Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.823
Filtrar
1.
Clin Chim Acta ; 564: 119916, 2025 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-39153653

RESUMEN

Laryngeal cancer remains a significant global health concern, with poor prognosis for advanced-stage disease highlighting the need for novel diagnostic, prognostic, and therapeutic approaches. Circular RNAs (circRNAs), a class of covalently closed non-coding RNAs, have emerged as important regulators of gene expression and cellular processes in various cancers, including laryngeal cancer. This review summarizes the current understanding of circRNAs in laryngeal cancer, covering their biogenesis, regulatory mechanisms, and potential clinical applications. We explore the diverse functions of circRNAs, including their roles as miRNA sponges, protein interactors, and direct mRNA regulators, and their influence on key cellular processes such as proliferation, invasion, and metastasis. The review highlights promising circRNAs as diagnostic and prognostic biomarkers, as well as potential therapeutic targets. We also outline current strategies for circRNA modulation, including suppression techniques like RNA interference and CRISPR/Cas systems, and overexpression methods using vectors and synthetic circRNAs.


Asunto(s)
Neoplasias Laríngeas , ARN Circular , Humanos , ARN Circular/genética , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/diagnóstico , Biomarcadores de Tumor/genética
2.
Head Face Med ; 20(1): 62, 2024 Oct 26.
Artículo en Inglés | MEDLINE | ID: mdl-39456084

RESUMEN

BACKGROUND: Activated regulatory T cells (aTregs) play a vital role in promoting a tumor immunosuppressive microenvironment in laryngeal squamous cell carcinoma (LSCC). However, the regulatory factors that induce the generation of aTregs are not clear. Herein, we investigated the effect of amphiregulin (AREG) on the production of aTregs in the tumor microenvironment of LSCC. METHODS: Immunohistochemical (IHC) analysis was conducted to examine the expression of AREG and FOXP3, and their association with clinical parameters and patient outcomes was demonstrated. The expression level of EGFRs in three functional subsets of Tregs was assessed, and the induction of CD4+ T cells into aTregs in the presence or absence of AREG or Gefitinib was analyzed using flow cytometry. RESULTS: Our results showed a higher expression level of AREG was significantly related to advanced clinical stage and worse survival, particularly with increased infiltration of Tregs in LSCC tumor tissue. The in vitro study showed that AREG significantly promoted the differentiation of aTregs, and enhanced the inhibitory effect of Tregs on T cell proliferation, which could be reversed by epidermal growth factor receptor (EGFR) inhibitors. In addition, we found that EGFR was highly expressed in aTregs, but not in other subsets of Tregs. It is suggested that AREG might induce aTregs, and enhance the immunosuppressive function of Tregs via the AREG/EGFR signal pathway. CONCLUSIONS: Collectively, this study revealed the role and mechanism of AREG in negative immune regulation, and targeting AREG might be a novel immunotherapy for LSCC.


Asunto(s)
Anfirregulina , Carcinoma de Células Escamosas , Receptores ErbB , Neoplasias Laríngeas , Linfocitos T Reguladores , Microambiente Tumoral , Anfirregulina/metabolismo , Humanos , Linfocitos T Reguladores/inmunología , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/inmunología , Neoplasias Laríngeas/genética , Receptores ErbB/metabolismo , Masculino , Femenino , Persona de Mediana Edad , Microambiente Tumoral/inmunología , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/inmunología , Carcinoma de Células Escamosas/genética , Transducción de Señal , Anciano , Inmunohistoquímica , Citometría de Flujo
3.
Cancer Med ; 13(20): e70312, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39428922

RESUMEN

BACKGROUND: Laryngeal squamous cell carcinoma (LSCC) is a type of cancer of the respiratory tract that often presents with subtle symptoms at the early stage and is susceptible to recurrence and metastasis. MATERIALS AND METHODS: To find out key regulatory genes involved in LSCC development, we downloaded LSCC-related sequencing datasets for bioinformatics analysis. WGCNA was performed on GSE142083 and differential analysis was conducted on GSE51985 and TCGA-HNSC. Intersection genes were taken from the above three datasets. To confirm the function of genes, we overexpressed and knocked down genes in cells and treated them with autophagy agonist Rapamycin and PI3K-AKT pathway inhibitor. At the cellular level, the expression of CH25H, autophagy-related proteins (LC3 I, LC3 II, p62, and Beclin 1), and PI3K-AKT pathway-related proteins (PI3K, AKT, and p-AKT) were assessed via Western blot; the mRNA level of CH25H was evaluated through qRT-PCR; the cell activity was examined by CCK8; the apoptosis was assessed through flow cytometry; and the cell migration and invasion were assessed through wound healing and Transwell assays. RESULTS: Through bioinformatics analysis, we screened 7 genes (CH25H, NELL2, STC2, TMEM158, ZIC2, HOXD11, and HOXD10). Ultimately, CH25H was selected for follow-up experiments. By detecting CH25H expression in human immortalized keratinocytes (HaCaT) and LSCC cells (Tu-686, SNU899, and AMC-HN-8), it was found out that CH25H expression was higher in HaCaT cells than in LSCC cells. To elucidate the role of CH25H in LSCC development, we overexpressed CH25H in Tu-686 cells and downregulated its expression in AMC-HN-8 cells. CH25H was revealed to reduce the proliferation, activity, invasion, and migration of LSCC cells while increasing their apoptosis levels. Significant changes were also observed in the expressions of autophagy- and PI3K-AKT pathway-related proteins. To further investigate the roles of autophagy and the PI3K-AKT pathway in LSCC development, we respectively employed autophagy agonists and inhibitors targeting the PI3K-AKT pathway to intervene the cells, and found that CH25H regulated the PI3K-AKT pathway to promote autophagy, thus enhancing the apoptosis of LSCC cells. We further investigated CH25H's impact on tumor growth, autophagy, and the PI3K-AKT pathway at the animal level and found that CH25H promoted autophagy of LSCC cells and inhibited the PI3K-AKT pathway, and ultimately inhibiting the progression of LSCC. CONCLUSIONS: In summary, CH25H promotes autophagy and affects the malignant progression of LSCC through the PI3K-AKT pathway.


