Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 227
Filter
1.
J Infect Dis ; 230(Supplement_2): S87-S94, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39255395

ABSTRACT

Periodontitis is a common chronic inflammatory disease, affecting approximately 19% of the global adult population. A relationship between periodontal disease and Alzheimer disease has long been recognized, and recent evidence has been uncovered to link these 2 diseases mechanistically. Periodontitis is caused by dysbiosis in the subgingival plaque microbiome, with a pronounced shift in the oral microbiota from one consisting primarily of Gram-positive aerobic bacteria to one predominated by Gram-negative anaerobes, such as Porphyromonas gingivalis. A common phenomenon shared by all bacteria is the release of membrane vesicles to facilitate biomolecule delivery across long distances. In particular, the vesicles released by P gingivalis and other oral pathogens have been found to transport bacterial components across the blood-brain barrier, initiating the physiologic changes involved in Alzheimer disease. In this review, we summarize recent data that support the relationship between vesicles secreted by periodontal pathogens to Alzheimer disease pathology.


Subject(s)
Alzheimer Disease , Periodontitis , Porphyromonas gingivalis , Alzheimer Disease/microbiology , Alzheimer Disease/metabolism , Humans , Periodontitis/microbiology , Porphyromonas gingivalis/pathogenicity , Dysbiosis/microbiology , Bacterial Infections/microbiology , Blood-Brain Barrier/microbiology , Animals , Microbiota
2.
J Infect Dis ; 230(Supplement_2): S117-S127, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39255391

ABSTRACT

Dysbiosis within microbiomes has been increasingly implicated in many systemic illnesses, such as cardiovascular disease, metabolic syndrome, respiratory infections, and Alzheimer disease (Ad). The correlation between Ad and microbial dysbiosis has been repeatedly shown, yet the etiologic cause of microbial dysbiosis remains elusive. From a neuropathology perspective, abnormal (often age-related) changes in the brain, associated structures, and bodily lumens tend toward an accumulation of oxygen-depleted pathologic structures, which are anaerobically selective niches. These anaerobic environments may promote progressive change in the microbial community proximal to the brain and thus deserve further investigation. In this review, we identify and explore what is known about the anaerobic niche near or associated with the brain and the anaerobes that it is harbors. We identify the anaerobe stakeholders within microbiome communities and the impacts on the neurodegenerative processes associated with Ad. Chronic oral dysbiosis in anaerobic dental pockets and the composition of the gut microbiota from fecal stool are the 2 largest anaerobic niche sources of bacterial transference to the brain. At the blood-brain barrier, cerebral atherosclerotic plaques are predominated by anaerobic species intimately associated with the brain vasculature. Focal cerebritis/brain abscess and corpora amylacea may also establish chronic anaerobic niches in direct proximity to brain parenchyma. In exploring the anaerobic niche proximal to the brain, we identify research opportunities to explore potential sources of microbial dysbiosis associated with Ad.


Subject(s)
Alzheimer Disease , Bacteria, Anaerobic , Brain , Dysbiosis , Gastrointestinal Microbiome , Humans , Alzheimer Disease/microbiology , Alzheimer Disease/pathology , Alzheimer Disease/etiology , Dysbiosis/microbiology , Bacteria, Anaerobic/pathogenicity , Brain/pathology , Brain/microbiology , Blood-Brain Barrier/microbiology , Microbiota
3.
J Infect Dis ; 230(Supplement_2): S95-S108, 2024 Sep 10.
Article in English | MEDLINE | ID: mdl-39255397

ABSTRACT

BACKGROUND: Klebsiella pneumoniae is infamous for hospital-acquired infections and sepsis, which have also been linked to Alzheimer disease (AD)-related neuroinflammatory and neurodegenerative impairment. However, its causative and mechanistic role in AD pathology remains unstudied. METHODS: A preclinical model of K. pneumoniae enteric infection and colonization is developed in an AD model (3xTg-AD mice) to investigate whether and how K. pneumoniae pathogenesis exacerbates neuropathogenesis via the gut-blood-brain axis. RESULTS: K. pneumoniae, particularly under antibiotic-induced dysbiosis, was able to translocate from the gut to the bloodstream by penetrating the gut epithelial barrier. Subsequently, K. pneumoniae infiltrated the brain by breaching the blood-brain barrier. Significant neuroinflammatory phenotype was observed in mice with K. pneumoniae brain infection. K. pneumoniae-infected mice also exhibited impaired neurobehavioral function and elevated total tau levels in the brain. Metagenomic analyses revealed an inverse correlation of K. pneumoniae with gut biome diversity and commensal bacteria, highlighting how antibiotic-induced dysbiosis triggers an enteroseptic "pathobiome" signature implicated in gut-brain perturbations. CONCLUSIONS: The findings demonstrate how infectious agents following hospital-acquired infections and consequent antibiotic regimen may induce gut dysbiosis and pathobiome and increase the risk of sepsis, thereby increasing the predisposition to neuroinflammatory and neurobehavioral impairments via breaching the gut-blood-brain barrier.


