Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Cell Commun Signal ; 21(1): 292, 2023 10 18.
Article in English | MEDLINE | ID: mdl-37853464

ABSTRACT

BACKGROUND: Hypopharyngeal squamous cell carcinoma (HPSCC) has the worst prognosis among all head-and-neck cancers, and treatment options are limited. Tumor microenvironment (TME) analysis can help identify new therapeutic targets and combined treatment strategies. METHODS: Six primary HPSCC tissues and two adjacent normal mucosae from six treatment-naïve patients with HPSCC were analyzed using scRNA-seq. Cell types were curated in detail, ecosystemic landscapes were mapped, and cell-cell interactions were inferred. Key results were validated with The Cancer Genome Atlas and cell biology experiments. RESULTS: Malignant HPSCC epithelial cells showed significant intratumor heterogeneity. Different subtypes exhibited distinct histological features, biological behaviors, and spatial localization, all affecting treatment selection and prognosis. Extracellular matrix cancer-associated fibroblasts (mCAFs) expressing fibroblast activation protein were the dominant CAFs in HPSCC tumors. mCAFs, constituting an aggressive CAF subset, promoted tumor cell invasion, activated endothelial cells to trigger angiogenesis, and synergized with SPP1+ tumor associated macrophages to induce tumor progression, ultimately decreasing the overall survival of patients with HPSCC. Moreover, the LAMP3+ dendritic cell subset was identified in HPSCC and formed an immunosuppressive TME by recruiting Tregs and suppressing CD8+ T cell function. CONCLUSIONS: mCAFs, acting as the communication center of the HPSCC TME, enhance the invasion ability of HPSCC cells, mobilizing surrounding cells to construct a tumor-favorable microenvironment. Inhibiting mCAF activation offers a new anti-HPSCC therapeutic strategy. Video Abstract.


Subject(s)
Cancer-Associated Fibroblasts , Carcinoma, Squamous Cell , Head and Neck Neoplasms , Hypopharyngeal Neoplasms , Humans , Squamous Cell Carcinoma of Head and Neck/pathology , Carcinoma, Squamous Cell/metabolism , Cancer-Associated Fibroblasts/metabolism , Endothelial Cells/metabolism , Hypopharyngeal Neoplasms/genetics , Hypopharyngeal Neoplasms/pathology , Head and Neck Neoplasms/metabolism , Sequence Analysis, RNA , Tumor Microenvironment
2.
Front Biosci (Landmark Ed) ; 28(8): 170, 2023 08 17.
Article in English | MEDLINE | ID: mdl-37664923

ABSTRACT

BACKGROUND: The role of Pin2 telomeric repeat factor 1-interacting telomerase inhibitor 1 (PinX1) in tumorigenesis and development has been extensively studied. As we previously demonstrated, PinX1 plays an important role in modulating epithelial-mesenchymal transition (EMT), stemness, cell proliferation, and apoptosis in nasopharyngeal carcinoma (NPC). However, the relationship between PinX1, autophagy, and cell function in NPC remains unclear. This study aimed to investigate the mechanisms by which PinX1 regulates autophagy in NPC, and to explore its biological role and clinical significance in disease progression. METHODS: The proliferative capacity of NPC cells was assessed by MTT and xenograft tumorigenicity assays. Autophagic flux was monitored using a tandem monomeric DAPI-FITC-LC3 reporter assay. The rates of apoptosis and the cell cycle in NPC cells were analyzed using flow cytometry. The activation of autophagy and the signaling status of the AKT/mTOR and NF-κB/p65 pathways were evaluated by Western blot analysis. RESULTS: In addition to promoting autophagy and apoptosis, PinX1 overexpression suppressed proliferation, migration, invasion, and decelerated cell-cycle progression in NPC cells. These effects were reversed by inhibiting autophagy with 3-methyladenine. Mechanistic investigations clarified that PinX1 overexpression significantly reduced the expression of p-AKT, p-mTOR, p65, and p-p65. Chloroquine treatment in PinX1-overexpressing cells did not significantly alter p-AKT and p-mTOR levels, whereas 3-MA treatment in PinX1-overexpressing cells resulted in increased p65 and p-p65 expression, relative to untreated PinX1-overexpressing cells. CONCLUSIONS: It appears that PinX1 promotes autophagy by inhibiting the AKT/mTOR signaling pathway, which then inhibits NF-κB/p65 pathways, and consequently inhibiting cell proliferation and causing cell apoptosis in NPC cells.


Subject(s)
NF-kappa B , Nasopharyngeal Neoplasms , Humans , Apoptosis , Autophagy , Cell Proliferation , Nasopharyngeal Carcinoma/genetics , Nasopharyngeal Neoplasms/genetics , Proto-Oncogene Proteins c-akt , Signal Transduction , TOR Serine-Threonine Kinases , Animals
3.
J Otolaryngol Head Neck Surg ; 52(1): 40, 2023 May 29.
Article in English | MEDLINE | ID: mdl-37248502

ABSTRACT

BACKGROUND: For recurrent laryngeal cancer, the feasibility of salvage transoral laser microsurgery (TLM) remains controversial. This study compared the efficacy of TLM and open partial laryngectomy (OPL) for treatment of early local recurrence of glottic squamous cell cancer (GSCC) and confirm the effectiveness of salvage TLM as a treatment option. METHODS: This retrospective study involved 55 patients with early local recurrent GSCC treated with TLM, and the oncologic outcomes, functional outcomes, hospitalization time and complications were compared with a group of 40 recurrent GSCC patients matched for clinical variables of TLM group, treated by OPL by the same team of surgeons. RESULTS: The 5-year overall survival and disease-specific survival rates were 65.8% and 91.5%, respectively, for 55 patients with rTis-rT2 stage treated by TLM and 77.1% and 94.7%, respectively, for 40 patients with rTis-rT2 stage treated by OPL (OPL group). In the TLM and OPL groups, the local control rates after 5 years were 77.5% and 79.3%, respectively, and the laryngeal preservation rates were 94.4% and 83.6%, respectively (p > 0.05). Compared with the OPL group, the complication rate (1.82%) and hospitalization duration (5.42 ± 2.26 days) were significantly lower in the TLM group (p < 0.05). Compared with the OPL group, postsurgical health-related quality of life and quality of voice were significantly better in the TLM group (p < 0.001). CONCLUSION: Salvage TLM can be used as an effective treatment option for suitable patients after a full, comprehensive, and careful assessment of the characteristics of early locally recurrent glottic carcinoma.


Subject(s)
Head and Neck Neoplasms , Laryngeal Neoplasms , Laser Therapy , Neoplasms, Squamous Cell , Humans , Retrospective Studies , Microsurgery , Quality of Life , Neoplasm Recurrence, Local/pathology , Squamous Cell Carcinoma of Head and Neck/surgery , Squamous Cell Carcinoma of Head and Neck/pathology , Treatment Outcome , Laryngeal Neoplasms/pathology , Glottis/surgery , Head and Neck Neoplasms/surgery , Neoplasms, Squamous Cell/pathology , Neoplasms, Squamous Cell/surgery , Lasers , Neoplasm Staging
4.
BMC Cancer ; 22(1): 577, 2022 May 24.
Article in English | MEDLINE | ID: mdl-35610596

ABSTRACT

BACKGROUND: Mast cells can reshape the tumour immune microenvironment and greatly affect tumour occurrence and development. However, mast cell gene prognostic and predictive value in head and neck squamous cell carcinoma (HNSCC) remains unclear. This study was conducted to identify and establish a prognostic mast cell gene signature (MCS) for assessing the prognosis and immunotherapy response of patients with HNSCC. METHODS: Mast cell marker genes in HNSCC were identified using single-cell RNA sequencing analysis. A dataset from The Cancer Genome Atlas was divided into a training cohort to construct the MCS model and a testing cohort to validate the model. Fluorescence in-situ hybridisation was used to evaluate the MCS model gene expression in tissue sections from patients with HNSCC who had been treated with programmed cell death-1 inhibitors and further validate the MCS. RESULTS: A prognostic MCS comprising nine genes (KIT, RAB32, CATSPER1, SMYD3, LINC00996, SOCS1, AP2M1, LAT, and HSP90B1) was generated by comprehensively analysing clinical features and 47 mast cell-related genes. The MCS effectively distinguished survival outcomes across the training, testing, and entire cohorts as an independent prognostic factor. Furthermore, we identified patients with favourable immune cell infiltration status and immunotherapy responses. Fluorescence in-situ hybridisation supported the MCS immunotherapy response of patients with HNSCC prediction, showing increased high-risk gene expression and reduced low-risk gene expression in immunotherapy-insensitive patients. CONCLUSIONS: Our MCS provides insight into the roles of mast cells in HNSCC prognosis and may have applications as an immunotherapy response predictive indicator in patients with HNSCC and a reference for immunotherapy decision-making.


Subject(s)
Head and Neck Neoplasms , Mast Cells , Biomarkers, Tumor/genetics , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/therapy , Histone-Lysine N-Methyltransferase , Humans , Prognosis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/therapy , Tumor Microenvironment/genetics
5.
Mol Carcinog ; 61(1): 45-58, 2022 01.
Article in English | MEDLINE | ID: mdl-34644425

ABSTRACT

The 5-year survival rate of laryngeal cancer continues to decline, and the laryngeal particularity of the anatomy adversely affects the patient's quality of life. Emerging evidence suggests that long noncoding RNAs (lncRNAs) are closely correlated to key steps in the malignant progression of cancer cells. In this study, we report the role of lncRNA SBF2-AS1/miR-302b-3p/TGFBR2 interactions in the metastasis of laryngeal squamous cell carcinoma (LSCC). We verified that SBF2-AS1 was significantly downregulated in LSCC tissues and cell lines using qRT-PCR analysis. Its low expression was correlated to lymph node metastasis and an advanced clinical stage. More importantly, LSCC patients with low expression of SBF2-AS1 tended to have a poor prognosis. Based on this, we performed gain-of-function and loss-of-function experiments in LSCC cell lines. The results confirmed that knocking down SBF2-AS1 can promote the metastasis of LSCC cells and enhance epithelial-mesenchymal transition phenotype, while the upregulation of SBF2-AS1 expression resulted in the opposite. Our in vivo model verified that SBF2-AS1 overexpression could inhibit LSCC cell metastasis. Subsequent mechanistic studies revealed that SBF2-AS1 acted as a competing endogenous RNA that upregulated the expression of TGFBR2 by endogenous sponging for miR-302b-3p in LSCC cell lines. Moreover, miR-302b-3p overexpression reversed the inhibitory effects on LSCC metastasis induced by upregulation of SBF2-AS1 expression, and inhibition of TGFBR2 expression reversed the effect of SBF2-AS1 on metastasis. Our study proposes SBF2-AS1 as a biomarker to predict the prognosis of LSCC patients and a novel potential therapeutic target.


Subject(s)
Down-Regulation , Laryngeal Neoplasms/pathology , MicroRNAs/genetics , RNA, Long Noncoding/genetics , Receptor, Transforming Growth Factor-beta Type II/genetics , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Laryngeal Neoplasms/genetics , Male , Mice , Neoplasm Metastasis , Neoplasm Staging , Neoplasm Transplantation
6.
Laryngoscope ; 132(9): 1807-1816, 2022 09.
Article in English | MEDLINE | ID: mdl-34873700

ABSTRACT

OBJECTIVES/HYPOTHESIS: This study aimed to elucidate the role of elective neck dissection (END) in improving the outcome of T3cN0M0 glottic squamous cell cancer (GSCC). STUDY DESIGN: Retrospective population-based database analysis. METHODS: Patients with T3cN0M0 GSCC in the Surveillance, Epidemiology, and End Results database (SEER) were extracted and stratified into END and non-END cohorts. Propensity score matching (PSM) was used to eliminate the baseline variations. The Kaplan-Meier method was performed to access the association between END and survival. RESULTS: We retrospectively analyzed 1,589 T3cN0M0 GSCC patients in the SEER database from 2004 to 2015, and found that only 22% to 58% T3cN0M0 GSCC were performed with END. After PSM, END cohort had better overall survival (OS) (median survival time: 93 vs. 55 months, respectively; P = .0047) and cancer-specific survival (CSS) (hazard ratio [HR] 0.48, 95% confidence interval [CI] 0.29-0.77, P = .003) than non-END cohort. In addition, Subgroup analysis also indicated END cohort had better OS or CSS than non-END cohort. CONCLUSION: This study demonstrated that in patients with T3cN0M0 GSCC, END significantly associated with better survival outcomes compared with non-END. LEVEL OF EVIDENCE: 3 Laryngoscope, 132:1807-1816, 2022.


Subject(s)
Head and Neck Neoplasms , Neck Dissection , Epithelial Cells , Head and Neck Neoplasms/surgery , Humans , Neck Dissection/methods , Neoplasm Staging , Prognosis , Propensity Score , Retrospective Studies , SEER Program , Squamous Cell Carcinoma of Head and Neck
7.
Bioengineered ; 12(1): 2311-2325, 2021 12.
Article in English | MEDLINE | ID: mdl-34167440

ABSTRACT

The tumor immune microenvironment plays an important role in head and neck squamous cell carcinoma (HNSCC). Reliable prognostic signatures able to accurately predict the immune landscape and survival rate of HNSCC patients are crucial to ensure an individualized/effective treatment. Here, we used HNSCC transcriptomic and clinical data retrieved from The Cancer Genome Atlas and identified differentially expressed immune-related long non-coding RNAs (DEirlncRNAs). DEirlncRNA pairs were recognized using univariate analysis. Cox and Lasso regression analyses were used to determine the association between DEirlncRNA pairs and the patients' overall survival and build the prediction model. Receiver operating characteristic curves and Kaplan-Meier survival curves were used to validate the prediction model. We then reevaluated the model based on the clinical factors, tumor-infiltrating immune cells, chemotherapeutic efficacy, and immunosuppression biomarkers. We built a risk score model based on 18 DEirlncRNA pairs, closely related to the overall survival of patients (hazard ratio: 1.376; 95% confidence interval: 1.302-1.453; P < 0.0001). Compared with two recently published lncRNA signatures, our DEirlncRNA pair signature had a higher area under the curve, indicating better prognostic performance. Additionally, the signature score positively correlated with aggressive HNSCC outcomes (low immunity score, significantly reduced CD8 + T cell infiltration, and low expression of immunosuppression biomarkers). However, high-risk patients might have high chemosensitivity. Overall, the lncRNAs signature established here shows promising clinical prediction and the effective disclosure of the tumor immune microenvironment in HNSCC patients; therefore, such signature might help distinguish patients that could benefit from immunotherapy.


Subject(s)
Head and Neck Neoplasms , RNA, Long Noncoding/immunology , Squamous Cell Carcinoma of Head and Neck , Databases, Genetic , Head and Neck Neoplasms/diagnosis , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/immunology , Head and Neck Neoplasms/mortality , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Prognosis , RNA, Long Noncoding/genetics , Squamous Cell Carcinoma of Head and Neck/diagnosis , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/immunology , Squamous Cell Carcinoma of Head and Neck/mortality , Transcriptome/genetics , Transcriptome/immunology
8.
J Exp Clin Cancer Res ; 39(1): 31, 2020 Feb 07.
Article in English | MEDLINE | ID: mdl-32028978

ABSTRACT

BACKGROUND: Recurrence and distant metastasis are still the main factors leading to treatment failure for malignant tumors including nasopharyngeal carcinoma (NPC). Therefore, elucidating the molecular mechanisms underlying nasopharyngeal carcinoma metastasis is of great clinical significance for targeted gene therapy and prognostic evaluation. PinX1, a tumor suppressor gene, was previously demonstrated to be a powerful tool for targeting telomerase in order to resist malignant tumor proliferation and migration. The aim of this study was to explore the mechanism through which PinX1 regulates epithelial-mesenchymal transition (EMT) and tumor metastasis in NPC and investigate its clinical significance and biological role with respect to disease progression. METHODS: Cell Counting Kit-8 (CCK8), Transwell assays, Colony formation analysis and Xenograft tumorigenicity assay were used to measure the nasopharyngeal CD133+ cancer stem cell proliferation, migration, and invasion abilities. Reverse transcription-quantitative polymerase chain reaction (RT-qPCR) and western blot assays were conducted to investigate the underlying mechanism that PinX1 inhibits cell proliferation, migration, and invasion via regulating EMT in nasopharyngeal CD133+ CSCs. RESULTS: We found that the overexpression of PinX1 and P53 inhibited cell proliferation, migration, and invasion, but that the inhibition of miR-200b blocked these effects, in nasopharyngeal CD133+ cancer stem cells (CSCs). Mechanistic investigations elucidated that PinX1 inhibits cell proliferation, migration, and invasion by regulating the P53/miR-200b-mediated transcriptional suppression of Snail1, Twist1, and Zeb1, consequently inhibiting EMT in nasopharyngeal CD133+ CSCs. CONCLUSIONS: Our findings indicate that PinX1 inhibits cell proliferation, migration, and invasion via P53/miR-200b-regulated EMT in the malignant progression of human NPC, which might suggest novel clinical implications for disease treatment.


Subject(s)
Biomarkers, Tumor , Cell Cycle Proteins/genetics , Nasopharyngeal Neoplasms/genetics , Neoplastic Stem Cells/metabolism , Tumor Suppressor Proteins/genetics , Animals , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Mice , MicroRNAs/genetics , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/pathology , Neoplastic Stem Cells/pathology , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...