Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 1.855
Filtrar
1.
Int J Mol Med ; 53(4)2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38426543

RESUMO

Laryngeal squamous cell carcinoma (LSCC) is a common malignant tumor with a poor prognosis. Fascin actin­bundling protein 1 (FSCN1) has been reported to play a crucial role in the development and progression of LSCC; however, the underlying molecular mechanisms remain unknown. Herein, a whole transcriptome microarray analysis was performed to screen for differentially expressed genes (DEGs) in cells in which FSCN1 was knocked down. A total of 462 up and 601 downregulated mRNA transcripts were identified. Functional annotation analysis revealed that these DEGs were involved in multiple biological functions, such as transcriptional regulation, response to radiation, focal adhesion, extracellular matrix­receptor interaction, steroid biosynthesis and others. Through co­expression and protein­protein interaction analysis, FSCN1 was linked to novel functions, including defense response to virus and steroid biosynthesis. Furthermore, crosstalk analysis with FSCN1­interacting proteins revealed seven DEGs, identified as FSCN1­interacting partners, in LSCC cells, three of which were selected for further validation. Co­immunoprecipitation validation confirmed that FSCN1 interacted with prostaglandin reductase 1 and 24­dehydrocholesterol reductase (DHCR24). Of note, DHCR24 is a key enzyme involved in cholesterol biosynthesis, and its overexpression promotes the proliferation and migration of LSCC cells. These findings suggest that DHCR24 is a novel molecule associated with FSCN1 in LSCC, and that the FSCN1­DHCR24 interaction may promote LSCC progression by regulating cholesterol metabolism­related signaling pathways.


Assuntos
Carcinoma de Células Escamosas , Proteínas de Transporte , Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , MicroRNAs , Proteínas dos Microfilamentos , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Actinas/metabolismo , Neoplasias Laríngeas/metabolismo , Carcinoma de Células Escamosas/metabolismo , Perfilação da Expressão Gênica , Neoplasias de Cabeça e Pescoço/genética , Colesterol , Oxirredutases/genética , Oxirredutases/metabolismo , Esteroides , Regulação Neoplásica da Expressão Gênica , MicroRNAs/genética , Linhagem Celular Tumoral , Proliferação de Células
2.
Front Biosci (Landmark Ed) ; 29(3): 125, 2024 Mar 22.
Artigo em Inglês | MEDLINE | ID: mdl-38538265

RESUMO

BACKGROUND: The prevalence of laryngeal squamous cell carcinoma (LSCC) is increasing, and it poses a significant threat to human health; therefore, identifying specific targets for LSCC remains crucial. METHODS: Bioinformatics analysis was used to compare the different expression genes expressed in LSCC. Immunohistochemical assay and western blotting were used to analysis protein expression. Cell viability was measured by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl-2H-tetrazolium bromide)((4,5 Dimethyl thiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide)4,5 Dimethyl thiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide (MTT) and 5-ethynyl 2'-deoxyuridine (Edu) assay. Flow cytometry was used to measure the cell cycle. Cell migration was measured by wound healing assay and transwell assay. RESULTS: Our analysis revealed 36 upregulated and 65 downregulated differentially expressed genes (DEGs) when comparing LSCC tumors to adjacent tissues, with cornulin (CRNN) identified as a key hub gene connecting these DEGs. We observed a consistent downregulation of CRNN expression in LSCC cell lines and tissues and was associated with poor patient survival and the tumor microenvironment. CRNN overexpression was found to significantly inhibit cell growth, cell cycle progression, migration and invasion, while CRNN knockdown had the opposite effects. Additionally, in vivo experiments demonstrated that CRNN overexpression suppressed tumor growth in nude mice. CONCLUSIONS: CRNN functions as a potential tumor suppressor and regulates important aspects of LSCC, providing valuable insights into the role of CRNN in LSCC pathogenesis and potential for targeted therapeutic interventions.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Laríngeas , MicroRNAs , Carcinoma de Células Escamosas de Cabeça e Pescoço , Animais , Humanos , Camundongos , Brometos/metabolismo , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Camundongos Nus , MicroRNAs/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Microambiente Tumoral
3.
Cancer Lett ; 587: 216735, 2024 Apr 10.
Artigo em Inglês | MEDLINE | ID: mdl-38369001

RESUMO

As the second most prevalent malignant tumor of head and neck, laryngeal squamous cell carcinoma (LSCC) imposes a substantial health burden on patients worldwide. Within recent years, resistance to oxidative stress and N6-methyladenosine (m6A) of RNA have been proved to be significantly involved in tumorigenesis. In current study, we investigated the oncogenic role of m6A modified long non coding RNAs (lncRNAs), specifically HOXA10-AS, and its downstream signaling pathway in the regulation of oxidative resistance in LSCC. Bioinformatics analysis revealed that heightened expression of HOXA10-AS was associated with the poor prognosis in LSCC patients, and N (6)-Methyladenosine (m6A) methyltransferase-like 3 (METTL3) was identified as a factor in promoting m6A modification of HOXA10-AS and further intensify its RNA stability. Mechanistically, HOXA10-AS was found to play as a competitive endogenous RNA (ceRNA) by sequestering miR-29 b-3p and preventing its downregulation of Integrin subunit alpha 6 (ITGA6), ultimately enhancing the oxidative resistance of tumor cells and promoting the malignant progression of LSCC. Furthermore, our research elucidated the mechanism by which ITGA6 accelerates Keap1 proteasomal degradation via enhancing TRIM25 expression, leading to increased Nrf2 stability and exacerbating its aberrant activation. Additionally, we demonstrated that ITGA6 enhances γ-secretase-mediated Notch signaling activation, ultimately promoting RBPJ-induced TRIM25 transcription. The current study provides the evidence supporting the effect of m6A modified HOXA10-AS and its downstream miR-29 b-3p/ITGA6 axis on regulating oxidative resistance and malignant progression in LSCC through the Notch and Keap1/Nrf2 pathways, and proposed that targeting this axis holds promise as a potential therapeutic approach for treating LSCC.


Assuntos
Adenina/análogos & derivados , Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Proteínas Homeobox A10 , Integrina alfa6 , Neoplasias Laríngeas , MicroRNAs , RNA Longo não Codificante , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , MicroRNAs/genética , Fator 2 Relacionado a NF-E2/genética , Fator 2 Relacionado a NF-E2/metabolismo , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Carcinoma de Células Escamosas/genética , Proteína 1 Associada a ECH Semelhante a Kelch/genética , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Neoplasias de Cabeça e Pescoço/genética , Estresse Oxidativo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proliferação de Células , RNA Longo não Codificante/genética , Metiltransferases/metabolismo
4.
Recent Pat Anticancer Drug Discov ; 19(2): 176-187, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38214357

RESUMO

BACKGROUND: Laryngeal squamous cell carcinoma (LSCC) is the most common cancer of head and neck cancer. Y-box binding protein-1 (YBX1) has tumor-promoting effects in some types of cancers. However, its role in LSCC remains unknown. This study set out to identify the role of YBX1 in LSCC. METHODS: Bioinformatics analysis of the Gene Expression Omnibus (GEO) database and our cohort data were used to explore the association of YBX1 expression with clinicopathological factors in LSCC. Then, cells with stably or transiently transfected with plasmid or siRNA were constructed to assess the effect of loss and gain of YBX1 on the biological phenotypes of LSCC cells in vitro. In addition, subcutaneous xenograft and orthotopic liver tumor mouse models were constructed for validation. The interrogated miRNA databases and subsequent luciferase reporter assays were used to confirm the miR-382-5p target of YBX1. At last, KEGG enrichment annotation from TGCA data was used for downstream analyses of miR-382-5p/YBX1 and verified by PCR and Western immunoblotting. RESULTS: The results showed that significant upregulation of YBX1 in LSCC tumors was correlated with advanced TNM stage and poor prognosis. Knockdown of YBX1 markedly impaired the proliferative, invasive, and migratory activity of Tu212 cells. We confirmed that miR-382-5p targets YBX1 to mediate LSCC progression both in vitro and in vivo. We further confirmed that miR-382-5p/YBX1 modulated the Ras/MAPK signaling axis to regulate the progression of LSCC. CONCLUSION: Together, our results indicated that YBX1 is an important promoter of LSCC progression. And miR-382-5p/YBX1/RAS/MAPK signaling pathway can be perceived as a promising target in the treatment of LSCC.


Assuntos
Neoplasias Laríngeas , MicroRNAs , Carcinoma de Células Escamosas de Cabeça e Pescoço , Proteína 1 de Ligação a Y-Box , Animais , Humanos , Camundongos , Linhagem Celular Tumoral , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Proteína 1 de Ligação a Y-Box/genética , Proteína 1 de Ligação a Y-Box/metabolismo
5.
Cell Signal ; 114: 111002, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38048860

RESUMO

Laryngeal squamous cell carcinoma (LSCC) is one of the common malignant tumors in the head and neck region, and its high migration and invasion seriously threaten the survival and health of patients. In cancer development, m6A RNA modification plays a crucial role in regulating gene expression and signaling. This study delved into the function and mechanism of the m6A reading protein YTHDF1 in LSCC. It was found that YTHDF1 was highly expressed in the GEO database and LSCC tissues. Cell function experiments confirmed that the downregulation of YTHDF1 significantly inhibited the proliferation, migration, and invasion ability of LSCC cells. Further studies revealed that EIF4A3 was a downstream target gene of YTHDF1, and knockdown of EIF4A3 similarly significantly inhibited the malignant progression of LSCC in both in vivo and in vitro experiments. The molecular mechanism studies suggested that YTHDF1-EIF4A3 may promote the malignant development of LSCC by activating the EMT signaling pathway. This study provides important clues for an in-depth understanding of the pathogenesis of LSCC and is a solid foundation for the discovery of new therapeutic targets and approaches.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , MicroRNAs , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço , Carcinoma de Células Escamosas/patologia , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Proliferação de Células/genética , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , MicroRNAs/metabolismo , Fator de Iniciação 4A em Eucariotos/genética , Fator de Iniciação 4A em Eucariotos/metabolismo , RNA Helicases DEAD-box/metabolismo , Proteínas de Ligação a RNA/metabolismo
6.
Eur J Med Res ; 28(1): 591, 2023 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-38102653

RESUMO

BACKGROUND: Although great progress has been made in anti-cancer therapy, the prognosis of laryngeal squamous cell carcinoma (LSCC) patients remains unsatisfied. Quantities of studies demonstrate that glycolytic reprograming is essential for the progression of cancers, where triosephosphate isomerase 1 (TPI1) serves as a catalytic enzyme. However, the clinicopathological significance and potential biological functions of TPI1 underlying LSCC remains obscure. METHODS: We collected in-house 82 LSCC tissue specimens and 56 non-tumor tissue specimens. Tissue microarrays (TMA) and immunohistochemical (IHC) experiments were performed. External LSCC microarrays and bulk RNA sequencing data were integrated to evaluate the expression of TPI1. We used a log-rank test and the CIBERSORT algorithm to assess the prognostic value of TPI1 and its association with the LSCC microenvironment. Malignant laryngeal epithelial cells and immune-stromal cells were identified using inferCNV and CellTypist. We conducted a comprehensive analysis to elucidate the molecular functions of TPI1 in LSCC tissue and single cells using Pearson correlation analysis, high dimensional weighted gene co-expression analysis, gene set enrichment analysis, and clustered regularly interspaced short palindromic repeats (CRISPR) screen. We explored intercellular communication patterns between LSCC single cells and immune-stromal cells and predicted several therapeutic agents targeting TPI1. RESULTS: Based on the in-house TMA and IHC analysis, TPI1 protein was found to have a strong positive expression in the nucleus of LSCC cells but only weakly positive activity in the cytoplasm of normal laryngeal cells (p < 0.0001). Further confirmation of elevated TPI1 mRNA expression was obtained from external datasets, comparing 251 LSCC tissue samples to 136 non-LSCC tissue samples (standardized mean difference = 1.06). The upregulated TPI1 mRNA demonstrated a high discriminative ability between LSCC and non-LSCC tissue (area under the curve = 0.91; sensitivity = 0.87; specificity = 0.79), suggesting its potential as a predictive marker for poor prognosis (p = 0.037). Lower infiltration abundance was found for plasma cells, naïve B cells, monocytes, and neutrophils in TPI-high expression LSCC tissue. Glycolysis and cell cycle were significantly enriched pathways for both LSCC tissue and single cells, where heat shock protein family B member 1, TPI1, and enolase 1 occupied a central position. Four outgoing communication patterns and two incoming communication patterns were identified from the intercellular communication networks. TPI1 was predicted as an oncogene in LSCC, with CRISPR scores less than -1 across 71.43% of the LSCC cell lines. TPI1 was positively correlated with the half maximal inhibitory concentration of gemcitabine and cladribine. CONCLUSIONS: TPI1 is dramatically overexpressed in LSCC than in normal tissue, and the high expression of TPI1 may promote LSCC deterioration through its metabolic and non-metabolic functions. This study contributes to advancing our knowledge of LSCC pathogenesis and may have implications for the development of targeted therapies in the future.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , RNA/genética , Triose-Fosfato Isomerase/genética , Triose-Fosfato Isomerase/metabolismo , Imuno-Histoquímica , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Prognóstico , RNA Mensageiro/genética , Neoplasias de Cabeça e Pescoço/genética , Regulação Neoplásica da Expressão Gênica , Microambiente Tumoral
7.
Cancer Biol Ther ; 24(1): 2274143, 2023 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-37948132

RESUMO

Laryngeal squamous cell carcinoma (LSCC), is a prevalent malignant tumor, belongs to the category of head and neck tumors. N-acetyltransferase 10 (NAT10) can alter mRNA stability through N4- acetylcytidine (ac4C) modification. This study aimed to make an investigation into the role of NAT10-mediated ac4C modification in the malignant processes of LSCC cells. The NAT10 expression in LSCC tissues and cells was detected RT-qPCR and western blot. The ac4C dot blot was performed to detect ac4C level. Besides, the cell viability, migration, and invasion abilities were detected by CCK-8 and transwell assays. AcRIP-qPCR was performed to measure the abundance of ac4C on FOXM1 mRNA. RIP and Luciferase reporter assays were performed to demonstrate the interaction between NAT10 and FOXM1. Finally, the xenograft model was established to explore the role of NAT10 in vivo. NAT1 levels were significantly increased in the LSCC tissues and cells. Knockdown of NAT10 could significantly suppress the proliferation, migration, and invasion of LSCC cells. Additionally, NAT10 recognized the ac4C-modified sites in the 3'-untranslated regions (3' UTR) of forkhead box M1 (FOXM1) to enhance the ability of FOXM1 mRNA. Furthermore, FOXM1 overexpression reversed the suppressing effects of NAT10 knockdown on the proliferation, migration, and invasion of LSCC cells, according to the results of rescue assays. Finally, results of animal experiments showed that NAT10 promoted in vivo tumorigenesis of LSCC cells through upregulating FOXM1. Our current study demonstrated that NAT10-mediated ac4C modification of FOXM1 mRNA promoted the malignant processes of LSCC cells.


Assuntos
Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , MicroRNAs , Animais , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , MicroRNAs/genética , RNA Mensageiro/genética , Neoplasias Laríngeas/metabolismo , Linhagem Celular Tumoral , Carcinogênese/genética , Transformação Celular Neoplásica , Proteína Forkhead Box M1/genética , Acetiltransferases N-Terminal
8.
Int J Mol Sci ; 24(19)2023 Sep 30.
Artigo em Inglês | MEDLINE | ID: mdl-37834257

RESUMO

Obstructive sleep apnea (OSA), characterized by intermittent hypoxia (IH), may increase the risk of cancer development and a poor cancer prognosis. TAMs of the M2 phenotype, together with the intermittent hypoxic environment within the tumor, drive tumor aggressiveness. However, the mechanism of TAMs in IH remains unclear. In our study, IH induced the recruitment of macrophages, and IH-induced M2-like TAMs promoted glycolysis in laryngeal cancer cells through hexokinase 1. The hexokinase inhibitor 2-deoxy-D-glucose and HK1 shRNA were applied to verify this finding, confirming that M2-like TAMs enhanced glycolysis in laryngeal cancer cells through HK1 under intermittent hypoxic conditions. Comprehensive RNA-seq analysis disclosed a marked elevation in the expression levels of the transcription factor ZBTB10, while evaluation of a laryngeal cancer patient tissue microarray demonstrated a positive correlation between ZBTB10 and HK1 expression in laryngeal carcinoma. Knockdown of ZBTB10 decreased HK1 expression, and overexpression of ZBTB10 increased HK1 expression in both laryngeal cancer cells and 293T cells. The luciferase reporter assay and Chromatin immunoprecipitation assay confirmed that ZBTB10 directly bound to the promoter region of HK1 and regulated the transcriptional activity of HK1. Finally, the CLEC3B level of the M2 supernatant is significantly higher in the IH group and showed a protumor effect on Hep2 cells. As ZBTB10-mediated regulation of HK1 affects glycolysis in laryngeal cancer, our findings may provide new potential therapeutic targets for laryngeal cancer.


Assuntos
Glicólise , Hexoquinase , Neoplasias Laríngeas , Proteínas Repressoras , Apneia Obstrutiva do Sono , Humanos , Hexoquinase/genética , Hexoquinase/metabolismo , Hipóxia , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Proteínas Repressoras/metabolismo , RNA Interferente Pequeno/metabolismo , Apneia Obstrutiva do Sono/complicações
9.
Tissue Cell ; 84: 102187, 2023 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-37536262

RESUMO

Laryngeal squamous cell carcinoma (LSCC) is the second most common head and neck cancer. To identify the link between ferroptosis and LSCC, we targeted the dual oxidase 1 (DUOX1) gene. This study aimed to reveal the intrinsic mechanism by which the DUOX1-zinc-finger CCCH domain-containing protein 13 (ZC3H13) ferroptosis axis affected the LSCC process. GEPIA was used to investigate the expression of DUOX1 in LSCC, and the expression levels of DUOX1 and ZC3H13 were manipulated by overexpression and RNA interference. MTT assay was used to detect cell proliferation. Chromatin immunoprecipitation (CHIP) detected the binding of DUOX1 and ZC3H13, and ROS assessment and intracellular Fe2+ content determination were performed to examine the ferroptosis. MeRIP was used to analyze the m6A methylation of DUOX1. Ferroptosis-related proteins were detected by qRT-PCR. DUOX1 was found to be poorly expressed in LSCC cells, low DUOX1 level promoted LSCC cell proliferation, and low ZC3H13 level decreased LSCC cell proliferation. Besides, there was an interaction between DUOX1 and ZC3H13. DUOX1 could inhibit the expression levels of ferroptosis-related genes GPX4 and F1H1 in LSCC cells DUOX1 inhibited the expression levels of ROS and ferroptosis-related genes GPX4 and F1H1 and increased intracellular iron content in LSCC cells, but ZC3H13 reversed this phenomenon by inhibiting DUOX1 gene through m6A methylation modification. ZC3H13 reduced DUOX1-mediated ferroptosis in LSCC cells through m6A-dependent modification. The regulatory pathway of DUOX1 and ferroptosis are potential targets for designing diagnostic and combination therapeutic strategies for LSCC patients.


Assuntos
Carcinoma de Células Escamosas , Ferroptose , Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , Carcinoma de Células Escamosas de Cabeça e Pescoço , Humanos , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Proliferação de Células/genética , Oxidases Duais/genética , Oxidases Duais/metabolismo , Ferroptose/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Laríngeas/metabolismo , Proteínas Nucleares/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Proteínas de Ligação a RNA/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética
10.
Mol Biol Rep ; 50(9): 7245-7252, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37418079

RESUMO

BACKGROUND: Laryngeal squamous cell carcinoma (LSCC) is the major pathological subtype of laryngeal cancer. It has been shown that alterations of the expression of non-classical human leukocyte antigens (HLA) and the chain-related MIC molecules by malignant cells can lead to escape from the immune system control and certain allele variants may participate in immune editing and therefore be associated with modulation of cancer risk. The aim of the present study was to investigate the role of non-classical HLA class Ib and chain-related MIC polymorphisms, determined at the allelic level by next-generation sequencing (NGS), in patients from the Bulgarian population, diagnosed with LSCC. MATERIALS AND METHODS: In the present study DNA samples from 48 patients with LSCC were used. Data was compared to 63 healthy controls analysed in previous studies. HLA genotyping was performed by using the AlloSeq Tx17 early pooling protocol and the library preparation AlloSeq Tx17 kit (CareDx). Sequencing was performed on MiniSeq sequencing platform (Illumina) and HLA genotypes were assigned with the AlloSeq Assign analysis software v1.0.3 (CareDx) and the IPD-IMGT/HLA database 3.45.1.2. RESULTS: The HLA disease association tests revealed a statistically significant predisposing association of HLA-F*01:01:02 (Pc = 0.0103, OR = 24.0194) with LSCC, while HLA-F*01:01:01 (Pc = 8.21e-04, OR = 0.0485) has a possible protective association. Additionally we observed several haplotypes with statistically significant protective and predisposing associations. The strongest association was observed for F*01:01:01-H*01:01:01 (P = 0.0054, haplotype score=-2.7801). CONCLUSION: Our preliminary study suggests the involvement of HLA class Ib in cancer development and the possible role of the shown alleles as biomarkers of LSCC.


Assuntos
Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Antígenos de Histocompatibilidade Classe II/genética , Polimorfismo Genético/genética , Antígenos HLA/genética , Antígenos de Histocompatibilidade Classe I/genética , Haplótipos/genética , Neoplasias de Cabeça e Pescoço/genética , Alelos , Frequência do Gene/genética , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo
11.
Int J Biol Sci ; 19(5): 1382-1400, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37056932

RESUMO

Translation machinery associated 7 homolog (TMA7) is closely related to proliferation-related diseases. However, the function and regulatory mechanism of TMA7 in laryngeal squamous cell carcinoma (LSCC) remain unclear. The present study aimed to investigate the effect of TMA7 on the occurrence and development of LSCC and to study the mechanism of TMA7. TMA7 is upregulated in LSCC tissues and associated with poor prognosis. After TMA7 downregulation, the autophagy level was increased, and the proliferation, migration, and invasion of LSCC cells were inhibited. The m6A methylated reader IGF2BP3 enhanced the stability of TMA7 and reduced the level of autophagy. TMA7 interacted directly with UBA2. Furthermore, the activation of the IGF2BP3-regulated TMA7-UBA2-PI3K pathway is the primary mechanism by which TMA7 inhibits autophagy and promotes the progression of LSCC. The current study revealed that IGF2BP3-mediated TMA7 m6A modification promotes LSCC progression and cisplatin-resistance through UBA2-PI3K pathway, providing new insights into the autophagy-related mechanism, potential biomarkers, and therapeutic targets for LSCC.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , MicroRNAs , Humanos , Autofagia/genética , Carcinoma de Células Escamosas/genética , Linhagem Celular Tumoral , Proliferação de Células/genética , Cisplatino/farmacologia , Cisplatino/uso terapêutico , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias de Cabeça e Pescoço/genética , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Metilação , MicroRNAs/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Enzimas Ativadoras de Ubiquitina/genética , Enzimas Ativadoras de Ubiquitina/metabolismo , Proteínas de Ligação a RNA/metabolismo
12.
Biomed Res Int ; 2023: 1733100, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36718148

RESUMO

Laryngeal squamous cell cancer (LSCC) is a common malignant tumor with a high degree of malignancy, and its etiology remains unclear. Therefore, screening potential biomarkers is necessary to facilitate the treatment and diagnosis of LSCC. Robust rank aggregation (RRA) analysis was used to integrate two gene expression datasets of LSCC patients from the Gene Expression Omnibus (GEO) database and identify differentially expressed genes (DEGs) between LSCC and nonneoplastic tissues. A gene coexpression network was constructed using weighted gene correlation network analysis (WGCNA) to explore potential associations between the module genes and clinical features of LSCC. Combining differential gene expression analysis and survival analysis, we screened potential hub genes, including CDK1, SPC24, HOXB7, and SELENBP1. Subsequently, western blotting and immunohistochemistry were used to test the protein levels in clinical specimens to verify our findings. Finally, four candidate diagnostic and prognostic biomarkers (CDK1, SPC24, HOXB7, and SELENBP1) were identified. We propose, for the first time, that SPC24 is a gene that may associate with LSCC malignancy and is a novel therapeutic target. These findings may provide important mechanistic insight of LSCC.


Assuntos
Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , Neoplasias de Células Escamosas , Humanos , Neoplasias Laríngeas/diagnóstico , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Prognóstico , Carcinoma de Células Escamosas de Cabeça e Pescoço/diagnóstico , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/genética , Ciclo Celular/genética , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Proteínas Associadas aos Microtúbulos/genética , Proteínas de Homeodomínio/genética
13.
Int J Mol Sci ; 24(2)2023 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-36675308

RESUMO

Head and neck squamous cell carcinoma (HNSCC) arises from the mucosal epithelium in the oral cavity, pharynx, sino-nasal region, and larynx. Laryngeal squamous cell carcinoma (LSCC) represents one-third of all head and neck cancers. Dysregulated RNA-related pathways define an important molecular signature in this aggressive carcinoma. The Survival Motor Neuron (SMN) protein regulates fundamental aspects of the RNA metabolism but, curiously, its role in cancer is virtually unknown. For the first time, here, we focus on the SMN in the cancer context. We conducted a pilot study in a total of 20 patients with LSCC where the SMN was found overexpressed at both the protein and transcript levels. By a cellular model of human laryngeal carcinoma, we demonstrated that the SMN impacts cancer-relevant behaviors and perturbs key players of cell migration, invasion, and adhesion. Furthermore, in LSCC we showed a physical interaction between the SMN and the epidermal growth factor receptor (EGFR), whose overexpression is an important feature in these tumors. This study proposes the SMN protein as a novel therapeutic target in LSSC and likely in the whole spectrum of HNSCC. Overall, we provide the first analysis of the SMN in human cancer.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas/patologia , Projetos Piloto , Neoplasias de Cabeça e Pescoço/genética , Neoplasias Laríngeas/metabolismo , RNA , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Proliferação de Células/genética
14.
Mol Biotechnol ; 65(4): 655-667, 2023 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-36214976

RESUMO

LINC00467 was reported as an oncogenic gene in different types of human cancers. In this study, we investigated the molecular mechanisms of LINC00467 in the tumorigenesis of laryngeal squamous cell cancer (LSCC). RT-qPCR was utilized to detect the mRNA expression of genes, and western blot assay was used to determine the protein levels of TNF alpha-induced protein 3 (TNFAIP3). The cell viability was detected by CCK-8 assay. Transwell assays were conducted to determine the cell migration and invasion of LSCC cells, and the cell cycle was assessed by flow cytometry. The association between paired box 5 (PAX5), LINC00467, miR-4735-3p, and TNFAIP3 was verified using ChIP, RNA pull-down, or luciferase reporter assays. In our study, we found that LINC00467 was upregulated in LSCC, and knockdown of LINC00467 suppressed cell viability and metastasis of LSCC. Besides, LINC00467 transcription could be activated by PAX5 in LSCC. Furthermore, LINC00467 acted as competitive endogenous RNA (ceRNA) for miR-4735-3p to accelerate LSCC progression. In the meantime, TNFAIP3 was identified as a downstream gene of miR-4735-3p. Finally, TNFAIP3 overexpression could overturn the effects of miR-4735-3p mimic on LSCC cellular activities. In conclusion, our results demonstrated that PAX5-induced LINC00467 facilitated LSCC progression by inhibiting miR-4735-3p to increase TNFAIP3 expression.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , MicroRNAs , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Regulação para Cima , Fator de Necrose Tumoral alfa/genética , Carcinoma de Células Escamosas/metabolismo , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Linhagem Celular Tumoral , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica
15.
Pathol Res Pract ; 241: 154229, 2023 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-36509010

RESUMO

Advanced laryngeal squamous cell carcinoma (LSCC) has a high mortality rate, and the prognosis is poor. However, the underlying molecular biological mechanisms bringing about the development and progression of advanced LSCC are not entirely clarified. This study aimed to find out the potential biomarkers to predict the prognosis in advanced LSCC patients who had undergone postoperative radiotherapy alone. The next-generation sequencing of RNA was performed to detect the mRNAs expression profiling in 10 advanced LSCC samples, comprised of 5 samples from LSCC patients with favorable outcome and 5 samples from paired patients with poor outcome. Then bioinformatics analysis including Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway were used to find out functional core genes that were significantly different between the two groups. 1630 differentially expressed genes (DEGs) were confirmed to have significant differences between the two groups. 53 GO terms and 19 pathways which were closely related to the DEGs were identified. Finally, 52 intersection DEGs which were both related to the top three GO terms and pathways were identified. The expression of several core genes was confirmed with RT-qPCR in tissues from another 75 patients. RT-qPCR confirmed that the genes of c-JUN, LYN, PIK3R2, and TNFAIP3 were significantly differentially expressed between the two groups, which was in accordance with the RNA sequencing data. The DEGs identified above may be potential prognostic markers for advanced LSCC patients with postoperative radiotherapy, and may provide essential guidance for following-up.


Assuntos
Carcinoma de Células Escamosas , Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , Humanos , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/radioterapia , Prognóstico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Carcinoma de Células Escamosas/metabolismo , Perfilação da Expressão Gênica , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/radioterapia , Neoplasias Laríngeas/metabolismo , Redes Reguladoras de Genes , Neoplasias de Cabeça e Pescoço/genética , Regulação Neoplásica da Expressão Gênica , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo
16.
Cell Mol Biol (Noisy-le-grand) ; 69(14): 197-200, 2023 Dec 20.
Artigo em Inglês | MEDLINE | ID: mdl-38279443

RESUMO

The present study aimed to elucidate the role of MicroRNA-203b-3p (miRNA-203b-3p) in protecting the deterioration of laryngeal carcinoma through targeting ZNF324. Relative levels of miRNA-203b-3p and ZNF324 in laryngeal carcinoma tissues with different tumor node metastasis (TNM) staging and pathological grades were detected. Regulatory effects of miRNA-203b-3p on clonality, viability and 5-Ethynyl-2'- deoxyuridine (EdU)-positive ratio in M2E and TU212 cells were assessed. The binding relationship between miRNA-203b-3p and ZNF324 was evaluated by dual-luciferase reporter gene assay. The involvement of ZNF324 in cell phenotype changes of laryngeal carcinoma regulated by miRNA-203b-3p was explored by rescue experiments. MiRNA-203b-3p was downregulated in laryngeal carcinoma, especially in those with advanced TNM staging or pathological grade. Overexpression of miRNA-203b-3p reduced clonality, viability and EdU-positive ratio in M2E and TU212 cells. In addition, ZNF324 was upregulated in laryngeal carcinoma, which was negatively regulated by miRNA-203b-3p. ZNF324 was verified to be the target binding miRNA-203b-3p. Notably, overexpression of ZNF324 could partially reverse the inhibitory effects of miRNA-203b-3p on laryngeal carcinoma proliferation. MiRNA-203b-3p is downregulated in laryngeal carcinoma, which blocks laryngeal carcinoma cells to proliferate through targeting ZNF324 and thus alleviates cancer progression.


Assuntos
Carcinoma , Proteínas de Ligação a DNA , Neoplasias Laríngeas , MicroRNAs , Humanos , Carcinoma/metabolismo , Carcinoma/patologia , Linhagem Celular Tumoral , Proliferação de Células , Regulação Neoplásica da Expressão Gênica , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , MicroRNAs/genética , MicroRNAs/metabolismo , Proteínas de Ligação a DNA/metabolismo
17.
Curr Pharm Des ; 29(43): 3467-3477, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38163971

RESUMO

BACKGROUND: Growth differentiation factor-10 (GDF-10), a member of the TGF-ß superfamily, plays a crucial role in cell proliferation and differentiation. In some tumors, GDF-10 can act as a tumor suppressor to inhibit tumor progression, but its role in posterior squamous cell carcinoma has not been reported yet. METHODS: The aim of this study was to investigate the effect of GDF-10 on the epithelial-mesenchymal transition of laryngeal squamous cell carcinoma, and to provide new ideas for future targets in the treatment of laryngeal squamous carcinoma. RESULTS: The effect of GDF-10 on tumor growth was detected; bioinformatics analysis was performed to predict the downstream targets of GDF-10, and RT-PCR and western blot were performed to detect the expression levels of target genes and proteins, respectively. CONCLUSION: Our findings support that GDF-10 can inhibit the proliferation, migration, and invasion, and promote apoptosis of laryngeal carcinoma AMC-HN-8 cells. GDF-10 inhibits the EMT of laryngeal carcinoma through LRP4 and thus inhibits the progression of laryngeal carcinoma.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Laríngeas , Humanos , Neoplasias Laríngeas/tratamento farmacológico , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Fator 10 de Diferenciação de Crescimento/genética , Fator 10 de Diferenciação de Crescimento/metabolismo , Carcinoma de Células Escamosas/tratamento farmacológico , Carcinoma de Células Escamosas/genética , Fator de Crescimento Transformador beta/metabolismo , Transição Epitelial-Mesenquimal/genética , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Regulação Neoplásica da Expressão Gênica
18.
Acta Biochim Pol ; 69(4): 753-759, 2022 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-36269869

RESUMO

Laryngeal cancer is detected commonly worldwide, and it ranks second highest in incidence among the respiratory tract neoplasms following only head and neck squamous cell cancer. In the present study salicin dimethyl ether was synthesized and evaluated against laryngeal cancers cells for anticancer property. MTT assay was used for the measurement of changes in TU686 and Tu212 cell proliferation while as induced apoptosis was detected by flow cytometry. Protein expression was determined by western blotting and expression of mRNA by RT-PCR assay. In the present study salicin dimethyl ether was synthesized by the reaction of salicin with methyl iodide using sodium hydride as base. Salicin dimethyl ether treatment led to a significant decrease in TU686 and Tu212 cell proliferation in a dose-dependent manner. In TU686 and Tu212 cells salicin dimethyl ether treatment caused a significant increase in cell apoptosis and elevated caspase-3 activity. Treatment with salicin dimethyl ether led to a prominent reduction in Bcl-2 protein expression in TU686 and Tu212 cells at 72 h. Salicin dimethyl ether treatment led to a prominent decrease in p-PI3K and p-Akt protein expression in TU686 and Tu212 cells, compared to the untreated cells. A significant increase in miR­15a expression in TU686 and Tu212 cells was observed on treatment with salicin dimethyl ether at 72 h. In summary, the current study demonstrates that salicin dimethyl ether, a synthetic derivative of salicin, suppresses proliferation of TU686 and Tu212 cells. The underlying mechanism involves induction of apoptosis, inhibition of PI3K/Akt pathway and promotion of miR-15a expression. Therefore, salicin dimethyl ether may be used for inhibition of laryngeal cancer growth, however, in vivo studies need to be conducted to confirm the effect.


Assuntos
Neoplasias de Cabeça e Pescoço , Neoplasias Laríngeas , MicroRNAs , Humanos , Neoplasias Laríngeas/tratamento farmacológico , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , Fosfatidilinositol 3-Quinases , Proteínas Proto-Oncogênicas c-akt , Linhagem Celular Tumoral , Apoptose , Proliferação de Células/genética , MicroRNAs/genética
19.
J Food Biochem ; 46(12): e14465, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-36226832

RESUMO

The objective of this study was to examine Nerolidol (NER) and Cisplatin (CIS) performed against human laryngeal carcinoma (Hep 2) cells. We evaluated the effect of NER, CIS, and NER + CIS on cell viability, cell migration, oxidative stress, mitochondrial membrane depolarization, nuclear condensation, apoptotic induction, and DNA damage in Hep 2 cells. We used the MTT assay to assess the cytotoxicity effect of NER and CIS on Hep 2 cells in terms of morphological alterations. Present results demonstrated that IC50 values of NER and CIS have potential cytotoxicity against Hep 2 cells. NER effectively inhibited cell viability, increased reactive oxygen species generation, apoptotic induction, and DNA damage in Hep 2 cells. In addition, the docking study evaluated the structural binding interaction of NER with PI3K/Akt and PCNA protein. Furthermore, NER with PI3K/Akt, PCNA has a higher crucial score and affinity. Present results infer that NER could be used to target signaling molecules in anticancer studies. PRACTICAL APPLICATIONS: Nerolidol is a dietary phytochemical with high biological activity that can find in a variety of plants. Many researchers focused on Nerolidol to treat various diseases including cancer. However, there is no studies exist on laryngeal cancer. This study uses Nerolidol and Cisplatin to generate oxidative stress and stimulate apoptosis and DNA damage in human laryngeal cancer cells. Based on present findings, Nerolidol could be a choice of anticancer medication, either alone or in combination against oral squamous cell carcinomas in both in vitro and in vivo experimental systems.


Assuntos
Carcinoma de Células Escamosas , Neoplasias Laríngeas , Humanos , Cisplatino/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Neoplasias Laríngeas/tratamento farmacológico , Neoplasias Laríngeas/metabolismo , Neoplasias Laríngeas/patologia , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Antígeno Nuclear de Célula em Proliferação/farmacologia , Antígeno Nuclear de Célula em Proliferação/uso terapêutico , Linhagem Celular Tumoral , Apoptose , Carcinoma de Células Escamosas/tratamento farmacológico
20.
Cell Mol Biol (Noisy-le-grand) ; 68(6): 79-83, 2022 Jun 30.
Artigo em Inglês | MEDLINE | ID: mdl-36227673

RESUMO

Laryngeal cancer has become the focus of research because of its high incidence rate and mortality rate. However, the research on miR-194-5p in laryngeal cancer is quite rare. The purpose of this study is to explore the effect of miR-194-5p on the proliferation and invasion of laryngeal cancer cells in order to find an effective way to treat laryngeal cancer. The results showed that miR-194-5p could activate the mTOR signaling pathway by regulating the expression of Smurf1, which had an effect on laryngeal cancer cells. The results showed that the absorbance of the miR-194-5p group was 0.38 lower than that of the NC group, which indicated that up-regulation of mir-194-5p could weaken the proliferation of laryngeal cancer cells. In addition, the average number of laryngeal cancer cells in NC and the miR-194-5p groups was 125.2 and 53.8, respectively, which indicated that miR-194-5p could reduce the number of laryngeal cancer cells passing through the basement membrane and their invasion ability.


Assuntos
Neoplasias Laríngeas , MicroRNAs , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células/genética , Regulação Neoplásica da Expressão Gênica , Humanos , Neoplasias Laríngeas/genética , Neoplasias Laríngeas/metabolismo , MicroRNAs/genética , MicroRNAs/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo , Ubiquitina-Proteína Ligases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...