Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 722
Filter
1.
BMC Oral Health ; 24(1): 518, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698370

ABSTRACT

BACKGROUND: Fusobacterium nucleatum (F. nucleatum) is a microbial risk factor whose presence increases the risk of oral squamous cell carcinoma (OSCC) progression. However, whether it can promote the proliferation of OSCC cells remains unknown. METHODS: In this study, we investigated F. nucleatum effect on OSCC cell proliferation using in vitro and in vivo experiments. RESULTS: Our results showed that F. nucleatum promoted OSCC cell proliferation, doubling the cell count after 72 h (CCK-8 assay). Cell cycle analysis revealed G2/M phase arrest. F. nucleatum interaction with CDH1 triggered phosphorylation, upregulating downstream protein ß-catenin and activating cyclinD1 and Myc. Notably, F. nucleatum did not affect noncancerous cells, unrelated to CDH1 expression levels in CAL27 cells. Overexpression of phosphorylated CDH1 in 293T cells did not upregulate ß-catenin and cycle-related genes. In vivo BALB/c nude experiments showed increased tumor volume and Ki-67 proliferation index after F. nucleatum intervention. CONCLUSION: Our study suggests that F. nucleatum promotes OSCC cell proliferation through the CDH1/ß-catenin pathway, advancing our understanding of its role in OSCC progression and highlighting its potential as a therapeutic target.


Subject(s)
Cadherins , Carcinoma, Squamous Cell , Cell Proliferation , Fusobacterium nucleatum , Mice, Inbred BALB C , Mice, Nude , Mouth Neoplasms , beta Catenin , Cadherins/metabolism , Mouth Neoplasms/pathology , Mouth Neoplasms/metabolism , Mouth Neoplasms/microbiology , beta Catenin/metabolism , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Humans , Animals , Mice , Cell Line, Tumor , Antigens, CD/metabolism , Signal Transduction
2.
Arch Microbiol ; 206(6): 244, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38702412

ABSTRACT

Aggregatibacter actinomycetemcomitans is an opportunistic Gram-negative periodontopathogen strongly associated with periodontitis and infective endocarditis. Recent evidence suggests that periodontopathogens can influence the initiation and progression of oral squamous cell carcinoma (OSCC). Herein we aimed to investigate the effect of A. actinomycetemcomitans-derived extracellular vesicles (EVs) on OSCC cell behavior compared with EVs from periodontopathogens known to associate with carcinogenesis. EVs were isolated from: A. actinomycetemcomitans and its mutant strains lacking the cytolethal distending toxin (CDT) or lipopolysaccharide (LPS) O-antigen; Porphyromonas gingivalis; Fusobacterium nucleatum; and Parvimonas micra. The effect of EVs on primary and metastatic OSCC cells was assessed using cell proliferation, apoptosis, migration, invasion, and tubulogenesis assays. A. actinomycetemcomitans-derived EVs reduced the metastatic cancer cell proliferation, invasion, tubulogenesis, and increased apoptosis, mostly in CDT- and LPS O-antigen-dependent manner. EVs from F. nucleatum impaired the metastatic cancer cell proliferation and induced the apoptosis rates in all OSCC cell lines. EVs enhanced cancer cell migration regardless of bacterial species. In sum, this is the first study demonstrating the influence of A. actinomycetemcomitans-derived EVs on oral cancer in comparison with other periodontopathogens. Our findings revealed a potential antitumorigenic effect of these EVs on metastatic OSCC cells, which warrants further in vivo investigations.


Subject(s)
Aggregatibacter actinomycetemcomitans , Apoptosis , Cell Proliferation , Extracellular Vesicles , Mouth Neoplasms , Aggregatibacter actinomycetemcomitans/genetics , Extracellular Vesicles/metabolism , Mouth Neoplasms/microbiology , Mouth Neoplasms/pathology , Humans , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Movement , Fusobacterium nucleatum/physiology , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Porphyromonas gingivalis/genetics
3.
J Cancer Res Clin Oncol ; 150(4): 206, 2024 Apr 21.
Article in English | MEDLINE | ID: mdl-38644421

ABSTRACT

PURPOSE: Periodontitis-associated bacteria, such as Porphyromonas gingivalis and Fusobacterium nucleatum, are closely linked to the risk of oral squamous cell carcinoma (OSCC). Emerging studies have indicated that another common periodontal pathogen, Prevotella intermedia (P. intermedia), is enriched in OSCC and could affect the occurrence and progression of OSCC. Our aim is to determine the effects of P. intermedia on the progression of OSCC and the role of antibiotics in reversing these effects. METHODS: In this study, a murine xenograft model of OSCC was established, and the mice were injected intratumorally with PBS (control group), P. intermedia (P.i group), or P. intermedia combined with an antibiotic cocktail administration (P.i + ABX group), respectively. The effects of P. intermedia and ABX administration on xenograft tumor growth, invasion, angiogenesis, and metastasis were investigated by tumor volume measurement and histopathological examination. Enzyme-linked immunosorbent assay (ELISA) was used to investigate the changes in serum cytokine levels. Immunohistochemistry (IHC) was adopted to analyze the alterations in the levels of inflammatory cytokines and infiltrated immune cells in OSCC tissues of xenograft tumors. Transcriptome sequencing and analysis were conducted to determine differential expression genes among various groups. RESULTS: Compared with the control treatment, P. intermedia treatment significantly promoted tumor growth, invasion, angiogenesis, and metastasis, markedly affected the levels of inflammatory cytokines, and markedly altered M2 macrophages and regulatory T cells (Tregs) infiltration in the tumor microenvironment. However, ABX administration clearly abolished these effects of P. intermedia. Transcriptome and immunohistochemical analyses revealed that P. intermedia infection increased the expression of interferon-stimulated gene 15 (ISG15). Correlation analysis indicated that the expression level of ISG15 was positively correlated with the Ki67 expression level, microvessel density, serum concentrations and tissue expression levels of inflammatory cytokines, and quantities of infiltrated M2 macrophages and Tregs. However, it is negatively correlated with the quantities of infiltrated CD4+ and CD8+ T cells. CONCLUSION: In conclusion, intratumoral P. intermedia infection aggravated OSCC progression, which may be achieved through upregulation of ISG15. This study sheds new light on the possible pathogenic mechanism of intratumoral P. intermedia in OSCC progression, which could be a prospective target for OSCC prevention and treatment.


Subject(s)
Cytokines , Disease Progression , Mouth Neoplasms , Prevotella intermedia , Ubiquitins , Up-Regulation , Animals , Mice , Cytokines/metabolism , Humans , Mouth Neoplasms/pathology , Mouth Neoplasms/microbiology , Ubiquitins/metabolism , Squamous Cell Carcinoma of Head and Neck/microbiology , Squamous Cell Carcinoma of Head and Neck/pathology , Squamous Cell Carcinoma of Head and Neck/metabolism , Squamous Cell Carcinoma of Head and Neck/drug therapy , Xenograft Model Antitumor Assays , Mice, Nude , Bacteroidaceae Infections/microbiology , Cell Line, Tumor , Mice, Inbred BALB C , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/drug therapy , Anti-Bacterial Agents/pharmacology
4.
mSphere ; 9(4): e0055523, 2024 Apr 23.
Article in English | MEDLINE | ID: mdl-38530017

ABSTRACT

Human cutaneous squamous cell carcinomas (SCCs) and actinic keratoses (AK) display microbial dysbiosis with an enrichment of staphylococcal species, which have been implicated in AK and SCC progression. SCCs are common in both felines and canines and are often diagnosed at late stages leading to high disease morbidity and mortality rates. Although recent studies support the involvement of the skin microbiome in AK and SCC progression in humans, there is no knowledge of this in companion animals. Here, we provide microbiome data for SCC in cats and dogs using culture-independent molecular profiling and show a significant decrease in microbial alpha diversity on SCC lesions compared to normal skin (P ≤ 0.05). Similar to human skin cancer, SCC samples had an elevated abundance of staphylococci relative to normal skin-50% (6/12) had >50% staphylococci, as did 16% (4/25) of perilesional samples. Analysis of Staphylococcus at the species level revealed an enrichment of the pathogenic species Staphylococcus felis in cat SCC samples, a higher prevalence of Staphylococcus pseudintermedius in dogs, and a higher abundance of Staphylococcus aureus compared to normal skin in both companion animals. Additionally, a comparison of previously published human SCC and perilesional samples against the present pet samples revealed that Staphylococcus was the most prevalent genera across human and companion animals for both sample types. Similarities between the microbial profile of human and cat/dog SCC lesions should facilitate future skin cancer research. IMPORTANCE: The progression of precancerous actinic keratosis lesions (AK) to cutaneous squamous cell carcinoma (SCC) is poorly understood in humans and companion animals, despite causing a significant burden of disease. Recent studies have revealed that the microbiota may play a significant role in disease progression. Staphylococcus aureus has been found in high abundance on AK and SCC lesions, where it secretes DNA-damaging toxins, which could potentiate tumorigenesis. Currently, a suitable animal model to investigate this relationship is lacking. Thus, we examined the microbiome of cutaneous SCC in pets, revealing similarities to humans, with increased staphylococci and reduced commensals on SCC lesions and peri-lesional skin compared to normal skin. Two genera that were in abundance in SCC samples have also been found in human oral SCC lesions. These findings suggest the potential suitability of pets as a model for studying microbiome-related skin cancer progression.


Subject(s)
Carcinoma, Squamous Cell , Cat Diseases , Dog Diseases , Microbiota , Skin Neoplasms , Skin , Staphylococcus , Cats , Dogs , Animals , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/veterinary , Skin Neoplasms/microbiology , Skin Neoplasms/veterinary , Skin Neoplasms/pathology , Skin/microbiology , Skin/pathology , Cat Diseases/microbiology , Staphylococcus/isolation & purification , Staphylococcus/genetics , Staphylococcus/classification , Staphylococcus/pathogenicity , Dog Diseases/microbiology , Keratosis, Actinic/microbiology , Keratosis, Actinic/veterinary , Keratosis, Actinic/pathology
5.
Infect Immun ; 92(3): e0053623, 2024 Mar 12.
Article in English | MEDLINE | ID: mdl-38289109

ABSTRACT

Oral streptococci, key players in oral biofilm formation, are implicated in oral dysbiosis and various clinical conditions, including dental caries, gingivitis, periodontal disease, and oral cancer. Specifically, Streptococcus anginosus is associated with esophageal, gastric, and pharyngeal cancers, while Streptococcus mitis is linked to oral cancer. However, no study has investigated the mechanistic links between these Streptococcus species and cancer-related inflammatory responses. As an initial step, we probed the innate immune response triggered by S. anginosus and S. mitis in RAW264.7 macrophages. These bacteria exerted time- and dose-dependent effects on macrophage morphology without affecting cell viability. Compared with untreated macrophages, macrophages infected with S. anginosus exhibited a robust proinflammatory response characterized by significantly increased levels of inflammatory cytokines and mediators, including TNF, IL-6, IL-1ß, NOS2, and COX2, accompanied by enhanced NF-κB activation. In contrast, S. mitis-infected macrophages failed to elicit a robust inflammatory response. Seahorse Xfe96 analysis revealed an increased extracellular acidification rate in macrophages infected with S. anginosus compared with S. mitis. At the 24-h time point, the presence of S. anginosus led to reduced extracellular itaconate, while S. mitis triggered increased itaconate levels, highlighting distinct metabolic profiles in macrophages during infection in contrast to aconitate decarboxylase expression observed at the 6-h time point. This initial investigation highlights how S. anginosus and S. mitis, two Gram-positive bacteria from the same genus, can prompt distinct immune responses and metabolic shifts in macrophages during infection.IMPORTANCEThe surge in head and neck cancer cases among individuals devoid of typical risk factors such as Human Papilloma Virus (HPV) infection and tobacco and alcohol use sparks an argumentative discussion around the emerging role of oral microbiota as a novel risk factor in oral squamous cell carcinoma (OSCC). While substantial research has dissected the gut microbiome's influence on physiology, the oral microbiome, notably oral streptococci, has been underappreciated during mucosal immunopathogenesis. Streptococcus anginosus, a viridans streptococci group, has been linked to abscess formation and an elevated presence in esophageal cancer and OSCC. The current study aims to probe the innate immune response to S. anginosus compared with the early colonizer Streptococcus mitis as an important first step toward understanding the impact of distinct oral Streptococcus species on the host immune response, which is an understudied determinant of OSCC development and progression.


Subject(s)
Carcinoma, Squamous Cell , Dental Caries , Mouth Neoplasms , Succinates , Humans , Streptococcus anginosus , Carcinoma, Squamous Cell/microbiology , Streptococcus , Macrophages
6.
Microbiome ; 11(1): 171, 2023 08 05.
Article in English | MEDLINE | ID: mdl-37542310

ABSTRACT

BACKGROUND: Treating oral squamous cell carcinoma (OSCC) introduces new ecological environments in the oral cavity. This is expected to cause changes in the oral microbiome. The purpose of this study was to gain new information on the salivary microbiome of OSCC patients in order to improve the aftercare of OSCC patients. The aims of this study were to investigate possible changes in the salivary microbiome profiles of OSCC patients before and after cancer treatment and to compare these changes with the profiles of healthy controls. PATIENTS AND METHODS: Paraffin-stimulated whole saliva samples were collected, and the salivary flow rate was measured from 99 OSCC patients prior to surgical resection of the tumor and other adjuvant therapy. After treatment, 28 OSCC patients were re-examined with a mean follow-up time of 48 months. In addition, 101 healthy controls were examined and sampled. After DNA extraction and purification, the V4 hypervariable region of the 16S rRNA gene was amplified and sequenced using Illumina MiSeq. The merged read pairs were denoised using UNOISE3, mapped to zero-radius operational taxonomic units (zOTUs), and the representative zOTU sequences were assigned a taxonomy using HOMD. Descriptive statistics were used to study the differences in the microbial profiles of OSCC patients before and after treatment and in comparison to healthy controls. RESULTS: At baseline, the OSCC patients showed a higher relative abundance of zOTUs classified as Streptococcus anginosus, Abiotrophia defectiva, and Fusobacterium nucleatum. The microbial profiles differed significantly between OSCC patients and healthy controls (F = 5.9, p < 0.001). Alpha diversity of the salivary microbiome of OSCC patients was decreased at the follow-up, and the microbial profiles differed significantly from the pre-treatment (p < 0.001) and from that of healthy controls (p < 0.001). CONCLUSIONS: OSCC patients' salivary microbiome profile had a higher abundance of potentially pathogenic bacteria compared to healthy controls. Treatment of the OSCC caused a significant decrease in alpha diversity and increase in variability of the salivary microbiome, which was still evident after several years of follow-up. OSCC patients may benefit from preventive measures, such as the use of pre- or probiotics, salivary substitutes, or dietary counseling. Video Abstract.


Subject(s)
Carcinoma, Squamous Cell , Microbiota , Mouth Neoplasms , Humans , Mouth Neoplasms/therapy , Mouth Neoplasms/microbiology , Carcinoma, Squamous Cell/therapy , Carcinoma, Squamous Cell/microbiology , RNA, Ribosomal, 16S/genetics , Saliva/microbiology , Microbiota/genetics
7.
Technol Health Care ; 31(S1): 145-167, 2023.
Article in English | MEDLINE | ID: mdl-37038789

ABSTRACT

BACKGROUND: Differential protein expression of the oral microbiome is related to human diseases, including cancer. OBJECTIVE: In order to reveal the potential relationship between oral bacterial protein expression in oral squamous cell carcinoma (OSCC), we designed this study. METHODS: We obtained samples of the same patient from cancer lesion and anatomically matched normal site. Then, we used the label free quantitative technique based on liquid chromatography tandem mass spectrometry (LC-MS/MS) to analyze the bacteria in the samples of oral squamous cell carcinoma at the protein level, so as to detect the functional proteins. RESULTS: Protein diversity in the cancer samples was significantly greater than in the normal samples. We identified a substantially higher number of the taxa than those detected in previous studies, demonstrating the presence of a remarkable number of proteins in the groups. In particular, proteins involved in energy production and conversion, proton transport, hydrogen transport and hydrogen ion transmembrane transport, ATP-binding cassette (ABC) transporter, PTS system, and L-serine dehydratase were enriched significantly in the experimental group. Moreover, some proteins associated with Actinomyces and Fusobacterium were highly associated with OSCC and provided a good diagnostic outcome. CONCLUSION: The present study revealed considerable changes in the expression of bacterial proteins in OSCC and enrich our understanding in this point.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Mouth Neoplasms , Humans , Mouth Neoplasms/metabolism , Mouth Neoplasms/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Squamous Cell Carcinoma of Head and Neck , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Bacteria
8.
Otolaryngol Head Neck Surg ; 168(6): 1443-1452, 2023 06.
Article in English | MEDLINE | ID: mdl-36939272

ABSTRACT

OBJECTIVE: To examine the oral microbiome in the context of oral cavity squamous cell carcinoma. STUDY DESIGN: Basic science research. SETTING: Academic medical center. METHODS: Oral swabs were collected from patients presenting to the operating room for management of oral cavity squamous cell carcinoma and from age- and sex-matched control patients receiving surgery for unrelated benign conditions. 16S ribosomal RNA (rRNA) sequencing was performed on genetic material obtained from swabs. A bacterial rRNA gene library was created and sequence reads were sorted into taxonomic units. RESULTS: Thirty-one control patients (17 males) and 35 cancer patients (21 males) were enrolled. Ages ranged from 23 to 89 (median 63) for control patients and 35 to 86 (median 66) for cancer patients. Sixty-one percent of control patients and 63% of cancer patients were smokers. 16S analyses demonstrated a significant decrease in Streptococcus genera in oral cancer patients (34.11% vs 21.74% of the population, p = .04). Increases in Fusobacterium, Peptostreptococcus, Parvimonas, and Neisseria were also found. The abundance of these bacteria correlated with tumor T-stage. CONCLUSION: 16S rRNA sequencing demonstrated changes in bacterial populations in oral cavity cancer and its progression compared to noncancer controls. We found increases in bacteria genera that correspond with tumor stage-Fusobacteria, Peptostreptococcus, Parvimonas, Neisseria, and Treponema. These data suggest that oral cancer creates an environment to facilitate foreign bacterial growth, rather than implicating a specific bacterial species in carcinogenesis. These bacteria can be employed as a potential marker for tumor progression or interrogated to better characterize the tumor microenvironment.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Humans , Male , Bacteria , Carcinoma, Squamous Cell/microbiology , Head and Neck Neoplasms , Mouth Neoplasms/microbiology , RNA, Ribosomal, 16S/genetics , Squamous Cell Carcinoma of Head and Neck , Tumor Microenvironment
9.
Nature ; 611(7937): 810-817, 2022 11.
Article in English | MEDLINE | ID: mdl-36385528

ABSTRACT

The tumour-associated microbiota is an intrinsic component of the tumour microenvironment across human cancer types1,2. Intratumoral host-microbiota studies have so far largely relied on bulk tissue analysis1-3, which obscures the spatial distribution and localized effect of the microbiota within tumours. Here, by applying in situ spatial-profiling technologies4 and single-cell RNA sequencing5 to oral squamous cell carcinoma and colorectal cancer, we reveal spatial, cellular and molecular host-microbe interactions. We adapted 10x Visium spatial transcriptomics to determine the identity and in situ location of intratumoral microbial communities within patient tissues. Using GeoMx digital spatial profiling6, we show that bacterial communities populate microniches that are less vascularized, highly immuno­suppressive and associated with malignant cells with lower levels of Ki-67 as compared to bacteria-negative tumour regions. We developed a single-cell RNA-sequencing method that we name INVADEseq (invasion-adhesion-directed expression sequencing) and, by applying this to patient tumours, identify cell-associated bacteria and the host cells with which they interact, as well as uncovering alterations in transcriptional pathways that are involved in inflammation, metastasis, cell dormancy and DNA repair. Through functional studies, we show that cancer cells that are infected with bacteria invade their surrounding environment as single cells and recruit myeloid cells to bacterial regions. Collectively, our data reveal that the distribution of the microbiota within a tumour is not random; instead, it is highly organized in microniches with immune and epithelial cell functions that promote cancer progression.


Subject(s)
Carcinoma, Squamous Cell , Colorectal Neoplasms , Host Microbial Interactions , Microbiota , Mouth Neoplasms , Humans , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Microbiota/genetics , Microbiota/immunology , Microbiota/physiology , Mouth Neoplasms/genetics , Mouth Neoplasms/immunology , Mouth Neoplasms/microbiology , Mouth Neoplasms/pathology , Myeloid Cells/immunology , Tumor Microenvironment , Host Microbial Interactions/genetics , Host Microbial Interactions/immunology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/immunology , Colorectal Neoplasms/microbiology , Colorectal Neoplasms/pathology , Sequence Analysis, RNA , Gene Expression Profiling , Ki-67 Antigen/metabolism , Disease Progression
10.
Microbiol Spectr ; 10(6): e0273722, 2022 12 21.
Article in English | MEDLINE | ID: mdl-36445134

ABSTRACT

Oral microbial dysbiosis contributes to the development of oral squamous cell carcinoma (OSCC). Numerous studies have focused on variations in the oral bacterial microbiota of patients with OSCC. However, similar studies on fungal microbiota, another integral component of the oral microbiota, are scarce. Moreover, there is an evidence gap regarding the role that microecosystems play in different niches of the oral cavity at different stages of oral carcinogenesis. Here, we catalogued the microbial communities in the human oral cavity by profiling saliva, gingival plaque, and mucosal samples at different stages of oral carcinogenesis. We analyzed the oral bacteriome and mycobiome along the health-premalignancy-carcinoma sequence. Some species, including Prevotella intermedia, Porphyromonas endodontalis, Acremonium exuviarum, and Aspergillus fumigatus, were enriched, whereas others, such as Streptococcus salivarius subsp. salivarius, Scapharca broughtonii, Mortierella echinula, and Morchella septimelata, were depleted in OSCC. These findings suggest that an array of signature species, including bacteria and fungi, are closely associated with oral carcinogenesis. OSCC-associated diversity differences, species distinction, and functional alterations were most remarkable in mucosal samples, not in gingival plaque or saliva samples, suggesting an urgent need to define oral carcinogenesis-associated microbial dysbiosis based on the spatial microbiome. IMPORTANCE Abundant oral microorganisms constitute a complex microecosystem within the oral environment of the host, which plays a critical role in the adjustment of various physiological and pathological states of the oral cavity. In this study, we demonstrated that variations in the "core microbiome" may be used to predict carcinogenesis. In addition, sample data collected from multiple oral sites along the health-premalignancy-carcinoma sequence increase our understanding of the microecosystems of different oral niches and their specific changes during oral carcinogenesis. This work provides insight into the roles of bacteria and fungi in OSCC and may contribute to the development of early diagnostic assays and novel treatments.


Subject(s)
Carcinoma, Squamous Cell , Mouth Neoplasms , Mycobiome , Humans , Mouth Neoplasms/complications , Mouth Neoplasms/microbiology , Carcinoma, Squamous Cell/complications , Carcinoma, Squamous Cell/microbiology , Dysbiosis/microbiology , Bacteria/genetics , Fungi/genetics
11.
J BUON ; 26(5): 1862-1870, 2021.
Article in English | MEDLINE | ID: mdl-34761593

ABSTRACT

PURPOSE: The characteristics of pathogenic microbes are useful for understanding the microbe-driven tumorigenesis. There is a lack of studies on the lung microecology for lung cancer (LC) patients without any respiratory infection. In this work, we aimed to describe the profiles of pathogenic microbes in lung microenvironment of non-small cell lung cancer (NSCLC) patients using pathogen targeted sequencing and 16S rDNA sequencing. METHODS: A total of 22 NSCLC patients (13 adenocarcinomas and 9 squamous cell carcinomas) without any pulmonary infection were enrolled. Among them, we collected 15 pieces of tumor tissues, 5 pieces of peritumoral tissues, 6 blood serum samples, and 5 broncho-alveolar lavage fluid (BALF) samples. Pathogen targeted sequencingand16S rDNA sequencing was performed for microbial classification. RESULTS: The pathogen targeted sequencing results showed that 33, 14, 11, and 27 pathogenic microorganisms were detected in tumor tissues, peritumoral tissues, blood samples, and BALF, respectively. No common microorganisms were shared by four sample types. However, some common elements were shared by three sets: Streptococcus cristatus, Enterococcus, Staphylococcus haemolyticus, Corynebacterium pseudodiphtheria, Acinetobacter jungii, Haemophilus haemolyticus and Haemophilus parainfluenzae. Based on the 16S rDNA sequencing of two BALF samples, there were 104 OTUs found in one BALF sample and 127 OTUs in the other BALF sample; among them, there were 82 common ones, such as OTU1, OTU10, OTU101, OTU105, OTU106, and so on. Based on the above microbial classification and abundance, there might be enriched function in COG terms like COG1132, COG0438 and COG0745, and KEGG terms like K06147, K02029, and K09687. CONCLUSION: This study emphasizes the role of the microbiome in LC patients without respiratory infection. These potential biomarkers of LC based on the taxonomic composition of pathogenic microorganisms might have clinical application.


Subject(s)
Adenocarcinoma/microbiology , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Squamous Cell/microbiology , Lung Neoplasms/microbiology , Tumor Microenvironment , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Respiratory Tract Infections
12.
Genes (Basel) ; 12(9)2021 09 21.
Article in English | MEDLINE | ID: mdl-34573440

ABSTRACT

This study was designed to characterize the microbiomes of the lung tissues of lung cancer patients. RNA-sequencing was performed on lung tumor samples from 49 patients with lung cancer. Metatranscriptomics data were analyzed using SAMSA2 and Kraken2 software. 16S rRNA sequencing was also performed. The heterogeneous cellular landscape and immune repertoires of the lung samples were examined using xCell and TRUST4, respectively. We found that nine bacteria were significantly enriched in the lung tissues of cancer patients, and associated with reduced overall survival (OS). We also found that subjects with mutations in the epidermal growth factor receptor gene were less likely to experience the presence of Pseudomonas. aeruginosa. We found that the presence of CD8+ T-cells, CD4+ naive T-cells, dendritic cells, and CD4+ central memory T cells were associated with a good prognosis, while the presence of pro B-cells was associated with a poor prognosis. Furthermore, high clone numbers were associated with a high ImmuneScore for all immune receptor repertoires. Clone numbers and diversity were significantly higher in unpresented subjects compared to presented subjects. Our results provide insight into the microbiota of human lung cancer, and how its composition is linked to the tumor immune microenvironment, immune receptor repertoires, and OS.


Subject(s)
Lung Neoplasms/genetics , Lung Neoplasms/microbiology , Tumor Microenvironment/immunology , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/immunology , Adenocarcinoma of Lung/microbiology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/microbiology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/microbiology , Gene Expression Profiling , Humans , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Metagenome , Mutation , Pilot Projects , RNA, Ribosomal, 16S , Receptors, Antigen, B-Cell/immunology , Receptors, Antigen, T-Cell/immunology , Sequence Analysis, RNA
13.
Braz J Microbiol ; 52(3): 1287-1302, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34002353

ABSTRACT

There is increasing evidence showing positive association between changes in oral microbiome and the occurrence of oral squamous cell carcinoma (OSCC). Alcohol- and nicotine-related products can induce microbial changes but are still unknown if these changes are related to cancerous lesion sites. In an attempt to understand how these changes can influence the OSCC development and maintenance, the aim of this study was to investigate the oral microbiome linked with OSCC as well as to identify functional signatures and associate them with healthy or precancerous and cancerous sites. Our group used data of oral microbiomes available in public repositories. The analysis included data of oral microbiomes from electronic cigarette users, alcohol consumers, and precancerous and OSCC samples. An R-based pipeline was used for taxonomic and functional prediction analysis. The Streptococcus spp. genus was the main class identified in the healthy group. Haemophilus spp. predominated in precancerous lesions. OSCC samples revealed a higher relative abundance compared with the other groups, represented by an increased proportion of Fusobacterium spp., Prevotella spp., Haemophilus spp., and Campylobacter spp. Venn diagram analysis showed 52 genera exclusive of OSCC samples. Both precancerous and OSCC samples seemed to present a specific associated functional pattern. They were menaquinone-dependent protoporphyrinogen oxidase pattern enhanced in the former and both 3',5'-cyclic-nucleotide phosphodiesterase (purine metabolism) and iron(III) transport system ATP-binding protein enhanced in the latter. We conclude that although precancerous and OSCC samples present some differences on microbial profile, both microbiomes act as "iron chelators-like" potentially contributing to tumor growth.


Subject(s)
Carcinoma, Squamous Cell , Iron/metabolism , Microbiota , Mouth Neoplasms , Tumor Microenvironment , Alcohol Drinking , Carcinoma, Squamous Cell/microbiology , Electronic Nicotine Delivery Systems , Ferric Compounds/metabolism , Humans , Mouth Neoplasms/microbiology , Precancerous Conditions/microbiology
14.
PLoS Pathog ; 17(2): e1009357, 2021 02.
Article in English | MEDLINE | ID: mdl-33617591

ABSTRACT

Successful intestinal infection by Salmonella requires optimized invasion of the gut epithelium, a function that is energetically costly. Salmonella have therefore evolved to intricately regulate the expression of their virulence determinants by utilizing specific environmental cues. Here we show that a powerful repressor of Salmonella invasion, a cis-2 unsaturated long chain fatty acid, is present in the murine large intestine. Originally identified in Xylella fastidiosa as a diffusible signal factor for quorum sensing, this fatty acid directly interacts with HilD, the master transcriptional regulator of Salmonella, and prevents hilA activation, thus inhibiting Salmonella invasion. We further identify the fatty acid binding region of HilD and show it to be selective and biased in favour of signal factors with a cis-2 unsaturation over other intestinal fatty acids. Single mutation of specific HilD amino acids to alanine prevented fatty acid binding, thereby alleviating their repressive effect on invasion. Together, these results highlight an exceedingly sensitive mechanism used by Salmonella to colonize its host by detecting and exploiting specific molecules present within the complex intestinal environment.


Subject(s)
Bacterial Proteins/metabolism , Fatty Acids, Unsaturated/metabolism , Intestines/microbiology , Laryngeal Neoplasms/microbiology , Salmonella Infections/microbiology , Salmonella typhimurium/pathogenicity , Trans-Activators/metabolism , Transcription Factors/metabolism , Animals , Bacterial Proteins/genetics , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Gene Expression Regulation, Bacterial , Humans , Intestines/physiology , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/pathology , Mice , Mice, Inbred C57BL , Salmonella Infections/metabolism , Salmonella Infections/pathology , Salmonella typhimurium/genetics , Salmonella typhimurium/metabolism , Trans-Activators/genetics , Transcription Factors/genetics , Tumor Cells, Cultured , Virulence
15.
J Mycol Med ; 31(1): 101105, 2021 Mar.
Article in English | MEDLINE | ID: mdl-33422739

ABSTRACT

Chromoblastomycosis is a subcutaneous, chronic, granulomatous mycosis that occurs more frequently in tropical and subtropical countries. Herein, we describe a case of a 90-year-old female patient with diagnosis of chromoblastomycosis by Exophiala jeanselmei with a 22-year evolution who developed a squamous cell carcinoma. In the meantime, She underwent two misdiagnoses and an unnecessary operation. This case is also the fifth case of E. jeanselmei caused CBM in history.


Subject(s)
Carcinoma, Squamous Cell/microbiology , Chromoblastomycosis/complications , Chromoblastomycosis/diagnosis , Exophiala/pathogenicity , Aged, 80 and over , Antifungal Agents/therapeutic use , Chromoblastomycosis/drug therapy , Chromoblastomycosis/microbiology , Exophiala/drug effects , Female , Humans
16.
EMBO J ; 40(4): e105202, 2021 02 15.
Article in English | MEDLINE | ID: mdl-33410511

ABSTRACT

Cytotoxic necrotizing factors (CNFs) are bacterial single-chain exotoxins that modulate cytokinetic/oncogenic and inflammatory processes through activation of host cell Rho GTPases. To achieve this, they are secreted, bind surface receptors to induce endocytosis and translocate a catalytic unit into the cytosol to intoxicate host cells. A three-dimensional structure that provides insight into the underlying mechanisms is still lacking. Here, we determined the crystal structure of full-length Yersinia pseudotuberculosis CNFY . CNFY consists of five domains (D1-D5), and by integrating structural and functional data, we demonstrate that D1-3 act as export and translocation module for the catalytic unit (D4-5) and for a fused ß-lactamase reporter protein. We further found that D4, which possesses structural similarity to ADP-ribosyl transferases, but had no equivalent catalytic activity, changed its position to interact extensively with D5 in the crystal structure of the free D4-5 fragment. This liberates D5 from a semi-blocked conformation in full-length CNFY , leading to higher deamidation activity. Finally, we identify CNF translocation modules in several uncharacterized fusion proteins, which suggests their usability as a broad-specificity protein delivery tool.


Subject(s)
Bacterial Toxins/chemistry , Bacterial Toxins/metabolism , Carcinoma, Squamous Cell/pathology , Cytosol/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Laryngeal Neoplasms/pathology , Yersinia pseudotuberculosis/metabolism , rhoA GTP-Binding Protein/metabolism , Biological Transport , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Crystallization , Crystallography, X-Ray , Humans , Laryngeal Neoplasms/metabolism , Laryngeal Neoplasms/microbiology , Protein Conformation , Tumor Cells, Cultured
17.
APMIS ; 129(5): 265-270, 2021 May.
Article in English | MEDLINE | ID: mdl-33205550

ABSTRACT

The carcinogenic effects of microorganisms have been discovered in multiple cancer types. In urology, the development of squamous cell carcinoma of the bladder due to the parasitic infection with Schistosoma Mansoni is widely accepted. The oncogenic potential of biofilms has been studied in colorectal cancer and experimental studies have shown that bacteria such as Escherichia coli drive the development of colorectal cancer. Notably, Escherichia coli is responsible for 80% of all urinary tract infections. Recent findings suggest an altered urinary microbiome in patients with bladder cancer compared to healthy subjects. In this case series, we demonstrate our findings of biofilm formation in human bladder cancer tissue. Tissue samples from ten patients that underwent routine Transurethral Resection of Bladder Tumor (TURBT) were obtained from the Danish National Biobank. Pathological tissue was examined for presence of bacterial aggregates by Fluorescence in situ Hybridization. In two of ten patients, analysis showed abundant bacterial aggregation on the surface epithelium. Both positive cases had pT2 urothelial bladder cancer. Our findings suggest that biofilm occurs in urothelial cancer tissue indicating an association between biofilm formation and bladder cancer.


Subject(s)
Bacteria/isolation & purification , Biofilms/growth & development , Muscle Neoplasms/microbiology , Urinary Bladder Neoplasms/microbiology , Aged , Aged, 80 and over , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/surgery , Cystoscopy , Female , Humans , Male , Muscle Neoplasms/pathology , Muscle Neoplasms/surgery , Neoplasm Invasiveness , Urinary Bladder Neoplasms/pathology , Urinary Bladder Neoplasms/surgery
18.
Int J Mol Sci ; 21(21)2020 Oct 29.
Article in English | MEDLINE | ID: mdl-33137960

ABSTRACT

Oral squamous cell carcinoma (OSCC) is one of the leading presentations of head and neck cancer (HNC). The first part of this review will describe the highlights of the oral microbiome in health and normal development while demonstrating how both the oral and gut microbiome can map OSCC development, progression, treatment and the potential side effects associated with its management. We then scope the dynamics of the various microorganisms of the oral cavity, including bacteria, mycoplasma, fungi, archaea and viruses, and describe the characteristic roles they may play in OSCC development. We also highlight how the human immunodeficiency viruses (HIV) may impinge on the host microbiome and increase the burden of oral premalignant lesions and OSCC in patients with HIV. Finally, we summarise current insights into the microbiome-treatment axis pertaining to OSCC, and show how the microbiome is affected by radiotherapy, chemotherapy, immunotherapy and also how these therapies are affected by the state of the microbiome, potentially determining the success or failure of some of these treatments.


Subject(s)
Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/therapy , Microbiota , Mouth Neoplasms/microbiology , Mouth Neoplasms/therapy , Animals , Carcinoma, Squamous Cell/pathology , Disease Progression , Drug Therapy , Humans , Immunotherapy , Mouth Neoplasms/pathology , Radiotherapy
19.
PLoS Pathog ; 16(10): e1008881, 2020 10.
Article in English | MEDLINE | ID: mdl-33002094

ABSTRACT

Epidemiological studies reveal significant associations between periodontitis and oral cancer. However, knowledge about the contribution of periodontal pathogens to oral cancer and potential regulatory mechanisms involved is limited. Previously, we showed that nisin, a bacteriocin and commonly used food preservative, reduced oral cancer tumorigenesis and extended the life expectancy in tumor-bearing mice. In addition, nisin has antimicrobial effects on key periodontal pathogens. Thus, the purpose of this study was to test the hypothesis that key periodontal pathogens (Porphyromonas gingivalis, Treponema denticola, and Fusobacterium nucleatum) promote oral cancer via specific host-bacterial interactions, and that bacteriocin/nisin therapy may modulate these responses. All three periodontal pathogens enhanced oral squamous cell carcinoma (OSCC) cell migration, invasion, tumorsphere formation, and tumorigenesis in vivo, without significantly affecting cell proliferation or apoptosis. In contrast, oral commensal bacteria did not affect OSCC cell migration. Pathogen-enhanced OSCC cell migration was mediated via integrin alpha V and FAK activation, since stably blocking alpha V or FAK expression abrogated these effects. Nisin inhibited these pathogen-mediated processes. Further, Treponema denticola induced TLR2 and 4 and MyD88 expression. Stable suppression of MyD88 significantly inhibited Treponema denticola-induced FAK activation and abrogated pathogen-induced migration. Together, these data demonstrate that periodontal pathogens contribute to a highly aggressive cancer phenotype via crosstalk between TLR/MyD88 and integrin/FAK signaling. Nisin can modulate these pathogen-mediated effects, and thus has therapeutic potential as an antimicrobial and anti-tumorigenic agent.


Subject(s)
Bacteroidaceae Infections/drug therapy , Carcinoma, Squamous Cell/drug therapy , Mouth Neoplasms/drug therapy , Periodontitis/drug therapy , Porphyromonas gingivalis/drug effects , Probiotics/pharmacology , Animals , Apoptosis , Bacteroidaceae Infections/metabolism , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Cell Movement , Cell Proliferation , Focal Adhesion Kinase 1/genetics , Focal Adhesion Kinase 1/metabolism , Humans , Integrins/genetics , Integrins/metabolism , Mice , Mice, Nude , Mouth Neoplasms/metabolism , Mouth Neoplasms/microbiology , Mouth Neoplasms/pathology , Myeloid Differentiation Factor 88/genetics , Myeloid Differentiation Factor 88/metabolism , Periodontitis/metabolism , Periodontitis/microbiology , Periodontitis/pathology , Porphyromonas gingivalis/pathogenicity , Toll-Like Receptor 2/genetics , Toll-Like Receptor 2/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
20.
Int J Gynecol Cancer ; 30(8): 1151-1156, 2020 08.
Article in English | MEDLINE | ID: mdl-32675252

ABSTRACT

INTRODUCTION: We characterized the cervical 16S rDNA microbiome of patients in Botswana with high-grade cervical dysplasia and locally advanced cervical cancer. METHODS: This prospective study included 31 patients: 21 with dysplasia and 10 with cancer. The Shannon diversity index was used to evaluate alpha (intra-sample) diversity, while the UniFrac (weighted and unweighted) and Bray-Curtis distances were employed to evaluate beta (inter-sample) diversity. The relative abundance of microbial taxa was compared among samples using linear discriminant analysis effect size. RESULTS: Alpha diversity was significantly higher in patients with cervical cancer than in patients with cervical dysplasia (P<0.05). Beta diversity also differed significantly (weighted UniFrac Bray-Curtis, P<0.01). Neither alpha diversity (P=0.8) nor beta diversity (P=0.19) varied by HIV status. The results of linear discriminant analysis effect size demonstrated that multiple taxa differed significantly between patients with cervical dysplasia vs cancer. Lachnospira bacteria (in the Clostridia class) were particularly enriched among cervical dysplasia patients, while Proteobacteria (members of the Firmicutes phyla and the Comamonadaceae family) were enriched in patients with cervical cancer. DISCUSSION: The results of our study suggest that differences exist in the diversity and composition of the cervical microbiota between patients with cervical dysplasia and patients with cervical cancer in Botswana. Additional studies are warranted to validate these findings and elucidate their clinical significance among women living in sub-Saharan Africa, as well as other regions of the world.


Subject(s)
Carcinoma, Squamous Cell/microbiology , Cervix Uteri/microbiology , Uterine Cervical Dysplasia/microbiology , Uterine Cervical Neoplasms/microbiology , Adult , Botswana , Carcinoma, Squamous Cell/pathology , Clostridiales , Comamonadaceae , Female , Gardnerella , Humans , Lactobacillus , Microbiota , Middle Aged , Neoplasm Grading , Prospective Studies , Proteobacteria , Uterine Cervical Dysplasia/pathology , Uterine Cervical Neoplasms/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...