Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 732
Filter
1.
World J Gastroenterol ; 30(29): 3479-3487, 2024 Aug 07.
Article in English | MEDLINE | ID: mdl-39156503

ABSTRACT

BACKGROUND: Helicobacter pylori (H. pylori) is a widespread microorganism related to gastric adenocarcinoma (AC). In contrast, it has been reported that an inverse association exists between H. pylori infection and esophageal carcinoma. The mechanisms underlying this supposedly protective effect remain controversial. AIM: To determine the prevalence of H. pylori infection in esophageal carcinoma patients, we performed a retrospective observational study of esophageal tumors diagnosed in our hospital. METHODS: We retrospectively reviewed the prevalence of H. pylori infection in a cohort of patients diagnosed with esophageal carcinoma. Concomitant or previous proton pump inhibitor (PPI) usage was also recorded. RESULTS: A total of 89 patients with esophageal carcinoma (69 males, 77.5%), with a mean age of 66 years (range, 26-93 years) were included. AC was the most frequent pathological variant (n = 47, 52.8%), followed by squamous cell carcinoma (n = 37, 41.6%). Fourteen ACs (29.8%) originated in the gastroesophageal junction and 33 (70.2%) in the esophageal body. Overall, 54 patients (60.7%) presented at stages III and IV. Previous H. pylori infection occurred only in 4 patients (4.5%), 3 with AC (6.3% of all ACs) and 1 with squamous cell carcinoma (2.7% of all squamous cell tumors). All patients with previous H. pylori infection had stage III-IV. Only one patient had received prior H. pylori eradication therapy, whereas 86 (96.6%) had received previous or concomitant PPI treatment. CONCLUSION: In our cohort of patients, and after histologic evaluation of paraffin-embedded primary tumors, we found a very low prevalence of previous H. pylori infection. We also reviewed the medical history of the patients, concluding that the majority had received or were on PPI treatment. The minimal prevalence of H. pylori infection found in this cohort of patients with esophageal carcinoma suggests a protective role.


Subject(s)
Esophageal Neoplasms , Helicobacter Infections , Helicobacter pylori , Proton Pump Inhibitors , Humans , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/microbiology , Male , Helicobacter Infections/epidemiology , Helicobacter Infections/diagnosis , Helicobacter Infections/microbiology , Helicobacter Infections/drug therapy , Retrospective Studies , Female , Aged , Helicobacter pylori/isolation & purification , Helicobacter pylori/pathogenicity , Middle Aged , Prevalence , Aged, 80 and over , Adult , Proton Pump Inhibitors/therapeutic use , Adenocarcinoma/epidemiology , Adenocarcinoma/microbiology , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/microbiology , Neoplasm Staging
2.
Zhonghua Zhong Liu Za Zhi ; 46(8): 746-754, 2024 Aug 23.
Article in Chinese | MEDLINE | ID: mdl-39143797

