Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Commun Biol ; 7(1): 551, 2024 May 08.
Article in English | MEDLINE | ID: mdl-38720110

ABSTRACT

Fusobacterium nucleatum, a gram-negative oral bacterium, has been consistently validated as a strong contributor to the progression of several types of cancer, including colorectal (CRC) and pancreatic cancer. While previous in vitro studies have shown that intracellular F. nucleatum enhances malignant phenotypes such as cell migration, the dependence of this regulation on features of the tumor microenvironment (TME) such as oxygen levels are wholly uncharacterized. Here we examine the influence of hypoxia in facilitating F. nucleatum invasion and its effects on host responses focusing on changes in the global epigenome and transcriptome. Using a multiomic approach, we analyze epigenomic alterations of H3K27ac and global transcriptomic alterations sustained within a hypoxia and normoxia conditioned CRC cell line HCT116 at 24 h following initial infection with F. nucleatum. Our findings reveal that intracellular F. nucleatum activates signaling pathways and biological processes in host cells similar to those induced upon hypoxia conditioning in the absence of infection. Furthermore, we show that a hypoxic TME favors F. nucleatum invasion and persistence and therefore infection under hypoxia may amplify malignant transformation by exacerbating the effects induced by hypoxia alone. These results motivate future studies to investigate host-microbe interactions in tumor tissue relevant conditions that more accurately define parameters for targeted cancer therapies.


Subject(s)
Colorectal Neoplasms , Epigenome , Fusobacterium Infections , Fusobacterium nucleatum , Oxygen , Transcriptome , Humans , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/physiology , Fusobacterium nucleatum/pathogenicity , Colorectal Neoplasms/genetics , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , HCT116 Cells , Fusobacterium Infections/genetics , Fusobacterium Infections/microbiology , Fusobacterium Infections/metabolism , Oxygen/metabolism , Tumor Microenvironment/genetics , Gene Expression Regulation, Neoplastic
2.
J Control Release ; 363: 43-56, 2023 11.
Article in English | MEDLINE | ID: mdl-37734673

ABSTRACT

The gut microbiota is closely associated with the progression of colorectal cancer (CRC) in which Fusobacterium nucleatum (F. nucleatum) was found to induce cancer resistance to chemotherapeutics. To relieve F. nucleatum-induced drug resistance, herein, we found that short-chain fatty acid butyrate can inhibit the growth, enrichment and adhesion of F. nucleatum in colorectal cancer tissues by downregulating the expression of adhesion-associated outer membrane proteins, including RadD, FomA, and FadA, to reduce the colonization and invasion of F. nucleatum and relieve the chemoresistance induced by F. nucleatum. Leveraging the killing effect of butyrate on F. nucleatum, sodium butyrate (NaBu) was encapsulated in liposomes or prepared as NaBu tablets with Eudragit S100 coating and administered by intravenous injection or oral administration, respectively. Interestingly, both intravenous administration of NaBu liposomes and oral delivery of NaBu tablets could effectively inhibit the proliferation of F. nucleatum and significantly improve the therapeutic efficacy of oxaliplatin in mice with subcutaneous colorectal tumors, orthotopic colorectal tumors and even spontaneously formed colorectal tumors. Thus, our work provides a simple but effective formulation of NaBu to relieve F. nucleatum-induced chemoresistance, exhibiting ideal clinical application prospects.


Subject(s)
Colorectal Neoplasms , Fusobacterium Infections , Animals , Mice , Fusobacterium nucleatum/metabolism , Butyrates , Drug Resistance, Neoplasm , Liposomes/metabolism , Fusobacterium Infections/complications , Fusobacterium Infections/metabolism , Fusobacterium Infections/microbiology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism
3.
Infect Immun ; 91(8): e0010223, 2023 08 16.
Article in English | MEDLINE | ID: mdl-37404144

ABSTRACT

Fusobacterium nucleatum colonization contributes to the occurrence of portal vein thrombosis in patients with gastric cancer (GC). However, the underlying mechanism by which F. nucleatum promotes thrombosis remains unclear. In this study, we recruited a total of 91 patients with GC and examined the presence of F. nucleatum in tumor and adjacent non-tumor tissues by fluorescence in situ hybridization and quantitative PCR. Neutrophil extracellular traps (NETs) were detected by immunohistochemistry. Extracellular vesicles (EVs) were extracted from the peripheral blood and proteins in the EVs were identified by mass spectrometry (MS). HL-60 cells differentiated into neutrophils were used to package engineered EVs to imitate the EVs released from NETs. Hematopoietic progenitor cells (HPCs) and K562 cells were used for megakaryocyte (MK) in vitro differentiation and maturation to examine the function of EVs. We observed that F. nucleatum-positive patients had increased NET and platelet counts. EVs from F. nucleatum-positive patients could promote the differentiation and maturation of MKs and had upregulated 14-3-3 proteins, especially 14-3-3ε. 14-3-3ε upregulation promoted MK differentiation and maturation in vitro. HPCs and K562 cells could receive 14-3-3ε from the EVs, which interacted with GP1BA and 14-3-3ζ to trigger PI3K-Akt signaling. In conclusion, we identified for the first time that F. nucleatum infection promotes NET formation, which releases EVs containing 14-3-3ε. These EVs could deliver 14-3-3ε to HPCs and promote their differentiation into MKs via activation of PI3K-Akt signaling.


Subject(s)
Extracellular Vesicles , Fusobacterium Infections , Stomach Neoplasms , Humans , Fusobacterium nucleatum/metabolism , In Situ Hybridization, Fluorescence , 14-3-3 Proteins/genetics , 14-3-3 Proteins/metabolism , Megakaryocytes/metabolism , Megakaryocytes/pathology , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Fusobacterium Infections/metabolism , Fusobacterium Infections/microbiology , Fusobacterium Infections/pathology , Extracellular Vesicles/metabolism
4.
Int J Oral Sci ; 15(1): 8, 2023 02 08.
Article in English | MEDLINE | ID: mdl-36754953

ABSTRACT

Fusobacterium nucleatum (F. nucleatum) is an early pathogenic colonizer in periodontitis, but the host response to infection with this pathogen remains unclear. In this study, we built an F. nucleatum infectious model with human periodontal ligament stem cells (PDLSCs) and showed that F. nucleatum could inhibit proliferation, and facilitate apoptosis, ferroptosis, and inflammatory cytokine production in a dose-dependent manner. The F. nucleatum adhesin FadA acted as a proinflammatory virulence factor and increased the expression of interleukin(IL)-1ß, IL-6 and IL-8. Further study showed that FadA could bind with PEBP1 to activate the Raf1-MAPK and IKK-NF-κB signaling pathways. Time-course RNA-sequencing analyses showed the cascade of gene activation process in PDLSCs with increasing durations of F. nucleatum infection. NFκB1 and NFκB2 upregulated after 3 h of F. nucleatum-infection, and the inflammatory-related genes in the NF-κB signaling pathway were serially elevated with time. Using computational drug repositioning analysis, we predicted and validated that two potential drugs (piperlongumine and fisetin) could attenuate the negative effects of F. nucleatum-infection. Collectively, this study unveils the potential pathogenic mechanisms of F. nucleatum and the host inflammatory response at the early stage of F. nucleatum infection.


Subject(s)
Fusobacterium Infections , Fusobacterium nucleatum , Humans , Fusobacterium nucleatum/metabolism , NF-kappa B/metabolism , Periodontal Ligament/metabolism , Signal Transduction , Fusobacterium Infections/metabolism , Fusobacterium Infections/microbiology , Fusobacterium Infections/pathology , Stem Cells/metabolism
5.
Ann Med ; 54(1): 989-1003, 2022 12.
Article in English | MEDLINE | ID: mdl-35435776

ABSTRACT

BACKGROUND: To analyse the regulatory effect of Fusobacterium nucleatum (Fn) on NOD-like receptor protein 3 (NLRP3) and myeloid-derived suppressor cells (MDSCs) in oesophageal squamous cell carcinoma (ESCC) as well as its effect on cisplatin (CDDP) therapy and to explore its clinical significance. METHODS: Fn infection, NLRP3 expression and MDSCs infiltration in ESCC tissues were detected by RNAscope and immunohistochemistry (IHC). The correlation between these three factors and the clinicopathological features and survival of ESCC patients was analysed. A coculture system of human peripheral blood monocytes (PBMCs) and ESCC cells was established to simulate the tumour microenvironment. In vitro and in vivo models were used to analyse the effects of Fn on the percentage of MDSCs in the coculture system and the NLRP3 expression level and CDDP sensitivity of ESCC cells. RESULTS: Fn infection was consistent with high NLRP3 expression and MDSCs enrichment in ESCC tissues. Moreover, the survival time of ESCC patients was significantly shortened under Fn infection, high NLRP3 expression and MDSCs enrichment. In the in vitro and in vivo models, Fn induced abundant enrichment of MDSCs by inducing high expression of NLRP3 in ESCC cells and reducing the sensitivity of ESCC cells to CDDP. CONCLUSIONS: Fn infection can induce high expression of NLRP3 in ESCC, lead to MDSCs enrichment, weaken the body's antitumour immunity, and lead to CDDP treatment resistance. The effective elimination of Fn and the inhibition of MDSCs enrichment may provide new strategies and treatments for ESCC.HighlightsThe survival of ESCC patients with Fn infection, high NLRP3 expression and MDSCs enrichment was significantly shortened.Fn infection could cause CDDP resistance in ESCC.Fn could induce the enrichment of MDSCs in the tumour microenvironment by activating NLRP3 in ESCC cells.


Subject(s)
Esophageal Neoplasms , Fusobacterium Infections , Myeloid-Derived Suppressor Cells , Cisplatin/pharmacology , Esophageal Neoplasms/drug therapy , Fusobacterium Infections/metabolism , Fusobacterium Infections/microbiology , Fusobacterium nucleatum , Humans , Myeloid-Derived Suppressor Cells/metabolism , Myeloid-Derived Suppressor Cells/pathology , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , NLR Proteins/metabolism , Tumor Microenvironment
6.
Adv Sci (Weinh) ; 9(12): e2105222, 2022 04.
Article in English | MEDLINE | ID: mdl-35170250

ABSTRACT

Fusobacterium nucleatum is a critical microbe that contributes to colorectal cancer progression and chemoresistance. However, whether and how F. nucleatum regulates colorectal cancer stem-like cells (CCSCs) remains unknown. Here, the authors show that F. nucleatum promotes CCSC self-renewal, and non-CCSCs to acquire CCSC features by manipulating cellular lipid accumulation. F. nucleatum infection decreases lipid accumulation in CCSCs by enhancing fatty acid oxidation, thus promoting CCSC self-renewal. In contrast, F. nucleatum increases lipid accumulation in non-CCSCs by promoting fatty acid formation. Lipids are deposited as lipid droplets, which recruits Numb, a key cell fate regulator, through the AP2A/ACSL3 complex, and MDM2, an E3 ubiquitin ligase, though VCP and UBXD8. On lipid droplets, Numb is degraded by MDM2, activating Notch signaling, thus promoting gain of stem-like cell features. Their findings demonstrate that F. nucleatum directly manipulates colorectal cancer cell fate and reveal the mechanism of lipid droplet-mediated Numb degradation for activating Notch signaling.


Subject(s)
Colorectal Neoplasms , Fusobacterium Infections , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Fatty Acids , Fusobacterium Infections/metabolism , Fusobacterium Infections/microbiology , Fusobacterium nucleatum/physiology , Humans , Lipid Droplets/metabolism , Lipids , Membrane Proteins , Nerve Tissue Proteins , Stem Cells/metabolism
7.
Cancer Lett ; 530: 59-67, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35033591

ABSTRACT

Fusobacterium nucleatum, found in the oral cavity, influences the progression of gastrointestinal cancers. Additionally, our previous results suggested that F. nucleatum is associated with poor patient prognosis in esophageal squamous cell carcinoma (ESCC). However, the mechanism by which F. nucleatum affects aggressive tumor behavior has yet to be elucidated. We have conducted this clinical, in vitro, and in vivo study to clarify the mechanism of ESCC progression induced by F. nucleatum. Transmission electron microscopy revealed that F. nucleatum invaded and occupied ESCC cells and impacted gene and protein expression. Comprehensive mRNA expression and pathway enrichment analyses of F. nucleatum-treated ESCC cells identified the "NF-κB" and "NOD-like receptor" signaling pathways as enriched. We confirmed the relationship between the presence of F. nucleatum and NF-κB activation in resected ESCC tissues. Furthermore, F. nucleatum-treated ESCC cells demonstrated enhanced growth ability, and NF-κB activation, as well as overexpression of NOD1 and phosphorylated RIPK2. Furthermore, treated cells showed accelerated tumor growth, with NF-κB activation in xenograft models. F. nucleatum invaded ESCC cells and induced the NF-κB pathway through the NOD1/RIPK2 pathway, leading to tumor progression.


Subject(s)
Esophageal Neoplasms/microbiology , Esophageal Squamous Cell Carcinoma/microbiology , Fusobacterium Infections/metabolism , Fusobacterium nucleatum/pathogenicity , NF-kappa B/metabolism , Nod1 Signaling Adaptor Protein/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/metabolism , Animals , Cell Line, Tumor , Disease Progression , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Female , Humans , Mice , Mice, Inbred BALB C , Mice, Nude , Signal Transduction/physiology
8.
Gut Microbes ; 13(1): 1980347, 2021.
Article in English | MEDLINE | ID: mdl-34632963

ABSTRACT

Colorectal cancer (CRC) is one of the most common malignant tumors and is associated with Fusobacterium nucleatum (F. nucleatum, Fn) infection. In this study, we explored the role of F. nucleatum in the CRC metastasis. Our results showed that the abundance of F. nucleatum was enriched in the feces and tumors of patients with CRC and tended to increase in stage IV compared to stage I in patients with metastatic CRC. Tumor-derived CCL20 activated by F. nucleatum not only increases CRC metastasis, but also participates in the reprograming of the tumor microenvironment. F. nucleatum promoted macrophage infiltration through CCL20 activation and simultaneously induced M2 macrophage polarization, enhancing the metastasis of CRC. In addition, we identified using database prediction and luciferase activity hat miR-1322, a candidate regulatory micro-RNA, could bind to CCL20 directly. F. nucleatum infection decreased the expression of miR-1322 by activating the NF-κB signaling pathway in CRC cells. In conclusion, F. nucleatum promotes CRC metastasis through the miR-1322/CCL20 axis and M2 polarization.


Subject(s)
Chemokine CCL20/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Fusobacterium nucleatum/physiology , Macrophages/cytology , MicroRNAs/metabolism , Animals , Cell Movement , Cell Polarity , Chemokine CCL20/genetics , Colorectal Neoplasms/pathology , Colorectal Neoplasms/physiopathology , Feces/microbiology , Female , Fusobacterium Infections/metabolism , Fusobacterium Infections/microbiology , Fusobacterium Infections/pathology , Fusobacterium Infections/physiopathology , Gastrointestinal Microbiome , Humans , Macrophages/metabolism , Male , Mice , MicroRNAs/genetics , NF-kappa B/genetics , NF-kappa B/metabolism , Neoplasm Metastasis
9.
mBio ; 12(5): e0206221, 2021 10 26.
Article in English | MEDLINE | ID: mdl-34700376

ABSTRACT

Fusobacterium nucleatum is a ubiquitous opportunistic pathogen with an emerging role as an oncomicrobe in colorectal cancer and other cancer settings. F. nucleatum can adhere to and invade host cells in a manner that varies across F. nucleatum strains and host cell phenotypes. Here, we performed pairwise cocultures between three F. nucleatum strains and two immortalized primary host cell types (human colonic epithelial [HCE] cells and human carotid artery endothelial [HCAE] cells) followed by transcriptome sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) to investigate transcriptional and epigenetic host cell responses. We observed that F. nucleatum-induced host cell transcriptional modulation involves strong upregulation of genes related to immune migration and inflammatory processes, such as TNF, CXCL8, CXCL1, and CCL20. Furthermore, we identified genes strongly upregulated in a cell line-specific manner. In HCE cells, overexpressed genes included UBD and DUOX2/DUOXA2, associated with p53 degradation-mediated proliferation and intestinal reactive oxygen species (ROS) production, respectively. In HCAE cells, overexpressed genes included EFNA1 and LIF, two genes commonly upregulated in colorectal cancer and associated with poor patient outcomes, and PTGS2 (COX2), a gene associated with the protective effect of aspirin in the colorectal cancer setting. Interestingly, we also observed downregulation of numerous histone modification genes upon F. nucleatum exposure. We used the ChIP-seq data to annotate chromatin states genome wide and found significant chromatin remodeling following F. nucleatum exposure in HCAE cells, with increased frequencies of active enhancer and low-signal/quiescent states. Thus, our results highlight increased inflammation and chemokine gene expression as conserved host cell responses to F. nucleatum exposure and extensive host cell epigenomic changes specific to host cell type. IMPORTANCE Fusobacterium nucleatum is a bacterium normally found in the healthy oral cavity but also has an emerging role in colorectal cancer and other cancer settings. The host-microbe interactions of F. nucleatum and its involvement in tumor initiation, progression, and treatment resistance are not fully understood. We explored host cell changes that occur in response to F. nucleatum. We identified key genes differentially expressed in response to various conditions of F. nucleatum exposure and determined that the conserved host cell response to F. nucleatum was dominated by increased inflammation and chemokine gene expression. Additionally, we found extensive host cell epigenomic changes as a novel aspect of host modulation associated with F. nucleatum exposure. These results extend our understanding of F. nucleatum as an emerging pathogen and highlight the importance of considering strain heterogeneity and host cell phenotypic variation when exploring pathogenic mechanisms of F. nucleatum.


Subject(s)
Epigenome , Fusobacterium Infections/genetics , Fusobacterium Infections/microbiology , Fusobacterium nucleatum/physiology , Transcriptome , Cell Line , Chemokine CCL20/genetics , Chemokine CCL20/metabolism , Dual Oxidases/genetics , Dual Oxidases/metabolism , Endothelial Cells/metabolism , Endothelial Cells/microbiology , Epithelial Cells/metabolism , Epithelial Cells/microbiology , Fusobacterium Infections/metabolism , Fusobacterium nucleatum/genetics , Host-Pathogen Interactions , Humans
10.
Cancer Res ; 81(17): 4485-4498, 2021 09 01.
Article in English | MEDLINE | ID: mdl-34162680

ABSTRACT

Emerging research has revealed regulation of colorectal cancer metabolism by bacteria. Fusobacterium nucleatum (Fn) plays a crucial role in the development of colorectal cancer, however, whether Fn infection modifies metabolism in patients with colorectal cancer remains unknown. Here, LC-MS/MS-based lipidomics identified the upregulation of cytochrome P450 monooxygenases, primarily CYP2J2, and their mediated product 12,13-EpOME in patients with colorectal cancer tumors and mouse models, which increased the invasive and migratory ability of colorectal cancer cells in vivo and in vitro by regulating the epithelial-mesenchymal transition (EMT). Metagenomic sequencing indicated a positive correlation between increased levels of fecal Fn and serum 12,13-EpOME in patients with colorectal cancer. High levels of CYP2J2 in tumor tissues also correlated with high Fn levels and worse overall survival in patients with stage III/IV colorectal cancer. Moreover, Fn was found to activate TLR4/AKT signaling, downregulating Keap1 and increasing NRF2 to promote transcription of CYP2J2. Collectively, these data identify that Fn promotes EMT and metastasis in colorectal cancer by activating a TLR4/Keap1/NRF2 axis to increase CYP2J2 and 12,13-EpOME, which could serve as clinical biomarkers and therapeutic targets for Fn-infected patients with colorectal cancer. SIGNIFICANCE: This study uncovers a mechanism by which Fusobacterium nucleatum regulates colorectal cancer metabolism to drive metastasis, suggesting the potential biomarker and therapeutic utility of the CYP2J2/12,13-EpOME axis in Fn-infected patients.


Subject(s)
Colorectal Neoplasms/metabolism , Cytochrome P-450 Enzyme System/metabolism , Fusobacterium Infections/metabolism , Kelch-Like ECH-Associated Protein 1/metabolism , NF-E2-Related Factor 2/metabolism , Oleic Acids/metabolism , Toll-Like Receptor 4/metabolism , Aged , Animals , Carcinogenesis , Cell Line, Tumor , Cell Movement , Colorectal Neoplasms/complications , Colorectal Neoplasms/microbiology , Cytochrome P-450 CYP2J2/genetics , Epithelial-Mesenchymal Transition , Female , Fusobacterium Infections/complications , Fusobacterium Infections/microbiology , Fusobacterium nucleatum/metabolism , HCT116 Cells , HEK293 Cells , Humans , Male , Metabolomics , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Middle Aged , Neoplasm Metastasis , Signal Transduction
11.
Proc Natl Acad Sci U S A ; 118(23)2021 06 08.
Article in English | MEDLINE | ID: mdl-34074747

ABSTRACT

A gram-negative colonizer of the oral cavity, Fusobacterium nucleatum not only interacts with many pathogens in the oral microbiome but also has the ability to spread to extraoral sites including placenta and amniotic fluid, promoting preterm birth. To date, however, the molecular mechanism of interspecies interactions-termed coaggregation-by F. nucleatum and how coaggregation affects bacterial virulence remain poorly defined. Here, we employed genome-wide transposon mutagenesis to uncover fusobacterial coaggregation factors, revealing the intertwined function of a two-component signal transduction system (TCS), named CarRS, and a lysine metabolic pathway in regulating the critical coaggregation factor RadD. Transcriptome analysis shows that CarR modulates a large regulon including radD and lysine metabolic genes, such as kamA and kamD, the expression of which are highly up-regulated in the ΔcarR mutant. Significantly, the native culture medium of ΔkamA or ΔkamD mutants builds up abundant amounts of free lysine, which blocks fusobacterial coaggregation with streptococci. Our demonstration that lysine-conjugated beads trap RadD from the membrane lysates suggests that lysine utilizes RadD as its receptor to act as a metabolic inhibitor of coaggregation. Lastly, using a mouse model of preterm birth, we show that fusobacterial virulence is significantly attenuated with the ΔkamA and ΔcarR mutants, in contrast to the enhanced virulence phenotype observed upon diminishing RadD (ΔradD or ΔcarS mutant). Evidently, F. nucleatum employs the TCS CarRS and environmental lysine to modulate RadD-mediated interspecies interaction, virulence, and nutrient acquisition to thrive in the adverse environment of oral biofilms and extraoral sites.


Subject(s)
Bacterial Proteins , Fusobacterium Infections , Fusobacterium nucleatum , Signal Transduction/genetics , Virulence Factors , Animals , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Fusobacterium Infections/genetics , Fusobacterium Infections/metabolism , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/pathogenicity , Genome-Wide Association Study , Humans , Mice , Premature Birth/genetics , Premature Birth/metabolism , Premature Birth/microbiology , Virulence Factors/genetics , Virulence Factors/metabolism
12.
Int J Mol Med ; 47(1): 125-136, 2021 01.
Article in English | MEDLINE | ID: mdl-33236145

ABSTRACT

Autophagy plays a dual role in the responses to the gut microflora. The present study aimed to examine the effects of Lactobacillus rhamnosus (L. rhamnosus) on Fusobacterium nucleatum (F. nucleatum)­induced intestinal dysfunction and to elucidate the underlying mechanisms, with particular focus on autophagy. Inflammatory models were established by treatment with L. rhamnosus following F. nucleatum intervention using cells or a mouse model of dextran sulfate sodium (DSS)­induced acute colitis. Autophagosomes were visualized by confocal microscopy following transfection with a tandem GFP­mCherry­LC3 construct and also by transmission electron microscopy. Autophagy­associated protein levels were examined by western blot analysis and immunohistochemistry. It was observed that F. nucleatum induced the production of pro­inflammatory cytokines in Caco­2 cells and aggravated DSS­induced acute colitis. The autophagic flux was impaired following infection with F. nucleatum. L. rhamnosus treatment attenuated the inflammation induced by F. nucleatum infection and effectively recovered the impaired autophagic flux. In addition, the production of pro­inflammatory cytokines induced by F. nucleatum was enhanced with autophagy inhibitors or the RNA interference of autophagy­related gene 16 like 1 (Atg16L1) in Caco­2 cells. Notably, this inhibition of autophagy weakened the effects of L. rhamnosus. Finally, the PI3K/AKT/mTOR pathway was found to be involved in this process. On the whole, the present study demonstrates that the mediation of autophagy by L. rhamnosus may be involved in the protective effects against F. nucleatum­related intestinal inflammation. Thus, L. rhamnosus treatment may prove to be a novel therapeutic strategy for F. nucleatum­realated gut disorders.


Subject(s)
Autophagy , Colitis/metabolism , Fusobacterium Infections/metabolism , Fusobacterium nucleatum/metabolism , Lacticaseibacillus rhamnosus/metabolism , Caco-2 Cells , Colitis/chemically induced , Colitis/pathology , Dextran Sulfate/toxicity , Fusobacterium Infections/chemically induced , Fusobacterium Infections/pathology , Humans
13.
Sci Signal ; 13(641)2020 07 21.
Article in English | MEDLINE | ID: mdl-32694172

ABSTRACT

Fusobacterium nucleatum is implicated in accelerating colorectal cancer (CRC) and is found within metastatic CRC cells in patient biopsies. Here, we found that bacterial invasion of CRC cells and cocultured immune cells induced a differential cytokine secretion that may contribute to CRC metastasis. We used a modified galactose kinase markerless gene deletion approach and found that F. nucleatum invaded cultured HCT116 CRC cells through the bacterial surface adhesin Fap2. In turn, Fap2-dependent invasion induced the secretion of the proinflammatory cytokines IL-8 and CXCL1, which are associated with CRC progression and promoted HCT116 cell migration. Conditioned medium from F. nucleatum-infected HCT116 cells caused naïve cells to migrate, which was blocked by depleting CXCL1 and IL-8 from the conditioned medium. Cytokine secretion from HCT116 cells and cellular migration were attenuated by inhibiting F. nucleatum host-cell binding and entry using galactose sugars, l-arginine, neutralizing membrane protein antibodies, or fap2 deletion. F. nucleatum also induces the mobilization of immune cells in the tumor microenvironment. However, in neutrophils and macrophages, the bacterial-induced secretion of cytokines was Fap2 independent. Thus, our findings show that F. nucleatum both directly and indirectly modulates immune and cancer cell signaling and migration. Because increased IL-8 and CXCL1 production in tumors is associated with increased metastatic potential and cell seeding, poor prognosis, and enhanced recruitment of tumor-associated macrophages and fibroblasts, we propose that inhibition of host-cell binding and invasion, potentially through vaccination or novel galactoside compounds, could be an effective strategy for reducing F. nucleatum-associated CRC metastasis.


Subject(s)
Chemokine CXCL1/metabolism , Fusobacterium Infections/metabolism , Fusobacterium nucleatum/metabolism , Interleukin-8/metabolism , Colorectal Neoplasms/etiology , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Fusobacterium Infections/complications , Fusobacterium Infections/pathology , HCT116 Cells , Humans
14.
Anaerobe ; 63: 102184, 2020 Jun.
Article in English | MEDLINE | ID: mdl-32247918

ABSTRACT

Fusobacterium necrophorum is a Gram negative, spore-free, anaerobic bacterium that can cause pyogenic and necrotic infections in animals and humans. It is a major bovine pathogen and causes hepatic abscesses, foot rot, and necrotic laryngitis. The 43K OMP of F. necrophorum is an outer membrane protein with molecular weight of 43 kDa, exhibiting similarity to pore-forming proteins of other Fusobacterium species that plays an important role in bacterial infections. However, the role of 43K OMP in F. necrophorum adhesion remains unknown. In this study, we evaluated whether the 43K OMP of F. necrophorum mediates adhesion to BHK-21 cells and performed a preliminary screen of the proteins that interact with 43K OMP of F. necrophorum by immunoprecipitation-mass spectrometry. The results showed that the natural 43K OMP and recombinant 43K OMP could bind to BHK-21 cells, and preincubation of F. necrophorum with an antibody against the recombinant 43K OMP of F. necrophorum decreased binding to BHK-21 cells. Seventy differential interacting proteins were successfully screened by immunoprecipitation-mass spectrometry. Among these seventy differential interacting proteins, seven cell membrane proteins and four extracellular matrix proteins shown to be relevant to bacteria adhesion through subcellular localization and single-molecule function analysis. These data increase our understanding of the pathogenesis of F. necrophorum and provide a new theoretical basis for the design of antimicrobial drugs against F. necrophorum.


Subject(s)
Bacterial Adhesion , Bacterial Outer Membrane Proteins/metabolism , Carrier Proteins , Fusobacterium necrophorum/metabolism , Animals , Antibodies, Neutralizing , Carrier Proteins/chemistry , Carrier Proteins/immunology , Carrier Proteins/metabolism , Cattle , Cell Line , Fusobacterium Infections/metabolism , Humans , Immunoprecipitation , Mass Spectrometry , Recombinant Proteins/metabolism
15.
FEBS J ; 287(18): 4032-4047, 2020 09.
Article in English | MEDLINE | ID: mdl-31997506

ABSTRACT

Fusobacterium nucleatum, an anaerobic oral opportunistic pathogen associated with periodontitis, has been considered to be associated with the development of oral squamous cell carcinoma (OSCC). However, the initial host molecular alterations induced by F. nucleatum infection which may promote predisposition to malignant transformation through epithelial-mesenchymal transition (EMT) have not yet been clarified. In the present study, we monitored the ability of F. nucleatum to induce EMT-associated features, and our results showed that F. nucleatum infection promoted cell migration in either noncancerous human immortalized oral epithelial cells (HIOECs) or the two OSCC cell lines SCC-9 and HSC-4, but did not accelerate cell proliferation or cell cycle progression. Mesenchymal markers, including N-cadherin, Vimentin, and SNAI1, were upregulated, while E-cadherin was decreased and was observed to translocate to the cytoplasm. Furthermore, FadA adhesin and heat-inactivated F. nucleatum were found to cause a similar effect as the viable bacterial cells. The upregulated lncRNA MIR4435-2HG identified by the high-throughput sequencing was demonstrated to negatively regulate the expression of miR-296-5p, which was downregulated in F. nucleatum-infected HIOECs and SCC-9 cells. The binding of MIR4435-2HG and miR-296-5p was validated via a dual-luciferase reporter assay. Additionally, knockdown of MIR4435-2HG with siRNA leads to a decrease in SNAI1 expression, while miR-296-5p could further negatively and indirectly regulate SNAI1 expression via Akt2. Therefore, our study demonstrated that F. nucleatum infection could trigger EMT via lncRNA MIR4435-2HG/miR-296-5p/Akt2/SNAI1 signaling pathway, and EMT process may be a probable link between F. nucleatum infection and initiation of oral epithelial carcinomas.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , MicroRNAs/genetics , Proto-Oncogene Proteins c-akt/genetics , Signal Transduction/genetics , Snail Family Transcription Factors/genetics , Cadherins/genetics , Cadherins/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Cell Line, Tumor , Fusobacterium Infections/genetics , Fusobacterium Infections/metabolism , Fusobacterium Infections/microbiology , Fusobacterium nucleatum/physiology , Host-Pathogen Interactions , Humans , Mouth Neoplasms/genetics , Mouth Neoplasms/metabolism , Mouth Neoplasms/microbiology , Proto-Oncogene Proteins c-akt/metabolism , RNA, Long Noncoding , Snail Family Transcription Factors/metabolism
16.
J Pathol ; 250(2): 170-182, 2020 02.
Article in English | MEDLINE | ID: mdl-31610014

ABSTRACT

Accumulating evidence links Fusobacterium nucleatum with ulcerative colitis (UC). The mechanism by which F. nucleatum promotes intestinal inflammation in UC remains poorly defined. Here, we first examined the abundance and impact of F. nucleatum on disease activity in UC tissues. Next, we isolated a strain of F. nucleatum from UC tissues and explored whether F. nucleatum aggravates the intestinal inflammatory response in vitro and in vivo. We also examined whether F. nucleatum infection involves the NF-κB or IL-17F signaling pathways. Our data showed that F. nucleatum was enriched in 51.78% of UC tissues and was correlated with the clinical course, clinical activity and refractory behavior of UC (p < 0.05). Furthermore, we demonstrated that F. nucleatum promoted intestinal epithelial damage and the expression of the inflammatory cytokines IL-1ß, Il-6, IL-17F and TNF-α. Mechanistically, F. nucleatum targeted caspase activation and recruitment domain 3 (CARD3) through NOD2 to activate the IL-17F/NF-κB pathway in vivo and in vitro. Thus, F. nucleatum orchestrates a molecular network involving CARD3 and IL-17F to control the UC process. Measuring and targeting F. nucleatum and its associated pathways will yield valuable insight into the prevention and treatment of UC. © 2019 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.


Subject(s)
Colitis, Ulcerative/microbiology , Fusobacterium Infections/complications , Fusobacterium nucleatum/pathogenicity , Interleukin-17/metabolism , Receptor-Interacting Protein Serine-Threonine Kinase 2/biosynthesis , Adolescent , Adult , Aged , Animals , Case-Control Studies , Cells, Cultured , Colitis, Ulcerative/chemically induced , Colitis, Ulcerative/metabolism , Colitis, Ulcerative/pathology , Dextran Sulfate , Disease Models, Animal , Female , Fusobacterium Infections/metabolism , Fusobacterium nucleatum/isolation & purification , Humans , Male , Mice, Knockout , Middle Aged , NF-kappa B/metabolism , RNA, Messenger/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/deficiency , Receptor-Interacting Protein Serine-Threonine Kinase 2/genetics , Receptor-Interacting Protein Serine-Threonine Kinase 2/physiology , Severity of Illness Index , Signal Transduction/physiology , Up-Regulation/physiology , Young Adult
17.
Oxid Med Cell Longev ; 2019: 1681972, 2019.
Article in English | MEDLINE | ID: mdl-31737164

ABSTRACT

Fusobacterium nucleatum (F. nucleatum) plays key roles in the initiation and progression of periodontitis. However, the pathogenic effect of F. nucleatum on human oral tissues and cells has not been fully evaluated. In this study, we aimed to analyze the pathogenic effects of F. nucleatum on human gingival fibroblasts (GFs) and clarify the potential mechanisms. RNA-sequencing analysis confirmed that F. nucleatum significantly altered the gene expression of GF as the stimulation time increased. Cell counting and EdU-labeling assays indicated that F. nucleatum inhibited GF proliferation and promoted cell apoptosis in a time- and dose-dependent manner. In addition, cell apoptosis, intracellular reactive oxygen species (ROS) generation, and proinflammatory cytokine production were dramatically elevated after F. nucleatum stimulation. Furthermore, we found that the AKT/MAPK and NF-κB signaling pathways were significantly activated by F. nucleatum infection and that a large number of genes related to cellular proliferation, apoptosis, ROS, and inflammatory cytokine production downstream of AKT/MAPK and NF-κB signaling pathways were significantly altered in F. nucleatum-stimulated GFs. These findings suggest that F. nucleatum inhibits GF proliferation and promotes cell apoptosis, ROS generation, and inflammatory cytokine production partly by activating the AKT/MAPK and NF-κB signaling pathways. Our study opens a new window for understanding the pathogenic effects of periodontal pathogens on the host oral system.


Subject(s)
Fibroblasts/metabolism , Fusobacterium Infections/metabolism , Fusobacterium nucleatum/physiology , Gingiva/pathology , Periodontitis/metabolism , Adult , Apoptosis , Cells, Cultured , Cytokines/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Fibroblasts/pathology , Humans , Inflammation Mediators/metabolism , Male , NF-kappa B/metabolism , Oncogene Protein v-akt/metabolism , Reactive Oxygen Species/metabolism , Signal Transduction
18.
J Proteomics ; 195: 125-137, 2019 03 20.
Article in English | MEDLINE | ID: mdl-30634002

ABSTRACT

Fusobacterium nucleatum is a Gram-negative bacterium commonly found in the oral cavity and is often involved in periodontal diseases. Recent studies have shown increased F. nucleatum prevalence in colorectal cancer (CRC) tissues, and causal data has linked this bacterium to CRC tumorigenesis. Immune-based approaches to contain, reduce or eradicate its gut colonization may prevent CRC. Outer membrane vesicles (OMVs) are naturally produced by Gram-negative bacteria, typically contain multiple putative virulence factors and may elicit protective immune responses if used as vaccines. Here, OMVs were isolated from F. nucleatum cultures and purified using gradient centrifugation. Proteins contained within the OMVs were identified by nano LC/MS/MS analysis. Of 98 proteins consistently identified from duplicate analyses, 60 were predicted to localize to the outer membrane or periplasm via signal peptide driven translocation. Of these, six autotransporter proteins, which constitute the majority of protein mass of OMVs, were associated with Type V secretion system. In addition, other putative virulence factor proteins with functional domains, including FadA, MORN2 and YadA-like domain, were identified with multiple exposed epitope sites as determined by in silico analysis. Altogether, the non-replicative OMVs of F. nucleatum contain multiple antigenic virulence factors that may play important roles in the design and development of vaccines against F. nucleatum. SIGNIFICANCE: Fusobacterium nulceatum has been proved playing significant role in colorectal carcinogenesis. Outer membrane vesicles are nanoparticles that naturally secreted by Gram-negative bacterial containing various antigenic components, which provides new insight in vaccine development. Understanding the constituents of F. nucleatum OMVs will provide fundamental information and potential strategies for OMV-based F. nucleatum vaccines design. Based on our knowledge this is the first proteomic study of OMVs from F. nucleatum.


Subject(s)
Bacterial Proteins/metabolism , Extracellular Vesicles/metabolism , Fusobacterium nucleatum , Intestinal Mucosa/microbiology , Virulence Factors/metabolism , Bacterial Vaccines/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/prevention & control , Fusobacterium Infections/metabolism , Fusobacterium Infections/pathology , Fusobacterium Infections/prevention & control , Fusobacterium nucleatum/metabolism , Fusobacterium nucleatum/pathogenicity , Humans
19.
Microbes Infect ; 20(9-10): 615-625, 2018.
Article in English | MEDLINE | ID: mdl-29024797

ABSTRACT

NOD-like receptors (NLRs) play a large role in regulation of host innate immunity, yet their role in periodontitis remains to be defined. NLRX1, a member of the NLR family that localizes to mitochondria, enhances mitochondrial ROS (mROS) generation. mROS can activate the NLRP3 inflammasome, yet the role of NLRX1 in NLRP3 inflammasome activation has not been examined. In this study, we revealed the mechanism by which NLRX1 positively regulates ATP-induced NLRP3 inflammasome activation through mROS in gingival epithelial cells (GECs). We found that depletion of NLRX1 by shRNA attenuated ATP-induced mROS generation and redistribution of the NLRP3 inflammasome adaptor protein, ASC. Furthermore, depletion of NLRX1 inhibited Fusobacterium nucleatum infection-activated caspase-1, suggesting that it also inhibits the NLRP3 inflammasome. Conversely, NLRX1 also acted as a negative regulator of NF-κB signaling and IL-8 expression. Thus, NLRX1 stimulates detection of the pathogen F. nucleatum via the inflammasome, while dampening cytokine production. We expect that commensals should not activate the inflammasome, and NLRX1 should decrease their ability to stimulate expression of pro-inflammatory cytokines such as IL-8. Therefore, NLRX1 may act as a potential switch with regards to anti-microbial responses in healthy or diseased states in the oral cavity.


Subject(s)
Fusobacterium Infections/metabolism , Inflammasomes/metabolism , Mitochondrial Proteins/metabolism , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Signal Transduction , Transcription Factor RelA/metabolism , CARD Signaling Adaptor Proteins/metabolism , Caspase 1/metabolism , Cell Nucleus/metabolism , Cells, Cultured , Epithelial Cells/metabolism , Fusobacterium nucleatum/physiology , Gene Expression , Gingiva , Humans , Interleukin-8/genetics , Mitochondria/metabolism , Mitochondrial Proteins/deficiency , Mitochondrial Proteins/genetics , RNA, Small Interfering , Reactive Oxygen Species/metabolism
20.
Sci Rep ; 6: 34520, 2016 10 03.
Article in English | MEDLINE | ID: mdl-27694921

ABSTRACT

Fusobacterium nucleatum has been associated with both periodontal disease and inflammatory bowel disease. This Gram-negative bacterium possesses a high inflammatory potential that may contribute to the disease process. We hypothesized that green and black tea polyphenols attenuate the inflammatory response of monocytes/macrophages mediated by F. nucleatum. We first showed that the tea extracts, EGCG and theaflavins reduce the NF-κB activation induced by F. nucleatum in monocytes. Since NF-κB is a key regulator of genes coding for inflammatory mediators, we tested the effects of tea polyphenols on secretion of IL-1ß, IL-6, TNF-α, and CXCL8 by macrophages. A pre-treatment of macrophages with the tea extracts, EGCG, or theaflavins prior to a stimulation with F. nucleatum significantly inhibited the secretion of all four cytokines and reduced the secretion of MMP-3 and MMP-9, two tissue destructive enzymes. TREM-1 expressed by macrophages is a cell-surface receptor involved in the propagation of the inflammatory response to bacterial challenges. Interestingly, tea polyphenols inhibited the secretion/shedding of soluble TREM-1 induced by a stimulation of macrophages with F. nucleatum. The anti-inflammatory properties of tea polyphenols identified in the present study suggested that they may be promising agents for the prevention and/or treatment of periodontal disease and inflammatory bowel disease.


Subject(s)
Collagenases/metabolism , Cytokines/metabolism , Fusobacterium Infections/drug therapy , Fusobacterium nucleatum/metabolism , Macrophages/metabolism , Polyphenols/pharmacology , Tea/chemistry , Fusobacterium Infections/metabolism , Fusobacterium Infections/pathology , Humans , Macrophages/microbiology , Macrophages/pathology , Polyphenols/chemistry , U937 Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...