Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 299
Filter
1.
Genome Res ; 34(6): 967-978, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39038849

ABSTRACT

The human gut microbiota is of increasing interest, with metagenomics a key tool for analyzing bacterial diversity and functionality in health and disease. Despite increasing efforts to expand microbial gene catalogs and an increasing number of metagenome-assembled genomes, there have been few pan-metagenomic association studies and in-depth functional analyses across different geographies and diseases. Here, we explored 6014 human gut metagenome samples across 19 countries and 23 diseases by performing compositional, functional cluster, and integrative analyses. Using interpreted machine learning classification models and statistical methods, we identified Fusobacterium nucleatum and Anaerostipes hadrus with the highest frequencies, enriched and depleted, respectively, across different disease cohorts. Distinct functional distributions were observed in the gut microbiomes of both westernized and nonwesternized populations. These compositional and functional analyses are presented in the open-access Human Gut Microbiome Atlas, allowing for the exploration of the richness, disease, and regional signatures of the gut microbiota across different cohorts.


Subject(s)
Gastrointestinal Microbiome , Metagenome , Metagenomics , Humans , Gastrointestinal Microbiome/genetics , Metagenomics/methods , Machine Learning , Fusobacterium nucleatum/genetics , Bacteria/classification , Bacteria/genetics
2.
BMC Infect Dis ; 24(1): 675, 2024 Jul 06.
Article in English | MEDLINE | ID: mdl-38971721

ABSTRACT

Pleural empyema can lead to significant morbidity and mortality despite chest drainage and antibiotic treatment, necessitating novel and minimally invasive interventions. Fusobacterium nucleatum is an obligate anaerobe found in the human oral and gut microbiota. Advances in sequencing and puncture techniques have made it common to detect anaerobic bacteria in empyema cases. In this report, we describe the case of a 65-year-old man with hypertension who presented with a left-sided encapsulated pleural effusion. Initial fluid analysis using metagenomic next-generation sequencing (mNGS) revealed the presence of Fusobacterium nucleatum and Aspergillus chevalieri. Unfortunately, the patient experienced worsening pleural effusion despite drainage and antimicrobial therapy. Ultimately, successful treatment was achieved through intrapleural metronidazole therapy in conjunction with systemic antibiotics. The present case showed that intrapleural antibiotic therapy is a promising measure for pleural empyema.


Subject(s)
Anti-Bacterial Agents , Empyema, Pleural , Fusobacterium nucleatum , Salvage Therapy , Humans , Male , Aged , Empyema, Pleural/drug therapy , Empyema, Pleural/microbiology , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/therapeutic use , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/isolation & purification , Fusobacterium nucleatum/genetics , Fusobacterium Infections/drug therapy , Fusobacterium Infections/complications , Fusobacterium Infections/microbiology , Metronidazole/therapeutic use , Metronidazole/administration & dosage , High-Throughput Nucleotide Sequencing , Treatment Outcome
3.
Forensic Sci Int ; 362: 112147, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39067179

ABSTRACT

The identification of biological fluids at crime scenes contributes to crime scene reconstruction and provides investigative leads. Traditional methods for body fluid identification are limited in terms of sensitivity and are mostly presumptive. Emerging methods based on mRNA and DNA methylation require high quality template source. An exploitable characteristic of body fluids is their distinct microbial profiles allowing for the discrimination of body fluids based on microbiome content. Microbial DNA is highly abundant within the body, robust and stable and can persist in the environment long after human DNA has degraded. 16S rRNA sequencing is the gold standard for microbial analysis; however, NGS is costly, and requires intricate workflows and interpretation. Also, species level resolution is not always achievable. Based on the current challenges, the first objective of this study was to develop a multiplex conventional PCR assay to identify vaginal fluid and saliva by targeting species-specific 16S rRNA microbial markers. The second objective was to employ droplet digital PCR (ddPCR) as a novel approach to quantify bacterial species alone and in a mixture of body fluids. Lactobacillus crispatus and Streptococcus salivarius were selected because of high abundance within vaginal fluid and saliva respectively. While Fusobacterium nucleatum and Gardnerella vaginalis, though present in healthy humans, are also frequently found in oral and vaginal infections, respectively. The multiplex PCR assay detected L. crispatus and G. vaginalis in vaginal fluid while F. nucleatum and S. salivarius was detected in saliva. Multiplex PCR detected F. nucleatum, S. salivarius and L. crispatus in mixed body fluid samples while, G. vaginalis was undetected in mixtures containing vaginal fluid. For samples exposed at room temperature for 65 days, L. crispatus and G. vaginalis were detected in vaginal swabs while only S. salivarius was detected in saliva swabs. The limit of detection was 0.06 copies/µl for F. nucleatum (2.5 ×10-9 ng/µl) and S. salivarius (2.5 ×10-6 ng/µl). L. crispatus and G. vaginalis had detection limits of 0.16 copies/µl (2.5 ×10-4 ng/µl) and 0.48 copies/µl (2.5 ×10-7 ng/µl). All 4 bacterial species were detected in mixtures and aged samples by ddPCR. No significant differences were observed in quantity of bacterial markers in saliva and vaginal fluid. The present research reports for the first time the combination of the above four bacterial markers for the detection of saliva and vaginal fluid and highlights the sensitivity of ddPCR for bacterial quantification in pure and mixed body fluids.


Subject(s)
DNA, Bacterial , Multiplex Polymerase Chain Reaction , RNA, Ribosomal, 16S , Saliva , Vagina , Humans , Saliva/microbiology , Saliva/chemistry , Female , DNA, Bacterial/analysis , Vagina/microbiology , Streptococcus salivarius/genetics , Lactobacillus/isolation & purification , Lactobacillus/genetics , Gardnerella vaginalis/isolation & purification , Gardnerella vaginalis/genetics , Cervix Mucus/microbiology , Fusobacterium nucleatum/isolation & purification , Fusobacterium nucleatum/genetics
4.
Sci Rep ; 14(1): 13739, 2024 06 14.
Article in English | MEDLINE | ID: mdl-38877111

ABSTRACT

The study aimed to develop a quantitative colorimetric loop-mediated isothermal amplification technique using the phenol red indicator (QLAMP-PhR) for detecting Fusobacterium nucleatum (Fn) levels in colorectal cancer (CRC) patients and healthy individuals. QLAMP-PhR assays were conducted on 251 stool samples specific for the Fn FadA gene. Six primers were synthesized and utilized with master mix reagents, and a phenol red indicator was employed to enhance the QLAMP-PhR technique. A standard quantitative analysis curve was generated using a logarithmic function (absorbance vs. concentration) by serially diluting the copy number of genomic DNA templates (Fn ATCC25586). The CRC group exhibited a significantly higher abundance of Fn compared to the healthy control group (P < 0.001). These findings suggest that the QLAMP-PhR technique effectively identifies Fn specifically by its gene for the key virulence factor FadA. Additionally, ideas for developing a real-time QLAMP-PhR test were presented. Compared to the traditional polymerase chain reaction (PCR) technique, QLAMP-PhR offers several advantages including rapidity, simplicity, specificity, sensitivity, and cost-effectiveness method that can quantitatively screen for Fn presence in normal populations. The QLAMP-PhR method represents a sensitive and specific amplification assay for the rapid detection of the Fn pathogen. To the best of our knowledge, this study is the first to report the application of QLAMP-PhR for detecting FadA in Fn.


Subject(s)
Colorectal Neoplasms , Colorimetry , Feces , Fusobacterium nucleatum , Nucleic Acid Amplification Techniques , Humans , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/diagnosis , Colorectal Neoplasms/genetics , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/isolation & purification , Feces/microbiology , Nucleic Acid Amplification Techniques/methods , Colorimetry/methods , Male , Female , Phenolsulfonphthalein , Molecular Diagnostic Techniques/methods , Middle Aged , Aged , Fusobacterium Infections/microbiology , Fusobacterium Infections/diagnosis , Sensitivity and Specificity , Adult
5.
Forensic Sci Int ; 361: 112112, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38917506

ABSTRACT

Body fluid detection is an important component in the toolbox of forensic scientists, with saliva playing a particularly critical role in forensic evidence. Given that each body fluid possesses a distinct microbiome, the identification of body fluid based on specific representatives of the microbiota presents an appealing approach for forensic applications. In this study, we have developed a real-time polymerase chain reaction (RT-PCR)-based method for the precise identification of saliva, focusing on three bacteria highly associated with saliva but not with other tested body fluids -Porphyromonas gingivalis, Fusobacterium nucleatum, and Streptococcus salivarius. The inclusion of these three bacterial species enhances the accuracy of detection and reinforces validation. Notably, specific identification of saliva was achievable even at low concentrations where Phadebas, a commonly used method for saliva detection, proved ineffective. Importantly, bacteria-based saliva detection utilizes DNA generated for small tandem repeats (STR) profiling, facilitating seamless integration into forensic laboratories and optimizing DNA sample utilization. This study collectively proposes an effective bacterial DNA-based approach for saliva identification, demonstrating promising potential for forensic applications.


Subject(s)
DNA, Bacterial , Fusobacterium nucleatum , Porphyromonas gingivalis , Real-Time Polymerase Chain Reaction , Saliva , Streptococcus salivarius , Saliva/microbiology , Humans , Fusobacterium nucleatum/isolation & purification , Fusobacterium nucleatum/genetics , Porphyromonas gingivalis/isolation & purification , Porphyromonas gingivalis/genetics , Streptococcus salivarius/isolation & purification , Streptococcus salivarius/genetics , Microsatellite Repeats , DNA Fingerprinting , RNA, Ribosomal, 16S/genetics
6.
Appl Microbiol Biotechnol ; 108(1): 336, 2024 May 18.
Article in English | MEDLINE | ID: mdl-38761182

ABSTRACT

To investigate the cell-cell interactions of intergeneric bacterial species, the study detected the survival of Enterococcus faecalis (Ef) under monospecies or coaggregation state with Fusobacterium nucleatum subsp. polymorphum (Fnp) in environmental stress. Ef and Fnp infected the human macrophages with different forms (Ef and Fnp monospecies, Ef-Fnp coaggregates, Ef + Fnp cocultures) for exploring the immunoregulatory effects and the relevant molecular mechanisms. Meanwhile, the transcriptomic profiles of coaggregated Ef and Fnp were analyzed. Ef was shown to coaggregate with Fnp strongly in CAB within 90 min by forming multiplexes clumps. Coaggregation with Fnp reinforced Ef resistance against unfavorable conditions including alkaline, hypertonic, nutrient-starvation, and antibiotic challenges. Compared with monospecies and coculture species, the coaggregation of Ef and Fnp significantly facilitates both species to invade dTHP-1 cells and aid Ef to survive within the cells. Compared with coculture species, dual-species interaction of Ef and Fnp significantly decreased the levels of pro-inflammatory cytokines IL-6, TNF-α, and chemokines MCP-1 secreted by dTHP-1 cells and lessened the phosphorylation of p38, JNK, and p65 signaling pathways. The transcriptome sequencing results showed that 111 genes were differentially expressed or Ef-Fnp coaggregated species compared to Ef monospecies; 651 genes were differentially expressed for Fnp when coaggregation with Ef. The analysis of KEGG pathway showed that Ef differentially expressed genes (DEGs) were enriched in quorum sensing and arginine biosynthesis pathway; Fnp DEGs were differentially concentrated in lipopolysaccharide (LPS) biosynthesis, biofilm formation, and lysine degradation pathway compared to monospecies. KEY POINTS: • Coaggregated with Fnp aids Ef's survival in environmental stress, especially in root canals after endodontic treatment. • The coaggregation of Ef and Fnp may weaken the pro-inflammatory response and facilitate Ef to evade killed by macrophages. • The coaggregation between Ef and Fnp altered interspecies transcriptional profiles.


Subject(s)
Enterococcus faecalis , Fusobacterium nucleatum , Macrophages , Stress, Physiological , Fusobacterium nucleatum/physiology , Fusobacterium nucleatum/genetics , Enterococcus faecalis/genetics , Enterococcus faecalis/physiology , Humans , Macrophages/microbiology , Macrophages/immunology , Cytokines/metabolism , Cytokines/genetics , Bacterial Adhesion , Coculture Techniques , Gene Expression Profiling , Transcriptome , Cell Line , Interleukin-6/genetics , Interleukin-6/metabolism , Tumor Necrosis Factor-alpha/metabolism , Tumor Necrosis Factor-alpha/genetics , Inflammation
7.
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
10.
ACS Infect Dis ; 10(5): 1612-1623, 2024 05 10.
Article in English | MEDLINE | ID: mdl-38597503

ABSTRACT

Fusobacterium nucleatum, a pathobiont inhabiting the oral cavity, contributes to opportunistic diseases, such as periodontal diseases and gastrointestinal cancers, which involve microbiota imbalance. Broad-spectrum antimicrobial agents, while effective against F. nucleatum infections, can exacerbate dysbiosis. This necessitates the discovery of more targeted narrow-spectrum antimicrobial agents. We therefore investigated the potential for the fusobacterial enoyl-ACP reductase II (ENR II) isoenzyme FnFabK (C4N14_ 04250) as a narrow-spectrum drug target. ENRs catalyze the rate-limiting step in the bacterial fatty acid synthesis pathway. Bioinformatics revealed that of the four distinct bacterial ENR isoforms, F. nucleatum specifically encodes FnFabK. Genetic studies revealed that fabK was indispensable for F. nucleatum growth, as the gene could not be deleted, and silencing of its mRNA inhibited growth under the test conditions. Remarkably, exogenous fatty acids failed to rescue growth inhibition caused by the silencing of fabK. Screening of synthetic phenylimidazole analogues of a known FabK inhibitor identified an inhibitor (i.e., 681) of FnFabK enzymatic activity and F. nucleatum growth, with an IC50 of 2.1 µM (1.0 µg/mL) and a MIC of 0.4 µg/mL, respectively. Exogenous fatty acids did not attenuate the activity of 681 against F. nucleatum. Furthermore, FnFabK was confirmed as the intracellular target of 681 based on the overexpression of FnFabK shifting MICs and 681-resistant mutants having amino acid substitutions in FnFabK or mutations in other genetic loci affecting fatty acid biosynthesis. 681 had minimal activity against a range of commensal flora, and it was less active against streptococci in physiologic fatty acids. Taken together, FnFabK is an essential enzyme that is amenable to drug targeting for the discovery and development of narrow-spectrum antimicrobial agents.


Subject(s)
Anti-Bacterial Agents , Fusobacterium nucleatum , Fusobacterium nucleatum/enzymology , Fusobacterium nucleatum/drug effects , Fusobacterium nucleatum/genetics , Anti-Bacterial Agents/pharmacology , Anti-Bacterial Agents/chemistry , Humans , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/genetics , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/antagonists & inhibitors , Enoyl-(Acyl-Carrier-Protein) Reductase (NADH)/metabolism , Microbial Sensitivity Tests , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/antagonists & inhibitors , Fatty Acids/chemistry , Fusobacterium Infections/microbiology , Fusobacterium Infections/drug therapy , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/chemistry
11.
mBio ; 15(4): e0353623, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38436569

ABSTRACT

The oral commensal Fusobacterium nucleatum can spread to extra-oral sites, where it is associated with diverse pathologies, including pre-term birth and cancer. Due to the evolutionary distance of F. nucleatum to other model bacteria, we lack a deeper understanding of the RNA regulatory networks that allow this bacterium to adapt to its various niches. As a first step in that direction, we recently showed that F. nucleatum harbors a global stress response governed by the extracytoplasmic function sigma factor, σE, which displays a striking functional conservation with Proteobacteria and includes a noncoding arm in the form of a regulatory small RNA (sRNA), FoxI. To search for putative additional σE-dependent sRNAs, we comprehensively mapped the 5' and 3' ends of transcripts in the model strain ATCC 23726. This enabled the discovery of FoxJ, a ~156-nucleotide sRNA previously misannotated as the 5' untranslated region (UTR) of ylmH. FoxJ is tightly controlled by σE and activated by the same stress conditions as is FoxI. Both sRNAs act as mRNA repressors of the abundant porin FomA, but FoxJ also regulates genes that are distinct from the target suite of FoxI. Moreover, FoxJ differs from other σE-dependent sRNAs in that it also positively regulates genes at the post-transcriptional level. We provide preliminary evidence for a new mode of sRNA-mediated mRNA activation, which involves the targeting of intra-operonic terminators. Overall, our study provides an important resource through the comprehensive annotation of 5' and 3' UTRs in F. nucleatum and expands our understanding of the σE response in this evolutionarily distant bacterium.IMPORTANCEThe oral microbe Fusobacterium nucleatum can colonize secondary sites, including cancer tissue, and likely deploys complex regulatory systems to adapt to these new environments. These systems are largely unknown, partly due to the phylogenetic distance of F. nucleatum to other model organisms. Previously, we identified a global stress response mediated by σE that displays functional conservation with the envelope stress response in Proteobacteria, comprising a coding and noncoding regulatory arm. Through global identification of transcriptional start and stop sites, we uncovered the small RNA (sRNA) FoxJ as a novel component of the noncoding arm of the σE response in F. nucleatum. Together with its companion sRNA FoxI, FoxJ post-transcriptionally modulates the synthesis of envelope proteins, revealing a conserved function for σE-dependent sRNAs between Fusobacteriota and Proteobacteria. Moreover, FoxJ activates the gene expression for several targets, which is a mode of regulation previously unseen in the noncoding arm of the σE response.


Subject(s)
Neoplasms , RNA, Small Untranslated , Humans , RNA, Messenger/genetics , RNA, Messenger/metabolism , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/metabolism , Transcriptome , Phylogeny , RNA, Small Untranslated/genetics , RNA, Small Untranslated/metabolism , Bacteria/genetics , RNA, Bacterial/genetics , RNA, Bacterial/metabolism , Gene Expression Regulation, Bacterial
12.
Cell Host Microbe ; 32(4): 479-488.e4, 2024 Apr 10.
Article in English | MEDLINE | ID: mdl-38479393

ABSTRACT

The ubiquitous inflammophilic oral pathobiont Fusobacterium nucleatum (Fn) is widely recognized for its strong association with inflammatory dysbiotic diseases and cancer. Fn is subdivided into four subspecies, which are historically considered functionally interchangeable in the oral cavity. To test this assumption, we analyzed patient-matched dental plaque and odontogenic abscess clinical specimens and examined whether an inflammatory environment selects for/against particular Fn subspecies. Dental plaque harbored a greater diversity of fusobacteria, with Fn. polymorphum dominating, whereas odontogenic abscesses were exceptionally biased for the largely uncharacterized organism Fn. animalis. Comparative genomic analyses revealed significant genotypic distinctions among Fn subspecies that correlate with their preferred ecological niches and support a taxonomic reassignment of each as a distinct Fusobacterium species. Despite originating as a low-abundance organism in dental plaque, Fn. animalis typically outcompetes other oral fusobacteria within the inflammatory abscess environment, which may explain its prevalence in other oral and extraoral diseases.


Subject(s)
Dental Plaque , Fusobacterium nucleatum , Fusobacterium , Humans , Fusobacterium nucleatum/genetics , Abscess , Mouth
13.
Nature ; 628(8007): 424-432, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38509359

ABSTRACT

Fusobacterium nucleatum (Fn), a bacterium present in the human oral cavity and rarely found in the lower gastrointestinal tract of healthy individuals1, is enriched in human colorectal cancer (CRC) tumours2-5. High intratumoural Fn loads are associated with recurrence, metastases and poorer patient prognosis5-8. Here, to delineate Fn genetic factors facilitating tumour colonization, we generated closed genomes for 135 Fn strains; 80 oral strains from individuals without cancer and 55 unique cancer strains cultured from tumours from 51 patients with CRC. Pangenomic analyses identified 483 CRC-enriched genetic factors. Tumour-isolated strains predominantly belong to Fn subspecies animalis (Fna). However, genomic analyses reveal that Fna, considered a single subspecies, is instead composed of two distinct clades (Fna C1 and Fna C2). Of these, only Fna C2 dominates the CRC tumour niche. Inter-Fna analyses identified 195 Fna C2-associated genetic factors consistent with increased metabolic potential and colonization of the gastrointestinal tract. In support of this, Fna C2-treated mice had an increased number of intestinal adenomas and altered metabolites. Microbiome analysis of human tumour tissue from 116 patients with CRC demonstrated Fna C2 enrichment. Comparison of 62 paired specimens showed that only Fna C2 is tumour enriched compared to normal adjacent tissue. This was further supported by metagenomic analysis of stool samples from 627 patients with CRC and 619 healthy individuals. Collectively, our results identify the Fna clade bifurcation, show that specifically Fna C2 drives the reported Fn enrichment in human CRC and reveal the genetic underpinnings of pathoadaptation of Fna C2 to the CRC niche.


Subject(s)
Colorectal Neoplasms , Fusobacterium nucleatum , Animals , Humans , Mice , Adenoma/microbiology , Case-Control Studies , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Feces/microbiology , Fusobacterium nucleatum/classification , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/isolation & purification , Fusobacterium nucleatum/pathogenicity , Gastrointestinal Tract/metabolism , Gastrointestinal Tract/microbiology , Genome, Bacterial/genetics , Mouth/microbiology , Female
14.
Nat Commun ; 15(1): 1688, 2024 Feb 24.
Article in English | MEDLINE | ID: mdl-38402201

ABSTRACT

Fusobacterium nucleatum (F. nucleatum) promotes intestinal tumor growth and its relative abundance varies greatly among patients with CRC, suggesting the presence of unknown, individual-specific effectors in F. nucleatum-dependent carcinogenesis. Here, we identify that F. nucleatum is enriched preferentially in KRAS p.G12D mutant CRC tumor tissues and contributes to colorectal tumorigenesis in Villin-Cre/KrasG12D+/- mice. Additionally, Parabacteroides distasonis (P. distasonis) competes with F. nucleatum in the G12D mouse model and human CRC tissues with the KRAS mutation. Orally gavaged P. distasonis in mice alleviates the F. nucleatum-dependent CRC progression. F. nucleatum invades intestinal epithelial cells and binds to DHX15, a protein of RNA helicase family expressed on CRC tumor cells, mechanistically involving ERK/STAT3 signaling. Knock out of Dhx15 in Villin-Cre/KrasG12D+/- mice attenuates the CRC phenotype. These findings reveal that the oncogenic effect of F. nucleatum depends on somatic genetics and gut microbial ecology and indicate that personalized modulation of the gut microbiota may provide a more targeted strategy for CRC treatment.


Subject(s)
Colorectal Neoplasms , Fusobacterium nucleatum , Animals , Humans , Mice , Carcinogenesis/genetics , Colorectal Neoplasms/pathology , Fusobacterium nucleatum/genetics , Proto-Oncogene Proteins p21(ras)/genetics , RNA Helicases
15.
J Microbiol ; 62(2): 63-73, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38402337

ABSTRACT

Cisplatin resistance is the main cause of colorectal cancer (CRC) treatment failure, and the cause has been reported to be related to Fusobacterium nucleatum (Fn) infection. In this study, we explored the role of Fn in regulating cisplatin resistance of CRC cells and its underlying mechanism involved. The mRNA and protein expressions were examined by qRT-PCR and western blot. Cell proliferation and cell apoptosis were assessed using CCK8 and flow cytometry assays, respectively. Dual-luciferase reporter gene assay was adopted to analyze the molecular interactions. Herein, our results revealed that Fn abundance and miR-135b expression were markedly elevated in CRC tissues, with a favorable association between the two. Moreover, Fn infection could increase miR-135b expression via a concentration-dependent manner, and it also enhanced cell proliferation but reduced apoptosis and cisplatin sensitivity by upregulating miR-135b. Moreover, KLF13 was proved as a downstream target of miR-135b, of which overexpression greatly diminished the promoting effect of miR-135b or Fn-mediated cisplatin resistance in CRC cells. In addition, it was observed that upstream 2.5 kb fragment of miR-135b promoter could be interacted by ß-catenin/TCF4 complex, which was proved as an effector signaling of Fn. LF3, a blocker of ß-catenin/TCF4 complex, was confirmed to diminish the promoting role of Fn on miR-135b expression. Thus, it could be concluded that Fn activated miR-135b expression through TCF4/ß-catenin complex, thereby inhibiting KLF13 expression and promoting cisplatin resistance in CRC.


Subject(s)
Colorectal Neoplasms , MicroRNAs , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Cisplatin/pharmacology , Fusobacterium nucleatum/genetics , beta Catenin , Colorectal Neoplasms/genetics , Colorectal Neoplasms/metabolism , Cell Proliferation/genetics , Repressor Proteins , Cell Cycle Proteins , Kruppel-Like Transcription Factors/genetics
16.
J Nanobiotechnology ; 22(1): 62, 2024 Feb 15.
Article in English | MEDLINE | ID: mdl-38360615

ABSTRACT

BACKGROUND: A large number of Fusobacterium nucleatum (Fn) are present in colorectal cancer (CRC) tissues of patients who relapse after chemotherapy, and Fn has been reported to promote oxaliplatin and 5-FU chemoresistance in CRC. Pathogens such as bacteria and parasites stimulate exosome production in tumor cells, and the regulatory mechanism of exosomal circRNA in the transmission of oxaliplatin and 5-FU chemotherapy resistance in Fn-infected CRC remains unclear. METHODS: Hsa_circ_0004085 was screened by second-generation sequencing of CRC tissues. The correlation between hsa_circ_0004085 and patient clinical response to oxaliplatin/5-FU was analyzed. Exosome tracing experiments and live imaging systems were used to test the effect of Fn infection in CRC on the distribution of hsa_circ_0004085. Colony formation, ER tracking analysis and immunofluorescence were carried out to verify the regulatory effect of exosomes produced by Fn-infected CRC cells on chemotherapeutic resistance and ER stress. RNA pulldown, LC-MS/MS analysis and RIP were used to explore the regulatory mechanism of downstream target genes by hsa_circ_0004085. RESULTS: First, we screened out hsa_circ_0004085 with abnormally high expression in CRC clinical samples infected with Fn and found that patients with high expression of hsa_circ_0004085 in plasma had a poor clinical response to oxaliplatin/5-FU. Subsequently, the circular structure of hsa_circ_0004085 was identified. Fn infection promoted hsa_circ_0004085 formation by hnRNP L and packaged hsa_circ_0004085 into exosomes by hnRNP A1. Exosomes produced by Fn-infected CRC cells transferred hsa_circ_0004085 between cells and delivered oxaliplatin/5-FU resistance to recipient cells by relieving ER stress. Hsa_circ_0004085 enhanced the stability of GRP78 mRNA by binding to RRBP1 and promoted the nuclear translocation of ATF6p50 to relieve ER stress. CONCLUSIONS: Plasma levels of hsa_circ_0004085 are increased in colon cancer patients with intracellular Fn and are associated with a poor response to oxaliplatin/5-FU. Fn infection promoted hsa_circ_0004085 formation by hnRNP L and packaged hsa_circ_0004085 into exosomes by hnRNP A1. Exosomes secreted by Fn-infected CRC cells deliver hsa_circ_0004085 between cells. Hsa_circ_0004085 relieves ER stress in recipient cells by regulating GRP78 and ATF6p50, thereby delivering resistance to oxaliplatin and 5-FU.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Exosomes , Heterogeneous-Nuclear Ribonucleoprotein L , MicroRNAs , Humans , Oxaliplatin/pharmacology , Oxaliplatin/therapeutic use , Oxaliplatin/metabolism , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/metabolism , Heterogeneous Nuclear Ribonucleoprotein A1/metabolism , Colorectal Neoplasms/metabolism , Exosomes/metabolism , Chromatography, Liquid , Endoplasmic Reticulum Chaperone BiP , Heterogeneous-Nuclear Ribonucleoprotein L/metabolism , Tandem Mass Spectrometry , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , MicroRNAs/metabolism , Cell Proliferation
17.
Nucleic Acids Res ; 52(7): 3950-3970, 2024 Apr 24.
Article in English | MEDLINE | ID: mdl-38281181

ABSTRACT

The common oral microbe Fusobacterium nucleatum has recently drawn attention after it was found to colonize tumors throughout the human body. Fusobacteria are also interesting study systems for bacterial RNA biology as these early-branching species encode many small noncoding RNAs (sRNAs) but lack homologs of the common RNA-binding proteins (RBPs) CsrA, Hfq and ProQ. To search for alternate sRNA-associated RBPs in F. nucleatum, we performed a systematic mass spectrometry analysis of proteins that co-purified with 19 different sRNAs. This approach revealed strong enrichment of the KH domain proteins KhpA and KhpB with nearly all tested sRNAs, including the σE-dependent sRNA FoxI, a regulator of several envelope proteins. KhpA/B act as a dimer to bind sRNAs with low micromolar affinity and influence the stability of several of their target transcripts. Transcriptome studies combined with biochemical and genetic analyses suggest that KhpA/B have several physiological functions, including being required for ethanolamine utilization. Our RBP search and the discovery of KhpA/B as major RBPs in F. nucleatum are important first steps in identifying key players of post-transcriptional control at the root of the bacterial phylogenetic tree.


Subject(s)
Bacterial Proteins , Fusobacterium nucleatum , RNA, Bacterial , RNA, Small Untranslated , RNA-Binding Proteins , Fusobacterium nucleatum/genetics , Fusobacterium nucleatum/metabolism , RNA-Binding Proteins/metabolism , RNA-Binding Proteins/genetics , Bacterial Proteins/metabolism , Bacterial Proteins/genetics , RNA, Small Untranslated/metabolism , RNA, Small Untranslated/genetics , RNA, Small Untranslated/chemistry , RNA, Bacterial/metabolism , RNA, Bacterial/genetics , Gene Expression Regulation, Bacterial , Protein Binding , Mass Spectrometry
18.
Mol Biol Rep ; 51(1): 124, 2024 Jan 16.
Article in English | MEDLINE | ID: mdl-38227097

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is a global health problem. The gut microbiome is now recognized as an important underlying factor to the initiation and progression of CRC. Fusobacterium nucleatum (FN) is one of the most studied bacteria in the aetiology of CRC. This study provided cohort evidence on the association of FN infection with clinicopathologic features in CRC patients. METHODS: We analysed the cancerous and adjacent non-cancerous formalin-fixed paraffin embedded (FFPE) tissue of 83 CRC patients from a single medical centre in Malaysia. TaqMan probe-based qPCR targeting the 16S rRNA gene was used to detect the presence of FN in the extracted FFPE DNA. The differences in FN expression between cancer and non-cancer tissues were evaluated. Association studies between FN infection in the tumour and relative FN abundance with available clinical data were conducted. RESULTS: FN was more abundant in the cancerous tissue compared to non-cancerous tissue (p = 0.0025). FN infection in the tumour was significantly associated with lymph node metastasis (p = 0.047) and cancer staging (p = 0.032), but not with other clinicopathologic variables. In double-positive patients where FN was detected in both cancerous and non-cancerous tissue, the expression fold-change of FN, calculated using 2-ΔΔCT formula, was significantly higher in patients with tumour size equal to or greater than 5 cm (p = 0.033) and in KRAS-mutated patients (p = 0.046). CONCLUSIONS: FN is enriched in CRC tumour tissue and is associated with tumour size, lymph node metastasis, cancer staging, and KRAS mutation in this single-centre small cohort study.


Subject(s)
Colorectal Neoplasms , Fusobacterium nucleatum , Humans , Cohort Studies , Fusobacterium nucleatum/genetics , Lymphatic Metastasis , Proto-Oncogene Proteins p21(ras)/genetics , RNA, Ribosomal, 16S/genetics , Colorectal Neoplasms/genetics
19.
mSystems ; 9(2): e0076423, 2024 Feb 20.
Article in English | MEDLINE | ID: mdl-38289043

ABSTRACT

The major oral odor compound methyl mercaptan (CH3SH) is strongly associated with halitosis and periodontitis. CH3SH production stems from the metabolism of polymicrobial communities in periodontal pockets and on the tongue dorsum. However, understanding of CH3SH-producing oral bacteria and their interactions is limited. This study aimed to investigate CH3SH production by major oral bacteria and the impact of interspecies interactions on its generation. Using a newly constructed large-volume anaerobic noncontact coculture system, Fusobacterium nucleatum was found to be a potent producer of CH3SH, with that production stimulated by metabolic interactions with Streptococcus gordonii, an early dental plaque colonizer. Furthermore, analysis of extracellular amino acids using an S. gordonii arginine-ornithine antiporter (ArcD) mutant demonstrated that ornithine excreted from S. gordonii is a key contributor to increased CH3SH production by F. nucleatum. Further study with 13C, 15N-methionine, as well as gene expression analysis, revealed that ornithine secreted by S. gordonii increased the demand for methionine through accelerated polyamine synthesis by F. nucleatum, leading to elevated methionine pathway activity and CH3SH production. Collectively, these findings suggest that interaction between S. gordonii and F. nucleatum plays a key role in CH3SH production, providing a new insight into the mechanism of CH3SH generation in oral microbial communities. A better understanding of the underlying interactions among oral bacteria involved in CH3SH generation can lead to the development of more appropriate prophylactic approaches to treat halitosis and periodontitis. An intervention approach like selectively disrupting this interspecies network could also offer a powerful therapeutic strategy.IMPORTANCEHalitosis can have a significant impact on the social life of affected individuals. Among oral odor compounds, CH3SH has a low olfactory threshold and halitosis is a result of its production. Recently, there has been a growing interest in the collective properties of oral polymicrobial communities, regarded as important for the development of oral diseases, which are shaped by physical and metabolic interactions among community participants. However, it has yet to be investigated whether interspecies interactions have an impact on the production of volatile compounds, leading to the development of halitosis. The present findings provide mechanistic insights indicating that ornithine, a metabolite excreted by Streptococcus gordonii, promotes polyamine synthesis by Fusobacterium nucleatum, resulting in a compensatory increase in demand for methionine, which results in elevated methionine pathway activity and CH3SH production. Elucidation of the mechanisms related to CH3SH production is expected to lead to the development of new strategies for managing halitosis.


Subject(s)
Halitosis , Periodontitis , Humans , Fusobacterium nucleatum/genetics , Halitosis/microbiology , Sulfhydryl Compounds/metabolism , Bacteria , Streptococcus gordonii , Ornithine/metabolism , Methionine/metabolism , Polyamines/metabolism
20.
Br J Cancer ; 130(5): 728-740, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38200234

ABSTRACT

BACKGROUND: This study aimed to investigate clinicopathological and molecular tumour features associated with intratumoral pks+ Escherichia coli (pks+E.coli+), pks+E.coli- (non-E.coli bacteria harbouring the pks island), Enterotoxigenic Bacteroides fragilis (ETBF) and Fusobacterium nucleatum (F. nucleatum). METHODS: We screened 1697 tumour-derived DNA samples from the Australasian Colorectal Cancer Family Registry, Melbourne Collaborative Cohort Study and the ANGELS study using targeted PCR. RESULTS: Pks+E.coli+ was associated with male sex (P < 0.01) and APC:c.835-8 A > G somatic mutation (P = 0.03). The association between pks+E.coli+ and APC:c.835-8 A > G was specific to early-onset CRCs (diagnosed<45years, P = 0.02). The APC:c.835-A > G was not associated with pks+E.coli- (P = 0.36). F. nucleatum was associated with DNA mismatch repair deficiency (MMRd), BRAF:c.1799T>A p.V600E mutation, CpG island methylator phenotype, proximal tumour location, and high levels of tumour infiltrating lymphocytes (Ps < 0.01). In the stratified analysis by MMRd subgroups, F. nucleatum was associated with Lynch syndrome, MLH1 methylated and double MMR somatic mutated MMRd subgroups (Ps < 0.01). CONCLUSION: Intratumoral pks+E.coli+ but not pks+E.coli- are associated with CRCs harbouring the APC:c.835-8 A > G somatic mutation, suggesting that this mutation is specifically related to DNA damage from colibactin-producing E.coli exposures. F. nucleatum was associated with both hereditary and sporadic MMRd subtypes, suggesting the MMRd tumour microenvironment is important for F. nucleatum colonisation irrespective of its cause.


Subject(s)
Brain Neoplasms , Colorectal Neoplasms , Fusobacterium nucleatum , Neoplastic Syndromes, Hereditary , Humans , Male , Fusobacterium nucleatum/genetics , Bacteroides fragilis/genetics , Escherichia coli/genetics , Cohort Studies , Colorectal Neoplasms/pathology , DNA Damage , DNA , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL