Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
Autophagy ; : 1-20, 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38497494

ABSTRACT

Streptococcus pneumoniae (S. pneumoniae) represents a major human bacterial pathogen leading to high morbidity and mortality in children and the elderly. Recent research emphasizes the role of extracellular vesicles (EVs) in bacterial pathogenicity. However, the contribution of S. pneumoniae EVs (pEVs) to host-microbe interactions has remained unclear. Here, we observed that S. pneumoniae infections in mice led to severe lung injuries and alveolar epithelial barrier (AEB) dysfunction. Infections of S. pneumoniae reduced the protein expression of tight junction protein OCLN (occludin) and activated macroautophagy/autophagy in lung tissues of mice and A549 cells. Mechanically, S. pneumoniae induced autophagosomal degradation of OCLN leading to AEB impairment in the A549 monolayer. S. pneumoniae released the pEVs that could be internalized by alveolar epithelial cells. Through proteomics, we profiled the cargo proteins inside pEVs and found that these pEVs contained many virulence factors, among which we identified a eukaryotic-like serine-threonine kinase protein StkP. The internalized StkP could induce the phosphorylation of BECN1 (beclin 1) at Ser93 and Ser96 sites, initiating autophagy and resulting in autophagy-dependent OCLN degradation and AEB dysfunction. Finally, the deletion of stkP in S. pneumoniae completely protected infected mice from death, significantly alleviated OCLN degradation in vivo, and largely abolished the AEB disruption caused by pEVs in vitro. Overall, our results suggested that pEVs played a crucial role in the spread of S. pneumoniae virulence factors. The cargo protein StkP in pEVs could communicate with host target proteins and even hijack the BECN1 autophagy initiation pathway, contributing to AEB disruption and bacterial pathogenicity.Abbreviations: AEB: alveolarepithelial barrier; AECs: alveolar epithelial cells; ATG16L1: autophagy related 16 like 1; ATP:adenosine 5'-triphosphate; BafA1: bafilomycin A1; BBB: blood-brain barrier; CFU: colony-forming unit; co-IP: co-immunoprecipitation; CQ:chloroquine; CTRL: control; DiO: 3,3'-dioctadecylox-acarbocyanineperchlorate; DOX: doxycycline; DTT: dithiothreitol; ECIS: electricalcell-substrate impedance sensing; eGFP: enhanced green fluorescentprotein; ermR: erythromycin-resistance expression cassette; Ery: erythromycin; eSTKs: eukaryotic-like serine-threoninekinases; EVs: extracellular vesicles; HA: hemagglutinin; H&E: hematoxylin and eosin; HsLC3B: human LC3B; hpi: hours post-infection; IP: immunoprecipitation; KD: knockdown; KO: knockout; LAMP1: lysosomal associated membrane protein 1; LC/MS: liquid chromatography-mass spectrometry; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MVs: membranevesicles; NC:negative control; NETs:neutrophil extracellular traps; OD: optical density; OMVs: outer membrane vesicles; PBS: phosphate-buffered saline; pEVs: S.pneumoniaeextracellular vesicles; protK: proteinase K; Rapa: rapamycin; RNAi: RNA interference; S.aureus: Staphylococcusaureus; SNF:supernatant fluid; sgRNA: single guide RNA; S.pneumoniae: Streptococcuspneumoniae; S.suis: Streptococcussuis; TEER: trans-epithelium electrical resistance; moi: multiplicity ofinfection; TEM:transmission electron microscope; TJproteins: tight junction proteins; TJP1/ZO-1: tight junction protein1; TSA: tryptic soy agar; WB: western blot; WT: wild-type.

2.
Heliyon ; 10(5): e26986, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38455548

ABSTRACT

The COVID-19 pandemic has greatly changed global practices of enterprise digital transformation (EDT). However, the impact of the pandemic on EDT research patterns remains unexplored. This study examines the overall development and research pattern shift of literature on EDT in the field of business and economics. A bibliometric analysis with CiteSpace was conducted on a total of 140 journal articles indexed the SSCI and SCIE databases on Web of Science prior to the pandemic and 621 articles published after the pandemic. The results suggest that following the outbreak of COVID-19 pandemic, there has been a significantly rapid growth of EDT-related publications, and the contributing role in EDT research of influential countries has undergone significant changes. Furthermore, the changes in keyword patterns were identified before and after the pandemic. Specifically, EDT research after the COVID-19 outbreak has been focusing on emerging topics, such as corporate governance, sustainable development, platform ecosystems, and dynamic capabilities. Finally, recommendations for future research are provided at individual, organizational, and ecosystem levels. Overall, this study is one of the first studies to uncover the dynamics of EDT research patterns due to the COVID-19 Pandemic, thus enhancing our understanding of the features and structures of digital transformation research in uncertain environment.

3.
ACS Infect Dis ; 10(3): 988-999, 2024 Mar 08.
Article in English | MEDLINE | ID: mdl-38317607

ABSTRACT

Escherichia coli continues to be the predominant Gram-negative pathogen causing neonatal meningitis worldwide. Inflammatory mediators have been implicated in the pathogenesis of meningitis and are key therapeutic targets. The role of interleukin-22 (IL-22) in various diseases is diverse, with both protective and pathogenic effects. However, little is understood about the mechanisms underlying the damaging effects of IL-22 on the blood-brain barrier (BBB) in E. coli meningitis. We observed that meningitic E. coli infection induced IL-22 expression in the serum and brain of mice. The tight junction proteins (TJPs) components ZO-1, Occludin, and Claudin-5 were degraded in the mouse brain and human brain microvascular endothelial cells (hBMEC) following IL-22 administration. Moreover, the meningitic E. coli-caused increase in BBB permeability in wild-type mice was restored by knocking out IL-22. Mechanistically, IL-22 activated the STAT3-VEGFA signaling cascade in E. coli meningitis, thus eliciting the degradation of TJPs to induce BBB disruption. Our data indicated that IL-22 is an essential host accomplice during E. coli-caused BBB disruption and could be targeted for the therapy of bacterial meningitis.


Subject(s)
Escherichia coli Infections , Meningitis, Bacterial , Meningitis, Escherichia coli , Humans , Mice , Animals , Blood-Brain Barrier , Meningitis, Escherichia coli/metabolism , Meningitis, Escherichia coli/microbiology , Meningitis, Escherichia coli/pathology , Escherichia coli/metabolism , Endothelial Cells , Interleukin-22 , Vascular Endothelial Growth Factor A/metabolism , Vascular Endothelial Growth Factor A/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , STAT3 Transcription Factor/pharmacology
4.
Cell Commun Signal ; 22(1): 123, 2024 02 15.
Article in English | MEDLINE | ID: mdl-38360663

ABSTRACT

BACKGROUND: Meningitic Escherichia coli (E. coli) is the major etiological agent of bacterial meningitis, a life-threatening infectious disease with severe neurological sequelae and high mortality. The major cause of central nervous system (CNS) damage and sequelae is the bacterial-induced inflammatory storm, where the immune response of the blood-brain barrier (BBB) is crucial. METHODS: Western blot, real-time PCR, enzyme-linked immunosorbent assay, immunofluorescence, and dual-luciferase reporter assay were used to investigate the suppressor role of transforming growth factor beta 1 (TGFß1) in the immune response of brain microvascular endothelial cells elicited by meningitic E. coli. RESULT: In this work, we showed that exogenous TGFß1 and induced noncanonical Hedgehog (HH) signaling suppressed the endothelial immune response to meningitic E. coli infection via upregulation of intracellular miR-155. Consequently, the increased miR-155 suppressed ERK1/2 activation by negatively regulating KRAS, thereby decreasing IL-6, MIP-2, and E-selectin expression. In addition, the exogenous HH signaling agonist SAG demonstrated promising protection against meningitic E. coli-induced neuroinflammation. CONCLUSION: Our work revealed the effect of TGFß1 antagonism on E. coli-induced BBB immune response and suggested that activation of HH signaling may be a potential protective strategy for future bacterial meningitis therapy. Video Abstract.


Subject(s)
Meningitis, Bacterial , Meningitis, Escherichia coli , MicroRNAs , Humans , Escherichia coli/genetics , Hedgehog Proteins/metabolism , Endothelial Cells/metabolism , Meningitis, Escherichia coli/metabolism , Brain/metabolism , Blood-Brain Barrier/microbiology , Meningitis, Bacterial/metabolism , Immunity , MicroRNAs/metabolism
5.
Mol Neurobiol ; 2024 Feb 19.
Article in English | MEDLINE | ID: mdl-38372957

ABSTRACT

Numerous studies have shown that exosomes play a regulatory role in a variety of biological processes as well as in disease development and progression. However, exosome-mediated intercellular communication between brain microvascular endothelial cells (BMECs) and astrocytes during meningitic Escherichia coli (E. coli)-induced neuroinflammation remains largely unknown. Here, by using in vivo and in vitro models, we demonstrate that exosomes derived from meningitic E. coli-infected BMECs can activate the inflammatory response of astrocytes. A label-free quantitation approach coupled with LC-MS/MS was used to compare the exosome proteomic profiles of human BMECs (hBMECs) in response to meningitic E. coli infection. A total of 57 proteins exhibited significant differences in BMEC-derived exosomes during the infection. Among these proteins, growth differentiation factor 15 (GDF15) was significantly increased in BMEC-derived exosomes during the infection, which triggered the Erk1/2 signaling pathway and promoted the activation of astrocytes. The identification and characterization of exosome protein profiles in BMECs during meningitic E. coli infection will contribute to the understanding of the underlying pathogenic mechanisms from the perspective of intercellular communication between BMECs and astrocytes, and provide new insights for future prevention and treatment of E. coli meningitis.

6.
Sensors (Basel) ; 24(2)2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38257627

ABSTRACT

Wireless sensor network (WSN) underpinning the smart-grid Internet of Things (SG-IoT) has been a popular research topic in recent years due to its great potential for enabling a wide range of important applications. However, the energy consumption (EC) characteristic of sensor nodes is a key factor that affects the operational performance (e.g., lifetime of sensors) and the total cost of ownership of WSNs. In this paper, to find the modulation techniques suitable for WSNs, we investigate the EC characteristic of continuous phase modulation (CPM), which is an attractive modulation scheme candidate for WSNs because of its constant envelope property. We first develop an EC model for the sensor nodes of WSNs by considering the circuits and a typical communication protocol that relies on automatic repeat request (ARQ)-based retransmissions to ensure successful data delivery. Then, we use this model to analyze the EC characteristic of CPM under various configurations of modulation parameters. Furthermore, we compare the EC characteristic of CPM with that of other representative modulation schemes, such as offset quadrature phase-shift keying (OQPSK) and quadrature amplitude modulation (QAM), which are commonly used in communication protocols of WSNs. Our analysis and simulation results provide insights into the EC characteristics of multiple modulation schemes in the context of WSNs; thus, they are beneficial for designing energy-efficient SG-IoT in the beyond-5G (B5G) and the 6G era.

7.
Front Public Health ; 11: 1273443, 2023.
Article in English | MEDLINE | ID: mdl-38035306

ABSTRACT

COVID-19 is an acute respiratory infectious disease caused by SARS-CoV-2. It was first reported in Wuhan, China in December 2019 and rapidly spread globally in early 2020, triggering a global pandemic. In December 2022, China adjusted the dynamic COVID-zero strategy that lasted for three years. The number of positive cases in China increased rapidly in the short term. Weihai was also affected during this period. We conducted genomic surveillance of SARS-CoV-2 variants in Weihai during this period, hoping to understand the changes in the genomic characteristics of SARS-CoV-2 before and after the adjustment of the epidemic policy. In this study,we collected SARS-CoV-2 positive samples from March 2022 to March 2023 in Weihai and performed SARS-CoV-2 whole genome sequencing on these samples using next-generation sequencing technology. we obtained a total of 704 SARS-CoV-2 whole genome sequences, and selected 581 high-quality sequences for further analysis. The analysis results showed that from March 2022 to November 2022, before the adjustment of epidemic policy, the COVID-19 cases in Weihai were mainly from four local clusters,which were caused by four variants, including BA.2,BA.1.1,P.1.15 and BA.5.2.1. Phylogenetic analysis showed that: In the same cluster,the sequences between each other were highly homologous, and the whole genome sequence were almost identical. After December 2022, the epidemic policy was adjusted, BF.7 and BA.5.2 became the dominant variants in Weihai, consistent with the main domestic strains in China during the same period. Phylodynamic analysis showed that BF.7 and BA.5.2 had a large amount of genetic diversities in December, and the effective population size of BF.7 and BA.5.2 also showed explosive growth in December. In conclusion, we reported the composition and dynamic trend of SARS-CoV-2 variants in Weihai from March 2022 to March 2023. We found that there have been significant changes in the variants and expansion patterns of SARS-CoV-2 before and after the adjustment of epidemic policies. But the dominant variants in Weihai were the same as the SARS-CoV-2 variants circulating globally at the same time and we found no persistently dominant variants or new lineages during this period.


Subject(s)
COVID-19 , SARS-CoV-2 , Humans , SARS-CoV-2/genetics , Phylogeny , COVID-19/epidemiology , Genomics , China/epidemiology , Pandemics
8.
Int J Mol Sci ; 24(13)2023 Jun 21.
Article in English | MEDLINE | ID: mdl-37445610

ABSTRACT

Meningitis is a major clinical manifestation of Escherichia coli (E. coli) infection characterized by inflammation of the meninges and subarachnoid space. Many chemokines are secreted during meningitic E. coli infection, of which C-X-C motif chemokine 3 (CXCL3) is the most highly expressed. However, it is unclear how CXCL3 plays a role in meningitic E. coli infection. Therefore, this study used in vitro and in vivo assays to clarify these contributions and to identify novel therapeutic targets for central nervous system inflammation. We found a significantly upregulated expression of CXCL3 in human brain microvascular endothelial cells and U251 cells after meningitic E. coli infection, and the CXCL3 receptor, C-X-C motif chemokine receptor 2 (CXCR2), was expressed in microglia. Furthermore, CXCL3 induced M1 microglia by selectively activating mitogen-activated protein kinases signaling and significantly upregulating tumor necrosis factor-α (TNF-α), interleukin (IL)-1ß, IL-6, nitric oxide synthase 2 (NOS2), and cluster of differentiation 86 (CD86) expression levels, promoting an inflammatory response. Our findings clarify the role of CXCL3 in meningitic E. coli-induced neuroinflammation and demonstrate that CXCL3 may be a potential therapeutic target for future investigation and prevention of E. coli-induced neuroinflammation.


Subject(s)
Escherichia coli Infections , Meningitis , Humans , Escherichia coli/metabolism , Microglia/metabolism , Neuroinflammatory Diseases , Endothelial Cells , Chemokines/metabolism , Inflammation/metabolism , Escherichia coli Infections/metabolism
9.
Head Neck Pathol ; 17(3): 864-870, 2023 Sep.
Article in English | MEDLINE | ID: mdl-37014573

ABSTRACT

BACKGROUND: Mucoepidermoid carcinoma is a malignant salivary gland tumor which, in most cases, is composed of variable proportions of mucous, epidermoid, and intermediate cells. METHODS: We report a case of parapharyngeal mucoepidermoid carcinoma with highly unusual ("monomorphic") light microscopic features as well as atypical immunohistochemical properties. Molecular analysis was performed using the TruSight RNA fusion panel. RESULTS: The tumor featured heretofore undescribed histopathological features: sheets and nests composed of monomorphic neoplastic (plump spindle to epithelioid) cells with no mucous, intermediate, glandular/columnar, or any other cell type identified. The neoplastic cells displayed variable clear cell change and only expressed cytokeratin 7. Despite this non-classical morphology, the presence of the classical CRTC1::MAML2 fusion was demonstrated. CONCLUSIONS: Mucoepidermoid carcinoma featuring a uniform ("monomorphic") population of neoplastic cells is a novel observation. A confident diagnosis of mucoepidermoid carcinoma can be made upon detection of the CRTC1/3::MAML2 fusion. Our case increases the spectrum of histopathological appearances that mucoepidermoid carcinoma may display.


Subject(s)
Carcinoma, Mucoepidermoid , Salivary Gland Neoplasms , Humans , DNA-Binding Proteins/genetics , Trans-Activators , Carcinoma, Mucoepidermoid/pathology , Transcription Factors/genetics , Salivary Gland Neoplasms/genetics , Salivary Gland Neoplasms/pathology
10.
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
11.
J Neuroinflammation ; 18(1): 114, 2021 May 13.
Article in English | MEDLINE | ID: mdl-33985523

ABSTRACT

BACKGROUND: Escherichia coli is the most common Gram-negative bacterium causing meningitis, and E. coli meningitis is associated with high mortality and morbidity throughout the world. Our previous study showed that E. coli can colonize the brain and cause neuroinflammation. Increasing evidence supports the involvement of miRNAs as key regulators of neuroinflammation. However, it is not clear whether these molecules participate in the regulation of meningitic E. coli-mediated neuroinflammation. METHODS: The levels of miR-155 and miR-146a, as well as their precursors, in E. coli-infected astrocytes were measured using quantitative real-time PCR (qPCR). Overexpression and knockdown studies of miR-155 and miR-146a were performed to observe the effects on bacterial loads, cytokines, chemokines, and NF-κB signaling pathways. Bioinformatics methods were utilized to predict the target genes, and these target genes were validated using qPCR, Western blotting, and luciferase reporter system. In vivo knockdown of miR-155 and miR-146a was carried out to observe the effects on bacterial loads, inflammatory genes, astrocyte activation, microglia activation, and survival in a mouse model. RESULTS: The levels of miR-155, miR-146a, and their precursors were significantly increased in astrocytes during E. coli infection. miR-155 and miR-146a were induced by the NF-κB-p65 signaling pathway upon infection. Overexpressing and inhibiting miR-155 and miR-146a in astrocytes did not affect the bacterial loads. Further, the in vitro overexpression of miR-155 and miR-146a suppressed the E. coli-induced inflammatory response, whereas the inhibition of miR-155 and miR-146a enhanced it. Mechanistically, miR-155 inhibited TAB2, and miR-146a targeted IRAK1 and TRAF6; therefore, they functioned collaboratively to modulate TLR-mediated NF-κB signaling. In addition, both miR-155 and miR-146a could regulate the EGFR-NF-κB signaling pathway. Finally, the in vivo suppression of E. coli-induced miR-155 and miR-146a further promoted the production of inflammatory cytokines, aggravated astrocyte and microglia activation, and decreased mouse survival time, without affecting the bacterial loads in the blood and brain. CONCLUSIONS: E. coli infection induced miR-155 and miR-146a, which collectively regulated bacteria-triggered neuroinflammatory responses through negative feedback regulation involving the TLR-mediated NF-κB and EGFR-NF-κB signaling pathways, thus protecting the central nervous system from further neuroinflammatory damage.


Subject(s)
Inflammation/microbiology , Meningitis, Escherichia coli/immunology , Meningitis, Escherichia coli/metabolism , MicroRNAs/immunology , MicroRNAs/metabolism , Adaptor Proteins, Signal Transducing/metabolism , Animals , Antagomirs , Astrocytes/immunology , Astrocytes/microbiology , Cell Line , Escherichia coli/immunology , Inflammation/metabolism , Interleukin-1 Receptor-Associated Kinases , Mice , NF-kappa B/metabolism , Signal Transduction , TNF Receptor-Associated Factor 6/metabolism
12.
ACS Infect Dis ; 7(4): 777-789, 2021 04 09.
Article in English | MEDLINE | ID: mdl-33723986

ABSTRACT

Meningitic Escherichia coli can infiltrate the central nervous system (CNS), consequently increasing the levels of proinflammatory cytokines and chemokines and deteriorating the integrity of the blood-brain barrier (BBB). Resveratrol has emerged in recent years as a compound with antioxidant and anti-inflammatory properties. However, it is still unknown how resveratrol affects meningitic E. coli-induced CNS dysfunction. Here, by using in vivo and in vitro BBB models, we demonstrated that resveratrol treatment significantly inhibited meningitic E. coli invasion of the BBB, protected the integrity of the BBB, and reduced neuroinflammation and lethality. In mechanism, resveratrol inhibited bacterial penetration of the BBB by attenuating the upregulation of caveolin-1 (CAV-1), a class of lipid rafts maintaining endothelial cell function. Resveratrol treatment also maintained BBB permeability by suppressing the ERK1/2-VEGFA signaling cascade. In vivo treatment of resveratrol decreased the production of inflammatory cytokines and improved the survival rate in mice challenged with meningitic E. coli. These findings collectively indicated that resveratrol could attenuate meningitic E. coli-induced CNS injury, which might constitute a new approach for future prevention and treatment of E. coli meningitis.


Subject(s)
Blood-Brain Barrier , Meningitis, Escherichia coli , Animals , Endothelial Cells , Escherichia coli , Mice , Resveratrol/pharmacology
13.
J Neuroinflammation ; 16(1): 101, 2019 May 15.
Article in English | MEDLINE | ID: mdl-31092253

ABSTRACT

BACKGROUND: Blood-brain barrier (BBB) disruption and neuroinflammation are considered key mechanisms of pathogenic Escherichia coli invasion of the brain. However, the specific molecules involved in meningitic E. coli-induced BBB breakdown and neuroinflammatory response remain unclear. Our previous RNA-sequencing data from human brain microvascular endothelial cells (hBMECs) revealed two important host factors: platelet-derived growth factor-B (PDGF-B) and intercellular adhesion molecule-1 (ICAM-1), which were significantly upregulated in hBMECs after meningitic E. coli infection. Whether and how PDGF-B and ICAM-1 contribute to the development of E. coli meningitis are still unclear. METHODS: The western blot, real-time PCR, enzyme-linked immunosorbent assay, immunohistochemistry, and immunofluorescence were applied to verify the significant induction of PDGF-B and ICAM-1 by meningitic E. coli in vivo and in vitro. Evan's blue assay and electric cell-substrate impedance sensing assay were combined to identify the effects of PDGF-B on BBB permeability. The CRISPR/Cas9 technology, cell-cell adhesion assay, and electrochemiluminescence assay were used to investigate the role of ICAM-1 in neuroinflammation subversion. RESULTS: We verified the significant induction of PDGF-B and ICAM-1 by meningitic E. coli in mouse as well as monolayer hBMECs models. Functionally, we showed that the increase of PDGF-B may directly enhance the BBB permeability by decreasing the expression of tight junction proteins, and the upregulation of ICAM-1 contributed to neutrophils or monocytes recruitment as well as neuroinflammation subversion in response to meningitic E. coli infection. CONCLUSIONS: Our findings demonstrated the roles of PDGF-B and ICAM-1 in mediating bacterial-induced BBB damage as well as neuroinflammation, providing new concepts and potential targets for future prevention and treatment of bacterial meningitis.


Subject(s)
Blood-Brain Barrier/metabolism , Escherichia coli Infections/metabolism , Inflammation Mediators/metabolism , Intercellular Adhesion Molecule-1/biosynthesis , Lymphokines/biosynthesis , Meningitis, Bacterial/metabolism , Platelet-Derived Growth Factor/biosynthesis , Animals , Blood-Brain Barrier/microbiology , Blood-Brain Barrier/pathology , Cells, Cultured , Escherichia coli , Escherichia coli Infections/pathology , Female , Meningitis, Bacterial/pathology , Mice , Tight Junctions/metabolism , Tight Junctions/microbiology , Up-Regulation/physiology
14.
Virulence ; 9(1): 1247-1262, 2018.
Article in English | MEDLINE | ID: mdl-30036124

ABSTRACT

Bacterial lipooligosaccharide (LOS) is an important virulence-associated factor, and its sialylation largely confers its ability to mediate cell adhesion, invasion, inflammation, and immune evasion. Here, we investigated the function of the Haemophilus parasuis α-2,3-sialyltransferase gene, lsgB, which determines the terminal sialylation of LOS, by generating a lsgB deletion mutant as well as a complementation strain. Our data indicate a direct effect of lsgB on LOS sialylation and reveal important roles of lsgB in promoting the pathogenicity of H. parasuis, including adhesion to and invasion of porcine cells in vitro, bacterial load and survival in vivo, as well as a contribution to serum resistance. These observations highlight the function of lsgB in mediating LOS sialylation and more importantly its role in H. parasuis infection. These findings provide a more profound understanding of the pathogenic mechanism of this disease-causing bacterium.


Subject(s)
Haemophilus parasuis/genetics , Haemophilus parasuis/pathogenicity , Lipopolysaccharides/chemistry , Sialyltransferases/genetics , Amino Acid Sequence , Animals , Cells, Cultured , Gene Deletion , Genetic Complementation Test , Haemophilus parasuis/enzymology , Mutation , Swine , Virulence , Virulence Factors/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...