Asunto(s)
Autofagia , Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Laríngeas , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Humanos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Autofagia/genética , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Apoptosis , Progresión de la Enfermedad , 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
4.
Int J Mol Sci ; 25(17)2024 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-39273632

RESUMEN

This article describes how the transcriptional alterations of the innate immune system divide dysplasias into aggressive forms that, despite the treatment, relapse quickly and more easily, and others where the progression is slow and more treatable. It elaborates on how the immune system can change the extracellular matrix, favoring neoplastic progression, and how infections can enhance disease progression by increasing epithelial damage due to the loss of surface immunoglobulin and amplifying the inflammatory response. We investigated whether these dysregulated genes were linked to disease progression, delay, or recovery. These transcriptional alterations were observed using the RNA-based next-generation sequencing (NGS) panel Oncomine Immune Response Research Assay (OIRRA) to measure the expression of genes associated with lymphocyte regulation, cytokine signaling, lymphocyte markers, and checkpoint pathways. During the analysis, it became apparent that certain alterations divide dysplasia into two categories: progressive or not. In the future, these biological alterations are the first step to provide new treatment modalities with different classes of drugs currently in use in a systemic or local approach, including classical chemotherapy drugs such as cisplatin and fluorouracile, older drugs like fenretinide, and new checkpoint inhibitor drugs such as nivolumab and pembrolizumab, as well as newer options like T cell therapy (CAR-T). Following these observed alterations, it is possible to differentiate which dysplasias progress or not or relapse quickly. This information could, in the future, be the basis for determining a close follow-up, minimizing surgical interventions, planning a correct and personalized treatment protocol for each patient and, after specific clinical trials, tailoring new drug treatments.


Asunto(s)
Transcriptoma , Humanos , Perfilación de la Expresión Génica/métodos , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patología , Secuenciación de Nucleótidos de Alto Rendimiento
5.
FASEB J ; 38(17): e70048, 2024 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-39259536

RESUMEN

The sensitivity of laryngeal squamous cell carcinoma (LSCC) to chemotherapy shows large heterogeneity. The role of miRNA in small extracellular vesicles (sEV) in chemotherapy resistance is under investigation. However, the regulation and sorting mechanism of sEV miRNAs remains unclear. In this study, small RNA sequencing was used to explore miRNA expression profiles in sEV of LSCC after cisplatin stimulation; RNA pull-down, mass spectrometry, and EMSA were used to clarify the binding of candidate RNA-binding protein (RBP) and candidate miRNA. Immunostaining and microRNA fluorescence in situ hybridization were performed to identify how candidate RBP affects miRNA stability and nuclear/cytoplasmic distribution. In vivo experiments were performed to verify the biological functions and response to cisplatin of candidate RBP. We found that cisplatin stimulation induced increased expression of miR-148a-3p and sEV sorting. ANXA11 binds to miR-148a-3p in a sequence-specific manner. ANXA11 inhibits tumor cell proliferation and drug resistance by binding to and retaining miR-148a-3p. Cisplatin stimulation reduced ANXA11 expression and promoted miR-148a-3p efflux through sEV pathways. ANXA11 overexpression reduced in vivo tumor proliferation and cisplatin-resistance. Taken together, ANXA11 mediates cisplatin resistance through sEV miRNA resorting. Mechanically, ANXA11 binds to miR-148a-3p in a sequence-specific manner to regulate its resorting and thus influences tumor proliferation and chemoresistance.


Asunto(s)
Cisplatino , Resistencia a Antineoplásicos , Vesículas Extracelulares , Neoplasias Laríngeas , Ratones Desnudos , MicroARNs , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Anexinas/metabolismo , Anexinas/genética , Antineoplásicos/farmacología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Vesículas Extracelulares/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/tratamiento farmacológico , Neoplasias Laríngeas/patología , Ratones Endogámicos BALB C , MicroARNs/metabolismo , MicroARNs/genética , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética
6.
Int J Mol Sci ; 25(18)2024 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-39337665

