Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Cancer Manag Res ; 15: 1165-1179, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37868687

RESUMEN

Purpose: Lysine crotonylation, an emerging posttranslational modification, has been implicated in the regulation of diverse biological processes. However, its involvement in oral squamous cell carcinoma (OSCC) remains elusive. This study aims to reveal the global crotonylome in OSCC under hypoxic conditions and explore the potential regulatory mechanism of crotonylation in OSCC. Methods: Liquid-chromatography fractionation, affinity enrichment of crotonylated peptides, and high-resolution mass spectrometry were employed to detect differential crotonylation in CAL27 cells cultured under hypoxia. The obtained data were further subjected to bioinformatics analysis to uncover the involved biological processes and pathways of the dysregulated crotonylated proteins. A site-mutated plasmid was utilized to investigate the effect of crotonylation on Heat Shock Protein 90 Alpha Family Class B Member 1 (HAP90AB1) function. Results: A large-scale crotonylome analysis revealed 1563 crotonylated modification sites on 605 proteins in CAL27 cells under hypoxia. Bioinformatics analysis revealed a significant decrease in histone crotonylation levels, while up-regulated crotonylated proteins were mainly concentrated in non-histone proteins. Notably, glycolysis-related proteins exhibited prominent up-regulation among the identified crotonylated proteins, with HSP90AB1 displaying the most significant changes. Subsequent experimental findings confirmed that mutating lysine 265 of HSP90AB1 into a silent arginine impaired its function in promoting glycolysis. Conclusion: Our study provides insights into the crotonylation modification of proteins in OSCC under hypoxic conditions and elucidates the associated biological processes and pathways. Crotonylation of HSP90AB1 in hypoxic conditions may enhance the glycolysis regulation ability in OSCC, offering novel perspectives on the regulatory mechanism of crotonylation in hypoxic OSCC and potential therapeutic targets for OSCC treatment.

2.
Biomed Res Int ; 2023: 4444869, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36814797

RESUMEN

Background: Lysine crotonylation (Kcr) is a newly identified posttranslational modification type regulated by various enzymes and coenzymes, including lysine crotonyltransferase, lysine decrotonylase, and binding proteins. However, the role of Kcr regulators in head and neck squamous cell carcinoma (HNSCC) remains unknown. The aim of this study was to establish and validate a Kcr-related prognostic signature of HNSCC and to assess the clinical predictive value of this signature. Methods: The mRNA expression profiles and clinicopathological data from The Cancer Genome Atlas (TCGA) database were downloaded to explore the clinical significance and prognostic value of these regulators in HNSCC. The least absolute shrinkage and selection operator (LASSO) Cox regression model was used to generate the Kcr-related prognostic signature for HNSCC. Subsequently, the GSE65858 dataset from the Gene Expression Omnibus (GEO) database was used to validate the signature. The prognostic value of the signature was evaluated using the Kaplan-Meier survival, receiver operating characteristic (ROC) curve, and univariate and multivariate Cox regression analyses. Results: We established a nine-gene risk signature associated with the prognosis of HNSCC based on Kcr regulators. High-risk patients demonstrated significantly poorer overall survival (OS) than low-risk patients in the training (TCGA) and validation (GEO) datasets. Then, the time-dependent receiver operating characteristic (ROC) curve analysis showed that the nine-gene risk signature was more accurate for predicting the 5-year OS than other clinical parameters, including age, gender, T stage, N stage, and histologic grade in the TCGA and GEO datasets. Moreover, the Cox regression analysis showed that the constructed risk signature was an independent risk factor for HNSCC. Conclusion: Our study identified and validated a nine-gene signature for HNSCC based on Kcr regulators. These results might contribute to prognosis stratification and treatment escalation for HNSCC patients.


Asunto(s)
Neoplasias de Cabeza y Cuello , Lisina , Humanos , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello , Relevancia Clínica
3.
Technol Cancer Res Treat ; 21: 15330338221118202, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35929142

RESUMEN

Background: Head and neck squamous cell carcinoma (HNSCC) is the 6th most common cancer worldwide. Heat shock protein 90 alpha family class B member 1 (HSP90AB1) is highly expressed in a variety of cancers and is associated with poor prognosis, however, its role in HNSCC is still poorly understood. This study aimed to explore the function HSP90AB1 played in HNSCC progression. Methods: The expression level of HSP90AB1 in HNSCC was analyzed by bioinformatics analysis and western blotting, and its relationship with clinicopathological parameters was analyzed by bioinformatics analysis and immunohistochemistry. Three stable HSP90AB1 knockdown HNSCC cell lines were constructed by lentiviral transfection. The effect of HSP90AB1 knockdown on the proliferation and migration of HNSCC cells was tested by CCK-8 assay, EdU incorporation assay, colony formation assay, nude mouse xenograft models, transwell migration assay, wound healing assay, and western blotting. The effect of HSP90AB1 knockdown on glycolysis in HNSCC cells was assessed by quantitative real-time PCR and related assay kits. Finally, the levels of Akt and phospho-Akt (Ser473) proteins after HSP90AB1 knockdown were detected by western blotting. Results: HSP90AB1 was highly expressed in HNSCC and associated with T grade, lymph node metastasis, and prognosis. Knockdown of HSP90AB1 inhibited the proliferation, migration, and glycolysis of HNSCC, and reduced the level of phospho-Akt. Conclusion: HSP90AB1 functions as an oncogene in HNSCC, and has the potential to become a prognostic factor and therapeutic target.


Asunto(s)
Proteínas HSP90 de Choque Térmico , Neoplasias de Cabeza y Cuello , Carcinoma de Células Escamosas de Cabeza y Cuello , Animales , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Glucólisis/genética , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/metabolismo , Humanos , Ratones , Pronóstico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo
4.
Cell Oncol (Dordr) ; 44(3): 661-671, 2021 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-33651283

RESUMEN

BACKGROUND: The tumor microenvironment (TME) is known to play a prominent role in the pathology of head and neck squamous cell carcinoma (HNSCC). Cancer-associated fibroblasts (CAFs) have been reported to regulate tumor progression, and serglycin (SRGN), one of the paracrine cytokines of CAFs, has been reported to play an important role in various signaling pathways. Hypoxia is a distinct feature of the HNSCC TME. Here, we investigated the mechanism underlying CAF-secreted SRGN leading to HNSCC progression under hypoxia. METHODS: Immunohistochemical staining was used to detect SRGN expression in clinical HNSCC samples, after which its relation with patient survival was assessed. CAFs were isolated and SRGN expression and secretion by CAFs under normoxia and hypoxia were confirmed using qRT-PCR and ELISA assays, respectively. HNSCC sphere-forming abilities, stemness-related gene expression, and chemoresistance were assessed with or without SRGN treatment. A Wnt/ß-catenin pathway inhibitor (PNU-75,654) was used to block its activation, after which nuclear translocation of ß-catenin in the presence of SRGN with or without PNU-75,654 was evaluated. shRNAs were used to stably knock down SRGN expression in CAFs. HNSCC tumor cells with or without (SRGN silenced) CAFs were inoculated submucosally in nude mice after which tumor weights and sizes were determined to assess the effects of CAFs and SRGN on tumor growth. RESULTS: We found that SRGN was expressed in both HNSCC tumor and stroma cells, and that high SRGN expression in the stroma cells, but not in the tumor cells, was significantly related to a poor patient survival. After the extraction of CAFs and normal fibroblasts (NFs) from paired tumor samples and adjacent normal tissues, respectively, we found that the expression of CAF-specific genes, including fibroblast activation protein (FAP) and alpha-smooth muscle actin (α-SMA), was clearly upregulated compared to the expression in NFs. The hypoxia marker HIF-1α was found to be expressed in tumor stroma cells. Hypoxyprobe immunofluorescence staining confirmed stromal hypoxia in an orthotopic tongue cancer mouse model. Using qRT-PCR and ELISA we found that a hypoxic TME upregulated SRGN expression and secretion by CAFs. SRGN markedly enhanced the sphere-forming ability, stemness-related gene expression and chemoresistance of HNSCC tumor cells. SRGN activated the Wnt/ß-catenin pathway and promoted ß-catenin nuclear translocation. An in vivo study confirmed that CAFs can accelerate HNSCC tumor growth, and that this effect can be counteracted by SRGN silencing. CONCLUSIONS: Our data indicate that a hypoxic tumor stroma can lead to upregulation of SRGN expression. SRGN secreted by CAFs can promote ß-catenin nuclear translocation to activate downstream signaling pathways, leading to enhanced HNSCC cell stemness, chemoresistance and accelerated tumor growth.


Asunto(s)
Fibroblastos Asociados al Cáncer/metabolismo , Hipoxia de la Célula/fisiología , Proteoglicanos/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Proteínas de Transporte Vesicular/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Proliferación Celular/fisiología , Xenoinjertos , Humanos , Ratones , Ratones Desnudos , Microambiente Tumoral/fisiología
5.
Bioact Mater ; 6(7): 2144-2157, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33511313

RESUMEN

Here, evodiamine (EVO) and the photosensitizer indocyanine green (ICG) were integrated into a liposomal nanoplatform for noninvasive diagnostic imaging and combinatorial therapy against oral squamous cell carcinoma (OSCC). EVO, as an active component extracted from traditional Chinese medicine, not only functioned as an antitumor chemotherapeutic agent but was also capable of 68Ga-chelation, thus working as a contrast agent for positron emission tomography/computed tomography (PET/CT) imaging. Moreover, EVO could exhibit peroxidase-like catalytic activity, converting endogenous tumor H2O2 into cytotoxic reactive oxygen species (ROS), enabling Chemo catalytic therapy beyond the well-known chemotherapy effect of EVO. As proven by in vitro and in vivo experiments, guided by optical imaging and PET/CT imaging, we show that the theragnostic liposomes have a significant inhibiting effect on in situ tongue tumor through photodynamic therapy combined with chemodynamic chemotherapy.

6.
Int J Clin Exp Pathol ; 13(1): 65-72, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32055274

RESUMEN

This study investigates the prognostic impact of the expression of hypoxia inducible factor (HIF)-1α and Toll-like receptor (TLR) 3 detected by immunohistochemistry in oral squamous cell carcinoma (OSCC). The study also evaluates the treatment outcome by inhibition of the HIF-1α and TLR 3 pathway (nuclear factor [NF]-κB) in an OSCC transplantation model in nude mice. Statistical analysis of immunohistochemical results with clinical findings that included overall survival outcomes was performed for 90 OSCC patients. Forty nude mice were divided into four groups (control; inhibition of HIF-1α; inhibition of NF-κB; and inhibition of HIF-1α and NF-κB). Tumor weight and immunohistochemical results of each group were compared. The results show that co-detection of low HIF-1α/TLR3 expression is significantly correlated with a better prognosis for OSCC patients. Use of an inhibitor of the HIF-1 and TLR3 pathway in an OSCC transplantation model shows a good treatment outcome.

7.
J Craniofac Surg ; 31(2): e189-e192, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-31934973

RESUMEN

OBJECTIVE: To describe the incidence, types, features, treatment and outcomes of head and neck sarcoma managed at a treatment center in eastern China. METHODS: Cases of head and neck soft tissue sarcoma and osteogenic sarcoma treated at the Stomatology Hospital of Nanjing University between 2008 and 2018 were retrospectively analyzed. Patient characteristics, site of lesion, main presenting symptoms, treatment, histology, local recurrence, development of metastatic disease, duration of follow-up and survival rates are described and compared. RESULTS: Sixty-three patients were diagnosed with head or neck sarcoma of which 42.9% had soft tissue sarcoma and 57.1% had osteogenic sarcoma. Of soft tissue sarcoma patients, the most frequently observed histologies were fibrosarcoma and malignant fibrous histiocytoma. Of 36 cases of osteogenic sarcoma, osteosarcoma, and fibrosarcoma of bone were most frequent. Mean latency period between initial symptoms and clinical presentation was 4.5 months. Radical resection was performed on 56 patients. For 33 patients, resection and radiotherapy were used and 10 patients received a triple combination of resection, chemotherapy and radiotherapy. Within the observation period, 17 patients died. CONCLUSIONS: Head and neck sarcomas, although rare, can represent a variety of pathological diagnoses. Surgery remains the main intervention although the data suggest chemotherapy, radical resection and irradiation as treatment. Outcomes are poor with high rates of local recurrence. Positive prognostic factors were tumor-free resection margins and choice of therapy. Due to the rarity of head and neck sarcoma, information remains limited and choice of treatment should be within the focus of clinical multi-center studies.


Asunto(s)
Neoplasias de Cabeza y Cuello/cirugía , Sarcoma/cirugía , Adolescente , Adulto , Anciano , Niño , Preescolar , Femenino , Neoplasias de Cabeza y Cuello/patología , Humanos , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Neoplasias de los Tejidos Blandos/patología , Neoplasias de los Tejidos Blandos/cirugía , Tasa de Supervivencia , Tomografía Computarizada por Rayos X , Adulto Joven
8.
Aging (Albany NY) ; 11(11): 3601-3623, 2019 06 06.
Artículo en Inglés | MEDLINE | ID: mdl-31170090

RESUMEN

Head and neck squamous cell carcinoma (HNSCC), a major histologic subtype of head and neck cancer, presents great mortality and morbidity worldwide. The aim of this study is to discover new potential biomarkers closely correlated with HNSCC progression. In this study, weighted gene co-expression network analysis was applied to construct a co-expression network, and the brown module was identified as the most correlated with HNSCC progression. Hub gene identification combined with survival analyses determined RHCG as a candidate biomarker for cancer progression and prognosis prediction. Further experimental results proved that RHCG was aberrantly downregulated in HNSCC tissues and cell lines. Moreover, decreased RHCG expression was shown to be associated with advanced stage and dismal prognosis in HNSCC patients. Functional assays revealed that RHCG could inhibit cell viability, clonogenicity, cell migration in vitro and suppress tumor formation in vivo. Further bioinformatics study demonstrated that DNA promoter hypermethylation of RHCG could lead to its downregulation and serve as potential prognostic maker in HNSCC. Our study reveals that RHCG acts as a tumor suppressor gene that plays a crucial role in inhibiting tumorigenicity and metastasis in HNSCC, which will shed light on the potential diagnostic and therapeutic strategies for HNSCC.


Asunto(s)
Carcinogénesis/metabolismo , Proteínas de Transporte de Catión/metabolismo , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/metabolismo , Glicoproteínas de Membrana/metabolismo , Metástasis de la Neoplasia/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Anciano , Carcinogénesis/genética , Carcinogénesis/patología , Proteínas de Transporte de Catión/genética , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular/genética , Genes Supresores de Tumor , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Glicoproteínas de Membrana/genética , Persona de Mediana Edad , Metástasis de la Neoplasia/genética , Estadificación de Neoplasias , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
9.
Cell Oncol (Dordr) ; 42(4): 459-475, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31001733

RESUMEN

BACKGROUND: Tumor-associated macrophages (TAMs) play an important role in drug resistance in many tumors, including head and neck squamous cell carcinoma (HNSCC). However, how TAMs interact with HNSCC cells to induce drug resistance, especially under hypoxic conditions, is unclear. In this study, we investigated the mechanism of TAM-induced gefitinib resistance in HNSCC cells and sought for novel therapeutic strategies. METHODS: The effects of hypoxia-treated HNSCC cells on the migration and polarization of macrophages were analyzed. Recombinant cytokine proteins and neutralizing antibodies were used as controls. In addition, we assessed the cytotoxic effects of gefitinib on HNSCC cells treated with M2-type macrophage conditioned medium, and carried out a cytokine antibody array analysis, thereby revealing the key factor CCL15. The relationship between serum CCL15 expression levels and prognosis in HNSCC patients was analyzed. In addition, we performed bioinformatic analyses to pursue the mechanisms of CCL15-induced gefitinib resistance. Finally, metformin was used to evaluate the sensitizing effects of gefitinib treatment on HNSCC cells in vitro and in vivo. RESULTS: We found that HNSCC cells recruited macrophages by secreting VEGF and polarized the macrophages to the M2 phenotype through IL-6. Conversely, we found that M2-type TAMs promoted HNSCC cell resistance to gefitinib through paracrine CCL15 signaling. The serum CCL15 levels in HNSCC patients showed a significant correlation with patient prognosis. Furthermore, we found that M2-type TAMs could suppress the sensitivity of HNSCC cells to gefitinib through the CCL15-CCR1-NF-κB pathway. In addition, we found that metformin not only inhibited CCL15 expression in M2-type TAMs enhanced by hypoxia, but also suppressed CCR1 surface expression in HNSCC cells. Encouragingly, we found that metformin sensitized HNSCC cells to gefitinib treatment in vitro and in vivo. CONCLUSIONS: Based on our data we conclude that we have identified a novel interaction between M2-type TAMs and HNSCC cells that contributes to gefitinib resistance. We also found that metformin inhibited the cross-talk between macrophages and tumor cells, thereby eliciting therapeutic effects both in vitro and in vivo.


Asunto(s)
Comunicación Celular , Gefitinib/uso terapéutico , Macrófagos/patología , Metformina/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Adulto , Anciano , Anciano de 80 o más Años , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Comunicación Celular/efectos de los fármacos , Línea Celular Tumoral , Polaridad Celular/efectos de los fármacos , Quimiocinas CC/metabolismo , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Gefitinib/farmacología , Humanos , Proteínas Inflamatorias de Macrófagos/metabolismo , Macrófagos/efectos de los fármacos , Metformina/farmacología , Persona de Mediana Edad , FN-kappa B/metabolismo , Receptores CCR1/metabolismo , Transducción de Señal/efectos de los fármacos , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Resultado del Tratamiento , Hipoxia Tumoral/efectos de los fármacos
10.
Biomed Res Int ; 2019: 5450617, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31032351

RESUMEN

BACKGROUND: Accumulating evidence has demonstrated the pivotal role of long noncoding RNAs (lncRNAs) in competing endogenous RNA (ceRNA) networks for predicting survival and evaluating prognosis in cancer patients. However, the pathogenesis of head and neck squamous cell carcinoma (HNSCC) remains unclear, and prognostic biomarkers for HNSCC are still lacking. METHODS: A total of 546 RNA sequencing profiles of HNSCC patients with clinical outcome data were obtained from the Cancer Genome Atlas (TCGA) database, providing a large sample of RNA sequencing data. From these, 71 Long noncoding RNAs lncRNAs, 8 microRNAs (miRNAs), and 16 messenger RNAs (mRNAs) were identified to construct a HNSCC-specific ceRNA network (fold change >2, P < 0.05). Univariate and multivariate Cox proportional regression models were used to assess independent indicators of prognosis. Then the expression of lncRNAs harboring prognostic value was validated in human HNSCC cell lines and tumor samples from our cohort and another two datasets from GEO (Gene Expression Omnibus) databases. RESULTS: As a result, a 3-mRNA signature and 6-lncRNA signature were identified. The six-lncRNA signature exhibited the highest prognostic value. Notably, in the six lncRNAs, HOTTIP showed the greatest prognostic value and was significantly correlated with clinical stage and histological grade of HNSCC patients. Furthermore, it was proved that HOTTIP was upregulated in HNSCC cell lines and cancerous tissues compared with corresponding normal cell lines and normal tissues. Functional assessment analysis revealed that HOTTIP might play a key role in the oncogenesis and progression of HNSCC. CONCLUSION: The present study deepened our understanding of the ceRNA-related regulatory mechanism in the pathogenesis of HNSCC and identified candidate prognostic biomarkers for clinical outcome prediction in HNSCC. HOTTIP may function as a key candidate biomarker in HNSCC and serve as a prognostic marker for HNSCC patients.


Asunto(s)
Biomarcadores de Tumor/genética , ARN Largo no Codificante/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Biología Computacional , Femenino , Regulación Neoplásica de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Estimación de Kaplan-Meier , Masculino , MicroARNs/genética , Pronóstico , Modelos de Riesgos Proporcionales , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
11.
Chem Sci ; 10(9): 2778-2784, 2019 Mar 07.
Artículo en Inglés | MEDLINE | ID: mdl-30996997

RESUMEN

Herein, a nano-integrated strategy was used to combine an anti-angiogenic agent sorafenib and a photosensitizer chlorin e6 to form carrier-free multifunctional nanoparticles (SC NPs) for synergetic anti-angiogenic therapy and phototherapy. SC NPs (diameter, ∼152 nm) presented excellent water dispersity and passive targeting ability towards tumor sites in vivo based on the enhanced permeability and retention (EPR) effect, which could be monitored by fluorescence imaging. Besides, SC NPs exhibited effective reactive oxygen species (ROS) generation and photothermal conversion abilities for both photodynamic therapy (PDT) and photothermal therapy (PTT). At a rather low dosage (200 µg kg-1) and illumination with laser (660 nm, 500 mW cm-2), SC NPs could attack tumor tissues by killing the internal tumor cells via mild phototherapy, simultaneously cutting off the external nutrient and oxygen supplements of the tumor cells via anti-angiogenesis. Besides, oxygen consumption in the PDT process may be combined with anti-angiogenic therapy to further cause cell apoptosis by tumor starvation. In addition to the highly efficient therapeutic effect in vivo, SC NPs possessed excellent biosafety and biocompatibility, making them promising for fluorescence imaging-guided synergetic anti-angiogenic therapy and phototherapy in clinic.

12.
Cancer Manag Res ; 10: 5785-5798, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30510448

RESUMEN

OBJECTIVES: The objectives of this study were to explore the mechanisms of metformin sensitization to hypoxia-induced gefitinib treatment in resistant head and neck squamous cell carcinoma (HNSCC) and evaluate the effects of this combined treatment strategy. METHODS: The effects of gefitinib treatment on HNSCC were measured under normoxic and hypoxic conditions. The relationship between hypoxia and cell cycle and epithelial-mesenchymal transition (EMT) in tumor cells were analyzed. Palbociclib and LY294002 were used in combination with gefitinib to evaluate the effects on HNSCC cell cytotoxicity during hypoxia. Finally, metformin was used to evaluate the sensitizing effects of gefitinib treatment on HNSCC in vivo and in vitro. RESULTS: Cell viability and apoptosis assays demonstrated a significant difference in HNSCC cells treated with gefitinib between the normoxia and hypoxia groups. Hypoxia induced the expression of cyclin D1, decreased the percentage of cells in G1, and promoted the EMT of tumor cells. Both palbociclib and LY294002 enhanced gefitinib-induced cytotoxicity of HNSCC cells under hypoxic conditions. Encouragingly, metformin sensitized HNSCC to gefitinib treatment in vivo and in vitro. CONCLUSION: Hypoxia promotes G1-S cell cycle progression and EMT in HNSCC, resulting in gefitinib treatment resistance. Metformin sensitizes HNSCC to gefitinib treatment, which might serve as a novel combined treatment strategy.

13.
Int J Nanomedicine ; 13: 1505-1524, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29559779

RESUMEN

BACKGROUND: Tumor microenvironment plays an important role in the chemoresistance of oral squamous cell carcinoma (OSCC). Hypoxia in the microenvironment is one of the important factors that contributes to OSCC chemoresistance; therefore overcoming hypoxia-mediated chemoresistance is one of the great challenges in clinical practice. METHODS: In this study, we developed a drug delivery system based on Pt-loaded, polyethylene glycol-modified graphene quantum dots via chemical oxidation and covalent reaction. RESULTS: Our results show that synthesized polyethylene glycol-graphene quantum dots-Pt (GPt) is about 5 nm in diameter. GPt sensitizes OSCC cells to its treatment in both normoxia and hypoxia conditions. Inductively coupled plasma-mass spectrometry assay shows that GPt enhances Pt accumulation in cells, which leads to a notable increase of S phase cell cycle arrest and apoptosis of OSCC cells in both normoxia and hypoxic conditions. Finally, compared with free cisplatin, GPt exhibits a strong inhibitory effect on the tumor growth with less systemic drug toxicity in an OSCC xenograft mouse tumor model. CONCLUSION: Taken together, our results show that GPt demonstrates superiority in combating hypoxia-induced chemoresistance. It might serve as a novel strategy for future microenvironment-targeted cancer therapy.


Asunto(s)
Antineoplásicos/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Resistencia a Antineoplásicos , Grafito/análisis , Neoplasias de la Boca/tratamiento farmacológico , Nanocompuestos/química , Platino (Metal)/uso terapéutico , Puntos Cuánticos/química , Animales , Antineoplásicos/farmacología , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/patología , Puntos de Control del Ciclo Celular/efectos de los fármacos , Hipoxia de la Célula/efectos de los fármacos , Línea Celular Tumoral , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Cisplatino/farmacología , Sistemas de Liberación de Medicamentos , Resistencia a Antineoplásicos/efectos de los fármacos , Grafito/química , Humanos , Concentración de Iones de Hidrógeno , Masculino , Ratones Endogámicos BALB C , Neoplasias de la Boca/patología , Platino (Metal)/farmacología , Microambiente Tumoral/efectos de los fármacos
14.
Sci Rep ; 7: 43907, 2017 03 06.
Artículo en Inglés | MEDLINE | ID: mdl-28262804

RESUMEN

Mucoepidermoid carcinoma (MEC) of the palate is a common malignancy of minor salivary glands. This study was designed to identify the prognostic factors for MEC of the palate. The medical records of patients diagnosed with MEC of the palate who visited the Department of Oral and Maxillofacial Surgery at Nanjing Stomatological Hospital and the Department of Stomatology at Central Hospital of Xuzhou were retrospectively studied. The prognostic factors were determined using a Cox proportional hazards model. Furthermore, the expression of cancer stem cell (CSC) markers CD44, CD133, Nanog and Sox2 were detected in neoplastic samples of these patients by immunohistochemistry. As a result, both univariate analysis and multivariate analysis proved a high histological grade and an advanced tumor stage as negative prognostic factors for overall survival. By immunohistochemistry staining and survival analysis, a combination of CD44/CD133/SOX2 was found to have the strongest prognostic value for palatal MEC patients. In conclusion, the proposed nomogram which include histological grade and tumor stage along with cancer stem cell markers provides a more accurate long-term prediction for palatal MEC patients.


Asunto(s)
Biomarcadores de Tumor/análisis , Carcinoma Mucoepidermoide/diagnóstico , Carcinoma Mucoepidermoide/patología , Neoplasias Palatinas/diagnóstico , Neoplasias Palatinas/patología , Antígeno AC133/análisis , China , Femenino , Humanos , Receptores de Hialuranos/análisis , Inmunohistoquímica , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Factores de Transcripción SOXB1/análisis , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...