ABSTRACT

Objective: To investigate the role of an inflammatory microenvironment induced by Porphyromonasgingivalis (P. gingivalis) in the occurrence of esophageal squamous cell carcinoma (ESCC) in mice. Methods: A total of 180 C57BL/6 mice were randomly divided into 6 groups, i.e. control group, P. gingivalis group, 4NQO group, 4NQO + P. gingivalis group, 4NQO + P. gingivalis + celecoxib group, and 4NQO + P. gingivalis + antibiotic cocktail (ABC, including metronidazole, neomycin, ampicillin, and vancomycin) group, with 30 mice in each group, using the random number table. All mice were normalized by treatment with ABC in drinking water for 2 weeks. In the following 2 weeks, the mice in the control group and the P. gingivalis group were given drinking water, while the other 4 groups were treated with 30 µg/ml 4NQO in the drinking water. In weeks 11-12, the mice in the P. gingivalis group, the 4NQO + P. gingivalis group, the 4NQO + P. gingivalis + celecoxib group, and the 4NQO + P. gingivalis + ABC group were subjected to ligation of the second molar in oral cavity followed by oral P. gingivalis infection thrice weekly for 24 weeks in weeks 11-34. In weeks 13-34, the mice in 4NQO + P. gingivalis+celecoxib group and 4NQO + P. gingivalis + ABC group were administered with celecoxib and ABC for 22 weeks, respectively. At the end of 34 weeks, gross and microscopic alterations were examined followed by RT-qPCR and immunohistochemistry to examine the expression profiles of inflammatory- and tumor-molecules in esophagi of mice. Results: At 34 weeks, 4NQO treatment alone did not affect the foci of papillary hyperproliferation, diseased area, and the thickness of the esophageal wall, but significantly enhanced the foci of hyperproliferation (median 1.00, P<0.05) and mild/moderate dysplasia (median 2.00, P<0.01). In addition, the expression levels of IL-6 [8.35(3.45,8.99)], IL-1ß [6.90(2.01,9.72)], TNF-α [12.04(3.31,14.08)], c-myc [2.21(1.80,3.04)], pSTAT3, Ki-67, and pH2AX were higher than those in the control group. The pathological changes of the esophageal mucosa were significantly more overt in the 4NQO + P. gingivalis group in terms of the foci of papillary hyperproliferation (median 2.00), diseased area (median 2.51 mm2), the thickness of the esophageal wall (median 172.52 µm), the foci of hyperproliferation (median 1.00, P<0.05), and mild/moderate dysplasia (median 1.00, P<0.01). In mice of the 4NQO + P. gingivalis group, the expression levels of IL-6 [12.27(5.35,22.08)], IL-1ß [13.89(10.04,15.96)], TNF-α [19.56(6.07,20.36)], IFN-γ [11.37(8.23,20.07)], c-myc [2.62(1.51,4.25)], cyclin D1 [4.52(2.68,7.83)], nuclear pSTAT3, COX-2, Ki-67, and pH2AX were significantly increased compared with the mice in the control group. In mice of the 4NQO + P. gingivalis group, the diseased area, invasive malignant foci as well as pSTAT3 and pH2AX expression were significantly blunted by celecoxib. Treatment with ABC markedly reduced the papillary hyperproliferative foci, invasive malignant foci, and pSTAT3 expression but not pH2AX. Conclusions: P. gingivalis promotes the occurrence of esophageal squamous cell carcinoma in mice by inducing an inflammatory microenvironment primed with 4NQO induced DNA damage. Clearance of P. gingivalis with ABC or anti-inflammatory intervention holds promise for prevention of esophageal squamous cell malignant pathogenesis via blockage of IL-6/STAT3 signaling and amelioration of inflammation.


Subject(s)
4-Nitroquinoline-1-oxide , Celecoxib , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Mice, Inbred C57BL , Porphyromonas gingivalis , Tumor Microenvironment , Animals , Mice , Esophageal Neoplasms/microbiology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/microbiology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Celecoxib/pharmacology , Inflammation , Bacteroidaceae Infections/microbiology , Interleukin-6/metabolism , Anti-Bacterial Agents/pharmacology , STAT3 Transcription Factor/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase 2/genetics , Esophagus/microbiology , Esophagus/pathology , Esophagitis/microbiology , Esophagitis/pathology , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism
3.
Oral Oncol ; 157: 106973, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39033683

ABSTRACT

The article "Characterization of oral microbiota in HPV and non-HPV head and neck squamous cell carcinoma and its association with patient outcomes" by Chan et al. investigates the relationship between oral microbiota, HPV infection, and patient outcomes in head and neck squamous cell carcinoma (HNSCC). This comprehensive study, involving 166 Chinese adults, utilized advanced sequencing techniques to profile bacterial and HPV regions in paired tumor and control tissues. The findings highlight the complex interplay between microbiota dysbiosis, HPV infection, and HNSCC progression. Despite the robustness of the study, limitations include potential biases in DNA extraction and PCR amplification, and unaccounted environmental factors. Recommendations for future research include increasing sequencing depth, comparing DNA extraction methods, using multiple bioinformatics pipelines, and controlling for environmental variables. Longitudinal studies and microbiota-targeted interventions are suggested to further elucidate the role of oral microbiota in HNSCC and improve patient outcomes.


