Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 216
Filtrar
1.
Int Immunopharmacol ; 142(Pt B): 113243, 2024 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-39340989

RESUMEN

BACKGROUND: Hypopharyngeal squamous cell carcinoma (HSCC) is a type of head and neck tumor with malignant behavior and poor prognosis. Spatial transcriptomics is a method that spatially analyzes gene expression patterns in tissues and has been used to discover tumor microenvironment and molecular markers in various tumors. However, there are no published reports on spatial transcriptomic analysis of HSCC. METHODS: In this study, spatial transcriptomic analysis was performed on tumor tissues in situ, peritumoral tissues, and lymphatic metastatic tissues of four patients with HSCC. Morphological markers, including panCK, SMA, and CD45, were used to identify epithelial, fibroblast, and immune cells, respectively. By analyzing the expression of more than 18, 000 genes within the transcriptome of all ROIs, differentially expressed genes of three cell types in different tissues were identified, and differentially expressed signaling pathways and immune infiltration were analyzed. RESULTS: The spatial distribution of cells suggests that fibroblast cells in tumor tissues may be involved in the genesis and development of tumors, and the immune infiltration of lymphatic tumor metastasis is lower than that of tumors in situ. For epithelial cells, SLCO2A1, which is a favorable prognosis marker in head and neck squamous cell carcinoma (HNSCC), was significantly down-regulated in tumor tissues and lymphatic metastatic tissues compared with adjacent normal tissues. For immune cells, KANK3, which is a favorable prognosis markers in HNSCC, was significantly down-regulated in lymphatic metastatic tissues compared with adjacent normal tissues. For fibroblast cells, AQP1, CLEC3B and SLCO2A1, which are favorable prognosis markers in HNSCC, were significantly down-regulated in tumor tissues compared with adjacent normal tissues. ITGA8, which is a favorable prognosis markers in HNSCC, was significantly down-regulated in lymphatic metastatic tissues compared with normal lymphatic tissues. CSRP1, DES, and SLCO2A1 positively correlate with immune infiltration in HNSCC. Moreover, SLCO2A1 overexpression suppressed Fadu cells proliferation and metastasis and significantly correlated with favorable survival overcome in HSCC. CONCLUSIONS: We investigated tumor and fibroblast heterogeneity, as well as the immune microenvironment in HSCC by using spatial transcriptomics. SLCO2A1 may be a tumor suppressor gene and correlates with immune infiltration for HSCC and could serve as a potential target for its diagnosis and treatment.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Neoplasias Hipofaríngeas , Transportadores de Anión Orgánico , Transcriptoma , Microambiente Tumoral , Humanos , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/patología , Neoplasias Hipofaríngeas/inmunología , Transportadores de Anión Orgánico/genética , Transportadores de Anión Orgánico/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Masculino , Biomarcadores de Tumor/genética , Perfilación de la Expresión Génica , Persona de Mediana Edad , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/patología , Femenino , Metástasis Linfática , Pronóstico , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/patología
2.
World J Surg Oncol ; 22(1): 254, 2024 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-39300515

RESUMEN

Hypopharyngeal carcinoma is one of the malignant tumors of the head and neck with a particularly poor prognosis. Recurrence and metastasis are important reasons for poor prognosis of hypopharyngeal cancer patients, and malignant proliferation, migration, and invasion of tumor cells are important factors for recurrence and metastasis of hypopharyngeal cancer. Therefore, elucidating hypopharyngeal cancer cells' proliferation, migration, and invasion mechanism is essential for improving diagnosis, treatment, and prognosis. Plasmacytoma Variant Translocation 1 (PVT1) is considered a potential diagnostic marker and therapeutic target for tumors. However, it remains unclear whether PVT1 is related to the occurrence and development of hypopharyngeal cancer and its specific mechanism. In this study, the promoting effect of PVT1 on the proliferation, migration, and invasion of hypopharyngeal carcinoma FaDu cells was verified by cell biology experiments and animal studies, and it was found that PVT1 inhibited the expression of TGF-ß, suggesting that PVT1 may regulate the occurrence and development of hypopharyngeal carcinoma FaDu cells through TGF-ß.


Asunto(s)
Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Hipofaríngeas , Invasividad Neoplásica , ARN Largo no Codificante , Animales , Humanos , Ratones , Apoptosis , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Neoplasias Hipofaríngeas/patología , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/metabolismo , Ratones Endogámicos BALB C , Ratones Desnudos , Pronóstico , ARN Largo no Codificante/genética , Factor de Crecimiento Transformador beta/metabolismo , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto , Femenino
3.
Sci Rep ; 14(1): 13796, 2024 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-38877096

RESUMEN

To explore the hub comorbidity genes and potential pathogenic mechanisms of hypopharyngeal carcinoma with esophageal carcinoma, and evaluate their diagnostic value for hypopharyngeal carcinoma with co-morbid esophageal carcinoma. We performed gene sequencing on tumor tissues from 6 patients with hypopharyngeal squamous cell carcinoma with esophageal squamous cell carcinoma (hereafter referred to as "group A") and 6 patients with pure hypopharyngeal squamous cell carcinoma (hereafter referred to as "group B"). We analyzed the mechanism of hub genes in the development and progression of hypopharyngeal squamous cell carcinoma with esophageal squamous cell carcinoma through bioinformatics, and constructed an ROC curve and Nomogram prediction model to analyze the value of hub genes in clinical diagnosis and treatment. 44,876 genes were sequenced in 6 patients with group A and 6 patients with group B. Among them, 76 genes showed significant statistical differences between the group A and the group B.47 genes were expressed lower in the group A than in the group B, and 29 genes were expressed higher. The top five hub genes were GABRG2, CACNA1A, CNTNAP2, NOS1, and SCN4B. GABRG2, CNTNAP2, and SCN4B in the hub genes have high diagnostic value in determining whether hypopharyngeal carcinoma patients have combined esophageal carcinoma (AUC: 0.944, 0.944, 0.972). These genes could possibly be used as potential molecular markers for assessing the risk of co-morbidity of hypopharyngeal carcinoma combined with esophageal carcinoma.


Asunto(s)
Neoplasias Esofágicas , Regulación Neoplásica de la Expresión Génica , Neoplasias Hipofaríngeas , Humanos , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/patología , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/diagnóstico , Masculino , Femenino , Persona de Mediana Edad , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Biomarcadores de Tumor/genética , Anciano , Análisis de Secuencia de ARN/métodos , Perfilación de la Expresión Génica , Biología Computacional/métodos , Nomogramas
4.
Int J Mol Sci ; 25(11)2024 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-38892156

RESUMEN

Hypopharyngeal squamous cell carcinoma (HSCC) is a kind of malignant tumor with a poor prognosis and low quality of life in the otolaryngology department. It has been found that microRNA (miRNA) plays an important role in the occurrence and development of various tumors. This study found that the expression level of miRNA-107 (miR-107) in HSCC was significantly reduced. Subsequently, we screened out the downstream direct target gene Neuronal Vesicle Trafficking Associated 1 (NSG1) related to miR-107 through bioinformatics analysis and found that the expression of NSG1 was increased in HSCC tissues. Following the overexpression of miR-107 in HSCC cells, it was observed that miR-107 directly suppressed NSG1 expression, leading to increased apoptosis, decreased proliferation, and reduced invasion capabilities of HSCC cells. Subsequent experiments involving the overexpression and knockdown of NSG1 in HSCC cells demonstrated that elevated NSG1 levels enhanced cell proliferation, migration, and invasion, while the opposite effect was observed upon NSG1 knockdown. Further investigations revealed that changes in NSG1 levels in the HSCC cells were accompanied by alterations in ERK signaling pathway proteins, suggesting a potential regulatory role of NSG1 in HSCC cell proliferation, migration, and invasion through the ERK pathway. These findings highlight the significance of miR-107 and NSG1 in hypopharyngeal cancer metastasis, offering promising targets for therapeutic interventions and prognostic evaluations for HSCC.


Asunto(s)
Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Hipofaríngeas , Sistema de Señalización de MAP Quinasas , MicroARNs , Humanos , Masculino , Apoptosis/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Progresión de la Enfermedad , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/patología , Neoplasias Hipofaríngeas/metabolismo , Sistema de Señalización de MAP Quinasas/genética , MicroARNs/genética , MicroARNs/metabolismo , Invasividad Neoplásica , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo
5.
FEBS Open Bio ; 14(7): 1166-1191, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38783639

RESUMEN

Hypopharyngeal cancer is a disease that is associated with EGFR-mutated lung adenocarcinoma. Here we utilized a bioinformatics approach to identify genetic commonalities between these two diseases. To this end, we examined microarray datasets from GEO (Gene Expression Omnibus) to identify differentially expressed genes, common genes, and hub genes between the selected two diseases. Our analyses identified potential therapeutic molecules for the selected diseases based on 10 hub genes with the highest interactions according to the degree topology method and the maximum clique centrality (MCC). These therapeutic molecules may have the potential for simultaneous treatment of these diseases.