Subject(s)
Alzheimer Disease , Blood-Brain Barrier , Disease Models, Animal , Dysbiosis , Gastrointestinal Microbiome , Klebsiella Infections , Klebsiella pneumoniae , Mice, Transgenic , Neuroinflammatory Diseases , Animals , Mice , Dysbiosis/microbiology , Dysbiosis/chemically induced , Alzheimer Disease/microbiology , Neuroinflammatory Diseases/microbiology , Gastrointestinal Microbiome/drug effects , Klebsiella Infections/microbiology , Blood-Brain Barrier/microbiology , Brain/pathology , Brain/microbiology , Anti-Bacterial Agents/pharmacology , Brain-Gut Axis , Male , Humans
4.
Gut Microbes ; 16(1): 2387800, 2024.
Article in English | MEDLINE | ID: mdl-39182226

ABSTRACT

The human gastrointestinal tract, boasting the most diverse microbial community, harbors approximately 100 trillion microorganisms comprising viruses, bacteria, fungi, and archaea. The profound genetic and metabolic capabilities of the gut microbiome underlie its involvement in nearly every facet of human biology, from health maintenance and development to aging and disease. Recent recognition of microbiota - gut - brain axis, referring to the bidirectional communication network between gut microbes and their host, has led to a surge in interdisciplinary research. This review begins with an overview of the current understandings regarding the influence of gut microbes on intestinal and blood-brain barrier integrity. Subsequently, we discuss the mechanisms of the microbiota - gut - brain axis, examining the role of gut microbiota-related neural transmission, metabolites, gut hormones and immunity. We propose the concept of microbiota-mediated multi-barrier modulation in the potential treatment in gastrointestinal and neurological disorders. Furthermore, the role of lymphatic network in the development and maintenance of barrier function is discussed, providing insights into lesser-known conduits of communication between the microbial ecosystem within the gut and the brain. In the final section, we conclude by describing the ongoing frontiers in understanding of the microbiota - gut - brain axis's impact on human health and disease.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/physiology , Brain-Gut Axis/physiology , Animals , Lymphatic System/physiology , Lymphatic System/microbiology , Brain/physiology , Brain/metabolism , Brain/microbiology , Blood-Brain Barrier/microbiology , Blood-Brain Barrier/metabolism , Bacteria/metabolism , Bacteria/genetics , Bacteria/classification , Gastrointestinal Tract/microbiology , Gastrointestinal Tract/physiology
5.
Fluids Barriers CNS ; 21(1): 66, 2024 Aug 16.
Article in English | MEDLINE | ID: mdl-39152442

ABSTRACT

BACKGROUND: Group B Streptococcus (GBS) is the leading cause of neonatal meningitis responsible for a substantial cause of death and disability worldwide. The vast majority of GBS neonatal meningitis cases are due to the CC17 hypervirulent clone. However, the cellular and molecular pathways involved in brain invasion by GBS CC17 isolates remain largely elusive. Here, we studied the specific interaction of the CC17 clone with the choroid plexus, the main component of the blood-cerebrospinal fluid (CSF) barrier. METHODS: The interaction of GBS CC17 or non-CC17 strains with choroid plexus cells was studied using an in vivo mouse model of meningitis and in vitro models of primary and transformed rodent choroid plexus epithelial cells (CPEC and Z310). In vivo interaction of GBS with the choroid plexus was assessed by microscopy. Bacterial invasion and cell barrier penetration were examined in vitro, as well as chemokines and cytokines in response to infection. RESULTS: GBS CC17 was found associated with the choroid plexus of the lateral, 3rd and 4th ventricles. Infection of choroid plexus epithelial cells revealed an efficient internalization of the bacteria into the cells with GBS CC17 displaying a greater ability to invade these cells than a non-CC17 strain. Internalization of the GBS CC17 strain involved the CC17-specific HvgA adhesin and occurred via a clathrin-dependent mechanism leading to transcellular transcytosis across the choroid plexus epithelial monolayer. CPEC infection resulted in the secretion of several chemokines, including CCL2, CCL3, CCL20, CX3CL1, and the matrix metalloproteinase MMP3, as well as immune cell infiltration. CONCLUSION: Our findings reveal a GBS strain-specific ability to infect the blood-CSF barrier, which appears to be an important site of bacterial entry and an active site of immune cell trafficking in response to infection.


Subject(s)
Choroid Plexus , Streptococcus agalactiae , Choroid Plexus/metabolism , Choroid Plexus/microbiology , Choroid Plexus/immunology , Animals , Streptococcus agalactiae/pathogenicity , Mice , Adhesins, Bacterial/metabolism , Virulence , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Blood-Brain Barrier/microbiology , Blood-Brain Barrier/metabolism , Disease Models, Animal , Streptococcal Infections/metabolism , Streptococcal Infections/microbiology , Streptococcal Infections/immunology , Mice, Inbred C57BL , Transcytosis/physiology , Female
6.
ACS Chem Neurosci ; 15(16): 2957-2965, 2024 Aug 21.
Article in English | MEDLINE | ID: mdl-39102500

ABSTRACT

The past decade has seen an explosion in our knowledge about the interactions between gut microbiota, the central nervous system, and the immune system. The gut-brain axis has recently gained much attention due to its role in regulating host physiology. This review explores recent findings concerning potential pathways linking the gut-brain axis to the initiation, pathophysiology, and development of neurological disorders. Our objective of this work is to uncover causative factors and pinpoint particular pathways and therapeutic targets that may facilitate the translation of experimental animal research into practical applications for human patients. We highlight three distinct yet interrelated mechanisms: (1) disruptions of both the intestinal and blood-brain barriers, (2) persistent neuroinflammation, and (3) the role of the vagus nerve.


Subject(s)
Brain-Gut Axis , Gastrointestinal Microbiome , Humans , Gastrointestinal Microbiome/physiology , Animals , Brain-Gut Axis/physiology , Vagus Nerve/physiology , Neuroinflammatory Diseases/microbiology , Neuroinflammatory Diseases/immunology , Nervous System Diseases/microbiology , Blood-Brain Barrier/microbiology , Blood-Brain Barrier/metabolism
7.
Vet Res ; 55(1): 104, 2024 Aug 29.
Article in English | MEDLINE | ID: mdl-39210406

ABSTRACT

Meningitis induced by Pasteurella multocida has been substantially described in clinical practice in both human and veterinary medicine, but the underlying mechanisms have not been previously reported. In this study, we investigated the influence of P. multocida infection on the permeability of the blood-brain barrier (BBB) using different models. Our in vivo tests in a mouse model and in vitro tests using human brain microvascular endothelial cell (hBMEC) model showed that P. multocida infection increased murine BBB permeability in mice and hBMEC monolayer permeability. Furthermore, we observed that P. multocida infection resulted in decreased expression of tight junctions (ZO1, claudin-5, occludin) and adherens junctions (E-cadherin) between neighboring hBMECs. Subsequent experiments revealed that P. multocida infection promoted the activation of hypoxia inducible factor-1α (HIF-1α)/vascular endothelial growth factor A (VEGFA) signaling and NF-κB signaling, and suppressed the HIF-1α/VEGFA significantly remitted the decrease in ZO1/E-cadherin induced by P. multocida infection (P < 0.001). NF-κB signaling was found to contribute to the production of chemokines such as TNF-1α, IL-ß, and IL-6. Additionally, transmission electron microscopy revealed that paracellular migration might be the strategy employed by P. multocida to cross the BBB. This study provides the first evidence of the migration strategy used by P. multocida to traverse the mammalian BBB. The data presented herein will contribute to a better understanding of the pathogenesis of the zoonotic pathogen P. multocida.


Subject(s)
Adherens Junctions , Blood-Brain Barrier , Endothelial Cells , Pasteurella Infections , Pasteurella multocida , Tight Junctions , Animals , Pasteurella multocida/physiology , Blood-Brain Barrier/microbiology , Mice , Adherens Junctions/metabolism , Pasteurella Infections/veterinary , Pasteurella Infections/microbiology , Endothelial Cells/microbiology , Endothelial Cells/physiology , Tight Junctions/metabolism , Humans , Brain/microbiology , Brain/blood supply
8.
Virulence ; 15(1): 2387180, 2024 Dec.
Article in English | MEDLINE | ID: mdl-39192572

ABSTRACT

Streptococcus pneumoniae is a common pathogen associated with community-acquired bacterial meningitis, characterized by high morbidity and mortality rates. While vaccination reduces the incidence of meningitis, many survivors experience severe brain damage and corresponding sequelae. The pathogenesis of pneumococcal meningitis has not been fully elucidated. Currently, meningitis requires bacterial disruption of the blood - brain barrier, a process that involves the interaction of bacterial surface components with host cells and various inflammatory responses. This review delineates the global prevalence, pathogenesis, and treatment strategies of pneumococcal meningitis. The objective is to enhance the thorough comprehension of the clinical manifestations and biological mechanisms of the disease, thereby enabling more efficient prevention, diagnosis, and therapeutic interventions.


Subject(s)
Meningitis, Pneumococcal , Streptococcus pneumoniae , Humans , Meningitis, Pneumococcal/microbiology , Meningitis, Pneumococcal/drug therapy , Meningitis, Pneumococcal/therapy , Streptococcus pneumoniae/pathogenicity , Blood-Brain Barrier/microbiology , Pneumococcal Vaccines/immunology , Animals , Community-Acquired Infections/microbiology , Anti-Bacterial Agents/therapeutic use
9.
Int J Mol Sci ; 25(16)2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39201303

ABSTRACT

Alzheimer's disease (AD) is a neurodegenerative disease that is known to accumulate amyloid-ß (Aß) and tau protein. Clinical studies have not identified pathogenesis mechanisms or produced an effective cure for AD. The Aß monoclonal antibody lecanemab reduces Aß plaque formation for the treatment of AD, but more studies are required to increase the effectiveness of drugs to reduce cognitive decline. The lack of AD therapy targets and evidence of an association with an acute neuroinflammatory response caused by several bacteria and viruses in some individuals has led to the establishment of the infection hypothesis during the last 10 years. How pathogens cross the blood-brain barrier is highly topical and is seen to be pivotal in proving the hypothesis. This review summarizes the possible role of the gut microbiome in the pathogenesis of AD and feasible therapeutic approaches and current research limitations.


Subject(s)
Alzheimer Disease , Gastrointestinal Microbiome , Alzheimer Disease/microbiology , Alzheimer Disease/drug therapy , Alzheimer Disease/metabolism , Humans , Animals , Amyloid beta-Peptides/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/microbiology , tau Proteins/metabolism
10.
Poult Sci ; 103(10): 104059, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39068696

ABSTRACT

Riemerella anatipestifer is a Gram-negative, rod-shaped bacterium that is flagellated, non-budded, and encapsulated, measuring approximately 0.4 µm × 0.7 µm. After infecting ducklings with R. anatipestifer, the hosts exhibited pathological changes, such as bacterial meningitis, fibrinous pericarditis, and fibrinous peripheral hepatitis. The pathogenesis of meningitis caused by R. anatipestifer has not yet been elucidated. To investigate the key molecules or proteins involved in R. anatipestifer's penetration of the blood-brain barrier (BBB) and the subsequent development of duck meningitis, a duck meningitis model was established and characterized. Duckling brain tissues were collected and analyzed using 4D label-free proteomic technology. Differentially expressed proteins were analyzed using a series of bioinformatics methods and verified using RT-qPCR and Western-Blot. The results showed that the differentially expressed proteins were primarily related to intracellular transport, transport protein activity, and transmembrane transport protein activity, and were mainly enriched in pathways associated with reducing intercellular connections and adhesion and increasing cell migration and apoptosis. Thus, it is suggested that R. anatipestifer may penetrate the BBB via transcellular and paracellular pathways, causing neurological diseases such as meningitis. This study is the first to analyze R. anatipestifer-infected duckling brain tissue using proteomics, thus providing a direction for further research into the mechanisms of R. anatipestifer's penetration of the BBB.


Subject(s)
Brain , Ducks , Flavobacteriaceae Infections , Poultry Diseases , Proteomics , Riemerella , Animals , Poultry Diseases/microbiology , Flavobacteriaceae Infections/veterinary , Flavobacteriaceae Infections/microbiology , Brain/microbiology , Brain/metabolism , Meningitis, Bacterial/veterinary , Meningitis, Bacterial/microbiology , Proteome , Avian Proteins/metabolism , Avian Proteins/genetics , Blood-Brain Barrier/microbiology
11.
Microbiol Spectr ; 12(8): e0061224, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38980021

ABSTRACT

Streptococcus suis type 2 (SS2) is a zoonotic pathogen capable of eliciting meningitis, presenting significant challenges to both the swine industry and public health. Suilysin (Sly), one of SS2 most potent virulence determinants, releases a surfeit of inflammatory agents following red blood cell lysis. Notably, while current research on Sly role in SS2-induced meningitis predominantly centers on its interaction with the blood-brain barrier (BBB), the repercussions of Sly hemolytic products on BBB function have largely been sidestepped. In this vein, our study delves into the ramifications of Sly-induced hemolysis on BBB integrity. We discern that Sly hemolytic derivatives exacerbate the permeability of Sly-induced in vitro BBB models. Within these Sly hemolytic products, Interleukin-33 (IL-33) disrupts the expression and distribution of Claudin-5 in brain microvascular endothelial cells, facilitating the release of Interleukin-6 (IL-6) and Interleukin-8 (IL-8), thereby amplifying BBB permeability. Preliminary mechanistic insights suggest that IL-33-driven expression of IL-6 and IL-8 is orchestrated by the p38-mitogen-activated protein kinase signaling, whereas matrix metalloproteinase 9 mediates IL-33-induced suppression of Claudin-5. To validate these in vitro findings, an SS2-infected mouse model was established, and upon intravenous administration of growth stimulation expressed gene 2 (ST2) antibodies, in vivo results further underscored the pivotal role of the IL-33/ST2 axis during SS2 cerebral invasion. In summation, this study pioneerly illuminates the involvement of Sly hemolytic products in SS2-mediated BBB compromise and spotlights the instrumental role and primary mechanism of IL-33 therein. These insights enrich our comprehension of SS2 meningitis pathogenesis, laying pivotal groundwork for therapeutic advancements against SS2-induced meningitis.IMPORTANCEThe treatment of meningitis caused by Streptococcus suis type 2 (SS2) has always been a clinical challenge. Elucidating the molecular mechanisms by which SS2 breaches the blood-brain barrier (BBB) is crucial for the development of meningitis therapeutics. Suilysin (Sly) is one of the most important virulence factors of SS2, which can quickly lyse red blood cells and release large amounts of damage-associated molecular patterns, such as hemoglobin, IL-33, cyclophilin A, and so on. However, the impact of these hemolytic products on the function of BBB is unknown and ignored. This study is the first to investigate the effect of Sly hemolytic products on BBB function. The data are crucial for the study of the pathogenesis of SS2 meningitis and can provide an important reference for the development of meningitis therapeutics.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Hemolysin Proteins , Hemolysis , Interleukin-33 , Streptococcus suis , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/microbiology , Animals , Mice , Interleukin-33/metabolism , Humans , Hemolysin Proteins/metabolism , Streptococcus suis/pathogenicity , Endothelial Cells/microbiology , Endothelial Cells/metabolism , Streptococcal Infections/microbiology , Interleukin-6/metabolism , Interleukin-6/genetics , Interleukin-8/metabolism , Swine , Matrix Metalloproteinase 9/metabolism
12.
Sci Signal ; 17(839): eadq7330, 2024 Jun 04.
Article in English | MEDLINE | ID: mdl-38833529
13.
Vet Microbiol ; 295: 110161, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38945021

ABSTRACT

Streptococcus suis (S. suis) type 2 (SS2) is an important zoonotic pathogen causing severe neural infections in pigs and causes serious threat to public health. Inflammasome activation plays an important role in the host against microbial infection but the role of inflammasome activation in the blood-brain barrier (BBB) integrity during S. suis infection is rarely studied. This study investigated the mechanism by which S. suis-induced NLRP3 inflammasome activation led to BBB disruption. Our results showed that S. suis infection activated NLRP3 inflammasome in brain microvascular endothelial cells (BMECs) leading to the secretion of pro-inflammatory cytokines (IL-1ß, IL-6 and TNF-α) and chemokines (CCL-2 and CXCL-2) as well as the cleavage of Gasdermin D (GSDMD) which were significantly attenuated by inflammasome inhibitor MCC950. Furthermore, S. suis infection significantly downregulated expression of tight junctions (TJs) proteins and trans-endothelial electrical resistance (TEER) while NLRP3 inhibition rescued S. suis-induced degradation of TJs proteins and significantly reduced the number of S. suis crossing BBB in transwell infection model. Moreover, recombinant IL-1ß exacerbated the reduction of TJs proteins in BMECs. In murine S. suis-infection model, MCC950 reduced the bacterial load and the excessive inflammatory response in mice brain. In addition, the integrity of the BBB was protected with increased TJ proteins expression and decreased pathological injury after the inhibition of NLRP3 inflammasome, indicating NLRP3 inflammasome plays a destructive role in meningitis induced by S. suis. Our study expands the understanding on the role of NLRP3 inflammasome in bacterial meningitis, which provide the valuable information for the development of anti-infective agents targeting NLRP3 to treat bacterial meningitis.


Subject(s)
Blood-Brain Barrier , Endothelial Cells , Inflammasomes , NLR Family, Pyrin Domain-Containing 3 Protein , Streptococcal Infections , Streptococcus suis , Animals , Blood-Brain Barrier/microbiology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Inflammasomes/metabolism , Inflammasomes/immunology , Mice , Streptococcal Infections/immunology , Streptococcal Infections/microbiology , Endothelial Cells/microbiology , Cytokines/metabolism , Cytokines/genetics , Mice, Inbred C57BL , Brain/microbiology , Brain/immunology , Female
14.
Gut Microbes ; 16(1): 2363014, 2024.
Article in English | MEDLINE | ID: mdl-38904096

ABSTRACT

Gut bacteria regulate brain pathology of Alzheimer's disease (AD) patients and animal models; however, the underlying mechanism remains unclear. In this study, 3-month-old APP-transgenic female mice with and without knock-out of Il-17a gene were treated with antibiotics-supplemented or normal drinking water for 2 months. The antibiotic treatment eradicated almost all intestinal bacteria, which led to a reduction in Il-17a-expressing CD4-positive T lymphocytes in the spleen and gut, and to a decrease in bacterial DNA in brain tissue. Depletion of gut bacteria inhibited inflammatory activation in both brain tissue and microglia, lowered cerebral Aß levels, and promoted transcription of Arc gene in the brain of APP-transgenic mice, all of which effects were abolished by deficiency of Il-17a. As possible mechanisms regulating Aß pathology, depletion of gut bacteria inhibited ß-secretase activity and increased the expression of Abcb1 and Lrp1 in the brain or at the blood-brain barrier, which were also reversed by the absence of Il-17a. Interestingly, a crossbreeding experiment between APP-transgenic mice and Il-17a knockout mice further showed that deficiency of Il-17a had already increased Abcb1 and Lrp1 expression at the blood-brain barrier. Thus, depletion of gut bacteria attenuates inflammatory activation and amyloid pathology in APP-transgenic mice via Il-17a-involved signaling pathways. Our study contributes to a better understanding of the gut-brain axis in AD pathophysiology and highlights the therapeutic potential of Il-17a inhibition or specific depletion of gut bacteria that stimulate the development of Il-17a-expressing T cells.


Subject(s)
Alzheimer Disease , Brain , Disease Models, Animal , Gastrointestinal Microbiome , Interleukin-17 , Mice, Transgenic , Animals , Alzheimer Disease/microbiology , Alzheimer Disease/metabolism , Alzheimer Disease/pathology , Interleukin-17/metabolism , Interleukin-17/genetics , Mice , Brain/pathology , Brain/metabolism , Female , Mice, Knockout , Amyloid Precursor Protein Secretases/metabolism , Amyloid Precursor Protein Secretases/genetics , Amyloid beta-Peptides/metabolism , Anti-Bacterial Agents/pharmacology , Mice, Inbred C57BL , Microglia/metabolism , Microglia/pathology , Microglia/microbiology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/microbiology , Humans , Low Density Lipoprotein Receptor-Related Protein-1
15.
J Bacteriol ; 206(6): e0008724, 2024 06 20.
Article in English | MEDLINE | ID: mdl-38771039

ABSTRACT

Bacterial meningitis is a life-threatening infection of the central nervous system (CNS) that occurs when bacteria are able to cross the blood-brain barrier (BBB) or the meningeal-cerebrospinal fluid barrier (mBCSFB). The BBB and mBCSFB comprise highly specialized brain endothelial cells (BECs) that typically restrict pathogen entry. Group B Streptococcus (GBS or Streptococcus agalactiae) is the leading cause of neonatal meningitis. Until recently, identification of GBS virulence factors has relied on genetic screening approaches. Instead, we here conducted RNA-seq analysis on GBS when interacting with induced pluripotent stem cell-derived BECs (iBECs) to pinpoint virulence-associated genes. Of the 2,068 annotated protein-coding genes of GBS, 430 transcripts displayed significant changes in expression after interacting with BECs. Notably, we found that the majority of differentially expressed GBS transcripts were downregulated (360 genes) during infection of iBECs. Interestingly, codY, encoding a pleiotropic transcriptional repressor in low-G + C Gram-positive bacteria, was identified as being highly downregulated. We conducted qPCR to confirm the codY downregulation observed via RNA-seq during the GBS-iBEC interaction and obtained codY mutants in three different GBS background parental strains. As anticipated from the RNA-seq results, the [Formula: see text]codY strains were more adherent and invasive in two in vitro BEC models. Together, this demonstrates the utility of RNA-seq during the BEC interaction to identify GBS virulence modulators. IMPORTANCE: Group B Streptococcus (GBS) meningitis remains the leading cause of neonatal meningitis. Research work has identified surface factors and two-component systems that contribute to GBS disruption of the blood-brain barrier (BBB). These discoveries often relied on genetic screening approaches. Here, we provide transcriptomic data describing how GBS changes its transcriptome when interacting with brain endothelial cells. Additionally, we have phenotypically validated these data by obtaining mutants of a select regulator that is highly down-regulated during infection and testing on our BBB model. This work provides the research field with a validated data set that can provide an insight into potential pathways that GBS requires to interact with the BBB and open the door to new discoveries.


Subject(s)
Brain , Endothelial Cells , Streptococcus agalactiae , Transcriptome , Streptococcus agalactiae/genetics , Streptococcus agalactiae/metabolism , Streptococcus agalactiae/pathogenicity , Endothelial Cells/microbiology , Humans , Brain/microbiology , Brain/metabolism , Blood-Brain Barrier/microbiology , Blood-Brain Barrier/metabolism , Gene Expression Regulation, Bacterial , Virulence Factors/genetics , Virulence Factors/metabolism , Virulence , Streptococcal Infections/microbiology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Meningitis, Bacterial/microbiology
16.
J Clin Periodontol ; 51(7): 818-839, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38414291

ABSTRACT

AIM: Blood-brain barrier (BBB) disorder is one of the early findings in cognitive impairments. We have recently found that Porphyromonas gingivalis bacteraemia can cause cognitive impairment and increased BBB permeability. This study aimed to find out the possible key virulence factors of P. gingivalis contributing to the pathological process. MATERIALS AND METHODS: C57/BL6 mice were infected with P. gingivalis or gingipains or P. gingivalis lipopolysaccharide (P. gingivalis LPS group) by tail vein injection for 8 weeks. The cognitive behaviour changes in mice, the histopathological changes in the hippocampus and cerebral cortex, the alternations of BBB permeability, and the changes in Mfsd2a and Cav-1 levels were measured. The mechanisms of Ddx3x-induced regulation on Mfsd2a by arginine-specific gingipain A (RgpA) in BMECs were explored. RESULTS: P. gingivalis and gingipains significantly promoted mice cognitive impairment, pathological changes in the hippocampus and cerebral cortex, increased BBB permeability, inhibited Mfsd2a expression and up-regulated Cav-1 expression. After RgpA stimulation, the permeability of the BBB model in vitro increased, and the Ddx3x/Mfsd2a/Cav-1 regulatory axis was activated. CONCLUSIONS: Gingipains may be one of the key virulence factors of P. gingivalis to impair cognition and enhance BBB permeability by the Ddx3x/Mfsd2a/Cav-1 axis.


Subject(s)
Blood-Brain Barrier , Gingipain Cysteine Endopeptidases , Mice, Inbred C57BL , Porphyromonas gingivalis , Virulence Factors , Animals , Porphyromonas gingivalis/pathogenicity , Blood-Brain Barrier/microbiology , Mice , Virulence Factors/metabolism , Adhesins, Bacterial/metabolism , Male , Disease Models, Animal , Permeability , Cognitive Dysfunction/microbiology , Cognitive Dysfunction/metabolism , Hippocampus/metabolism , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/complications
17.
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
18.
Cell Commun Signal ; 22(1): 44, 2024 01 17.
Article in English | MEDLINE | ID: mdl-38233877

ABSTRACT

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.