Subject(s)
Carcinoma, Squamous Cell , Head and Neck Neoplasms , Microbiota , Papillomavirus Infections , Humans , Head and Neck Neoplasms/microbiology , Head and Neck Neoplasms/virology , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/virology , Papillomavirus Infections/virology , Papillomavirus Infections/microbiology , Papillomavirus Infections/complications , Mouth/microbiology , Squamous Cell Carcinoma of Head and Neck/microbiology , Squamous Cell Carcinoma of Head and Neck/virology , Female , Male
4.
JCI Insight ; 9(16)2024 Jul 18.
Article in English | MEDLINE | ID: mdl-39024554

ABSTRACT

Anal squamous cell carcinoma (ASCC) is a rare gastrointestinal malignancy linked to high-risk human papillomavirus (HPV) infection, which develops from precursor lesions like low-grade squamous intraepithelial lesions and high-grade squamous intraepithelial lesions (HGSILs). ASCC incidence varies across populations and poses increased risk for people living with HIV. Our investigation focused on transcriptomic and metatranscriptomic changes from squamous intraepithelial lesions to ASCC. Metatranscriptomic analysis highlighted specific bacterial species (e.g., Fusobacterium nucleatum, Bacteroides fragilis) more prevalent in ASCC than precancerous lesions. These species correlated with gene-encoding enzymes (Acca, glyQ, eno, pgk, por) and oncoproteins (FadA, dnaK), presenting potential diagnostic or treatment markers. Unsupervised transcriptomic analysis identified distinct sample clusters reflecting histological diagnosis, immune infiltrate, HIV/HPV status, and pathway activities, recapitulating anal cancer progression's natural history. Our study unveiled molecular mechanisms in anal cancer progression, aiding in stratifying HGSIL cases based on low or high risk of progression to malignancy.


Subject(s)
Anus Neoplasms , Carcinoma, Squamous Cell , Transcriptome , Humans , Anus Neoplasms/genetics , Anus Neoplasms/immunology , Anus Neoplasms/pathology , Anus Neoplasms/virology , Anus Neoplasms/microbiology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/immunology , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/pathology , Microbiota/immunology , Male , Papillomavirus Infections/complications , Papillomavirus Infections/genetics , Papillomavirus Infections/virology , Papillomavirus Infections/immunology , Squamous Intraepithelial Lesions/genetics , Squamous Intraepithelial Lesions/pathology , Squamous Intraepithelial Lesions/virology , Female , Disease Progression , Middle Aged , HIV Infections/complications , HIV Infections/immunology
5.
Int J Mol Sci ; 25(11)2024 May 31.
Article in English | MEDLINE | ID: mdl-38892262

ABSTRACT

The impact of gut and oral microbiota on the clinical outcomes of patients with oral squamous cell carcinoma (OSCC) is unknown. We compared the bacterial composition of dental plaque and feces between patients with OSCC and healthy controls (HCs). Fecal and dental plaque samples were collected from 7 HCs and 18 patients with OSCC before treatment initiation. Terminal restriction fragment-length polymorphism analysis of 16S rRNA genes was performed. Differences in bacterial diversity between the HC and OSCC groups were examined. We compared the occupancy of each bacterial species in samples taken from patients with OSCC and HCs and analyzed the correlation between PD-L1 expression in the tumor specimens and the occupancy of each bacterial species. The gut and oral microbiota of patients with OSCC were more varied than those of HCs. Porphyromonas and Prevotella were significantly more abundant in patients with OSCC than in HCs. The abundance of Clostridium subcluster XIVa in the gut microbiota of the PD-L1-positive group was significantly greater than that in the PD-L1-negative group. The oral and gut microbiomes of patients with OSCC were in a state of dysbiosis. Our results suggest the possibility of new cancer therapies targeting these disease-specific microbiomes using probiotics and synbiotics.


Subject(s)
Carcinoma, Squamous Cell , Gastrointestinal Microbiome , Mouth Neoplasms , RNA, Ribosomal, 16S , Humans , Gastrointestinal Microbiome/genetics , Mouth Neoplasms/microbiology , Male , Female , Middle Aged , Carcinoma, Squamous Cell/microbiology , RNA, Ribosomal, 16S/genetics , Aged , Feces/microbiology , Mouth/microbiology , B7-H1 Antigen/metabolism , B7-H1 Antigen/genetics , Microbiota/genetics , Adult , Dysbiosis/microbiology , Dental Plaque/microbiology , Bacteria/classification , Bacteria/genetics , Bacteria/isolation & purification , Case-Control Studies
6.
Int J Mol Sci ; 25(10)2024 May 07.
Article in English | MEDLINE | ID: mdl-38791123

ABSTRACT

Periodontitis is linked to the onset and progression of oral squamous cell carcinoma (OSCC), an epidemiologically frequent and clinically aggressive malignancy. In this context, Fusobacterium (F.) nucleatum and Porphyromonas (P.) gingivalis, two bacteria that cause periodontitis, are found in OSCC tissues as well as in oral premalignant lesions, where they exert pro-tumorigenic activities. Since the two bacteria are present also in endodontic diseases, playing a role in their pathogenesis, here we analyze the literature searching for information on the impact that endodontic infection by P. gingivalis or F. nucleatum could have on cellular and molecular events involved in oral carcinogenesis. Results from the reviewed papers indicate that infection by P. gingivalis and/or F. nucleatum triggers the production of inflammatory cytokines and growth factors in dental pulp cells or periodontal cells, affecting the survival, proliferation, invasion, and differentiation of OSCC cells. In addition, the two bacteria and the cytokines they induce halt the differentiation and stimulate the proliferation and invasion of stem cells populating the dental pulp or the periodontium. Although most of the literature confutes the possibility that bacteria-induced endodontic inflammatory diseases could impact on oral carcinogenesis, the papers we have analyzed and discussed herein recommend further investigations on this topic.


Subject(s)
Fusobacterium Infections , Fusobacterium nucleatum , Mouth Neoplasms , Porphyromonas gingivalis , Humans , Porphyromonas gingivalis/pathogenicity , Fusobacterium nucleatum/pathogenicity , Mouth Neoplasms/microbiology , Mouth Neoplasms/pathology , Fusobacterium Infections/microbiology , Fusobacterium Infections/complications , Carcinogenesis , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/complications , Carcinoma, Squamous Cell/microbiology , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/pathology , Periodontitis/microbiology , Animals , Cytokines/metabolism
7.
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
8.
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
9.
J Oral Biosci ; 66(3): 594-604, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38782256

ABSTRACT

OBJECTIVES: Several studies have reported the effects of Fusobacterium nucleatum stimulation on oral cancer cells. However, given that these studies typically span a stimulation period of three days to eight days, the in vitro studies conducted to date may not fully mimic the oral cancer environment, which involves constant exposure to oral commensal bacteria. This study aimed to elucidate the effects of prolonged and persistent Fusobacterium nucleatum infection on oral cancer cells. METHODS: Human tongue squamous cell carcinoma (SCC) cells were continuously stimulated with Fusobacterium nucleatum for two or four weeks, then experimentally evaluated. RESULTS: Prolonged, persistent Fusobacterium nucleatum stimulation increased the cells' proliferative, invasive, and migratory capacities, decreased their expression of epithelial markers, and increased their expression of mesenchymal markers progressively with time. The cells also adopted a spindle-shaped morphology and cell-to-cell contact dependence was progressively lost, suggesting time-dependent occurrence of epithelial-mesenchymal transition. Furthermore, mRNA levels of CD44, a cancer stem cell marker, were time-dependently upregulated. When SCC cells were stimulated with Fusobacterium nucleatum for four weeks in the presence of dexamethasone, Fusobacterium nucleatum induced epithelial-mesenchymal transition was inhibited. CONCLUSIONS: Epithelial-mesenchymal transition in human tongue SCC cells was time-dependently induced by prolonged, persistent Fusobacterium nucleatum stimulation and inhibited by dexamethasone. Routine decontamination of the oral cavity may be crucial for controlling tumor invasion and metastasis.


Subject(s)
Carcinoma, Squamous Cell , Epithelial-Mesenchymal Transition , Fusobacterium nucleatum , Epithelial-Mesenchymal Transition/drug effects , Humans , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/microbiology , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Mouth Neoplasms/pathology , Mouth Neoplasms/microbiology , Tongue Neoplasms/pathology , Tongue Neoplasms/microbiology , Fusobacterium Infections/microbiology , Fusobacterium Infections/pathology , Dexamethasone/pharmacology , Hyaluronan Receptors/metabolism , Neoplasm Invasiveness
10.
Head Neck ; 46(7): 1660-1670, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38695435

ABSTRACT

OBJECTIVE: This study aimed to explore the potential predictive value of oral microbial signatures for oral squamous cell carcinoma (OSCC) risk based on machine learning algorithms. METHODS: The oral microbiome signatures were assessed in the unstimulated saliva samples of 80 OSCC patients and 179 healthy individuals using 16S rRNA gene sequencing. Four different machine learning classifiers were used to develop prediction models. RESULTS: Compared with control participants, OSCC patients had a higher microbial dysbiosis index (MDI, p < 0.001). Among four machine learning classifiers, random forest (RF) provided the best predictive performance, followed by the support vector machines, artificial neural networks and naive Bayes. After controlling the potential confounders using propensity score matching, the optimal RF model was further developed incorporating a minimal set of 20 bacteria genera, exhibiting better predictive performance than the MDI (AUC: 0.992 vs. 0.775, p < 0.001). CONCLUSIONS: The novel MDI and RF model developed in this study based on oral microbiome signatures may serve as noninvasive tools for predicting OSCC risk.


Subject(s)
Carcinoma, Squamous Cell , Machine Learning , Microbiota , Mouth Neoplasms , Saliva , Humans , Mouth Neoplasms/microbiology , Male , Female , Middle Aged , Saliva/microbiology , Carcinoma, Squamous Cell/microbiology , Case-Control Studies , Aged , Algorithms , Predictive Value of Tests , Adult , Dysbiosis/microbiology , Mouth/microbiology , RNA, Ribosomal, 16S/genetics , Support Vector Machine
11.
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
12.
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
13.
Cell Prolif ; 57(6): e13609, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38351596

ABSTRACT

The association between Porphyromonas gingivalis infection and oral squamous cell carcinoma (OSCC) has been established by numerous epidemiological studies. However, the underlying mechanism specific to this connection remains unclear. By bioinformatical analysis, we identified ZFP36 as a potentially significant co-expressed gene in both the OSCC gene database and the persistent infection model of P. gingivalis. To further investigate the role of ZFP36, we established a cell model that human immortalized oral epithelial cells (HIOECs) that were sustainedly infected by P. gingivalis (MOI = 1) for a duration of 30 weeks. Our findings indicated that sustained infection with P. gingivalis inhibited the expression of ZFP36 protein and induced changes in the biological behaviour of HIOECs. The mechanism investigation demonstrated the potential role of ZFP36 in regulating the cancer-related biological behaviour of HIOECs. Subsequent studies revealed that highly expressed CCAT1 could serve as a molecular scaffold in the formation of the ZFP36/CCAT1/MK2 complex. This complex formation enhanced the binding abundance of MK2 and ZFP36, thereby promoting the inhibition of ZFP36 protein phosphorylation. To summarize, low expression of ZFP36 protein under persistent P. gingivalis infection enhances the cancer-related biological behaviour of HIOECs.


Subject(s)
Bacteroidaceae Infections , Epithelial Cells , Porphyromonas gingivalis , Tristetraprolin , Humans , Porphyromonas gingivalis/pathogenicity , Epithelial Cells/microbiology , Epithelial Cells/metabolism , Epithelial Cells/pathology , Bacteroidaceae Infections/microbiology , Bacteroidaceae Infections/metabolism , Tristetraprolin/metabolism , Tristetraprolin/genetics , Mouth Neoplasms/pathology , Mouth Neoplasms/microbiology , Mouth Neoplasms/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Carcinoma, Squamous Cell/pathology , Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/microbiology , Carcinogenesis/metabolism , Carcinogenesis/pathology , Phosphorylation
15.
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
16.
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
17.
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
18.
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
19.
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
20.
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
SELECTION OF CITATIONS
SEARCH DETAIL