RESUMEN

According to recent research, inflammatory STAT4 and its protein impact may be important factors in developing cancerous diseases. Still unanalyzed is this effect in patients with laryngeal squamous cell carcinoma (LSCC). In the present study, we evaluated four single nucleotide variants (SNVs) of STAT4 (rs10181656, rs7574865, rs7601754, and rs10168266) and STAT4 serum levels to determine their link between LSCC development and its clinical manifestations. A total of 632 men (324 LSCC patients and 338 healthy individuals) were involved in this study. The genotyping was carried out using real-time PCR. Additionally, we measured 80 study subjects' (40 LSCC patients and 40 control subjects) STAT4 protein concentrations using an enzyme-linked immunosorbent assay (ELISA). In our study, the T allele of STAT4 rs7574865 significantly increases the likelihood of LSCC occurrence by 1.4-fold. Additionally, this SNV is associated with higher odds of early-stage disease, T1 size LSCC development, absence of metastasis to neck lymph nodes, and well-differentiated carcinoma. The G allele of rs10181656 is significantly associated with various clinical characteristics of LSCC, increasing the odds of early- and advanced-stage disease by 2.8-fold and 1.9-fold, respectively. Additionally, this allele is linked to an increased likelihood of developing tumors of different sizes and non-metastasized LSCC, as well as poorly differentiated carcinoma, highlighting its potential impact on the development and features of LSCC. Conclusion: The analysis of the STAT4 rs7574865 SNV revealed that the G allele is linked to a more favorable prognosis in LSCC. Additionally, it is hypothesized that the G allele of rs10181656 may be associated with the occurrence of LSCC but may not serve as a sensitive prognostic biomarker for distinguishing between disease stages, cell differentiation, or tumor size.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias Laríngeas , Polimorfismo de Nucleótido Simple , Factor de Transcripción STAT4 , Humanos , Factor de Transcripción STAT4/genética , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patología , Masculino , Persona de Mediana Edad , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Anciano , Predisposición Genética a la Enfermedad , Alelos , Estudios de Casos y Controles , Adulto , Genotipo , Biomarcadores de Tumor/genética , 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/sangre , Estadificación de Neoplasias , Femenino
7.
Eur J Pharmacol ; 983: 176991, 2024 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-39265883

RESUMEN

BACKGROUND: Laryngocarcinoma is a common malignancy in the upper respiratory tract. Enabled homolog (ENAH) is an actin-binding protein that is associated with the development of various cancers. However, its role and mechanism in laryngocarcinoma remain unknown. METHODS: The ENAH level in laryngocarcinoma was examined in silico, in vitro and in vivo. The prognostic analysis of the ENAH level was assessed on laryngocarcinoma patients. Gain- and loss-of-function assays were conducted in AMC-HN-8 and TU686 cells. Sh-ENAH-containing AMC-HN-8 cells were implanted into naked mice. The role and mechanism of ENAH in laryngocarcinoma were investigated by CCK-8, transwell, immunofluorescence, dual luciferase, RT-qPCR, immunohistochemistry, and western blotting experiments. RESULTS: The ENAH level was upregulated in laryngocarcinoma, which predicted a poor prognosis in laryngocarcinoma patients. Gain- and loss-of-function results showed that ENAH promoted proliferation, invasion and EMT of laryngocarcinoma cells. Moreover, ENAH was transcriptionally activated by YY1, and YY1/ENAH axis enhanced these malignant progresses of laryngocarcinoma cells. Besides, ENAH activated the PI3K/AKT pathway, and 740Y-P abolished the accelerative role of ENAH in proliferation, invasion and EMT of laryngocarcinoma cells. Furthermore, knockdown of ENAH reduced tumor size and weight, and the expression level of vimentin and PI3K/AKT pathway in tumor-bearing mice. CONCLUSION: ENAH transcriptionally activated by YY1 promotes cell growth, invasion and EMT of laryngocarcinoma through the activation of PI3K/AKT signaling.


Asunto(s)
Proliferación Celular , Neoplasias Laríngeas , Proteínas de Microfilamentos , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Transducción de Señal , Factor de Transcripción YY1 , Animales , Femenino , Humanos , Masculino , Ratones , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Transición Epitelial-Mesenquimal/genética , Regulación Neoplásica de la Expresión Génica , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Ratones Desnudos , Proteínas de Microfilamentos/genética , Proteínas de Microfilamentos/metabolismo , Invasividad Neoplásica , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Activación Transcripcional , Factor de Transcripción YY1/metabolismo , Factor de Transcripción YY1/genética
8.
Cell Signal ; 122: 111336, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-39121975

RESUMEN

Laryngeal squamous cell carcinoma (LSCC) is a prevalent human cancer with a complex pathogenesis that remains incompletely understood. Here, we unveil a long non-coding RNA (lncRNA) associated with LSCC tumorigenesis and progression. LOC730101 exhibits significant overexpression in human LSCC tissues, and elevated LOC730101 levels correlate with malignant clinicopathological characteristics. Moreover, we demonstrate that LOC730101 is encapsulated into exosomes in an hnRNPA2B1-dependent manner, serving as a promising plasma biomarker for discriminating LSCC patients from healthy individuals (AUC = 0.92 with 89.36% sensitivity and 86.36% specificity). Exosomes derived from LSCC cells enhance the viability, DNA synthesis rate, and invasiveness of normal nasopharynx epithelial cells, with pronounced effects observed upon LOC730101 overexpression. Additionally, exosomal LOC730101 promotes tumor growth in vivo. Mechanistically, exosomal LOC730101 internalization by normal nasopharynx epithelial cells leads to increased H3K4me3 levels on the p38 MAPK gamma (p38γ) promoter via direct interaction with hnRNPA2B1. This interaction activates p38γ transcription, ultimately driving LSCC tumorigenesis. Collectively, our findings uncover a novel exosomal lncRNA that mediates communication between normal and LSCC cells during LSCC carcinogenesis, suggesting that targeting LOC730101 may represent a promising therapeutic strategy for LSCC treatment.


Asunto(s)
Carcinogénesis , Exosomas , Neoplasias Laríngeas , ARN Largo no Codificante , Animales , Femenino , Humanos , Masculino , Ratones , Persona de Mediana Edad , Carcinogénesis/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Proliferación Celular , Exosomas/metabolismo , Regulación Neoplásica de la Expresión Génica , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo A-B/genética , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/genética , Ratones Endogámicos BALB C , Ratones Desnudos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Proteína Quinasa 12 Activada por Mitógenos
9.
Int Immunopharmacol ; 140: 112742, 2024 Oct 25.
Artículo en Inglés | MEDLINE | ID: mdl-39126735

RESUMEN

This study investigates the role of M2-exo-mediated HOXC13-AS in laryngeal squamous cell carcinoma (LSCC) by examining its transmission to tumor microenvironment (TME) macrophages. Exosomes from M2 macrophages were isolated and characterized using transmission electron microscopy, nanoparticle tracer analysis and western blot. Expression of HOXC13-AS, miR-485-5p, IGF2BP2, and PD-L1 was analyzed. Different interventions on LSCC cell function and immune escape were detected using molecular biological techniques. The study found that elevated HOXC13-AS were present in LSCC, and M2-exo expression was significantly increased in LSCC cells. Silencing HOXC13-AS in M2-exo inhibited LSCC malignant progression and immune escape in vivo and in vitro. M2-exo-mediated HOXC13-AS also regulated IGF2BP2 expression, impacting cellular biological function and immune escape process. The study concludes that M2-exo-mediated HOXC13-AS promotes LSCC malignancy and immune escape.


Asunto(s)
Exosomas , Proteínas de Homeodominio , Neoplasias Laríngeas , Macrófagos , ARN Largo no Codificante , Microambiente Tumoral , Animales , Humanos , Ratones , Antígeno B7-H1/metabolismo , Antígeno B7-H1/genética , Línea Celular Tumoral , Exosomas/metabolismo , Exosomas/genética , Regulación Neoplásica de la Expresión Génica , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Neoplasias Laríngeas/inmunología , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patología , Macrófagos/inmunología , Ratones Desnudos , MicroARNs/genética , MicroARNs/metabolismo , Proteínas de Unión al ARN/metabolismo , Proteínas de Unión al ARN/genética , Escape del Tumor/genética , Microambiente Tumoral/inmunología , ARN Largo no Codificante/genética
10.
Medicine (Baltimore) ; 103(33): e39231, 2024 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-39151523

RESUMEN

Nursing and physical examination early screening of multiple tumors is helpful to find tumors early, so as to improve the cure rate. Studying its molecular mechanisms is urgent. By logging into gene expression omnibus database, we found laryngeal cancer dataset GSE127165, bladder cancer dataset GSE65635, oral cancer dataset GSE146483, obtain differentially expressed genes, subsequently, weighted gene co-expression network analysis, protein-protein interaction networks, functional enrichment analysis, immune infiltration analysis, survival analysis, comparative toxicogenomics database analysis were conducted. Draw a heatmap of gene expression. Use targetScan to search for miRNA information about core DEG. Got 53 differentially expressed genes. In GOKEGG analysis, they were clustered in cell cycle processes, spindle poles, and protein serine/threonine/tyrosine kinase activity cell cycle, transcriptional dysregulation in cancer, RIG-I-like receptor signaling pathway, P53 signaling pathway. Protein-protein interaction analysis screened out 5 genes (NEK2, BUB1, HMMR, TTK, CCNB2). Cyclin B2 (CCNB2) and budding uninhibited by benzimidazole 1 (BUB1) were highly expressed in laryngeal cancer, bladder cancer, oral cancer. Comparative toxicogenomics database analysis found that core genes (CCNB2, BUB1) are associated with tumors, necrosis, and inflammation. Related miRNA of CCNB2 gene is hsa-miR-670-3p; related miRNAs of BUB1 gene are hsa-miR-5688, hsa-miR-495-3p. CCNB2 and BUB1 exhibit high expression in laryngeal cancer, bladder cancer, and oral cancer, suggesting their potential as molecular targets for precision therapy in these cancers.


Asunto(s)
Biomarcadores de Tumor , Detección Precoz del Cáncer , Humanos , Biomarcadores de Tumor/genética , Detección Precoz del Cáncer/métodos , Mapas de Interacción de Proteínas/genética , Examen Físico , MicroARNs/genética , Neoplasias de la Vejiga Urinaria/genética , Neoplasias de la Vejiga Urinaria/diagnóstico , Neoplasias de la Vejiga Urinaria/patología , Neoplasias de la Boca/genética , Neoplasias de la Boca/diagnóstico , Neoplasias Laríngeas/genética , Regulación Neoplásica de la Expresión Génica , Perfilación de la Expresión Génica , Neoplasias/genética
11.
Biochem Biophys Res Commun ; 730: 150366, 2024 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-38991254

RESUMEN

Laryngeal squamous cell carcinoma (LSCC) with a high incidence and mortality rate, has a serious impact worldwide. Phosphofructokinase-1 liver type (PFKL) is a major enzyme in glycolysis progress, but its role in modulating tumorigenesis and cisplatin (DDP) chemosensitivity of LSCC was still unclear. The mRNA and protein levels of PFKL were detected by qRT-PCR and immunohistochemical assay. Cell Counting Kit-8 assay and flow cytometry were carried out to observe cell viability, as well as apoptosis and mitochondrial reactive oxygen species (mito-ROS). Extracellular acidification rate and lactate content were measured using extracellular flux analysis and lactate assay kit. Immunofluorescent staining was used to evaluate the expression of γ-H2AX foci. DNA damage was detected via single-cell gel electrophoresis. Western blotting was introduced to evaluate the protein level of PFKL, LDHA, γ-H2AX, cleaved PARP, H3K27ac, and H3K9ac. Mice xenograft model of LSCC was built for in vivo validation. The PFKL expression was significantly increased in LSCC and associated with poor survival of LSCC patients. Knockdown of PFKL in LSCC cells significantly inhibited cell viability, ECAR, lactate content, and LDHA expression, but promoted mito-ROS level. Furthermore, knockdown of PFKL enhanced response of LSCC cells to DDP by increasing DDP-induced apoptosis, promoting DDP-induced mito-ROS level, γ-H2AX foci, tail DNA, and the expression of γ-H2AX and cleaved PARP. However, the overexpression of PFKL in LSCC cells had opposite experimental results. Nude mice tumor formation experiment proved that downregulation of PFKL significantly enhanced response of cells to DDP, demonstrated by reduced tumor volume, weight and increased TUNEL-positive cells. Suppression of CBP/EP300-mediated PFKL transcription inhibited cell viability and glycolysis and promoted mito-ROS in LSCC. PFKL promotes cell viability and DNA damage repair in DDP-treated LSCC through regulation of glycolysis pathway.


Asunto(s)
Antineoplásicos , Supervivencia Celular , Cisplatino , Glucólisis , Neoplasias Laríngeas , Ratones Desnudos , Cisplatino/farmacología , Glucólisis/efectos de los fármacos , Humanos , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/tratamiento farmacológico , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/genética , Animales , Supervivencia Celular/efectos de los fármacos , Ratones , Línea Celular Tumoral , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Fosfofructoquinasa-1/metabolismo , Fosfofructoquinasa-1/genética , Resistencia a Antineoplásicos/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/genética , Ratones Endogámicos BALB C , Daño del ADN/efectos de los fármacos
12.
Medicine (Baltimore) ; 103(27): e38362, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38968481

RESUMEN

Laryngeal carcinoma (LC) is reported to have a higher incidence rate among all types of head and neck cancers around the globe. Mechanisms resulting in the pathogenesis of LC are complicated due to involvement of invasion and metastasis and there is a need to understand this complicated multistep process. Numerous molecules including matrix metalloproteinases (MMPs) are involved in regulating metastatic mechanisms. Furthermore, activation and expression of different classes of MMPs have been observed in multiple pathological and physiological events including inflammation, invasion, and metastasis. Among all members of MMPs, matrix metalloproteinases-2 (MMP-2), and matrix metalloproteinases-9 (MMP-9) have been frequently reported to correlate with tumor pathogenesis. The present study is designed to check the involvement of MMP-2 and MMP-9 in LC pathogenesis. 184 laryngeal tumor samples along with adjacent uninvolved healthy sections were collected to check the expression deregulation of the above-mentioned gene in LC using real-time PCR and immunohistochemistry (IHC). Real-time PCR and IHC analyses showed the significant upregulation of MMP-2 (P < .0001) and MMP-9 (P < .0001) genes in laryngeal tumors compared to controls. Spearman correlation showed the positive correlation of expression deregulation of selected MMPs with advanced TNM stage [MMP-2, (P < .0001); MMP-9, P < .0001] and smoking status [MMP-2 (P < .0001); MMP-9 P < .0001] in laryngeal pathogenesis. Receiver operating curve (ROC) analysis showed the good diagnostic/prognostic value of said markers in laryngeal cancer patients. The present study showed that significant upregulation of selected MMPs was found associated with an increased risk of laryngeal cancer and can act as good diagnostic markers for the detection of said disease.


Asunto(s)
Neoplasias Laríngeas , Metaloproteinasa 2 de la Matriz , Metaloproteinasa 9 de la Matriz , Humanos , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/genética , Metaloproteinasa 2 de la Matriz/genética , Metaloproteinasa 2 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Metaloproteinasa 9 de la Matriz/genética , Estudios Retrospectivos , Masculino , Persona de Mediana Edad , Femenino , Anciano , Adulto , Biomarcadores de Tumor/metabolismo , Biomarcadores de Tumor/genética , Estadificación de Neoplasias , Inmunohistoquímica , Reacción en Cadena en Tiempo Real de la Polimerasa , Regulación Neoplásica de la Expresión Génica , Regulación hacia Arriba
13.
J Transl Med ; 22(1): 647, 2024 Jul 10.
Artículo en Inglés | MEDLINE | ID: mdl-38987822

RESUMEN

BACKGROUND: The growing understanding of cancer biology and the establishment of new treatment modalities has not yielded the expected results in terms of survival for Laryngeal Squamous Cell Cancer (LSCC). Early diagnosis, as well as prompt identification of patients with high risk of relapse would ensure greater chance of therapeutic success. However, this goal remains a challenge due to the absence of specific biomarkers for this neoplasm. METHODS: Serum samples from 45 LSCC patients and 23 healthy donors were collected for miRNA expression profiling by TaqMan Array analysis. Additional 20 patients and 42 healthy volunteers were included for the validation set, reaching an equal number of clinical samples for each group. The potential diagnostic ability of the such identified three-miRNA signature was confirmed by ROC analysis. Moreover, each miRNA was analyzed for the possible correlation with HNSCC patients' survival and TNM status by online databases Kaplan-Meier (KM) plotter and OncomiR. In silico analysis of common candidate targets and their network relevance to predict shared biological functions was finally performed by PANTHER and GeneMANIA software. RESULTS: We characterized serum miRNA profile of LSCC patients identifying a novel molecular signature, including miR-223, miR-93 and miR-532, as circulating marker endowed with high selectivity and specificity. The oncogenic effect and the prognostic significance of each miRNA was investigated by bioinformatic analysis, denoting significant correlation with OS. To analyse the molecular basis underlying the pro-tumorigenic role of the signature, we focused on the simultaneously regulated gene targets-IL6ST, GTDC1, MAP1B, CPEB3, PRKACB, NFIB, PURB, ATP2B1, ZNF148, PSD3, TBC1D15, PURA, KLF12-found by prediction tools and deepened for their functional role by pathway enrichment analysis. The results showed the involvement of 7 different biological processes, among which inflammation, proliferation, migration, apoptosis and angiogenesis. CONCLUSIONS: In conclusion, we have identified a possible miRNA signature for early LSCC diagnosis and we assumed that miR-93, miR-223 and miR-532 could orchestrate the regulation of multiple cancer-related processes. These findings encourage the possibility to deepen the molecular mechanisms underlying their oncogenic role, for the desirable development of novel therapeutic opportunities based on the use of short single-stranded oligonucleotides acting as non-coding RNA antagonists in cancer.


Asunto(s)
Carcinoma de Células Escamosas , Biología Computacional , Detección Precoz del Cáncer , Regulación Neoplásica de la Expresión Génica , Neoplasias Laríngeas , MicroARNs , Humanos , Neoplasias Laríngeas/sangre , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/diagnóstico , MicroARNs/sangre , MicroARNs/genética , Masculino , Femenino , Carcinoma de Células Escamosas/sangre , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/diagnóstico , Persona de Mediana Edad , Perfilación de la Expresión Génica , Curva ROC , Biomarcadores de Tumor/sangre , Biomarcadores de Tumor/genética , Estimación de Kaplan-Meier , Estudios de Casos y Controles , Redes Reguladoras de Genes , Anciano
14.
Biol Direct ; 19(1): 57, 2024 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-39039611

RESUMEN

Laryngeal carcinoma (LC) is a common cancer of the respiratory tract. This study aims to investigate the role of RNA-binding motif protein 15 (RBM15) in the cisplatin (DDP) resistance of LC cells. LC-DDP-resistant cells were constructed. RBM15, lysine-specific demethylase 5B (KDM5B), lncRNA Fer-1 like family member 4 (FER1L4), lncRNA KCNQ1 overlapping transcript 1 (KCNQ1OT1), glutathione peroxidase 4 (GPX4), and Acyl-CoA synthetase long-chain family (ACSL4) was examined. Cell viability, IC50, and proliferation were assessed after RBM15 downregulation. The enrichment of insulin-like growth factor 2 mRNA-binding protein 3 (IGF2BP3) and N6-methyladenosine (m6A) on KDM5B was analyzed. KDM5B mRNA stability was measured after actinomycin D treatment. A tumor xenograft assay was conducted to verify the role of RBM15 in LC. Results showed that RBM15 was upregulated in LC and its knockdown decreased IC50, cell viability, proliferation, glutathione, and upregulated iron ion content, ROS, malondialdehyde, ACSL4, and ferroptosis. Mechanistically, RBM15 improved KDM5B stability in an IGF2BP3-dependent manner, resulting in FER1L4 downregulation and GPX4 upregulation. KDM5B increased KCNQ1OT1 and inhibited ACSL4. KDM5B/KCNQ1OT1 overexpression or FER1L4 knockdown promoted DDP resistance in LC by inhibiting ferroptosis. In conclusion, RBM15 promoted KDM5B expression, and KDM5B upregulation inhibited ferroptosis and promoted DDP resistance in LC by downregulating FER1L4 and upregulating GPX4, as well as by upregulating KCNQ1OT1 and inhibiting ACSL4. Silencing RBM15 inhibited tumor growth in vivo.


Asunto(s)
Cisplatino , Resistencia a Antineoplásicos , Epigénesis Genética , Ferroptosis , Neoplasias Laríngeas , Proteínas de Unión al ARN , Ferroptosis/genética , Cisplatino/farmacología , Resistencia a Antineoplásicos/genética , Humanos , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/metabolismo , Línea Celular Tumoral , Ratones , Animales , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Ratones Desnudos , Regulación Neoplásica de la Expresión Génica , Proliferación Celular/efectos de los fármacos , Antineoplásicos/farmacología , Coenzima A Ligasas/genética , Coenzima A Ligasas/metabolismo
15.
BMC Cancer ; 24(1): 904, 2024 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-39068410

RESUMEN

The lncRNA NEAT1 has been shown to promote the progression of several cancers, containing laryngeal squamous cell carcinoma (LSCC). However, the precise mechanism by which it promotes LSCC progression remains unclear. In this study, we verified the high expression of lncRNA NEAT1 in LSCC tissues and cells using RT-qPCR. Analysis of clinical data exhibited that high expression of lncRNA NEAT1 was associated with a history of smoking, worse T stage, lymph node metastasis, and later TNM stage in patients with LSCC. The promotion effect of lncRNA NEAT1 on LSCC cell proliferation, migration, invasion, and tumor growth in vivo was verified by CCK-8, plate clone formation, Transwell, and nude mouse tumorigenicity assays. Bioinformatics prediction and double luciferase reporter gene assay verified the binding of miR-411-3p to lncRNA NEAT1 and FZD3 mRNA, and inhibition of miR-411-3p reversed the inhibitory effect of lncRNA NEAT1 on FZD3 expression in LSCC cells. We also verified that lncRNA NEAT1-mediated FZD3 activation in the Wnt pathway affects LSCC development. In conclusion, we demonstrate that lncRNA NEAT1 promotes the progression of LSCC, and propose that the lncRNA NEAT1/miR-411-3p/FZD3 axis may be an effective target for LSCC therapy.


Asunto(s)
Movimiento Celular , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Laríngeas , MicroARNs , ARN Largo no Codificante , Vía de Señalización Wnt , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , MicroARNs/genética , MicroARNs/metabolismo , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/metabolismo , Vía de Señalización Wnt/genética , Proliferación Celular/genética , Movimiento Celular/genética , Animales , Ratones , Masculino , Femenino , Línea Celular Tumoral , Invasividad Neoplásica/genética , Receptores Frizzled/genética , Receptores Frizzled/metabolismo , Ratones Desnudos , Persona de Mediana Edad , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas/metabolismo , 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
16.
J Radiat Res ; 65(4): 474-481, 2024 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-38950346

RESUMEN

Laryngeal squamous cell carcinoma (LSCC) is one of the most aggressive cancers that affect the head and neck region. Recent researches have confirmed that long non-coding RNAs (lncRNAs) present an emerging role in diversiform diseases including cancers. Prostate cancer-associated ncRNA transcript 6 (PCAT6) is an oncogene in lung cancer, cervical cancer, colon cancer and gastric cancer, but its role in LSCC is still unknown. In the current study, we attempted to figure out the role of PCAT6 in LSCC. RT-qPCR was to analyze PCAT6 expression in LSCC cells. Functional assays were to uncover the role of PCAT6 in LSCC. Mechanism assays were to explore the regulatory mechanism behind PCAT6 in LSCC. PCAT6 exhibited higher expression in LSCC cells and PCAT6 strengthened cell proliferation and inhibited cell apoptosis. Furthermore, lncRNA PCAT6 modulated notch receptor 3 expression and activated NOTCH signaling pathway via serving as a sponge for miR-4731-5p. Taken together, lncRNA PCAT6 was identified as an oncogene in LSCC, which revealed that PCAT6 might be used as potential therapeutic target for LSCC.


Asunto(s)
Apoptosis , Proliferación Celular , Regulación Neoplásica de la Expresión Génica , Neoplasias Laríngeas , MicroARNs , ARN Largo no Codificante , Receptor Notch3 , Transducción de Señal , Humanos , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/metabolismo , MicroARNs/metabolismo , MicroARNs/genética , Línea Celular Tumoral , Receptor Notch3/metabolismo , Receptor Notch3/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Secuencia de Bases
17.
Carcinogenesis ; 45(9): 642-657, 2024 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-38824399

RESUMEN

Laryngeal cancer (LC) is the second most common head and neck cancer and has a decreasing 5-year survival rate worldwide. Circular RNAs (circRNAs) regulate cancer development in diverse ways based on their distinct biogenesis mechanisms and expansive regulatory roles. However, currently, there is little research on how exosomal circRNAs are involved in the development of LC. Here, we demonstrated that circPVT1, a circRNA derived from the well-studied long noncoding RNA PVT1, is correlated with disease progression in LC and promotes angiogenesis both in vivo and in vitro. Mechanistically, circPVT1 is loaded into LC cell-secreted exosomes and taken up by vascular epithelium cells. By sponging miR-30c-5p, exosomal circPVT1 promotes Rap1b expression, which dramatically enhances vascular endothelial growth factor receptor 2 and the phosphatidylinositol 3-kinase (PI3K)/AKT pathway activation, ultimately resulting in the induction of angiogenesis. Furthermore, our xenograft models demonstrated that the combination of short hairpin RNA-circPVT1 and cetuximab showed high efficacy in inhibiting tumor growth and angiogenesis. Collectively, these findings uncover a novel mechanism of exosomal circRNA-mediated angiogenesis modulation and provide a preclinical rationale for testing this analogous combination in patients with LC.


Asunto(s)
Exosomas , Neoplasias Laríngeas , Neovascularización Patológica , ARN Circular , ARN Largo no Codificante , Transducción de Señal , Receptor 2 de Factores de Crecimiento Endotelial Vascular , Humanos , Neoplasias Laríngeas/patología , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , ARN Circular/genética , Neovascularización Patológica/genética , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Exosomas/metabolismo , Exosomas/genética , ARN Largo no Codificante/genética , Animales , Ratones , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo , Regulación Neoplásica de la Expresión Génica , Proteínas de Unión al GTP rap/metabolismo , Proteínas de Unión al GTP rap/genética , Proliferación Celular , Masculino , Ensayos Antitumor por Modelo de Xenoinjerto , Femenino , Línea Celular Tumoral , Ratones Desnudos , Angiogénesis
18.
Front Immunol ; 15: 1380982, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38915416

RESUMEN

Introduction: Viral infections have been implicated as a risk factor for laryngeal cancer. Given the possible effects of Corona virus disease 2019 (COVID-19) on the laryngeal tissue, we investigated the causal link between COVID-19 and laryngeal cancer using a two-sample Mendelian randomization (MR) approach. Methods: We utilized genetic data from the 5th Genome-wide association studies (GWAS) edition of the COVID-19 Host Genetics Initiative (published on January 18, 2021) and a large-scale laryngeal cancer GWAS comprising 180 cases and 218,612 controls of European ancestry. We applied inverse variance weighting, MR Egger, and weighted median methods to infer causality. We performed sensitivity analysis using the "leave-one-out" method to verify robustness. Results: We found no evidence of a causal association between gene-predicted COVID-19 and laryngeal cancer [Odds ratio (OR)=0.24 (95% Confidence intervals (CI), 0.05-1.26), P=0.09]. However, we observed significant inverse associations between gene-predicted COVID-19 hospitalization [OR=0.51 (95% CI, 0.28-0.95), P=0.03] and severe patients [OR=0.62 (95% CI, 0.43-0.90), P=0.01] and laryngeal cancer. Notably, the study detected important genetic variants, such as rs13050728, that modulate the expression of interferon alpha receptor 2 (IFNAR2), indicating possible roles for immune response pathways in both COVID-19 and cancer. Discussion: This study reveals a potential interaction between COVID-19 severity, genetic factors, and laryngeal cancer, underscoring the importance of investigating the immune response mechanisms in both conditions. These findings contribute to the understanding of the complex interactions between COVID-19 and laryngeal cancer and may guide future research on the role of immune response, particularly involving IFNAR2.


Asunto(s)
COVID-19 , Estudio de Asociación del Genoma Completo , Neoplasias Laríngeas , Análisis de la Aleatorización Mendeliana , Polimorfismo de Nucleótido Simple , SARS-CoV-2 , Humanos , COVID-19/genética , COVID-19/inmunología , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/virología , Neoplasias Laríngeas/inmunología , SARS-CoV-2/fisiología , Predisposición Genética a la Enfermedad , Factores de Riesgo
19.
Crit Rev Immunol ; 44(6): 75-85, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38848295

RESUMEN

Laryngeal cancer (LC) is a prevailing tumor with a high mortality rate. The pivotal role of mitophagy in LC is acknowledged; however, a comprehensive analysis of the corresponding genes has not been conducted. In the present study, we proposed a prognostic model consisting of mitophagy-related genes in LC. Clinical information and transcriptome profiling of patients with LC and mitophagy-related genes were retrieved from open-source databases. Gene set variation analysis (GSVA) and Weighted Gene Co-expression Network Analysis (WGCNA) were used to identify core mitophagy-related genes and construct gene co-expression networks. Functional enrichment analysis was employed to analyze the enriched regulatory pathways of the mitophagy-related genes. Kaplan-Meier curves (KM), Cox, and LASSO regression were applied to explore their prognostic effects. Finally, quantitative real-time PCR (RT-qPCR) further verified the bioinformatics prediction. A total of 45 genes related to mitochondrial pathways was collected. GSVA analysis demonstrated that these genes in tumor samples mainly referred to the mitochondrial pathway. Among these genes, five mitophagy-related-gene signatures (CERCAM, CHPF, EPHX3, EXT2, and MED15) were further identified to construct the prognostic model. KM and Cox regression analyses indicated that this model had an accurate prognostic prediction for LC. RT-qPCR showed that CERCAM, CHPF, EXT2, and MED15 expression were upregulated, and EPHX3 level was decreased in LC cells. The present study established a five-mitophagy-related-gene model that can predict the prognosis of LC patients, thus laying the foundation for a better understanding and potential advancements in clinical treatments for LC.


Asunto(s)
Biomarcadores de Tumor , Biología Computacional , Regulación Neoplásica de la Expresión Génica , Neoplasias Laríngeas , Mitofagia , Humanos , Mitofagia/genética , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/diagnóstico , Neoplasias Laríngeas/mortalidad , Biología Computacional/métodos , Pronóstico , Biomarcadores de Tumor/genética , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Transcriptoma
20.
J Cancer Res Clin Oncol ; 150(6): 295, 2024 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-38844723

RESUMEN

BACKGROUND: The DIAPH2 gene is one of the genes commonly associated with laryngeal squamous cell carcinoma (LSCC). In our study, we considered the four polymorphisms of this gene, i.e. rs5920828, rs4322175, rs12851931 and rs5921830 as potential genetic risk factors for LSCC. METHODS: We determined the genotyping of the genetic variants of DIAPH2 in 230 male patients with histologically confirmed LSCC compared to the European population. Demographic and environmental exposure data of each subject were examined. To conduct the genetic tests, extraction of total DNA was performed. We genotyped all four variants in each patient and determined their frequencies. RESULTS: In the case of the rs12851931 polymorphism in the DIAPH2 gene, a significant difference was observed in the distribution of the T stage depending on the polymorphism. Heterozygotes were more often associated with T2 stage, while homozygotes were more likely to have higher tumor stages. The rs12851931 homozygotes of DIAPH2 were statistically significantly more prevalent in smokers. The results suggested that rs12851931 polymorphism in DIAPH2 could increase the onset risk of LSCC. CONCLUSIONS: Our results provide further information on the role of the DIAPH2 gene in the pathogenesis of LSCC.


Asunto(s)
Forminas , Predisposición Genética a la Enfermedad , Neoplasias Laríngeas , Polimorfismo de Nucleótido Simple , Humanos , Masculino , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/epidemiología , Neoplasias Laríngeas/patología , Persona de Mediana Edad , Forminas/genética , Anciano , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Factores de Riesgo , Genotipo , Adulto
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...