Asunto(s)
Adenocarcinoma del Pulmón , Biología Computacional , Receptores ErbB , Redes Reguladoras de Genes , Neoplasias Hipofaríngeas , Neoplasias Pulmonares , Mutación , Humanos , Receptores ErbB/genética , Receptores ErbB/metabolismo , Redes Reguladoras de Genes/genética , Adenocarcinoma del Pulmón/genética , Neoplasias Hipofaríngeas/genética , Biología Computacional/métodos , Neoplasias Pulmonares/genética , Regulación Neoplásica de la Expresión Génica/genética , Perfilación de la Expresión Génica
6.
Front Immunol ; 15: 1310376, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38720887

RESUMEN

Introduction: Hypopharyngeal squamous cell carcinoma (HSCC) is one of the malignant tumors with the worst prognosis in head and neck cancers. The transformation from normal tissue through low-grade and high-grade intraepithelial neoplasia to cancerous tissue in HSCC is typically viewed as a progressive pathological sequence typical of tumorigenesis. Nonetheless, the alterations in diverse cell clusters within the tissue microenvironment (TME) throughout tumorigenesis and their impact on the development of HSCC are yet to be fully understood. Methods: We employed single-cell RNA sequencing and TCR/BCR sequencing to sequence 60,854 cells from nine tissue samples representing different stages during the progression of HSCC. This allowed us to construct dynamic transcriptomic maps of cells in diverse TME across various disease stages, and experimentally validated the key molecules within it. Results: We delineated the heterogeneity among tumor cells, immune cells (including T cells, B cells, and myeloid cells), and stromal cells (such as fibroblasts and endothelial cells) during the tumorigenesis of HSCC. We uncovered the alterations in function and state of distinct cell clusters at different stages of tumor development and identified specific clusters closely associated with the tumorigenesis of HSCC. Consequently, we discovered molecules like MAGEA3 and MMP3, pivotal for the diagnosis and treatment of HSCC. Discussion: Our research sheds light on the dynamic alterations within the TME during the tumorigenesis of HSCC, which will help to understand its mechanism of canceration, identify early diagnostic markers, and discover new therapeutic targets.


Asunto(s)
Carcinogénesis , Neoplasias Hipofaríngeas , Análisis de la Célula Individual , Carcinoma de Células Escamosas de Cabeza y Cuello , Humanos , Masculino , Biomarcadores de Tumor/genética , Carcinogénesis/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/patología , Neoplasias Hipofaríngeas/inmunología , Receptores de Antígenos de Linfocitos B/genética , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Análisis de Secuencia de ARN , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Transcriptoma , Microambiente Tumoral/inmunología , Microambiente Tumoral/genética
7.
BMC Genomics ; 25(1): 489, 2024 May 17.
Artículo en Inglés | MEDLINE | ID: mdl-38760729

RESUMEN

BACKGROUND: The cellular origin of hypopharyngeal diseases is crucial for further diagnosis and treatment, and the microenvironment in tissues may also be associated with specific cell types at the same time. Normal adjacent tissues (NATs) of hypopharyngeal carcinoma differ from non-tumor-bearing tissues, and can influenced by the tumor. However, the heterogeneity in kinds of disease samples remains little known, and the transcriptomic profile about biological information associated with disease occurrence and clinical outcome contained in it has yet to be fully evaluated. For these reasons, we should quickly investigate the taxonomic and transcriptomic information of NATs in human hypopharynx. RESULTS: Single-cell suspensions of normal adjacent tissues (NATs) of hypopharyngeal carcinoma were obtained and single-cell RNA sequencing (scRNA-seq) was performed. We present scRNA-seq data from 39,315 high-quality cells in the hypopharyngeal from five human donors, nine clusters of normal adjacent human hypopharyngeal cells were presented, including epithelial cells, endothelial cells (ECs), mononuclear phagocyte system cells (MPs), fibroblasts, T cells, plasma cells, B cells, mural cells and mast cells. Nonimmune components in the microenvironment, including epithelial cells, endothelial cells, fibroblasts and the subpopulations of them were performed. CONCLUSIONS: Our data provide a solid basis for the study of single-cell landscape in human normal adjacent hypopharyngeal tissues biology and related diseases.


Asunto(s)
Neoplasias Hipofaríngeas , Análisis de la Célula Individual , Transcriptoma , Microambiente Tumoral , Humanos , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/patología , Microambiente Tumoral/genética , Hipofaringe/patología , Hipofaringe/metabolismo , Perfilación de la Expresión Génica , Masculino , Análisis de Secuencia de ARN
8.
Antioxid Redox Signal ; 41(7-9): 505-521, 2024 09.
Artículo en Inglés | MEDLINE | ID: mdl-38661516

RESUMEN

Aims: Hypopharyngeal cancer (HPC) is associated with the worst prognosis of all head and neck cancers and is typically identified in an advanced stage at the time of diagnosis. While oxidative stress might contribute to the onset of HPC in patients using tobacco or alcohol, the extent of this influence and the characteristics of HPC cells in advanced stage remain to be investigated. In this study, we explored whether HPC cells survived from necrotic xenograft tumors at late stage would display increased tumor resistance along with altered tolerance to oxidative stress. Results: The remnant living HPC cells isolated from a late-stage xenograft tumor, named FaDu ex vivo cells, showed stronger chemo- and radioresistance, tumorigenesis, and invasiveness compared with parental FaDu cells. FaDu ex vivo cells also displayed increased angiogenic ability after re-transplantation in mice visualized by in vivo near infrared-II fluorescence imaging modality. Moreover, FaDu ex vivo cells exhibited significant tumor-initiating cell (TIC)-related properties accompanied by a reduction of the level of reactive oxygen species, which was associated with the upregulation of transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2). Interestingly, inhibition of Nrf2 by the RNA interference and the chemical inhibitor could reduce the TIC-related properties of FaDu ex vivo cells. Innovation: Oxidative stress potentially initiates HPC, but elevation of Nrf2-associated antioxidant mechanisms would be essential to mitigate this effect for promoting and sustaining the stemness of HPC at the advanced stage. Conclusion: Present data suggest that the antioxidant potency of advanced HPC would be a therapeutic target for the design of adjuvant treatment. Antioxid. Redox Signal. 41, 505-521.


Asunto(s)
Antioxidantes , Neoplasias Hipofaríngeas , Factor 2 Relacionado con NF-E2 , Células Madre Neoplásicas , Neovascularización Patológica , Especies Reactivas de Oxígeno , Factor 2 Relacionado con NF-E2/metabolismo , Animales , Neoplasias Hipofaríngeas/metabolismo , Neoplasias Hipofaríngeas/patología , Neoplasias Hipofaríngeas/genética , Humanos , Ratones , Neovascularización Patológica/metabolismo , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Antioxidantes/farmacología , Antioxidantes/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Estrés Oxidativo , Línea Celular Tumoral , Modelos Animales de Enfermedad , Angiogénesis
9.
Br J Cancer ; 130(10): 1725-1731, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38538728

RESUMEN

BACKGROUND: Advances in upper gastrointestinal endoscopic technology have enabled early detection and treatment of hypopharyngeal cancer. However, in-depth pharyngeal observations require sedation and are invasive. It is important to establish a minimally invasive and simple evaluation method to identify high-risk patients. METHODS: Eighty-seven patients with superficial hypopharyngeal cancer and 51 healthy controls were recruited. We assessed the methylation status of DCC, PTGDR1, EDNRB, and ECAD, in tissue and saliva samples and verified the diagnostic accuracy by methylation analyses of their promoter regions using quantitative methylation-specific PCR. RESULTS: Significant differences between cancer and their surrounding non-cancerous tissues were observed in the methylation values of DCC (p = 0.003), EDNRB (p = 0.001), and ECAD (p = 0.043). Using receiver operating characteristic analyses of the methylation values in saliva samples, DCC showed the highest area under the curve values for the detection of superficial hypopharyngeal cancer (0.917, 95% confidence interval = 0.864-0.970), compared with those for EDNRB (0.680) and ECAD (0.639). When the cutoff for the methylation values of DCC was set at ≥0.163, the sensitivity to detect hypopharyngeal cancer was 82.8% and the specificity was 90.2%. CONCLUSIONS: DCC methylation in saliva samples could be a non-invasive and efficient tool for early detection of hypopharyngeal cancer in high-risk patients.


Asunto(s)
Metilación de ADN , Neoplasias Hipofaríngeas , Saliva , Femenino , Humanos , Masculino , Biomarcadores de Tumor/genética , Estudios de Casos y Controles , Receptor DCC/genética , Detección Precoz del Cáncer/métodos , Genes DCC/genética , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/diagnóstico , Regiones Promotoras Genéticas , Receptor de Endotelina B/genética , Curva ROC , Saliva/química
10.
Carcinogenesis ; 45(8): 569-581, 2024 Aug 12.
Artículo en Inglés | MEDLINE | ID: mdl-38470063

