Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 46
Filtrar
1.
Cell Commun Signal ; 22(1): 44, 2024 01 17.
Artículo en Inglés | MEDLINE | ID: mdl-38233877

RESUMEN

Bacterial meningitis remains a leading cause of infection-related mortality worldwide. Although Escherichia coli (E. coli) is the most common etiology of neonatal meningitis, the underlying mechanisms governing bacterial blood-brain barrier (BBB) disruption during infection remain elusive. We observed that infection of human brain microvascular endothelial cells with meningitic E. coli triggers the activation of early growth response 1 (Egr-1), a host transcriptional activator. Through integrated chromatin immunoprecipitation sequencing and transcriptome analysis, we identified Egr-1 as a crucial regulator for maintaining BBB integrity. Mechanistically, Egr-1 induced cytoskeletal changes and downregulated tight junction protein expression by directly targeting VEGFA, PDGFB, and ANGPTL4, resulting in increased BBB permeability. Meanwhile, Egr-1 also served as a master regulator in the initiation of neuroinflammatory response during meningitic E. coli infection. Our findings support an Egr-1-dependent mechanism of BBB disruption by meningitic E. coli, highlighting a promising therapeutic target for bacterial meningitis.


Asunto(s)
Meningitis Bacterianas , Meningitis por Escherichia coli , Humanos , Recién Nacido , Barrera Hematoencefálica/microbiología , Células Endoteliales/metabolismo , Escherichia coli , Meningitis Bacterianas/metabolismo , Meningitis por Escherichia coli/metabolismo
2.
Cell Commun Signal ; 22(1): 123, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-38360663

RESUMEN

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.


Asunto(s)
Meningitis Bacterianas , Meningitis por Escherichia coli , MicroARNs , Humanos , Escherichia coli/genética , Proteínas Hedgehog/metabolismo , Células Endoteliales/metabolismo , Meningitis por Escherichia coli/metabolismo , Encéfalo/metabolismo , Barrera Hematoencefálica/microbiología , Meningitis Bacterianas/metabolismo , Inmunidad , MicroARNs/metabolismo
3.
Int J Mol Sci ; 25(6)2024 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-38542512

RESUMEN

Zoonoses are diseases and infections naturally transmitted between humans and vertebrate animals. They form the dominant group of diseases among emerging infectious diseases and represent critical threats to global health security. This dilemma is largely attributed to our insufficient knowledge of the pathogenesis regarding zoonotic spillover. Long non-coding RNAs (lncRNAs) are transcripts with limited coding capacity. Recent technological advancements have enabled the identification of numerous lncRNAs in humans, animals, and even pathogens. An increasing body of literature suggests that lncRNAs function as key regulators in zoonotic infection. They regulate immune-related epigenetic, transcriptional, and post-transcriptional events across a broad range of organisms. In this review, we discuss the recent research progress on the roles of lncRNAs in zoonoses. We address the classification and regulatory mechanisms of lncRNAs in the interaction between host and zoonotic pathogens. Additionally, we explore the surprising function of pathogen-derived lncRNAs in mediating the pathogenicity and life cycle of zoonotic bacteria, viruses, and parasites. Understanding how these lncRNAs influence the zoonotic pathogenesis will provide important therapeutic insights to the prevention and control of zoonoses.


Asunto(s)
Enfermedades Transmisibles Emergentes , ARN Largo no Codificante , Virus , Animales , Humanos , ARN Largo no Codificante/genética , Zoonosis/genética
4.
J Neuroinflammation ; 20(1): 173, 2023 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-37481642

RESUMEN

Neuroinflammation has been implicated in the initiation and progression of several central nervous system (CNS) disorders, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, multiple sclerosis, ischemic stroke, traumatic brain injury, spinal cord injury, viral encephalitis, and bacterial encephalitis. Microglia and astrocytes are essential in neural development, maintenance of synaptic connections, and homeostasis in a healthy brain. The activation of astrocytes and microglia is a defense mechanism of the brain against damaged tissues and harmful pathogens. However, their activation triggers neuroinflammation, which can exacerbate or induce CNS injury. Non-coding RNAs (ncRNAs) are functional RNA molecules that lack coding capabilities but can actively regulate mRNA expression and function through various mechanisms. ncRNAs are highly expressed in astrocytes and microglia and are potential mediators of neuroinflammation. We reviewed the recent research progress on the role of miRNAs, lncRNAs, and circRNAs in regulating neuroinflammation in various CNS diseases. Understanding how these ncRNAs affect neuroinflammation will provide important therapeutic insights for preventing and managing CNS dysfunction.


Asunto(s)
MicroARNs , ARN Largo no Codificante , Humanos , Astrocitos , Microglía , Enfermedades Neuroinflamatorias , ARN Largo no Codificante/genética
5.
Int J Mol Sci ; 24(13)2023 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-37445610

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli , Meningitis , Humanos , Escherichia coli/metabolismo , Microglía/metabolismo , Enfermedades Neuroinflamatorias , Células Endoteliales , Quimiocinas/metabolismo , Inflamación/metabolismo , Infecciones por Escherichia coli/metabolismo
6.
Int J Mol Sci ; 24(3)2023 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-36769171

RESUMEN

Despite advances in supportive care and antimicrobial treatment, bacterial meningitis remains the most serious infection of the central nervous system (CNS) that poses a serious risk to life. This clinical dilemma is largely due to our insufficient knowledge of the pathology behind this disease. By controlling the entry of molecules into the CNS microenvironment, the blood-brain barrier (BBB), a highly selective cellular monolayer that is specific to the CNS's microvasculature, regulates communication between the CNS and the rest of the body. A defining feature of the pathogenesis of bacterial meningitis is the increase in BBB permeability. So far, several contributing factors for BBB disruption have been reported, including direct cellular damage brought on by bacterial virulence factors, as well as host-specific proteins or inflammatory pathways being activated. Recent studies have demonstrated that targeting pathological factors contributing to enhanced BBB permeability is an effective therapeutic complement to antimicrobial therapy for treating bacterial meningitis. Hence, understanding how these meningitis-causing pathogens affect the BBB permeability will provide novel perspectives for investigating bacterial meningitis's pathogenesis, prevention, and therapies. Here, we summarized the recent research progress on meningitis-causing pathogens disrupting the barrier function of BBB. This review provides handy information on BBB disruption by meningitis-causing pathogens, and helps design future research as well as develop potential combination therapies.


Asunto(s)
Barrera Hematoencefálica , Meningitis Bacterianas , Humanos , Barrera Hematoencefálica/metabolismo , Meningitis Bacterianas/tratamiento farmacológico , Meningitis Bacterianas/metabolismo , Sistema Nervioso Central , Transporte Biológico , Bacterias
7.
J Neuroinflammation ; 19(1): 149, 2022 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-35705998

RESUMEN

BACKGROUND: The emergence of the novel, pathogenic severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) has caused a global health emergency. SARS-CoV-2 is highly contagious and has a high mortality rate in severe patients. However, there is very limited information on the effect of SARS-CoV-2 infection on the integrity of the blood-brain barrier (BBB). METHODS: RNA-sequencing profiling was performed to analyze the transcriptomic changes in human brain microvascular endothelial cells (hBMECs) after SARS-CoV-2 infection. Bioinformatic tools were used for differential analysis. Immunofluorescence, real-time quantitative PCR, and Western blotting analysis were used to explore biological phenotypes. RESULTS: A total of 927 differentially expressed genes were identified, 610 of which were significantly upregulated while the remaining 317 were downregulated. We verified the significant induction of cytokines, chemokines, and adhesion molecules in hBMECs by SARS-CoV-2, suggesting an activation of the vascular endothelium in brain. Moreover, we demonstrated that SARS-CoV-2 infection could increase the BBB permeability, by downregulating as well as remodeling the intercellular tight junction proteins. CONCLUSIONS: Our findings demonstrated that SARS-CoV-2 infection can cause BBB dysfunction, providing novel insights into the understanding of SARS-CoV-2 neuropathogenesis. Moreover, this finding shall constitute a new approach for future prevention and treatment of SARS-CoV-2-induced CNS infection.


Asunto(s)
COVID-19 , SARS-CoV-2 , Barrera Hematoencefálica/metabolismo , Encéfalo , Células Endoteliales , Humanos
8.
BMC Neurosci ; 23(1): 43, 2022 07 06.
Artículo en Inglés | MEDLINE | ID: mdl-35794518

RESUMEN

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is highly contagious, and the neurological symptoms of SARS-CoV-2 infection have already been reported. However, the mechanisms underlying the effect of SARS-CoV-2 infection on patients with central nervous system injuries remain unclear. METHODS: The high-throughput RNA sequencing was applied to analyze the transcriptomic changes in SK-N-SH cells after SARS-CoV-2 infection. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed to identify the functions of differentially expressed genes and related pathways. RESULTS: A total of 820 mRNAs were significantly altered, including 671 upregulated and 149 downregulated mRNAs (showing an increase of ≥ 2-fold or decrease to ≤ 0.5-fold, respectively; p ≤ 0.05). Moreover, we verified the significant induction of cytokines, chemokines, and their receptors, as well as the activation of NF-κB, p38, and Akt signaling pathways, in SK-N-SH by SARS-CoV-2. CONCLUSIONS: To our knowledge, this is the first time the transcriptional profiles of the host mRNAs involved in SARS-CoV-2 infection of SK-N-SH cells have been reported. These findings provide novel insight into the pathogenic mechanism of SARS-CoV-2 and might constitute a new approach for future prevention and treatment of SARS-CoV-2-induced central nervous system infection.


Asunto(s)
COVID-19 , Neuroblastoma , Citocinas , Humanos , FN-kappa B , ARN Mensajero/metabolismo , SARS-CoV-2
9.
Bioorg Med Chem Lett ; 67: 128745, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35447345

RESUMEN

Colorectal cancer (CRC) is one of the most commonly diagnosed cancer types and Traf2- and Nck-interacting kinase (TNIK) has been thought as a potential target for CRC treatment. Herein we report the discovery and structure-activity relationship (SAR) of benzo[d]oxazol-2(3H)-one derivatives as a new class of TNIK inhibitors. The most potent compound 8g showed an IC50 value of 0.050 µM against TNIK. It effectively suppressed proliferation and migration of colorectal cancer cells. Western blot analysis indicated that 8g could inhibit aberrant transcription activation of Wnt signaling. Collectively, this study provides a potential lead compound for subsequent drug discovery targeting TNIK.


Asunto(s)
Neoplasias Colorrectales , Proteínas Serina-Treonina Quinasas , Neoplasias Colorrectales/tratamiento farmacológico , Descubrimiento de Drogas , Humanos , Relación Estructura-Actividad , Vía de Señalización Wnt
10.
J Neuroinflammation ; 18(1): 114, 2021 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-33985523

RESUMEN

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.


Asunto(s)
Inflamación/microbiología , Meningitis por Escherichia coli/inmunología , Meningitis por Escherichia coli/metabolismo , MicroARNs/inmunología , MicroARNs/metabolismo , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Antagomirs , Astrocitos/inmunología , Astrocitos/microbiología , Línea Celular , Escherichia coli/inmunología , Inflamación/metabolismo , Quinasas Asociadas a Receptores de Interleucina-1 , Ratones , FN-kappa B/metabolismo , Transducción de Señal , Factor 6 Asociado a Receptor de TNF/metabolismo
11.
Bioorg Med Chem Lett ; 45: 128138, 2021 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-34044123

RESUMEN

The Rho-associated protein kinases (ROCKs) are associated with the pathology of glaucoma and discovery of ROCK inhibitors has attracted much attention in recent years. Herein, we report a series of 3,4-dihydrobenzo[f][1,4]oxazepin-5(2H)-one derivatives as a new class of ROCK inhibitors. Structure-activity relationship studies led to the discovery of compound 12b, which showed potent activities against ROCK I and ROCK Ⅱ with IC50 values of 93 nM and 3 nM, respectively. 12b also displayed considerable selectivity for ROCKs. The mean IOP-lowering effect of 12b in an ocular normotensive model was 34.3%, and no obvious hyperemia was observed. Overall, this study provides a good starting point for ROCK-targeting drug discovery against glaucoma.


Asunto(s)
Descubrimiento de Drogas , Glaucoma/tratamiento farmacológico , Oxazepinas/farmacología , Quinasas Asociadas a rho/antagonistas & inhibidores , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Glaucoma/metabolismo , Humanos , Simulación de Dinámica Molecular , Estructura Molecular , Oxazepinas/síntesis química , Oxazepinas/química , Relación Estructura-Actividad , Quinasas Asociadas a rho/metabolismo
12.
RNA Biol ; 18(sup1): 108-116, 2021 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-34241576

RESUMEN

The blood-brain barrier (BBB), which controls permeability into and out of the nervous system, is a tightly connected, structural, and functional separation between the central nervous system (CNS) and circulating blood. CNS diseases, such as Alzheimer's disease, multiple sclerosis, traumatic brain injury, stroke, meningitis, and brain cancers, often develop with the increased BBB permeability and further leads to irreversible CNS injury. Non-coding RNAs (ncRNAs) are functional RNA molecules that generally lack the coding abilities but can actively regulate the mRNA expression and function through different mechanisms. Various types of ncRNAs, including microRNAs (miRNAs), long ncRNAs (lncRNAs), and circular RNAs (circRNAs), are highly expressed in brain microvascular endothelial cells and are potential mediators of BBB permeability. Here, we summarized the recent research progress on miRNA, lncRNA, and circRNA roles regulating the BBB permeability in different CNS diseases. Understanding how these ncRNAs affect the BBB permeability shall provide important therapeutic insights into the prevention and control of the BBB dysfunction.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Encéfalo/metabolismo , Permeabilidad de la Membrana Celular , MicroARNs/genética , ARN Circular/genética , ARN no Traducido/genética , Animales , Humanos
13.
Int J Mol Sci ; 22(12)2021 Jun 14.
Artículo en Inglés | MEDLINE | ID: mdl-34198485

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Barrera Hematoencefálica/patología , Escherichia coli/fisiología , Metaloproteinasa 3 de la Matriz/metabolismo , Meningitis Bacterianas/genética , Meningitis Bacterianas/microbiología , MicroARNs/metabolismo , ARN Largo no Codificante/metabolismo , Secuencia de Bases , Citoplasma/metabolismo , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Redes Reguladoras de Genes , Humanos , MicroARNs/genética , Microvasos/patología , Modelos Biológicos , Permeabilidad , ARN Largo no Codificante/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Transducción de Señal , Uniones Estrechas/metabolismo , Transcripción Genética , Regulación hacia Arriba/genética
14.
J Neuroinflammation ; 16(1): 101, 2019 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-31092253

RESUMEN

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.


Asunto(s)
Barrera Hematoencefálica/metabolismo , Infecciones por Escherichia coli/metabolismo , Mediadores de Inflamación/metabolismo , Molécula 1 de Adhesión Intercelular/biosíntesis , Linfocinas/biosíntesis , Meningitis Bacterianas/metabolismo , Factor de Crecimiento Derivado de Plaquetas/biosíntesis , Animales , Barrera Hematoencefálica/microbiología , Barrera Hematoencefálica/patología , Células Cultivadas , Escherichia coli , Infecciones por Escherichia coli/patología , Femenino , Meningitis Bacterianas/patología , Ratones , Uniones Estrechas/metabolismo , Uniones Estrechas/microbiología , Regulación hacia Arriba/fisiología
15.
J Neuroinflammation ; 15(1): 291, 2018 Oct 19.
Artículo en Inglés | MEDLINE | ID: mdl-30340642

RESUMEN

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-1ß. 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.


Asunto(s)
Encéfalo/citología , Células Endoteliales/metabolismo , Células Endoteliales/microbiología , Regulación Bacteriana de la Expresión Génica/fisiología , Proteómica/métodos , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Células Cultivadas , Biología Computacional , Citocinas/genética , Citocinas/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Regulación Bacteriana de la Expresión Génica/efectos de los fármacos , Redes Reguladoras de Genes , Humanos , Oxidorreductasas Intramoleculares/química , Oxidorreductasas Intramoleculares/farmacología , Factores Inhibidores de la Migración de Macrófagos/química , Factores Inhibidores de la Migración de Macrófagos/farmacología , Meningitis por Escherichia coli/metabolismo , Meningitis por Escherichia coli/patología , FN-kappa B/metabolismo , Transducción de Señal/fisiología
16.
J Neuroinflammation ; 13(1): 274, 2016 10 19.
Artículo en Inglés | MEDLINE | ID: mdl-27756321

RESUMEN

BACKGROUND: Streptococcus suis serotype 2 (SS2) is an important zoonotic bacterial pathogen in both humans and animals, which can cause high morbidity and mortality. Meningitis is one of the major clinical manifestations of SS2 infection. However, the specific process of SS2 meningitis and its molecular mechanisms remain unclear. Epidermal growth factor receptor (EGFR) has been reported to initiate transduction of intracellular signals and regulate host inflammatory responses. Whether and how EGFR contributes to the development of S. suis meningitis are currently unknown. METHODS: The tyrosine phosphorylation of cellular proteins, the transactivation of EGFR, as well as its dimerization, and the associated signal transduction pathways were investigated by immunoprecipitation and western blotting. Real-time quantitative PCR was used to investigate the transcriptional level of the ErbB family members, EGFR-related ligands, cytokines, and chemokines. The secretion of cytokines and chemokines in the serum and brain were detected by Q-Plex™ Chemiluminescent ELISA. RESULTS: We found an important role of EGFR in SS2 strain SC19-induced meningitis. SC19 increasingly adhered to human brain microvascular endothelial cells (hBMEC) and caused inflammatory lesions in the brain tissues, with significant induction and secretion of proinflammatory cytokines and chemokines in the serum and brains. SC19 infection of hBMEC induced tyrosine phosphorylation of cellular EGFR in a ligand-dependent manner involving the EGF-like ligand HB-EGF, amphiregulin (AREG), and epiregulin (EREG) and led to heterodimerization of EGFR/ErbB3. The EGFR transactivation did not participate in SS2 strain SC19 adhesion of hBMEC, as well as in bacterial colonization in vivo. However, its transactivation contributed to the bacterial-induced neuroinflammation, via triggering the MAPK-ERK1/2 and NF-κB signaling pathways in hBMEC that promote the production of proinflammatory cytokines and chemokines. CONCLUSIONS: We investigated for the first time the tyrosine phosphorylation of cellular proteins in response to SS2 strain SC19 infection of hBMEC and demonstrated the contribution of EGFR to SS2-induced neuroinflammation. These observations propose a novel mechanism involving EGFR in SS2-mediated inflammatory responses in the brain, and therefore, EGFR might be an important host target for further investigation and prevention of neuroinflammation caused by SS2 strains.


Asunto(s)
Encéfalo/metabolismo , Receptores ErbB/metabolismo , Meningitis , Infecciones Estreptocócicas/complicaciones , Infecciones Estreptocócicas/fisiopatología , Streptococcus suis/fisiología , Activación Transcripcional/fisiología , Anfirregulina/metabolismo , Animales , Encéfalo/microbiología , Encéfalo/patología , Citocinas/genética , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Inhibidores Enzimáticos/farmacología , Receptores ErbB/genética , Femenino , Humanos , Meningitis/etiología , Meningitis/microbiología , Meningitis/fisiopatología , Ratones , Fosforilación/efectos de los fármacos , Quinazolinas/farmacología , Receptor ErbB-3/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/fisiología , Infecciones Estreptocócicas/microbiología , Porcinos , Tirosina/metabolismo , Tirfostinos/farmacología
17.
Mol Neurobiol ; 2024 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-38896157

RESUMEN

The blood-brain barrier (BBB) is a complex structure that separates the central nervous system (CNS) from the peripheral blood circulation. Effective communication between different cell types within the BBB is crucial for its proper functioning and maintenance of homeostasis. In this study, we demonstrate that meningitic Escherichia coli (E. coli)-induced WNT5B plays a role in facilitating intercellular communication between astrocytes and brain microvascular endothelial cells (BMECs). We discovered that astrocytes-derived WNT5B activates the non-canonical WNT signaling pathway JNK/c-JUN in BMECs through its receptor ROR1, leading to inhibition of ZO-1 expression and impairment of the tight junction integrity in BMECs. Notably, our findings reveal that c-JUN, a transcription factor, directly regulates ZO-1 expression. By employing a dual luciferase reporting system and chromatin immunoprecipitation techniques, we identified specific binding sites of c-JUN on the ZO-1 promoter region. Overall, our study highlights the involvement of WNT5B in mediating intercellular communication between astrocytes and BMECs, provides insights into the role of WNT5B in meningitic E. coli-induced disruption of BBB integrity, and suggests potential therapeutic targeting of WNT5B as a strategy to address BBB dysfunction.

18.
Sci Rep ; 14(1): 7582, 2024 Mar 30.
Artículo en Inglés | MEDLINE | ID: mdl-38555378

RESUMEN

With the continuous development of cloud computing, the application of cloud storage has become more and more popular. To ensure the integrity and availability of cloud data, scholars have proposed several cloud data auditing schemes. Still, most need help with outsourced data integrity, controlled outsourcing, and source file auditing. Therefore, we propose a controlled delegation outsourcing data integrity auditing scheme based on the identity-based encryption model. Our proposed scheme allows users to specify a dedicated agent to assist in uploading data to the cloud. These authorized proxies use recognizable identities for authentication and authorization, thus avoiding the need for cumbersome certificate management in a secure distributed computing system. While solving the above problems, our scheme adopts a bucket-based red-black tree structure to efficiently realize the dynamic updating of data, which can complete the updating of data and rebalancing of structural updates constantly and realize the high efficiency of data operations. We define the security model of the scheme in detail and prove the scheme's security under the difficult problem assumption. In the performance analysis section, the proposed scheme is analyzed experimentally in comparison with other schemes, and the results show that the proposed scheme is efficient and secure.

19.
ACS Infect Dis ; 10(3): 988-999, 2024 03 08.
Artículo en Inglés | MEDLINE | ID: mdl-38317607

RESUMEN

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.


Asunto(s)
Infecciones por Escherichia coli , Meningitis Bacterianas , Meningitis por Escherichia coli , Humanos , Ratones , Animales , Barrera Hematoencefálica , Meningitis por Escherichia coli/metabolismo , Meningitis por Escherichia coli/microbiología , Meningitis por Escherichia coli/patología , Escherichia coli/metabolismo , Células Endoteliales , Interleucina-22 , Factor A de Crecimiento Endotelial Vascular/metabolismo , Factor A de Crecimiento Endotelial Vascular/farmacología , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT3/farmacología
20.
Mol Neurobiol ; 2024 Feb 19.
Artículo en Inglés | MEDLINE | ID: mdl-38372957

RESUMEN

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.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA