Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
1.
J Transl Med ; 22(1): 401, 2024 Apr 30.
Artículo en Inglés | MEDLINE | ID: mdl-38689341

RESUMEN

BACKGROUND: The cancer microbiota was considered the main risk factor for cancer progression. We had proved that Fusobacterium periodonticum (F.p) was higher abundance in Esophageal cancer(EC)tissues. Bioinformation analysis found that BCT was a key virulence protein of F.p. However, little is known about the role and mechanism of BCT in EC. This study aimed to recognize the key virulence protein of F.p and explore the mechanism of BCT in promoting EC. METHODS: We constructed a eukaryotic expression vector and purified the recombinant protein BCT. CCK8 used to analyzed the activity of EC after treated by different concentration of BCT. UPLC-MS/MS and ELISA used to detect the metabonomics and metabolites. The ability of migration and invasion was completed by transwell assay. RT-QPCR, WB used to analyze the expression of relevant genes. RESULTS: Our data showed that BCT was higher expression in EC tumor tissues (p < 0.05) and BCT in 20 µg/mL promoted the survival, invasion and migration of EC cells (EC109) (p < 0.05). Meanwhile, UPLC-MS/MS results suggested that BCT resulted in an augmentation of hypotaurine metabolism, arachidonic acid metabolism, glycolysis/gluconeogenesis, tryptophan metabolism, citrate cycle activity in EC109. The metabolic changes resulted in decreasing in glucose and pyruvate levels but increase in lactate dehydrogenase (LDH) activity and lactic acid (LA) as well as the expression of glucose transporter 1, Hexokinase 2, LDH which regulated the glycolysis were all changed (p < 0.05). The BCT treatment upregulated the expression of TLR4, Akt, HIF-1α (p < 0.05) which regulated the production of LA. Furthermore, LA stimulation promoted the expression of GPR81, Wnt, and ß-catenin (p < 0.05), thereby inducing EMT and metastasis in EC109 cells. CONCLUSION: Altogether, these findings identified that impact of BCT in regulation of glycolysis in EC109 and its involves the TLR4/Akt/HIF-1α pathway. Meanwhile, glycolysis increasing the release of LA and promote the EMT of EC109 by GPR81/Wnt/ß-catenin signaling pathway. In summary, our findings underscore the potential of targeting BCT as an innovative strategy to mitigate the development of EC.


Asunto(s)
Movimiento Celular , Transición Epitelial-Mesenquimal , Neoplasias Esofágicas , Fusobacterium , Glucosa , Ácido Láctico , Humanos , Neoplasias Esofágicas/patología , Neoplasias Esofágicas/metabolismo , Ácido Láctico/metabolismo , Línea Celular Tumoral , Glucosa/metabolismo , Fusobacterium/metabolismo , Proteínas Bacterianas/metabolismo , Invasividad Neoplásica , Regulación Neoplásica de la Expresión Génica
2.
Neoplasia ; 31: 100813, 2022 09.
Artículo en Inglés | MEDLINE | ID: mdl-35834946

RESUMEN

Recently, increased number of studies have demonstrated a relationship between the oral microbiome and development of head and neck cancer, however, there are few studies to investigate the role of oral bacteria in the context of the tumor microenvironment in a single head and neck subsite. Here, paired tumor and adjacent normal tissues from thirty-seven oral tongue squamous cell carcinoma (SCC) patients were subjected to 16S rRNA gene sequencing and whole exome sequencing (WES), in addition to RNA sequencing for tumor samples. We observed that Fusobacterium was significantly enriched in oral tongue cancer and that Rothia and Streptococcus were enriched in adjacent normal tissues. A decrease in alpha diversity was found in tumor when compared to adjacent normal tissues. While increased Fusobacterium in tumor samples was not associated with changes in immune cell infiltration, it was associated with increased PD-L1 mRNA expression. Therefore, we examined the effects of Fusobacterium on PD-L1 expression in head and neck SCC cell lines. We demonstrated that infection with Fusobacterium species can increase both PD-L1 mRNA and surface PD-L1 protein expression on head and neck cancer cell lines. The correlation between Fusobacterium and PD-L1 expression in oral tongue SCC, in conjunction with the ability of the bacterium to induce PD-L1 expression in vitro suggests a potential role for Fusobacterium on modulation of the tumor immune microenvironment in head and neck cancer.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Neoplasias de la Lengua , Antígeno B7-H1/genética , Fusobacterium/genética , Fusobacterium/metabolismo , Humanos , Neoplasias de la Boca/genética , ARN Mensajero , ARN Ribosómico 16S/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Neoplasias de la Lengua/genética , Microambiente Tumoral/genética
3.
Front Immunol ; 12: 744184, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34659241

RESUMEN

Fusobacterium nucleatum is involved in the development of colorectal cancer (CRC) through innate immune cell modulation. However, the receptors of the interaction between F. nucleatum ssp. and immune cells remain largely undetermined. Here, we showed that F. nucleatum ssp. animalis interacts with Siglecs (sialic acid-binding immunoglobulin-like lectins) expressed on innate immune cells with highest binding to Siglec-7. Binding to Siglec-7 was also observed using F. nucleatum-derived outer membrane vesicles (OMVs) and lipopolysaccharide (LPS). F. nucleatum and its derived OMVs or LPS induced a pro-inflammatory profile in human monocyte-derived dendritic cells (moDCs) and a tumour associated profile in human monocyte-derived macrophages (moMϕs). Siglec-7 silencing in moDCs or CRISPR-cas9 Siglec-7-depletion of U-937 macrophage cells altered F. nucleatum induced cytokine but not marker expression. The molecular interaction between Siglec-7 and the LPS O-antigen purified from F. nucleatum ssp. animalis was further characterised by saturation transfer difference (STD) NMR spectroscopy, revealing novel ligands for Siglec-7. Together, these data support a new role for Siglec-7 in mediating immune modulation by F. nucleatum strains and their OMVs through recognition of LPS on the bacterial cell surface. This opens a new dimension in our understanding of how F. nucleatum promotes CRC progression through the generation of a pro-inflammatory environment and provides a molecular lead for the development of novel cancer therapeutic approaches targeting F. nucleatum-Siglec-7 interaction.


Asunto(s)
Antígenos de Diferenciación Mielomonocítica/inmunología , Neoplasias Colorrectales/inmunología , Células Dendríticas/inmunología , Fusobacterium/inmunología , Lectinas/inmunología , Macrófagos/inmunología , Antígenos de Diferenciación Mielomonocítica/metabolismo , Carcinogénesis/inmunología , Carcinogénesis/metabolismo , Línea Celular , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/microbiología , Células Dendríticas/metabolismo , Fusobacterium/metabolismo , Humanos , Inmunomodulación/inmunología , Lectinas/metabolismo , Macrófagos/metabolismo
4.
Gut Microbes ; 13(1): 1-23, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34057024

RESUMEN

Characterizing the metabolic functions of the gut microbiome in health and disease is pivotal for translating alterations in microbial composition into clinical insights. Two major analysis paradigms have been used to explore the metabolic functions of the microbiome but not systematically integrated with each other: statistical screening approaches, such as metabolome-microbiome association studies, and computational approaches, such as constraint-based metabolic modeling. To combine the strengths of the two analysis paradigms, we herein introduce a set of theoretical concepts allowing for the population statistical treatment of constraint-based microbial community models. To demonstrate the utility of the theoretical framework, we applied it to a public metagenomic dataset consisting of 365 colorectal cancer (CRC) cases and 251 healthy controls, shining a light on the metabolic role of Fusobacterium spp. in CRC. We found that (1) glutarate production capability was significantly enriched in CRC microbiomes and mechanistically linked to lysine fermentation in Fusobacterium spp., (2) acetate and butyrate production potentials were lowered in CRC, and (3) Fusobacterium spp. presence had large negative ecological effects on community butyrate production in CRC cases and healthy controls. Validating the model predictions against fecal metabolomics, the in silico frameworks correctly predicted in vivo species metabolite correlations with high accuracy. In conclusion, highlighting the value of combining statistical association studies with in silico modeling, this study provides insights into the metabolic role of Fusobacterium spp. in the gut, while providing a proof of concept for the validity of constraint-based microbial community modeling.


Asunto(s)
Bacterias/metabolismo , Butiratos/metabolismo , Heces/microbiología , Fusobacterium/metabolismo , Microbioma Gastrointestinal , Anciano , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Estudios de Casos y Controles , Neoplasias Colorrectales/microbiología , Heces/química , Femenino , Fusobacterium/genética , Fusobacterium/aislamiento & purificación , Humanos , Masculino , Metabolómica , Persona de Mediana Edad
5.
Food Chem ; 353: 129447, 2021 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-33714122

RESUMEN

In order to explore the correlation between the production of propionic acid (PA) and the succession of bacterial community during the fermentation of gray sufu, high-throughput sequencing and HPLC (High Performance Liquid Chromatography) were used to monitor the changes of bacterial community and metabolite content. The abundance and metabolite concentration of Propionibacterium increased rapidly in the early stage of fermentation. In the middle stage, the abundance of Lactobacillus began to increase, while the pH decreased rapidly. In the late stage, the concentration of PA began to decrease, but it remained at a high level at the end of fermentation. Correlation analysis showed that Lactobacillus and Bacillus had a strong negative correlation with PA and its precursor. The results showed that Fusobacterium, Providencia, Lactobacillus and Bacillus could be the key factors to reduce the PA content. This study provides a new idea for the quality control of traditional fermented food.


Asunto(s)
Bacterias/metabolismo , Alimentos Fermentados/microbiología , Propionatos/metabolismo , Alimentos de Soja/microbiología , Bacterias/genética , Cromatografía Líquida de Alta Presión , Fermentación , Microbiología de Alimentos , Fusobacterium/genética , Fusobacterium/metabolismo , Secuenciación de Nucleótidos de Alto Rendimiento , Lactobacillus/genética , Lactobacillus/metabolismo , Propionibacterium/genética , Propionibacterium/metabolismo
6.
Nat Commun ; 11(1): 4531, 2020 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-32913225

RESUMEN

RNAs begin to fold and function during transcription. Riboswitches undergo cotranscriptional switching in the context of transcription elongation, RNA folding, and ligand binding. To investigate how these processes jointly modulate the function of the folate stress-sensing Fusobacterium ulcerans ZTP riboswitch, we apply a single-molecule vectorial folding (VF) assay in which an engineered superhelicase Rep-X sequentially releases fluorescently labeled riboswitch RNA from a heteroduplex in a 5'-to-3' direction, at ~60 nt s-1 [comparable to the speed of bacterial RNA polymerase (RNAP)]. We demonstrate that the ZTP riboswitch is kinetically controlled and that its activation is favored by slower unwinding, strategic pausing between but not before key folding elements, or a weakened transcription terminator. Real-time single-molecule monitoring captures folding riboswitches in multiple states, including an intermediate responsible for delayed terminator formation. These results show how individual nascent RNAs occupy distinct channels within the folding landscape that controls the fate of the riboswitch.


Asunto(s)
Fusobacterium/genética , Regulación Bacteriana de la Expresión Génica , Pliegue del ARN/genética , ARN Bacteriano/genética , Riboswitch/genética , Aminoimidazol Carboxamida/metabolismo , Fusobacterium/metabolismo , Conformación de Ácido Nucleico , ARN Bacteriano/metabolismo , Ribonucleótidos/metabolismo , Imagen Individual de Molécula , Transcripción Genética
7.
Cell Chem Biol ; 27(10): 1241-1249.e4, 2020 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-32795418

RESUMEN

Riboswitches are mRNA domains that make gene-regulatory decisions upon binding their cognate ligands. Bacterial riboswitches that specifically recognize 5-aminoimidazole-4-carboxamide riboside 5'-monophosphate (ZMP) and 5'-triphosphate (ZTP) regulate genes involved in folate and purine metabolism. Now, we have developed synthetic ligands targeting ZTP riboswitches by replacing the sugar-phosphate moiety of ZMP with various functional groups, including simple heterocycles. Despite losing hydrogen bonds from ZMP, these analogs bind ZTP riboswitches with similar affinities as the natural ligand, and activate transcription more strongly than ZMP in vitro. The most active ligand stimulates gene expression ∼3 times more than ZMP in a live Escherichia coli reporter. Co-crystal structures of the Fusobacterium ulcerans ZTP riboswitch bound to synthetic ligands suggest stacking of their pyridine moieties on a conserved RNA nucleobase primarily determines their higher activity. Altogether, these findings guide future design of improved riboswitch activators and yield insights into how RNA-targeted ligand discovery may proceed.


Asunto(s)
Aminoimidazol Carboxamida/farmacología , Descubrimiento de Drogas , ARN Bacteriano/efectos de los fármacos , Riboswitch/efectos de los fármacos , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/química , Escherichia coli/química , Escherichia coli/metabolismo , Fusobacterium/química , Fusobacterium/metabolismo , Enlace de Hidrógeno , Ligandos , Estructura Molecular , ARN Bacteriano/química , ARN Bacteriano/metabolismo
8.
PLoS Biol ; 18(8): e3000788, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32841232

RESUMEN

Women with bacterial vaginosis (BV), an imbalance of the vaginal microbiome, are more likely to be colonized by potential pathogens such as Fusobacterium nucleatum, a bacterium linked with intrauterine infection and preterm birth. However, the conditions and mechanisms supporting pathogen colonization during vaginal dysbiosis remain obscure. We demonstrate that sialidase activity, a diagnostic feature of BV, promoted F. nucleatum foraging and growth on mammalian sialoglycans, a nutrient resource that was otherwise inaccessible because of the lack of endogenous F. nucleatum sialidase. In mice with sialidase-producing vaginal microbiotas, mutant F. nucleatum unable to consume sialic acids was impaired in vaginal colonization. These experiments in mice also led to the discovery that F. nucleatum may also "give back" to the community by reinforcing sialidase activity, a biochemical feature of human dysbiosis. Using human vaginal bacterial communities, we show that F. nucleatum supported robust outgrowth of Gardnerella vaginalis, a major sialidase producer and one of the most abundant organisms in BV. These results illustrate that mutually beneficial relationships between vaginal bacteria support pathogen colonization and may help maintain features of dysbiosis. These findings challenge the simplistic dogma that the mere absence of "healthy" lactobacilli is the sole mechanism that creates a permissive environment for pathogens during vaginal dysbiosis. Given the ubiquity of F. nucleatum in the human mouth, these studies also suggest a possible mechanism underlying links between vaginal dysbiosis and oral sex.


Asunto(s)
Proteínas Bacterianas/genética , Disbiosis/microbiología , Fusobacterium/metabolismo , Gardnerella vaginalis/metabolismo , Neuraminidasa/genética , Polisacáridos/metabolismo , Vaginosis Bacteriana/microbiología , Animales , Proteínas Bacterianas/metabolismo , Técnicas de Tipificación Bacteriana , Disbiosis/patología , Femenino , Fusobacterium/genética , Fusobacterium/aislamiento & purificación , Fusobacterium/patogenicidad , Gardnerella vaginalis/genética , Gardnerella vaginalis/aislamiento & purificación , Gardnerella vaginalis/patogenicidad , Expresión Génica , Humanos , Ratones , Ratones Endogámicos C57BL , Microbiota/genética , Neuraminidasa/metabolismo , ARN Ribosómico 16S/genética , Ácidos Siálicos/metabolismo , Simbiosis/genética , Vagina/microbiología , Vaginosis Bacteriana/patología
9.
J Bacteriol ; 201(23)2019 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-31501282

RESUMEN

Fusobacterium spp. are Gram-negative, anaerobic, opportunistic pathogens involved in multiple diseases, including a link between the oral pathogen Fusobacterium nucleatum and the progression and severity of colorectal cancer. The identification and characterization of virulence factors in the genus Fusobacterium has been greatly hindered by a lack of properly assembled and annotated genomes. Using newly completed genomes from nine strains and seven species of Fusobacterium, we report the identification and corrected annotation of verified and potential virulence factors from the type 5 secreted autotransporter, FadA, and MORN2 protein families, with a focus on the genetically tractable strain F. nucleatum subsp. nucleatum ATCC 23726 and type strain F. nucleatum subsp. nucleatum ATCC 25586. Within the autotransporters, we used sequence similarity networks to identify protein subsets and show a clear differentiation between the prediction of outer membrane adhesins, serine proteases, and proteins with unknown function. These data have identified unique subsets of type 5a autotransporters, which are key proteins associated with virulence in F. nucleatum However, we coupled our bioinformatic data with bacterial binding assays to show that a predicted weakly invasive strain of F. necrophorum that lacks a Fap2 autotransporter adhesin strongly binds human colonocytes. These analyses confirm a gap in our understanding of how autotransporters, MORN2 domain proteins, and FadA adhesins contribute to host interactions and invasion. In summary, we identify candidate virulence genes in Fusobacterium, and caution that experimental validation of host-microbe interactions should complement bioinformatic predictions to increase our understanding of virulence protein contributions in Fusobacterium infections and disease.IMPORTANCEFusobacterium spp. are emerging pathogens that contribute to mammalian and human diseases, including colorectal cancer. Despite a validated connection with disease, few proteins have been characterized that define a direct molecular mechanism for Fusobacterium pathogenesis. We report a comprehensive examination of virulence-associated protein families in multiple Fusobacterium species and show that complete genomes facilitate the correction and identification of multiple, large type 5a secreted autotransporter genes in previously misannotated or fragmented genomes. In addition, we use protein sequence similarity networks and human cell interaction experiments to show that previously predicted noninvasive strains can indeed bind to and potentially invade human cells and that this could be due to the expansion of specific virulence proteins that drive Fusobacterium infections and disease.


Asunto(s)
Adhesinas Bacterianas/genética , Fusobacterium/genética , Fusobacterium/patogenicidad , Genoma Bacteriano , Sistemas de Secreción Tipo V/genética , Factores de Virulencia/genética , Adhesinas Bacterianas/clasificación , Adhesinas Bacterianas/metabolismo , Secuencia de Aminoácidos , Adhesión Bacteriana , Línea Celular , Biología Computacional/métodos , Células Epiteliales/microbiología , Células Epiteliales/patología , Fusobacterium/clasificación , Fusobacterium/metabolismo , Infecciones por Fusobacterium/microbiología , Infecciones por Fusobacterium/patología , Expresión Génica , Encía/microbiología , Encía/patología , Células HCT116 , Humanos , Filogenia , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Sistemas de Secreción Tipo V/clasificación , Sistemas de Secreción Tipo V/metabolismo , Virulencia , Factores de Virulencia/clasificación , Factores de Virulencia/metabolismo
10.
Sci Rep ; 9(1): 3230, 2019 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-30824723

RESUMEN

Macrodomains constitute a conserved fold widely distributed that is not only able to bind ADP-ribose in its free and protein-linked forms but also can catalyse the hydrolysis of the latter. They are involved in the regulation of important cellular processes, such as signalling, differentiation, proliferation and apoptosis, and in host-virus response, and for this, they are considered as promising therapeutic targets to slow tumour progression and viral pathogenesis. Although extensive work has been carried out with them, including their classification into six distinct phylogenetically clades, little is known on bacterial macrodomains, especially if these latter are able to remove poly(ADP-ribose) polymer (PAR) from PARylated proteins, activity that only has been confirmed in human TARG1 (C6orf130) protein. To extend this limited knowledge, we demonstrate, after a comprehensive bioinformatic and phylogenetic analysis, that Fusobacterium mortiferum ATCC 9817 TARG1 (FmTARG1) is the first bacterial macrodomain shown to have high catalytic efficiency towards O-acyl-ADP-ribose, even more than hTARG1, and towards mono- and poly(ADPribosyl)ated proteins. Surprisingly, FmTARG1 gene is also inserted into a unique operonic context, only shared by the distantly related Fusobacterium perfoetens ATCC 29250 macrodomain, which include an immunity protein 51 domain, typical of bacterial polymorphic toxin systems.


Asunto(s)
Proteínas Bacterianas/química , Fusobacterium/metabolismo , Hidrolasas/química , N-Glicosil Hidrolasas/química , Poli Adenosina Difosfato Ribosa/metabolismo , Dominios Proteicos , Secuencia de Aminoácidos , Proteínas Bacterianas/clasificación , Proteínas Bacterianas/genética , Fusobacterium/genética , Humanos , Hidrolasas/genética , Hidrolasas/metabolismo , N-Glicosil Hidrolasas/clasificación , N-Glicosil Hidrolasas/genética , Filogenia , Poli(ADP-Ribosa) Polimerasa-1/química , Poli(ADP-Ribosa) Polimerasa-1/genética , Poli(ADP-Ribosa) Polimerasa-1/metabolismo , Procesamiento Proteico-Postraduccional , Estabilidad Proteica , Homología de Secuencia de Aminoácido , Temperatura , Tioléster Hidrolasas/química , Tioléster Hidrolasas/genética , Tioléster Hidrolasas/metabolismo
11.
Appl Biochem Biotechnol ; 187(1): 163-175, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29911265

RESUMEN

Accumulation of succinate as a fermentation product of Fusobacterium varium was enhanced when the anaerobic bacterium was grown on complex peptone medium supplemented with fumarate. Residual substrates and fermentation products were determined by proton NMR spectroscopy. Cells collected from the fumarate-supplemented medium (8-10 h after inoculation) supported the conversion of fumarate to succinate when suspended with fumarate and a co-substrate (glucose, sorbitol, or glycerol). Succinate production was limited by the availability of fumarate or reducing equivalents supplied by catabolism of a co-substrate via the Embden-Meyerhof-Parnas (EMP) pathway. The choice of reducing co-substrate influenced the yield of acetate and lactate as side products. High conversions of fumarate to succinate were achieved over pH 6.6-8.2 and initial fumarate concentrations up to 300 mM. However, at high substrate concentrations, intracellular retention of succinate reduced extracellular yields. Overall, the efficient utilization of fumarate (≤ 400 mM) combined with the significant extracellular accumulation of succinate (corresponding to ≥ 70% conversion) indicated the effective utilization of fumarate as a terminal electron acceptor by F. varium and the potential of the methodology for the bioproduction of succinate.


Asunto(s)
Fumaratos/metabolismo , Fusobacterium/metabolismo , Ácido Succínico/metabolismo , Oxidación-Reducción
12.
BMC Microbiol ; 17(1): 61, 2017 03 14.
Artículo en Inglés | MEDLINE | ID: mdl-28288582

RESUMEN

BACKGROUND: Hydrogen sulfide (H2S) is a toxic foul-smelling gas produced by subgingival biofilms in patients with periodontal disease and is suggested to be part of the pathogenesis of the disease. We studied the H2S-producing protein expression of bacterial strains associated with periodontal disease. Further, we examined the effect of a cysteine-rich growth environment on the synthesis of intracellular enzymes in F. nucleatum polymorphum ATCC 10953. The proteins were subjected to one-dimensional (1DE) and two-dimensional (2DE) gel electrophoresis An in-gel activity assay was used to detect the H2S-producing enzymes; Sulfide from H2S, produced by the enzymes in the gel, reacted with bismuth forming bismuth sulfide, illustrated as brown bands (1D) or spots (2D) in the gel. The discovered proteins were identified with liquid chromatography - tandem mass spectrometry (LC-MS/MS). RESULTS: Cysteine synthase and proteins involved in the production of the coenzyme pyridoxal 5'phosphate (that catalyzes the production of H2S) were frequently found among the discovered enzymes. Interestingly, a higher expression of H2S-producing enzymes was detected from bacteria incubated without cysteine prior to the experiment. CONCLUSIONS: Numerous enzymes, identified as cysteine synthase, were involved in the production of H2S from cysteine and the expression varied among Fusobacterium spp. and strains. No enzymes were detected with the in-gel activity assay among the other periodontitis-associated bacteria tested. The expression of the H2S-producing enzymes was dependent on environmental conditions such as cysteine concentration and pH but less dependent on the presence of serum and hemin.


Asunto(s)
Proteínas Bacterianas/metabolismo , Cisteína/metabolismo , Fusobacterium/enzimología , Fusobacterium/metabolismo , Sulfuro de Hidrógeno/metabolismo , Proteínas Bacterianas/análisis , Biopelículas , Bismuto/metabolismo , Cisteína Sintasa/metabolismo , Placa Dental , Electroforesis en Gel Bidimensional/métodos , Humanos , Concentración de Iones de Hidrógeno , Enfermedades Periodontales/microbiología , Proteómica , Sulfuros/metabolismo , Espectrometría de Masas en Tándem
13.
Glycobiology ; 26(10): 1107-1119, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27613803

RESUMEN

Sialic acids are nine-carbon backbone carbohydrates found in prominent outermost positions of glycosylated molecules in mammals. Mimicry of sialic acid (N-acetylneuraminic acid, Neu5Ac) enables some pathogenic bacteria to evade host defenses. Fusobacterium nucleatum is a ubiquitous oral bacterium also linked with invasive infections throughout the body. We employed multidisciplinary approaches to test predictions that F. nucleatum engages in de novo synthesis of sialic acids. Here we show that F. nucleatum sbsp. polymorphum ATCC10953 NeuB (putative Neu5Ac synthase) restores Neu5Ac synthesis to an Escherichia coli neuB mutant. Moreover, purified F. nucleatum NeuB participated in synthesis of Neu5Ac from N-acetylmannosamine and phosphoenolpyruvate in vitro Further studies support the interpretation that F. nucleatum ATCC10953 NeuA encodes a functional CMP-sialic acid synthetase and suggest that it may also contain a C-terminal sialic acid O-acetylesterase. We also performed BLAST queries of F. nucleatum genomes, revealing that only 4/31 strains encode a complete pathway for de novo Neu5Ac synthesis. Biochemical studies including mass spectrometry were consistent with the bioinformatic predictions, showing that F. nucleatum ATCC10953 synthesizes high levels of Neu5Ac, whereas ATCC23726 and ATCC25586 do not express detectable levels above background. While there are a number of examples of sialic acid mimicry in other phyla, these experiments provide the first biochemical and genetic evidence that a member of the phylum Fusobacterium can engage in de novo Neu5Ac synthesis.


Asunto(s)
Fusobacterium/metabolismo , Ácido N-Acetilneuramínico/biosíntesis , Cromatografía Líquida de Alta Presión , Espectrometría de Masas , Ácido N-Acetilneuramínico/química
14.
Appl Environ Microbiol ; 82(4): 1080-1089, 2016 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-26637604

RESUMEN

The gut microbiota of termites and cockroaches represents complex metabolic networks of many diverse microbial populations. The distinct microenvironmental conditions within the gut and possible interactions among the microorganisms make it essential to investigate how far the metabolic properties of pure cultures reflect their activities in their natural environment. We established the cockroach Shelfordella lateralis as a gnotobiotic model and inoculated germfree nymphs with two bacterial strains isolated from the guts of conventional cockroaches. Fluorescence microscopy revealed that both strains specifically colonized the germfree hindgut. In diassociated cockroaches, the facultatively anaerobic strain EbSL (a new species of Enterobacteriaceae) always outnumbered the obligately anaerobic strain FuSL (a close relative of Fusobacterium varium), irrespective of the sequence of inoculation, which showed that precolonization by facultatively anaerobic bacteria does not necessarily favor colonization by obligate anaerobes. Comparison of the fermentation products of the cultures formed in vitro with those accumulated in situ indicated that the gut environment strongly affected the metabolic activities of both strains. The pure cultures formed the typical products of mixed-acid or butyrate fermentation, whereas the guts of gnotobiotic cockroaches accumulated mostly lactate and acetate. Similar shifts toward more-oxidized products were observed when the pure cultures were exposed to oxygen, which corroborated the strong effects of oxygen on the metabolic fluxes previously observed in termite guts. Oxygen microsensor profiles of the guts of germfree, gnotobiotic, and conventional cockroaches indicated that both gut tissue and microbiota contribute to oxygen consumption and suggest that the oxygen status influences the colonization success.


Asunto(s)
Cucarachas , Microbioma Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/microbiología , Vida Libre de Gérmenes , Oxígeno , Aerobiosis , Anaerobiosis , Animales , Enterobacteriaceae/crecimiento & desarrollo , Enterobacteriaceae/metabolismo , Fusobacterium/crecimiento & desarrollo , Fusobacterium/metabolismo
15.
Curr Microbiol ; 70(2): 169-75, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25257648

RESUMEN

In this study, we classified the five strains (ChDC F128(T), ChDC F145, ChDC F174, ChDC F206, and ChDC F300) as a novel species of genus Fusobacterium by DNA-DNA hybridization and multi-locus phylogenetic analysis (MLPA), based on a single sequence (24,715 bp) of 22 concatenated housekeeping genes, with morphological and chemotaxonomic characteristics. DNA-DNA hybridization data showed that the values of genomic relatedness between ChDC F128(T) and each of the other novel strains were ranged from 79.0 to 82.6 %, while those of genomic relatedness between ChDC F128(T) and type strain of each of subspecies of F. nucleatum or Fusobacterium periodonticum were ranged from 40.9 to 54.4 %. MLPA revealed that the 5 strains were clustered as one group and clearly discriminated with F. nucleatum and F. periodonticum with 100 % bootstrap value. The DNA G+C content of the five novel strains were ranged from 26.9 to 27.0 mol%. The cellular fatty acid analysis of clinical isolates and type strains revealed C14:0, C16:0, and cis-9 C16:1 as the major fatty acids. The cell wall peptidoglycan of the 5 strains was comprised of meso-lanthionine. These results show that the 5 strains are novel species and belong to the genus Fusobacterium. Strain ChDC F128(T) (=KCOM 1249(T) = KCTC 5108(T) = JCM 30218(T)) is suggested to be the type strain of a novel species of genus Fusobacterium, for which the name Fusobacterium hwasookii sp. nov. is proposed.


Asunto(s)
Infecciones por Fusobacterium/microbiología , Fusobacterium , Periodontitis/microbiología , Composición de Base , ADN Bacteriano , Fusobacterium/clasificación , Fusobacterium/genética , Fusobacterium/metabolismo , Genes Esenciales , Humanos , Tipificación de Secuencias Multilocus , Filogenia
16.
Wei Sheng Wu Xue Bao ; 54(10): 1228-34, 2014 Oct 04.
Artículo en Chino | MEDLINE | ID: mdl-25803901

RESUMEN

OBJECTIVE: To compare the abundance of 16S rRNA gene of intestinal Fusobacterium and butyrate-producing bacteria in patients with colorectal adenomas patients and colorectal cancer and to reveal the correlation between the target bacteria and the development of colorectal cancer. METHODS: Feces were collected from colorectal cancer patients (n=19), colorectal adenomas patients (n=12) and healthy subjects (n=19). Bacteria genome DNA from the fecal samples was used to quantitate the Fusobacterium, two butyrate-producing bacteria Eubacterium rectal, Faecalibacterium prausnitzii and total bacteria by real-time polymerase chain reaction. Then the variation of the target bacteria among different groups were assayed using Mann-Whitney U test. RESULTS: The abundance of Fusobacterium was significantly higher in colorectal cancer patients than that in healthy subjects (P = 0.000) and colorectal adenomas patients (P = 0.013), and it was significantly higher in colorectal cancer patients than that in colorectal adenomas patients (P = 0.002). F. prausnitzii was significantly lower in colorectal adenomas patients compared to healthy subjects (P = 0.033). The total bacteria count was significantly lower in the colorectal adenomas samples than that in the healthy samples (P = 0.002). There was no significantly difference of E. rectal between the three groups. CONCLUSIONS: The shifts in the colonic bacterial population may potentially contribute to the development of colorectal cancer.


Asunto(s)
Adenoma/microbiología , Butiratos/metabolismo , Neoplasias Colorrectales/microbiología , Fusobacterium/metabolismo , Intestinos/microbiología , Bacterias/clasificación , Bacterias/genética , Bacterias/aislamiento & purificación , Bacterias/metabolismo , Heces/microbiología , Femenino , Fusobacterium/clasificación , Fusobacterium/genética , Fusobacterium/aislamiento & purificación , Humanos , Masculino , Persona de Mediana Edad
17.
Benef Microbes ; 4(3): 253-66, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23666100

RESUMEN

Gut bacteria influence host anatomy and physiology. It has been proposed that bacterial metabolites including polyamines are responsible for intestinal maturation and mucosal growth. We have hypothesised that bacterially produced polyamines act as trophic factors and thereby influence large intestinal crypt depth and thickness of the different gut layers. For that purpose, germ-free mice were associated with two different microbial consortia. One group was colonised with a simplified human microbiota (SIHUMI). The second group was associated with SIHUMI + Fusobacterium varium (SIHUMI + Fv), which is known to produce high amounts of polyamines. Polyamine concentrations were measured by HPLC and morphological parameters were determined microscopically. Germ-free and conventional mice served as controls. The caecal putrescine concentration of the SIHUMI + Fv was 61.8 µM (47.6-75.5 µM), whereas that of conventional and SIHUMI mice was 28.8 µM (1.3-41.7 µM) and 24.5 µM (16.8-29.1 µM), respectively. The caecal putrescine concentration of germ-free mice was only 0.6 µM (0-1.0 µM). Caecal crypt depth and thickness of the different caecal layers revealed no significant differences between SIHUMI and SIHUMI + Fv mice. However, the crypt depth in the caeca of conventional, SIHUMI and SIHUMI + Fv mice was increased by 48.6% (P<0.001), 39.7% (P<0.001) and 28.5% (P<0.05), respectively, compared to germ-free mice. These findings indicate that increased intestinal putrescine concentrations do not influence gut morphology in our gnotobiotic adolescent mice.


Asunto(s)
Ciego/microbiología , Ciego/fisiología , Fusobacterium/metabolismo , Putrescina/metabolismo , Animales , Ciego/anatomía & histología , Ciego/química , Cromatografía Líquida de Alta Presión , Vida Libre de Gérmenes , Ratones , Microbiota
18.
PLoS One ; 8(1): e53653, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23335968

RESUMEN

The human gut microbiota is increasingly recognized as a player in colorectal cancer (CRC). While particular imbalances in the gut microbiota have been linked to colorectal adenomas and cancer, no specific bacterium has been identified as a risk factor. Recent studies have reported a high abundance of Fusobacterium in CRC subjects compared to normal subjects, but this observation has not been reported for adenomas, CRC precursors. We assessed the abundance of Fusobacterium species in the normal rectal mucosa of subjects with (n = 48) and without adenomas (n = 67). We also confirmed previous reports on Fusobacterium and CRC in 10 CRC tumor tissues and 9 matching normal tissues by pyrosequencing. We extracted DNA from rectal mucosal biopsies and measured bacterial levels by quantitative PCR of the 16S ribosomal RNA gene. Local cytokine gene expression was also determined in mucosal biopsies from adenoma cases and controls by quantitative PCR. The mean log abundance of Fusobacterium or cytokine gene expression between cases and controls was compared by t-test. Logistic regression was used to compare tertiles of Fusobacterium abundance. Adenoma subjects had a significantly higher abundance of Fusobacterium species compared to controls (p = 0.01). Compared to the lowest tertile, subjects with high abundance of Fusobacterium were significantly more likely to have adenomas (OR 3.66, 95% CI 1.37-9.74, p-trend 0.005). Cases but not controls had a significant positive correlation between local cytokine gene expression and Fusobacterium abundance. Among cases, the correlation for local TNF-α and Fusobacterium was r = 0.33, p = 0.06 while it was 0.44, p = 0.01 for Fusobacterium and IL-10. These results support a link between the abundance of Fusobacterium in colonic mucosa and adenomas and suggest a possible role for mucosal inflammation in this process.


Asunto(s)
Adenoma/microbiología , Neoplasias Colorrectales/microbiología , Fusobacterium/genética , Fusobacterium/metabolismo , Adenoma/genética , Adenoma/patología , Adulto , Anciano , Biopsia , Estudios de Casos y Controles , Colon/metabolismo , Colon/microbiología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Citocinas/genética , Femenino , Expresión Génica , Humanos , Inflamación/metabolismo , Inflamación/microbiología , Inflamación/patología , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , ARN Bacteriano , ARN Ribosómico 16S , Reproducibilidad de los Resultados , Factores de Riesgo
19.
Antioxid Redox Signal ; 17(10): 1433-40, 2012 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-22607129

RESUMEN

Faecalibacterium prausnitzii is one of the most abundant commensal microbes in the human gut. It is an important supplier of butyrate to the colonic epithelium, and low numbers of faecalibacteria have been associated with severe inflammatory bowel disease. Previous studies revealed that F. prausnitzii shuttles electrons extracellularly to oxygen in systems containing flavins and thiols. Since this electron shuttling to oxygen strongly stimulates growth, the present studies were aimed at elucidating the role of riboflavin as an extracellular electronophore of F. prausnitzii. We show that F. prausnitzii can use riboflavin as a mediator for extracellular electron transfer (EET) to the anode of microbial fuel cell systems. However, this bacterium relies on exogenous riboflavin, since it does not secrete this compound as shown by the analysis of a spent growth medium using cyclic voltammetry (CV). Importantly, CV showed that riboflavin can undergo fully reversible redox cycling under physiologically relevant conditions. Lastly, riboflavin is shown to mediate the electrochemical oxidation of the main bacterial reducing equivalent NADH. Based on our present observations, we hypothesize that riboflavin is of major importance as a redox mediator for bacterial EET and growth in the human gut.


Asunto(s)
Transporte de Electrón/fisiología , Fusobacterium/metabolismo , Riboflavina/metabolismo , Reactores Biológicos , Oxidación-Reducción
20.
Anaerobe ; 18(4): 381-5, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22609780

RESUMEN

The occurrence of Porphyromonas gulae, Porphyromonas macacae, Fusobacterium nucleatum and Fusobacterium canifelinum in subgingival plaque from dogs with and without periodontitis as well as their antimicrobial susceptibility were evaluated. From 50 dogs with periodontitis were identified 38 P. gulae, 8 P. macacae, 26 F. nucleatum and 15 F. canifelinum, and from 50 dogs without periodontitis were identified 15 P. gulae, 12 F. nucleatum and 11 F. canifelinum. All strains were susceptible to most of the antibiotics tested, however, different resistance rates to clarithromycin, erythromycin and metronidazole among strains were observed. The role of P. gulae, P. macacae, F. nucleatum and F. canifelinum in periodontal disease of household pets needs to be defined to a better prevention and treatment of the canine periodontitis.


Asunto(s)
Enfermedades de los Perros/microbiología , Perros/microbiología , Fusobacterium/efectos de los fármacos , Fusobacterium/aislamiento & purificación , Periodontitis/microbiología , Porphyromonas/efectos de los fármacos , Porphyromonas/aislamiento & purificación , Animales , Antibacterianos/farmacología , Infecciones por Bacteroidaceae/microbiología , Claritromicina/farmacología , Placa Dental/microbiología , Farmacorresistencia Bacteriana , Eritromicina/farmacología , Femenino , Fusobacterium/metabolismo , Infecciones por Fusobacterium/microbiología , Pruebas de Inhibición de Hemaglutinación , Humanos , Masculino , Metronidazol/farmacología , Pruebas de Sensibilidad Microbiana , Porphyromonas/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...