RESUMEN

Previous studies have indicated that transmembrane protein 16A (TMEM16A) plays a crucial role in the pathogenesis and progression of various tumors by influencing multiple signaling pathways. However, the role of TMEM16A in regulating autophagy via the mammalian target of rapamycin (mTOR) pathway and its impact on the development of hypopharyngeal squamous cell carcinoma (HSCC) remain unclear. Immunohistochemistry and western blotting were used to assess the expression of TMEM16A in HSCC tissues and metastatic lymph nodes. Manipulation of TMEM16A expression levels was achieved in the FaDu cell line through overexpression or knockdown, followed by assessment of its biological effects using cell colony formation, wound healing, transwell and invasion assays. Additionally, apoptosis and autophagy-related proteins, as well as autophagosome formation, were evaluated through western blotting, transmission electron microscopy and immunofluorescence following TMEM16A knockdown or overexpression in FaDu cells. Our study revealed significantly elevated levels of TMEM16A in both HSCC tissues and metastatic lymph nodes compared with normal tissues. In vitro experiments demonstrated that silencing TMEM16A led to a notable suppression of HSCC cell proliferation, invasion and migration. Furthermore, TMEM16A silencing effectively inhibited tumor growth in xenografted mice. Subsequent investigations indicated that knockdown of TMEM16A in HSCC cells could suppress mTOR activation, thereby triggering autophagic cell death by upregulating sequestosome-1 (SQSTM1/P62) and microtubule-associated protein light chain 3 II (LC3II). This study highlights the crucial role of TMEM16A in modulating autophagy in HSCC, suggesting its potential as a therapeutic target for the treatment of this malignancy.


Asunto(s)
Anoctamina-1 , Autofagia , Movimiento Celular , Proliferación Celular , Neoplasias Hipofaríngeas , Invasividad Neoplásica , Proteínas de Neoplasias , Transducción de Señal , Serina-Treonina Quinasas TOR , Humanos , Serina-Treonina Quinasas TOR/metabolismo , Neoplasias Hipofaríngeas/patología , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/metabolismo , Animales , Ratones , Anoctamina-1/metabolismo , Anoctamina-1/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Línea Celular Tumoral , Masculino , Apoptosis , Regulación Neoplásica de la Expresión Génica , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Femenino , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/genética
11.
Hum Cell ; 37(2): 546-559, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-38280070

RESUMEN

Hypopharyngeal squamous cell carcinoma (HPSCC) has the worst prognosis among head and neck squamous cell carcinomas. The lack of available tumor cell lines poses a significant obstacle to the development of efficient treatments for HPSCC. In this study, we successfully established a novel cell line, named CZH1, from the postcricoid region of a Chinese male patient with a T3N0M0 HPSCC. Short tandem repeat analysis confirmed the uniqueness of CZH1. The cell line was characterized by its phenotypes, biomarkers, and genetics. Importantly, CZH1 cells retained the typical features of epithelial malignancy, similar to the primary tumor tissue. Furthermore, CZH1 demonstrated a greater capacity for invasion and increased susceptibility to irradiation in comparison to FaDu, which is the most commonly used HPSCC cell line. Whole-exome sequencing analysis revealed that CZH1 cells had typical genomic features of HNSCC, including mutations of TP53 and amplifications of multiple transcripts. Therefore, our newly developed CZH1 cell line could serve as an efficient tool for the in vitro investigation of the etiology, pathogenesis, and preclinical treatment of HPSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias Hipofaríngeas , Humanos , Masculino , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/terapia , Carcinoma de Células Escamosas/metabolismo , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/terapia , Neoplasias Hipofaríngeas/metabolismo , Línea Celular Tumoral
12.
Aging (Albany NY) ; 15(21): 12476-12496, 2023 11 08.
Artículo en Inglés | MEDLINE | ID: mdl-37944249

RESUMEN

Hypopharyngeal carcinoma is the most malignant type of head and neck squamous cell carcinoma, and lncRNAs play an important role in its formation and progression. However, the related specific mechanisms are rarely studied. lncRNAs closely associated with hypopharyngeal cancer were examined by lncRNA sequencing for in-depth exploration of the relationship between HOXC-AS2 and hypopharyngeal cancer pathogenesis. The mRNA expression of HOXC-AS2 and related genes was measured by qRT-PCR, and the biological function of HOXC-AS2 in hypopharyngeal carcinoma was demonstrated by gain- and loss-of-function experiments. RNA pulldown, RNA immunoprecipitation (RIP) and gene body truncation experiments and transcriptome sequencing were used to investigate the potential mechanism of HOXC-AS2 and its downstream genes, including P62, NF-KB and HMOX1. Finally, the biological function of HOXC-AS2 was confirmed in animal experiments. HOXC-AS2 and P62 expression was significantly upregulated in hypopharyngeal cancer tissues compared with normal hypopharyngeal tissues, while HMOX1 expression was decreased. Functionally, HOXC-AS2 overexpression can promote the viability, proliferation, migration and invasion of hypopharyngeal cancer cells and facilitate hypopharyngeal cancer progression. It was confirmed that HOXC-AS2 can bind to the P62 protein and activate the NF-KB signaling pathway, thereby affecting HMOX1 expression and regulating autophagy in hypopharyngeal cancer cells, ultimately regulating the formation and progression of hypopharyngeal cancer. In conclusion, our findings suggest that HOXC-AS2 regulates the progression of hypopharyngeal cancer by regulating autophagy and is abnormally highly expressed in hypopharyngeal cancer tissues. HOXC-AS2 may become a new target for the diagnosis and treatment of hypopharyngeal cancer.


Asunto(s)
Neoplasias de Cabeza y Cuello , Neoplasias Hipofaríngeas , MicroARNs , ARN Largo no Codificante , Animales , MicroARNs/metabolismo , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Línea Celular Tumoral , Neoplasias Hipofaríngeas/genética , FN-kappa B/genética , FN-kappa B/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/genética , Autofagia/genética , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Movimiento Celular/genética
13.
Cell Commun Signal ; 21(1): 292, 2023 10 18.
Artículo en Inglés | MEDLINE | ID: mdl-37853464

RESUMEN

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.


Asunto(s)
Fibroblastos Asociados al Cáncer , Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias Hipofaríngeas , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas/metabolismo , Fibroblastos Asociados al Cáncer/metabolismo , Células Endoteliales/metabolismo , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/patología , Neoplasias de Cabeza y Cuello/metabolismo , Análisis de Secuencia de ARN , Microambiente Tumoral
14.
Chin J Physiol ; 66(3): 162-170, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37322626

RESUMEN

Hypopharyngeal cancer is a subtype of the head and neck malignancies. We aimed to explore the role of lysine-specific demethylase 1 (LSD1/KDM1A) in the progression of hypopharyngeal cancer and to identify the potential mechanisms. First, LSD1 expression in head and neck squamous cell carcinoma (HNSCC) tissues and the correlation between LSD1 and the stage of HNSC were analyzed by the University of ALabama at Birmingham CANcer data analysis Portal (UALCAN). Following LSD1 silencing, proliferation of pharyngeal cancer cell line FaDu cells was evaluated by cell counting kit-8 and colony formation assays. Wounding healing and transwell assays were used to measure the capacities of migration and invasion. In addition, expression of proteins related to epithelial-to-mesenchymal transition (EMT), autophagy, and pyroptosis was tested by Western blot analysis or immunofluorescence. After treatment with autophagy inhibitor 3-methyladenine (3-MA) or NLR family pyrin domain containing 3 (NLRP3) inhibitor MCC950, the malignant biological properties were measured again. High LSD1 expression was observed in HNSC tissues, which was correlated with stage. LSD1 knockdown significantly suppressed the proliferation, migration, invasion, and EMT of hypopharyngeal cancer cells. Moreover, autophagy and pyroptosis were induced by LSD1 depletion, observed by the enhanced fluorescence intensity of LC3, gasdermin-D (GSDMD)-N, and apoptosis-associated speck-like protein containing a CARD (ASC), accompanied by upregulated expression of LC3II/LC3I, Beclin-1, NLRP3, cleaved-caspase 1, ASC, interleukin (IL)-1ß, and IL-18 and downregulated expression of p62. Importantly, 3-MA or MCC950 addition obviously reversed the inhibitory effects of LSD1 silencing on the proliferation, migration, invasion, and EMT of hypopharyngeal cancer cells. To sum up, LSD1 silencing could restrain the progression of hypopharyngeal cancer cells by inducing autophagy and pyroptosis.


Asunto(s)
Neoplasias Hipofaríngeas , Piroptosis , Humanos , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Neoplasias Hipofaríngeas/genética , Autofagia , Proliferación Celular , Histona Demetilasas/genética , Histona Demetilasas/farmacología
15.
Braz J Otorhinolaryngol ; 89(3): 401-409, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37116374

RESUMEN

OBJECTIVE: MicroRNA-29a-3p has been reported in a variety of cancers, but its role in hypopharyngeal cancer remains unclear. This study was to determine the role of microRNA-29a-3p in the occurrence and development of hypopharyngeal cancer. METHODS: 40 patients with hypopharyngeal cancer who underwent surgery in the Affiliated Hospital of Jining Medical University from April 2013 to November 2017 were selected for this study. The cancer tissue samples of the patients were collected, and the patients were followed up for three years. The expression of microRNA-29a-3p in tissue samples was detected by in situ hybridization with fluorescent probe, and the relationships among microRNA-29a-3p and clinicopathological factors, postoperative recurrent-metastasis, survival time were studied. Immunohistochemical was used to detect the expression of Ki67 and E-cadherin in tissue samples. RESULTS: Combined with HE staining results showed that microRNA-29a-3p expression was relatively high in non-cancer tissue cells (red blood cells and fibroblasts in tumor interstitial vessels), but was relatively low in cancer tissue and cells. According to the follow-up data of 40 patients with hypopharyngeal cancer, tumor size, T-stage, tumor differentiation, postoperative recurrent-metastasis of hypopharyngeal cancer patients were significantly negatively correlated with microRNA-29a-3p (p < 0.05). Immunohistochemica results further confirmed that microRNA-29a-3p was negatively correlated with the expression of Ki67 and E-cadherin. The survival time of patients positively related with microRNA-29a-3p expression (p < 0.05). Moreover, ROC curve analysis showed that the area under the curve of the combined detection of miRNA-29a-3p+Ki67+E-cadherin was larger than that of the single detection of the three indexes. CONCLUSIONS: The expression of microRNA-29a-3p is closely related to the occurrence, development and prognosis of hypopharyngeal cancer, and it affects the proliferation and invasion. This indicates that microRNA-29a-3p serves as a therapeutic target for the occurrence and development of hypopharyngeal cancer. The evidence of study designs of this study is IV using "Oxford Centre for Evidence-Based Medicine 2011 Levels of Evidence".


Asunto(s)
Neoplasias Hipofaríngeas , MicroARNs , Humanos , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/cirugía , Relevancia Clínica , Antígeno Ki-67 , Cadherinas/genética
16.
Nat Commun ; 14(1): 1466, 2023 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-36928331

RESUMEN

The efficacy of the first-line treatment for hypopharyngeal carcinoma (HPC), a predominantly male cancer, at advanced stage is only about 50% without reliable molecular indicators for its prognosis. In this study, HPC biopsy samples collected before and after the first-line treatment are classified into different groups according to treatment responses. We analyze the changes of HPC tumor microenvironment (TME) at the single-cell level in response to the treatment and identify three gene modules associated with advanced HPC prognosis. We estimate cell constitutions based on bulk RNA-seq of our HPC samples and build a binary classifier model based on non-malignant cell subtype abundance in TME, which can be used to accurately identify treatment-resistant advanced HPC patients in time and enlarge the possibility to preserve their laryngeal function. In summary, we provide a useful approach to identify gene modules and a classifier model as reliable indicators to predict treatment responses in HPC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Hipofaríngeas , Humanos , Masculino , Femenino , Carcinoma de Células Escamosas/patología , Transcriptoma/genética , Microambiente Tumoral/genética , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/terapia , Neoplasias Hipofaríngeas/patología
17.
Sci Rep ; 13(1): 107, 2023 01 03.
Artículo en Inglés | MEDLINE | ID: mdl-36596842

RESUMEN

The research on targeted therapy of hypopharyngeal cancer is very scarce. The discovery of new targeted driver genes will promote the progress of hypopharyngeal cancer therapy to a great extent. In our research, whole-exome sequencing in 10 patients with hypopharyngeal cancer was performed to identify single nucleotide variations (SNVs) and insertions and deletions (INDELs). American College of Medical Genetics and Genomics (ACMG) guidelines were used to evaluate the pathogenicity of the selected variants. 8113 mutation sites in 5326 genes were identified after strict screening. We identified 72 pathogenic mutations in 53 genes according to the ACMG guidelines. Gene Ontology (GO) annotation and KEGG enrichment analysis show the effect of these genes on cancer. Protein-protein interaction (PPI) was analyzed by string online software. The validation results of the ualcan database showed that 22 of the 53 genes may be related to the poor prognosis of patients with hypopharyngeal cancer. RBM20 has the most significant correlation with hypopharyngeal cancer, and it is likely to be the driver gene of hypopharyngeal cancer. In conclusion, we found possible therapeutic targets for hypopharyngeal cancer, especially RBM20 and KMT2C. Our study provides a basis for the pathogenesis and targeted therapy of hypopharyngeal cancer.


Asunto(s)
Neoplasias Hipofaríngeas , Humanos , Secuenciación del Exoma , Neoplasias Hipofaríngeas/genética , Detección Precoz del Cáncer , Mutación , Genómica
18.
Cancer Gene Ther ; 30(3): 462-471, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36460803

RESUMEN

Hypopharyngeal carcinoma is a cancer with the worst prognosis. We constructed the first single-cell transcriptome map for hypopharyngeal carcinoma and explored its underlying mechanisms. We systematically studied single-cell transcriptome data of 17,599 cells from hypopharyngeal carcinoma and paracancerous tissues. We identified categories of cells by dimensionality reduction and performed further subgroup analysis. Focusing on the potential mechanism in the cellular communication of hypopharyngeal carcinoma, we predicted ligand-receptor interactions and verified them via immunohistochemical and cellular experiments. In total, seven cell types were identified, including epithelial and myeloid cells. Subsequently, subgroup analysis showed significant tumor heterogeneity. Based on the pathological type of squamous cell carcinoma, we focused on intercellular communication between epithelial cells and various cells. We predicted the crosstalk and inferred the regulatory effect of cellular active ligands on the surface receptor of epithelial cells. From the top potential pairs, we focused on the BMPR2 receptor for further research, as it showed significantly higher expression in epithelial cancer tissue than in adjacent tissue. Further bioinformatics analysis, immunohistochemical staining, and cell experiments also confirmed its cancer-promoting effects. Overall, the single-cell perspective revealed complex crosstalk in hypopharyngeal cancer, in which BMPR2 promotes its proliferation and migration, providing a rationale for further study and treatment of this carcinoma.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Hipofaríngeas , Humanos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/terapia , Carcinoma de Células Escamosas/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/terapia , Neoplasias Hipofaríngeas/metabolismo , Inmunoterapia , Pronóstico , Análisis de Expresión Génica de una Sola Célula
19.
J Cell Mol Med ; 26(21): 5551-5561, 2022 11.
Artículo en Inglés | MEDLINE | ID: mdl-36254814

RESUMEN

Several members of the transmembrane protein family are associated with the biological processes of human malignancies; however, the expression pattern and biological function of one family member, TMEM184B, in hypopharyngeal squamous cell carcinoma (HPSCC) are not fully understood. The expression between HPSCC tumours and adjacent normal tissues was determined by the Immunohistochemistry (IHC). A bioinformatics analysis was performed to verify the expression pattern of TMEM184B in The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) databases. Furthermore, in vitro assays on cell proliferation, invasion, migration and in vivo experiments on tumour growth and apoptosis of TMEM184B in HPSCC were performed. We found that the HPSCC tissues had a significantly higher expression of TMEM184B than the adjacent normal tissues. Bioinformatics analysis confirmed the different expression of TMEM184B expression in HPSCC. Furthermore, in vitro and in vivo experiments demonstrated that TMEM184B promotes HPSCC cell growth, cell invasion and migration in FaDu cells, whereas flow cytometry assay showed that TMEM184B inhibited cell apoptosis. Our study revealed for the first time that TMEM184B might serve an oncogenic function in HPSCC and could be a potential diagnostic biomarker and therapeutic target for HPSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias Hipofaríngeas , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Neoplasias Hipofaríngeas/genética , Carcinoma de Células Escamosas/patología , Regulación Neoplásica de la Expresión Génica , Apoptosis/genética , Proliferación Celular/genética , Neoplasias de Cabeza y Cuello/genética , Movimiento Celular/genética , Línea Celular Tumoral
20.
Sci Rep ; 12(1): 17751, 2022 10 22.
Artículo en Inglés | MEDLINE | ID: mdl-36273237

RESUMEN

CD271 (also referred to as nerve growth factor receptor or p75NTR) is expressed on cancer stem cells in hypopharyngeal cancer (HPC) and regulates cell proliferation. Because elevated expression of CD271 increases cancer malignancy and correlates with poor prognosis, CD271 could be a promising therapeutic target; however, little is known about the induction of CD271 expression and especially its promoter activity. In this study, we screened transcription factors and found that RELA (p65), a subunit of nuclear factor kappaB (NF-κB), is critical for CD271 transcription in cancer cells. Specifically, we found that RELA promoted CD271 transcription in squamous cell carcinoma cell lines but not in normal epithelium and neuroblastoma cell lines. Within the CD271 promoter sequence, region + 957 to + 1138 was important for RELA binding, and cells harboring deletions in proximity to the + 1045 region decreased CD271 expression and sphere-formation activity. Additionally, we found that clinical tissue samples showing elevated CD271 expression were enriched in RELA-binding sites and that HPC tissues showed elevated levels of both CD271 and phosphorylated RELA. These data suggested that RELA increases CD271 expression and that inhibition of RELA binding to the CD271 promoter could be an effective therapeutic target.


Asunto(s)
Neoplasias Hipofaríngeas , Humanos , Adapaleno , Proliferación Celular/genética , Neoplasias Hipofaríngeas/genética , Neoplasias Hipofaríngeas/metabolismo , Neoplasias Hipofaríngeas/patología , FN-kappa B/genética , FN-kappa B/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo , Factor de Transcripción ReIA/genética , Factor de Transcripción ReIA/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA