans , Meningitis, Bacterial/genetics , Meningitis, Bacterial/metabolism , Meningitis, Bacterial/microbiology , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism
8.
J Infect Dis ; 224(12 Suppl 2): S194-S203, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34469556

ABSTRACT

BACKGROUND: As part of the global Invasive Bacterial Vaccine-Preventable Diseases Surveillance Network, 12 African countries referred cerebrospinal fluid (CSF) samples to South Africa's regional reference laboratory. We evaluated the utility of real-time polymerase chain reaction (PCR) in detecting and serotyping/grouping Haemophilus influenzae, Neisseria meningitidis, and Streptococcus pneumoniae (HNS). METHODS: From 2008 to 2017, CSF samples collected from children <5 years old with suspected meningitis underwent routine microbiology testing in-country, and 11 680 samples were submitted for HNS PCR at the regional reference laboratory. Unconditional logistic regression, with adjustment for geographic location, was performed to identify factors associated with PCR positivity. RESULTS: The overall HNS PCR positivity rate for all countries was 10% (1195 of 11 626 samples). In samples with both PCR and culture results, HNS PCR positivity was 11% (744 of 6747 samples), and HNS culture positivity was 3% (207 of 6747). Molecular serotype/serogroup was assigned in 75% of PCR-positive specimens (762 of 1016). Compared with PCR-negative CSF samples, PCR-positive samples were more often turbid (adjusted odds ratio, 6.80; 95% confidence interval, 5.67-8.17) and xanthochromic (1.72; 1.29-2.28), had elevated white blood cell counts (6.13; 4.71-7.99) and high protein concentrations (5.80; 4.34-7.75), and were more often HNS culture positive (32.70; 23.18-46.12). CONCLUSION: PCR increased detection of vaccine-preventable bacterial meningitis in countries where confirmation of suspected meningitis cases is impeded by limited culture capacity.


Subject(s)
Haemophilus influenzae/genetics , Meningitis, Bacterial/diagnosis , Neisseria meningitidis/genetics , Real-Time Polymerase Chain Reaction/methods , Streptococcus pneumoniae/genetics , Africa, Eastern/epidemiology , Africa, Southern/epidemiology , Bacterial Vaccines/therapeutic use , Child, Preschool , Female , Haemophilus influenzae/isolation & purification , Humans , Infant , Infant, Newborn , Male , Meningitis, Bacterial/epidemiology , Meningitis, Bacterial/genetics , Molecular Diagnostic Techniques , Neisseria meningitidis/isolation & purification , Public Health Surveillance , Streptococcus pneumoniae/isolation & purification
9.
Brain Res ; 1770: 147611, 2021 11 01.
Article in English | MEDLINE | ID: mdl-34403663

ABSTRACT

BACKGROUND: Bacterial meningitis (BM) is a serious infectious disease of the central nervous system that often occurs in children and adolescents. Many studies have suggested that microRNAs (miRNAs) are involved in BM. This study aimed to address the effects of miR-141-3p on astrocyte activation and inflammatory response in BM through HMGB1. METHODS: The 3-week-old rats were injected with Streptococcus pneumoniae (SP) into the lateral ventricle to establish a BM model. Loeffler scoring method was used to evaluate the recovery of neurological function. Brain pathological damage was observed by hematoxylin and eosin (H&E) staining. Primary astrocytes were isolated from brain tissues of BM or non-infected SD rats. The levels of TNF-α, IL-1ß, and IL-6 in brain tissues and astrocyte culture supernatant were measured by enzyme-linked immunosorbent assay (ELISA). The targeting relationship between miR-141-3p and HMGB1 was tested using dual-luciferase reporter assay. The expression of miR-141-3p, HMGB1, and the astrocytic marker glial fibrillary acidic protein (GFAP) were detected by quantitative real-time polymerase chain reaction (qRT-PCR) or western blotting. Methylation-specific PCR (MSP) analysis was performed to measure the methylation status of miR-141 promoter. RESULTS: The results showed that lower Loeffler scores were exhibited in rats with BM. The subarachnoid space of brain tissues of BM rats was widened, and obvious inflammatory cells were observed. miR-141-3p expression was reduced in BM rats and SP-treated astrocytes. Additionally, we found that overexpression of miR-141-3p led to the downregulation of HMGB1, GFAP, and inflammatory cytokines (TNF-α, IL-1ß, and IL-6) in astrocytes. Furthermore, the results of dual-luciferase reporter assay confirmed that miR-141-3p directly targeted HMGB1. Overexpression of miR-141-3p inhibited the levels of GFAP, TNF-α, IL-1ß, and IL-6 in astrocytes, which was eliminated by the up-regulation of HMGB1. The results of MSP analysis indicated that miR-141 promoter was highly methylated in brain tissues and astrocytes. DNMT1 was involved in the methylation of miR-141 promoter in BM. CONCLUSION: The present study verified that miR-141-3p affected inflammatory response by suppressing HMGB1 in SP-induced astrocytes and BM rat model.


Subject(s)
Astrocytes/metabolism , Cytokines/metabolism , Down-Regulation , HMGB1 Protein/metabolism , Meningitis, Bacterial/metabolism , MicroRNAs/metabolism , Animals , Brain/metabolism , HMGB1 Protein/genetics , Inflammation/metabolism , Interleukin-1beta/metabolism , Interleukin-6/metabolism , Meningitis, Bacterial/genetics , MicroRNAs/genetics , Rats , Rats, Sprague-Dawley , Tumor Necrosis Factor-alpha/metabolism
10.
Infect Genet Evol ; 93: 104984, 2021 09.
Article in English | MEDLINE | ID: mdl-34214672

ABSTRACT

Bacterial meningitis (BM) is a serious infectious disease of the central nervous system,which is mainly caused by Streptococcus pneumoniae, Neisseria meningitidis, Haemophilus influenzae, Group B Streptococcus and Listeria monocytogenes. Throughout the world, BM has become one of the most lethal diseases that commonly occurs in children. Toll like receptors (TLRs) are one of the most important immune defense lines in infectious diseases, and play an essential role in host defense. Accumulating evidence shows that genetic variations in TLRs are associated with host responses in BM. This review aims to summarize the role of different TLRs and their genetic variations in the susceptibility, severity and prognosis of BM and discuss the identified risk factors for better treatment and improvement of the course and outcome of BM.


Subject(s)
Genetic Predisposition to Disease , Genetic Variation , Meningitis, Bacterial/diagnosis , Toll-Like Receptors/genetics , Disease Susceptibility/microbiology , Humans , Meningitis, Bacterial/genetics , Meningitis, Bacterial/microbiology , Prognosis , Severity of Illness Index , Toll-Like Receptors/metabolism
11.
Microvasc Res ; 138: 104225, 2021 11.
Article in English | MEDLINE | ID: mdl-34256086

ABSTRACT

PURPOSE: Blood-brain barrier (BBB) damage is closely related to various neurological disorders, including bacterial meningitis (BM). Determining a reliable strategy to prevent BBB damage in the context of infection would be highly desirable. In the present study, we investigated the implications of the long non-coding RNA (lncRNA) nuclear paraspeckle assembly transcript 1 (NEAT1) in moderating BBB damage. METHODS: In vitro BBB models were developed by co-culturing hCMEC/D3 cells with glioma cells, whereupon the glioma-exposed endothelial cells (GECs) were treated with a series of mimics, inhibitors, overexpression plasmids, and shRNAs for evaluating whether NEAT1, microRNA-135a (miR-135a) and hypoxia-inducible factor 1α (HIF1α) mediated BBB integrity and permeability. Furthermore, the in vivo biological function of NEAT1 was validated in a mouse model of BBB damage. RESULTS: NEAT1 and HIF1α were determined to be up-regulated, while miR-135a was under-expressed in GECs. As demonstrated by chromatin immunoprecipitation and dual-luciferase reporter assays, NEAT1 could bind to miR-135a, and HIF1α was confirmed as a target of miR-135a. Either overexpression of NEAT1 or depletion of miR-135a impaired the integrity and augmented the permeability of BBB. However, HIF1α silencing could reverse the BBB damage induced by NEAT1 overexpression or by inhibition of miR-135a. In vivo experiments substantiated that knockdown of NEAT1 could alleviate BBB damage in living mice. CONCLUSIONS: Hence, NEAT1 knockdown prevents BBB disruption and exerts promise as a potential target for BM treatment.


Subject(s)
Blood-Brain Barrier/metabolism , Capillary Permeability , Meningitis, Bacterial/metabolism , RNA, Long Noncoding/metabolism , Animals , Blood-Brain Barrier/pathology , Cell Line, Tumor , Coculture Techniques , Disease Models, Animal , Gene Expression Regulation , HEK293 Cells , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lipopolysaccharides , Male , Meningitis, Bacterial/chemically induced , Meningitis, Bacterial/genetics , Meningitis, Bacterial/pathology , Mice, Inbred ICR , MicroRNAs/genetics , MicroRNAs/metabolism , RNA, Long Noncoding/genetics
12.
Int J Mol Sci ; 22(12)2021 Jun 14.
Article in English | MEDLINE | ID: mdl-34198485

ABSTRACT

Brain microvascular endothelial cells (BMECs) constitute the structural and functional basis for the blood-brain barrier (BBB) and play essential roles in bacterial meningitis. Although the BBB integrity regulation has been under extensive investigation, there is little knowledge regarding the roles of long non-coding RNAs (lncRNAs) in this event. The present study aimed to investigate the roles of one potential lncRNA, lncRSPH9-4, in meningitic E. coli infection of BMECs. LncRSPH9-4 was cytoplasm located and significantly up-regulated in meningitic E. coli-infected hBMECs. Electrical cell-substrate impedance sensing (ECIS) measurement and Western blot assay demonstrated lncRSPH9-4 overexpression in hBMECs mediated the BBB integrity disruption. By RNA-sequencing analysis, 639 mRNAs and 299 miRNAs were significantly differentiated in response to lncRSPH9-4 overexpression. We further found lncRSPH9-4 regulated the permeability in hBMECs by competitively sponging miR-17-5p, thereby increasing MMP3 expression, which targeted the intercellular tight junctions. Here we reported the infection-induced lncRSPH9-4 aggravated disruption of the tight junctions in hBMECs, probably through the miR-17-5p/MMP3 axis. This finding provides new insights into the function of lncRNAs in BBB integrity during meningitic E. coli infection and provides the novel nucleic acid targets for future treatment of bacterial meningitis.


Subject(s)
Blood-Brain Barrier/metabolism , Blood-Brain Barrier/pathology , Escherichia coli/physiology , Matrix Metalloproteinase 3/metabolism , Meningitis, Bacterial/genetics , Meningitis, Bacterial/microbiology , MicroRNAs/metabolism , RNA, Long Noncoding/metabolism , Base Sequence , Cytoplasm/metabolism , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Gene Regulatory Networks , Humans , MicroRNAs/genetics , Microvessels/pathology , Models, Biological , Permeability , RNA, Long Noncoding/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Signal Transduction , Tight Junctions/metabolism , Transcription, Genetic , Up-Regulation/genetics
13.
J Clin Invest ; 131(5)2021 03 01.
Article in English | MEDLINE | ID: mdl-33465054

ABSTRACT

Group B Streptococcus (GBS) is the major cause of human neonatal infections. A single clone, designated CC17-GBS, accounts for more than 80% of meningitis cases, the most severe form of the infection. However, the events allowing blood-borne GBS to penetrate the brain remain largely elusive. In this study, we identified the host transmembrane receptors α5ß1 and αvß3 integrins as the ligands of Srr2, a major CC17-GBS-specific adhesin. Two motifs located in the binding region of Srr2 were responsible for the interaction between CC17-GBS and these integrins. We demonstrated in a blood-brain-barrier cellular model that both integrins contributed to the adhesion and internalization of CC17-GBS. Strikingly, both integrins were overexpressed during the postnatal period in the brain vessels of the blood-brain barrier and blood-cerebrospinal fluid barrier and contributed to juvenile susceptibility to CC17 meningitis. Finally, blocking these integrins decreased the ability of CC17-GBS to cross into the CNS of juvenile mice in an in vivo model of meningitis. Our study demonstrated that CC17-GBS exploits integrins in order to cross the brain vessels, leading to meningitis. Importantly, it provides host molecular insights into neonate's susceptibility to CC17-GBS meningitis, thereby opening new perspectives for therapeutic and prevention strategies of GBS-elicited meningitis.


Subject(s)
Adhesins, Bacterial/metabolism , Blood-Brain Barrier/metabolism , Integrin alphaVbeta3/metabolism , Meningitis, Bacterial/metabolism , Receptors, Vitronectin/metabolism , Streptococcal Infections/metabolism , Streptococcus agalactiae/metabolism , Adhesins, Bacterial/genetics , Animals , Animals, Newborn , Bacterial Adhesion/genetics , Blood-Brain Barrier/microbiology , Cell Line , Humans , Integrin alphaVbeta3/genetics , Meningitis, Bacterial/genetics , Rats , Receptors, Vitronectin/genetics , Streptococcal Infections/genetics , Streptococcus agalactiae/genetics
14.
mBio ; 11(6)2020 11 10.
Article in English | MEDLINE | ID: mdl-33173000

ABSTRACT

Nutritional immunity is an elegant host mechanism used to starve invading pathogens of necessary nutrient metals. Calprotectin, a metal-binding protein, is produced abundantly by neutrophils and is found in high concentrations within inflammatory sites during infection. Group B Streptococcus (GBS) colonizes the gastrointestinal and female reproductive tracts and is commonly associated with severe invasive infections in newborns such as pneumonia, sepsis, and meningitis. Although GBS infections induce robust neutrophil recruitment and inflammation, the dynamics of GBS and calprotectin interactions remain unknown. Here, we demonstrate that disease and colonizing isolate strains exhibit susceptibility to metal starvation by calprotectin. We constructed a mariner transposon (Krmit) mutant library in GBS and identified 258 genes that contribute to surviving calprotectin stress. Nearly 20% of all underrepresented mutants following treatment with calprotectin are predicted metal transporters, including known zinc systems. As calprotectin binds zinc with picomolar affinity, we investigated the contribution of GBS zinc uptake to overcoming calprotectin-imposed starvation. Quantitative reverse transcriptase PCR (qRT-PCR) revealed a significant upregulation of genes encoding zinc-binding proteins, adcA, adcAII, and lmb, following calprotectin exposure, while growth in calprotectin revealed a significant defect for a global zinc acquisition mutant (ΔadcAΔadcAIIΔlmb) compared to growth of the GBS wild-type (WT) strain. Furthermore, mice challenged with the ΔadcAΔadcAIIΔlmb mutant exhibited decreased mortality and significantly reduced bacterial burden in the brain compared to mice infected with WT GBS; this difference was abrogated in calprotectin knockout mice. Collectively, these data suggest that GBS zinc transport machinery is important for combatting zinc chelation by calprotectin and establishing invasive disease.IMPORTANCE Group B Streptococcus (GBS) asymptomatically colonizes the female reproductive tract but is a common causative agent of meningitis. GBS meningitis is characterized by extensive infiltration of neutrophils carrying high concentrations of calprotectin, a metal chelator. To persist within inflammatory sites and cause invasive disease, GBS must circumvent host starvation attempts. Here, we identified global requirements for GBS survival during calprotectin challenge, including known and putative systems involved in metal ion transport. We characterized the role of zinc import in tolerating calprotectin stress in vitro and in a mouse model of infection. We observed that a global zinc uptake mutant was less virulent than the parental GBS strain and found calprotectin knockout mice to be equally susceptible to infection by wild-type (WT) and mutant strains. These findings suggest that calprotectin production at the site of infection results in a zinc-limited environment and reveals the importance of GBS metal homeostasis to invasive disease.


Subject(s)
Leukocyte L1 Antigen Complex/metabolism , Streptococcal Infections/metabolism , Streptococcus agalactiae/metabolism , Zinc/metabolism , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Female , Humans , Leukocyte L1 Antigen Complex/genetics , Meningitis, Bacterial/genetics , Meningitis, Bacterial/metabolism , Meningitis, Bacterial/microbiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/metabolism , Streptococcal Infections/genetics , Streptococcal Infections/microbiology , Streptococcus agalactiae/genetics , Streptococcus agalactiae/growth & development , Streptococcus agalactiae/pathogenicity , Virulence
15.
PLoS One ; 15(11): e0242628, 2020.
Article in English | MEDLINE | ID: mdl-33211777

ABSTRACT

Meningitis is one of the top ten causes of death among Ethiopian infants. Group B streptococcus (GBS) has emerged as a leading cause of meningitis in neonates and young infants, resulting in high mortality. Despite this, there is no report on GBS associated meningitis in Ethiopia where infant meningitis is common. Hence, the aim of this study was to determine the proportion of GBS associated meningitis among Ethiopian infants. PCR was prospectively used to detect GBS in culture-negative cerebrospinal fluid (CSF) samples, which were collected from infants suspected for meningitis, at Tikur Anbessa specialized hospital, Ethiopia, over a one-year period. GBS was detected by PCR in 63.9% of culture-negative CSF samples. Out of the 46 GBS positive infants, 10.9% (n = 5) of them died. The late onset of GBS (LOGBS) disease was noted to have a poor outcome with 3 LOGBS out of 5 GBS positive samples collected from patients with the final outcome of death. PCR was advantageous in the identification of GBS in culture-negative CSF samples. GBS was detected in 64% of the CSF samples from infants with meningitis compared with zero-detection rate by culture.


Subject(s)
Meningitis, Bacterial , Polymerase Chain Reaction , Streptococcal Infections , Streptococcus agalactiae/genetics , Cross-Sectional Studies , Ethiopia/epidemiology , Female , Humans , Infant , Infant, Newborn , Male , Meningitis, Bacterial/cerebrospinal fluid , Meningitis, Bacterial/epidemiology , Meningitis, Bacterial/genetics , Streptococcal Infections/cerebrospinal fluid , Streptococcal Infections/epidemiology , Streptococcal Infections/genetics
16.
Epidemiol Infect ; 148: e294, 2020 11 04.
Article in English | MEDLINE | ID: mdl-33143777

ABSTRACT

Some studies have suggested that the Toll-like receptor 9 polymorphism (TLR9 rs352140) is closely related to the risk of bacterial meningitis (BM), but this is subject to controversy. This study set out to estimate whether the TLR9 rs352140 polymorphism confers an increased risk of BM. Relevant literature databases were searched including PubMed, Embase, the Cochrane Library and China National Knowledge Infrastructure (CNKI) up to August 2020. Seven case-control studies from four publications were enrolled in the present meta-analysis. Odds ratios (OR) and confidence intervals (95% CI) were calculated to estimate associations between BM risk and the target polymorphism. Significant associations identified were allele contrast (A vs. G: OR 0.66, 95% CI 0.59-0.75, P = 0.000), homozygote comparison (AA vs. AG/GG: OR 0.62, 95% CI 0.49-0.78, P = 0.000), heterozygote comparison (A vs. G: OR 0.74, 95% CI 0.61-0.91, P = 0.005), recessive genetic model (AA vs. AG/GG: OR 0.78, 95% CI 0.65-0.93, P = 0.006) and dominant genetic model (AA vs. AG/GG: OR 0.70, 95% CI 0.57-0.85, P = 0.000). The findings indicate that, in contrast to some studies, the TLR9 rs352140 polymorphism is associated with a decreased risk for BM.


Subject(s)
Bacterial Infections/genetics , Genetic Predisposition to Disease , Meningitis, Bacterial/genetics , Toll-Like Receptor 9/metabolism , Bacteria/classification , Bacterial Infections/metabolism , Humans , Odds Ratio , Polymorphism, Single Nucleotide , Toll-Like Receptor 9/genetics
17.
CNS Neurosci Ther ; 26(12): 1288-1302, 2020 12.
Article in English | MEDLINE | ID: mdl-32790044

ABSTRACT

BACKGROUND: Specific highly polarized aquaporin-4 (AQP4) expression is reported to play a crucial role in blood-brain barrier (BBB) integrity and brain water transport balance. The upregulation of polymerase δ-interacting protein 2 (Poldip2) was involved in aggravating BBB disruption following ischemic stroke. This study aimed to investigate whether Poldip2-mediated BBB disruption and cerebral edema formation in mouse bacterial meningitis (BM) model occur via induction of AQP4 polarity loss. METHODS AND RESULTS: Mouse BM model was induced by injecting mice with group B hemolytic streptococci via posterior cistern. Recombinant human Poldip2 (rh-Poldip2) was administered intranasally at 1 hour after BM induction. Small interfering ribonucleic acid (siRNA) targeting Poldip2 was administered by intracerebroventricular (i.c.v) injection at 48 hours before BM induction. A specific inhibitor of matrix metalloproteinases (MMPs), UK383367, was administered intravenously at 0.5 hour before BM induction. Western blotting, immunofluorescence staining, quantitative real-time PCR, neurobehavioral test, brain water content test, Evans blue (EB) permeability assay, transmission electron microscopy (TEM), and gelatin zymography were carried out. The results showed that Poldip2 was upregulated and AQP4 polarity was lost in mouse BM model. Both Poldip2 siRNA and UK383367 improved neurobehavioral outcomes, alleviated brain edema, preserved the integrity of BBB, and relieved the loss of AQP4 polarity in BM model. Rh-Poldip2 upregulated the expression of MMPs and glial fibrillary acidic protein (GFAP) and downregulated the expression of ß-dystroglycan (ß-DG), zonula occludens-1 (ZO-1), occludin, and claudin-5; whereas Poldip2 siRNA downregulated the expression of MMPs and GFAP, and upregulated ß-DG, ZO-1, occludin, and claudin-5. Similarly, UK383367 downregulated the expression of GFAP and upregulated the expression of ß-DG, ZO-1, occludin, and claudin-5. CONCLUSION: Poldip2 inhibition alleviated brain edema and preserved the integrity of BBB partially by relieving the loss of AQP4 polarity via MMPs/ß-DG pathway.


Subject(s)
Aquaporin 4/biosynthesis , Blood-Brain Barrier/metabolism , Brain Edema/metabolism , Disease Models, Animal , Meningitis, Bacterial/metabolism , Mitochondrial Proteins/biosynthesis , Nuclear Proteins/biosynthesis , Administration, Intranasal , Animals , Aquaporin 4/genetics , Blood-Brain Barrier/pathology , Brain Edema/genetics , Brain Edema/pathology , Humans , Male , Meningitis, Bacterial/genetics , Meningitis, Bacterial/pathology , Mice , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , Nuclear Proteins/deficiency , Nuclear Proteins/genetics
18.
Int J Mol Sci ; 21(15)2020 Jul 25.
Article in English | MEDLINE | ID: mdl-32722502

ABSTRACT

Streptococcus suis is a zoonotic agent causing meningitis in pigs and humans. Neutrophils, as the first line of defense against S. suis infections, release neutrophil extracellular traps (NETs) to entrap pathogens. In this study, we investigated the role of the secreted nuclease A of S. suis (SsnA) as a NET-evasion factor in vivo and in vitro. Piglets were intranasally infected with S. suis strain 10 or an isogenic ssnA mutant. DNase and NET-formation were analyzed in cerebrospinal fluid (CSF) and brain tissue. Animals infected with S. suis strain 10 or S. suis 10ΔssnA showed the presence of NETs in CSF and developed similar clinical signs. Therefore, SsnA does not seem to be a crucial virulence factor that contributes to the development of meningitis in pigs. Importantly, DNase activity was detectable in the CSF of both infection groups, indicating that host nucleases, in contrast to bacterial nuclease SsnA, may play a major role during the onset of meningitis. The effect of DNase 1 on neutrophil functions was further analyzed in a 3D-cell culture model of the porcine blood-CSF barrier. We found that DNase 1 partially contributes to enhanced killing of S. suis by neutrophils, especially when plasma is present. In summary, host nucleases may partially contribute to efficient innate immune response in the CSF.


Subject(s)
Bacterial Proteins/metabolism , Deoxyribonuclease I/metabolism , Meningitis, Bacterial/enzymology , Neutrophils/enzymology , Streptococcal Infections/enzymology , Streptococcus suis/enzymology , Swine Diseases/enzymology , Animals , Meningitis, Bacterial/genetics , Meningitis, Bacterial/veterinary , Mutation , Streptococcal Infections/genetics , Streptococcal Infections/veterinary , Streptococcus suis/genetics , Swine , Swine Diseases/genetics
19.
J Neuroinflammation ; 16(1): 279, 2019 Dec 28.
Article in English | MEDLINE | ID: mdl-31883521

ABSTRACT

BACKGROUND: The complement system is a vital component of the inflammatory response occurring during bacterial meningitis. Blocking the complement system was shown to improve the outcome of experimental pneumococcal meningitis. Complement factor H (FH) is a complement regulatory protein inhibiting alternative pathway activation but is also exploited by the pneumococcus to prevent complement activation on its surface conferring serum resistance. METHODS: In a nationwide prospective cohort study of 1009 episodes with community-acquired bacterial meningitis, we analyzed whether genetic variations in CFH influenced FH cerebrospinal fluid levels and/or disease severity. Subsequently, we analyzed the role of FH in our pneumococcal meningitis mouse model using FH knock-out (Cfh-/-) mice and wild-type (wt) mice. Finally, we tested whether adjuvant treatment with human FH (hFH) improved outcome in a randomized investigator blinded trial in a pneumococcal meningitis mouse model. RESULTS: We found the major allele (G) of single nucleotide polymorphism in CFH (rs6677604) to be associated with low FH cerebrospinal fluid concentration and increased mortality. In patients and mice with bacterial meningitis, FH concentrations were elevated during disease and Cfh-/- mice with pneumococcal meningitis had increased mortality compared to wild-type mice due to C3 depletion. Adjuvant treatment of wild-type mice with purified human FH led to complement inhibition but also increased bacterial outgrowth which resulted in similar disease outcomes. CONCLUSION: Low FH levels contribute to mortality in pneumococcal meningitis but adjuvant treatment with FH at a clinically relevant time point is not beneficial.


Subject(s)
Complement Factor H/cerebrospinal fluid , Complement Factor H/genetics , Meningitis, Bacterial/genetics , Meningitis, Bacterial/immunology , Meningitis, Bacterial/mortality , Adult , Aged , Animals , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Polymorphism, Single Nucleotide
20.
J Transl Med ; 17(1): 282, 2019 08 23.
Article in English | MEDLINE | ID: mdl-31443725

ABSTRACT

BACKGROUND: Meningitis can be caused by several viruses and bacteria. Identifying the causative pathogen as quickly as possible is crucial to initiate the most optimal therapy, as acute bacterial meningitis is associated with a significant morbidity and mortality. Bacterial meningitis requires antibiotics, as opposed to enteroviral meningitis, which only requires supportive therapy. Clinical presentation is usually not sufficient to differentiate between viral and bacterial meningitis, thereby necessitating cerebrospinal fluid (CSF) analysis by PCR and/or time-consuming bacterial cultures. However, collecting CSF in children is not always feasible and a rather invasive procedure. METHODS: In 12 Belgian hospitals, we obtained acute blood samples from children with signs of meningitis (49 viral and 7 bacterial cases) (aged between 3 months and 16 years). After pathogen confirmation on CSF, the patient was asked to give a convalescent sample after recovery. 3' mRNA sequencing was performed to determine differentially expressed genes (DEGs) to create a host transcriptomic profile. RESULTS: Enteroviral meningitis cases displayed the largest upregulated fold change enrichment in type I interferon production, response and signaling pathways. Patients with bacterial meningitis showed a significant upregulation of genes related to macrophage and neutrophil activation. We found several significantly DEGs between enteroviral and bacterial meningitis. Random forest classification showed that we were able to differentiate enteroviral from bacterial meningitis with an AUC of 0.982 on held-out samples. CONCLUSIONS: Enteroviral meningitis has an innate immunity signature with type 1 interferons as key players. Our classifier, based on blood host transcriptomic profiles of different meningitis cases, is a possible strong alternative for diagnosing enteroviral meningitis.


Subject(s)
Enterovirus Infections/blood , Enterovirus Infections/genetics , Meningitis, Viral/diagnosis , Meningitis, Viral/genetics , Spinal Puncture , Transcriptome/genetics , Adolescent , Child , Child, Preschool , Enterovirus Infections/diagnosis , Gene Expression Regulation , Gene Ontology , Humans , Infant , Meningitis, Bacterial/genetics , Meningitis, Viral/blood , ROC Curve
SELECTION OF CITATIONS
SEARCH DETAIL
...