Subject(s)
Meningitis, Bacterial , Meningitis, Escherichia coli , Humans , Infant, Newborn , Blood-Brain Barrier/microbiology , Endothelial Cells/metabolism , Escherichia coli , Meningitis, Bacterial/metabolism , Meningitis, Escherichia coli/metabolism
19.
Front Cell Infect Microbiol ; 13: 1113528, 2023.
Article in English | MEDLINE | ID: mdl-37065199

ABSTRACT

The Gram-negative bacterium Neisseria meningitidis, which causes meningitis in humans, has been demonstrated to manipulate or alter host signalling pathways during infection of the central nervous system (CNS). However, these complex signalling networks are not completely understood. We investigate the phosphoproteome of an in vitro model of the blood-cerebrospinal fluid barrier (BCSFB) based on human epithelial choroid plexus (CP) papilloma (HIBCPP) cells during infection with the N. meningitidis serogroup B strain MC58 in presence and absence of the bacterial capsule. Interestingly, our data demonstrates a stronger impact on the phosphoproteome of the cells by the capsule-deficient mutant of MC58. Using enrichment analyses, potential pathways, molecular processes, biological processes, cellular components and kinases were determined to be regulated as a consequence of N. meningitidis infection of the BCSFB. Our data highlight a variety of protein regulations that are altered during infection of CP epithelial cells with N. meningitidis, with the regulation of several pathways and molecular events only being detected after infection with the capsule-deficient mutant. Mass spectrometry proteomics data are available via ProteomeXchange with identifier PXD038560.


Subject(s)
Neisseria meningitidis , Humans , Neisseria meningitidis/physiology , Choroid Plexus/microbiology , Epithelial Cells/microbiology , Blood-Brain Barrier/microbiology , Cell Line, Tumor
20.
Int J Oral Sci ; 15(1): 3, 2023 01 12.
Article in English | MEDLINE | ID: mdl-36631446

ABSTRACT

Bacteremia induced by periodontal infection is an important factor for periodontitis to threaten general health. P. gingivalis DNA/virulence factors have been found in the brain tissues from patients with Alzheimer's disease (AD). The blood-brain barrier (BBB) is essential for keeping toxic substances from entering brain tissues. However, the effect of P. gingivalis bacteremia on BBB permeability and its underlying mechanism remains unclear. In the present study, rats were injected by tail vein with P. gingivalis three times a week for eight weeks to induce bacteremia. An in vitro BBB model infected with P. gingivalis was also established. We found that the infiltration of Evans blue dye and Albumin protein deposition in the rat brain tissues were increased in the rat brain tissues with P. gingivalis bacteremia and P. gingivalis could pass through the in vitro BBB model. Caveolae were detected after P. gingivalis infection in BMECs both in vivo and in vitro. Caveolin-1 (Cav-1) expression was enhanced after P. gingivalis infection. Downregulation of Cav-1 rescued P. gingivalis-enhanced BMECs permeability. We further found P. gingivalis-gingipain could be colocalized with Cav-1 and the strong hydrogen bonding between Cav-1 and arg-specific-gingipain (RgpA) were detected. Moreover, P. gingivalis significantly inhibited the major facilitator superfamily domain containing 2a (Mfsd2a) expression. Mfsd2a overexpression reversed P. gingivalis-increased BMECs permeability and Cav-1 expression. These results revealed that Mfsd2a/Cav-1 mediated transcytosis is a key pathway governing BBB BMECs permeability induced by P. gingivalis, which may contribute to P. gingivalis/virulence factors entrance and the subsequent neurological impairments.


Subject(s)
Bacteremia , Blood-Brain Barrier , Caveolin 1 , Porphyromonas gingivalis , Animals , Rats , Bacteremia/complications , Bacteremia/metabolism , Blood-Brain Barrier/metabolism , Blood-Brain Barrier/microbiology , Caveolin 1/metabolism , Gingipain Cysteine Endopeptidases/metabolism , Permeability , Porphyromonas gingivalis/pathogenicity , Transcytosis , Virulence Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL