Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Protein Cell ; 2024 Apr 05.
Artículo en Inglés | MEDLINE | ID: mdl-38577810

RESUMEN

Aging has a profound impact on the gingiva and significantly increases its susceptibility to periodontitis, a worldwide prevalent inflammatory disease. However, a systematic characterization and comprehensive understanding of the regulatory mechanism underlying gingival aging is still lacking. Here, we systematically dissected the phenotypic characteristics of gingiva during aging in primates and constructed the first single-nucleus transcriptomic landscape of gingival aging, by which a panel of cell type-specific signatures were elucidated. Epithelial cells were identified as the most affected cell types by aging in the gingiva. Further analyses pinpointed the crucial role of YAP in epithelial self-renew and homeostasis, which declined during aging in epithelial cells, especially in basal cells. The decline of YAP activity during aging was confirmed in the human gingival tissues, and downregulation of YAP in human primary gingival keratinocytes recapitulated the major phenotypic defects observed in the aged primate gingiva while overexpression of YAP showed rejuvenation effects. Our work provides an in-depth understanding of gingival aging and serves as a rich resource for developing novel strategies to combat aging-associated gingival diseases, with the ultimate goal of advancing periodontal health and promoting healthy aging.

2.
Cell Stem Cell ; 30(11): 1452-1471.e10, 2023 11 02.
Artículo en Inglés | MEDLINE | ID: mdl-37832549

RESUMEN

Our understanding of the molecular basis for cellular senescence remains incomplete, limiting the development of strategies to ameliorate age-related pathologies by preventing stem cell senescence. Here, we performed a genome-wide CRISPR activation (CRISPRa) screening using a human mesenchymal precursor cell (hMPC) model of the progeroid syndrome. We evaluated targets whose activation antagonizes cellular senescence, among which SOX5 outperformed as a top hit. Through decoding the epigenomic landscapes remodeled by overexpressing SOX5, we uncovered its role in resetting the transcription network for geroprotective genes, including HMGB2. Mechanistically, SOX5 binding elevated the enhancer activity of HMGB2 with increased levels of H3K27ac and H3K4me1, raising HMGB2 expression so as to promote rejuvenation. Furthermore, gene therapy with lentiviruses carrying SOX5 or HMGB2 rejuvenated cartilage and alleviated osteoarthritis in aged mice. Our study generated a comprehensive list of rejuvenators, pinpointing SOX5 as a potent driver for rejuvenation both in vitro and in vivo.


Asunto(s)
Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Rejuvenecimiento , Humanos , Ratones , Animales , Proteína HMGB2/genética , Proteína HMGB2/metabolismo , Senescencia Celular/genética , Factores de Transcripción/genética , Factores de Transcripción SOXD/genética , Factores de Transcripción SOXD/metabolismo
3.
Oral Dis ; 2023 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-37213085

RESUMEN

OBJECTIVES: Radiation injury is common after radiotherapy for head and neck cancer. Radiotherapy can reshape the immune microenvironment and cause immunosuppression, including dysregulation of immune checkpoints (ICs). However, the relationship between oral ICs expression after radiation and the development of second primary tumors is unclear. METHODS: Clinical specimens of second primary oral squamous cell carcinoma after radiotherapy (s-OSCC) and primary OSCC (p-OSCC) were collected. The expression and prognostic value of PD-1, VISTA, and TIM-3 were analyzed using immunohistochemistry. To further clarify the relationship between radiation and ICs alteration, a rat model was constructed to explore the spatiotemporal changes of ICs in the oral mucosa after radiation. RESULTS: In carcinoma tissue, the expression of TIM-3 was higher in s-OSCC than in p-OSCC, while the expression of PD-1 and VISTA was similar between the groups. In para-carcinoma tissue, the expression of PD-1, VISTA, and TIM-3 was higher in s-OSCC. High ICs expression was associated with poor survival. In the rat model, ICs were locally upregulated in the irradiated tongue. Moreover, there was a bystander effect, in which the ICs were also upregulated in the unirradiated site. CONCLUSION: Radiation may upregulate ICs expression in oral mucosa and contribute to the development of s-OSCC.

4.
Front Genet ; 13: 1065320, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36437918

RESUMEN

Retinoids, natural and synthetic derivatives of vitamin A, have many regulatory functions in human body, including regulating cellular proliferation, differentiation, apoptosis. Moreover, retinoids have been used successfully for the treatment of certain malignancies, especially acute promyelocytic leukemia (APL) in adults and neuroblastoma in children. However, retinoids have not yet been translated into effective systemic treatments for most solid cancers. Some recent studies have shown that retinoids promote tumorigenesis. Therefore, we performed this meta-analysis to systematically evaluate the efficacy of retinoids in the chemoprevention and treatment of cancers. We performed literature search of several electronic databases, including PubMed, Embase and Cochrane Library from 2000 January to 2021 November. Various outcomes were applied to investigate the potential of retinoids for prevention and treatment of cancers. The primary outcomes in this study were disease recurrence and clinical response. The secondary outcomes included overall survival (OS), cancer development, disease progression and event-free survival. We identified 39 randomized controlled trials with 15,627 patients in this study. Our results showed that lower recurrence rate and better clinical response were obtained in retinoids treated patients with cancer or premalignancy as compared with control. The differences were statistically significant (RR = 0.85, 95% CI = 0.74-0.96, p = 0.01; RR = 1.24, 95% CI = 1.03-1.49, p = 0.02, respectively). Retinoids treatment was not associated with improvement in overall survival, cancer development, disease progression or event-free survival. Subgroup analysis conducted based on cancer type showed that patients benefited from retinoids treatment in APL, renal cell carcinoma, hepatocellular carcinoma, lung cancer, Kaposi sarcoma, and complete hydatidiform mole. No significant therapeutic effect was noted in head and neck cancer, acute myeloid leukemia (AML), melanoma, breast cancer, bladder cancer, cervical intraepithelial neoplasia (CIN) or cervical carcinoma. Subgroup analysis based on tumor classification demonstrated that retinoids group obtained a lower recurrence rate and better clinical response than control group in solid cancers. In conclusion, clinical application of retinoids was associated with reduction in disease recurrence and improvement in clinical response, illustrating that retinoids play a key role in cancer prevention and therapy. Further research is needed to broaden the utility of retinoids in other types of cancers. Systematic Review Registration: PROSPERO, identifier CRD42022296706.

5.
Signal Transduct Target Ther ; 7(1): 374, 2022 11 07.
Artículo en Inglés | MEDLINE | ID: mdl-36336680

RESUMEN

Aging is accompanied by the decline of organismal functions and a series of prominent hallmarks, including genetic and epigenetic alterations. These aging-associated epigenetic changes include DNA methylation, histone modification, chromatin remodeling, non-coding RNA (ncRNA) regulation, and RNA modification, all of which participate in the regulation of the aging process, and hence contribute to aging-related diseases. Therefore, understanding the epigenetic mechanisms in aging will provide new avenues to develop strategies to delay aging. Indeed, aging interventions based on manipulating epigenetic mechanisms have led to the alleviation of aging or the extension of the lifespan in animal models. Small molecule-based therapies and reprogramming strategies that enable epigenetic rejuvenation have been developed for ameliorating or reversing aging-related conditions. In addition, adopting health-promoting activities, such as caloric restriction, exercise, and calibrating circadian rhythm, has been demonstrated to delay aging. Furthermore, various clinical trials for aging intervention are ongoing, providing more evidence of the safety and efficacy of these therapies. Here, we review recent work on the epigenetic regulation of aging and outline the advances in intervention strategies for aging and age-associated diseases. A better understanding of the critical roles of epigenetics in the aging process will lead to more clinical advances in the prevention of human aging and therapy of aging-related diseases.


Asunto(s)
Envejecimiento , Epigénesis Genética , Animales , Humanos , Epigénesis Genética/genética , Envejecimiento/genética , Metilación de ADN/genética , Ensamble y Desensamble de Cromatina , Procesamiento Proteico-Postraduccional
6.
Oral Dis ; 2022 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-36444706

RESUMEN

PURPOSE: This study aims to clarify whether blood lipid profiles are indicators of prognosis in patients with head and neck squamous cell carcinoma (HNSCC). METHODS: This retrospective study included 512 T1/2N0M0 HNSCC patients. The correlation between blood lipid profiles and progression-free survival (PFS) and disease-specific survival (DSS) was analyzed by multivariate analysis. The data from TCGA was also analyzed to investigate the expression levels and prognostic values of different lipoprotein receptors essential for specific lipid uptake. RESULTS: A high level of low-density lipoprotein cholesterol (LDL-C) indicated better PFS and DSS, and a low level of apolipoprotein A-I (Apo A-I) indicated better PFS, while a high level of apolipoprotein B (Apo B) indicated poorer PFS and DSS. The Apo A-I receptor gene SCARB1 was upregulated and associated with poor survival in HNSCC patients. Activation of SCARB1 was implicated in a series of tumor-promoting pathways. There was no significant correlation between the expression of LDL-C and Apo B-related receptors and prognosis. CONCLUSION: A high level of LDL-C and a low level of Apo A-I are protective factors for HNSCC, while a high level of Apo B is a risk factor. The upregulation of SCARB1 may participate in the progression of HNSCC.

7.
World J Stem Cells ; 14(2): 146-162, 2022 Feb 26.
Artículo en Inglés | MEDLINE | ID: mdl-35432735

RESUMEN

Cancer stem cells (CSCs) comprise a subpopulation of cancer cells with stem cell properties, which exhibit the characteristics of high tumorigenicity, self-renewal, and tumor initiation and are associated with the occurrence, metastasis, therapy resistance, and relapse of cancer. Compared with differentiated cells, CSCs have unique metabolic characteristics, and metabolic reprogramming contributes to the self-renewal and maintenance of stem cells. It has been reported that CSCs are highly dependent on lipid metabolism to maintain stemness and satisfy the requirements of biosynthesis and energy metabolism. In this review, we demonstrate that lipid anabolism alterations promote the survival of CSCs, including de novo lipogenesis, lipid desaturation, and cholesterol synthesis. In addition, we also emphasize the molecular mechanism underlying the relationship between lipid synthesis and stem cell survival, the signal trans-duction pathways involved, and the application prospect of lipid synthesis reprogramming in CSC therapy. It is demonstrated that the dependence on lipid synthesis makes targeting of lipid synthesis metabolism a promising therapeutic strategy for eliminating CSCs. Targeting key molecules in lipid synthesis will play an important role in anti-CSC therapy.

8.
Cell Death Dis ; 12(10): 946, 2021 10 14.
Artículo en Inglés | MEDLINE | ID: mdl-34650054

RESUMEN

Although obesity has been associated with an increased risk and aggressiveness of many types of carcinoma, whether it promotes squamous cell carcinoma remains unclear. To reveal the role of obesity in oral squamous cell carcinoma (OSCC) initiation and development, we used 4NQO-induced OSCC model mice to examine the impact of dietary obesity on carcinogenesis. The results showed that high-fat diet (HFD)-induced obesity significantly promoted the incidence of OSCC and altered the local immune microenvironment with the expansion of CD11b+Gr1+ myeloid-derived suppressor cells (MDSCs). The underlying mechanism that induced an immunosuppressive local microenvironment in obesity was the recruitment of MDSCs through the CCL9/CCR1 axis and enhancement of MDSC immunosuppressive function via intracellular fatty acid uptake. Furthermore, clinical samples verified the increase in infiltrated CD33+ (a marker of human MDSCs) cells in obese OSCC patients, and data from the TCGA dataset confirmed that CD33 expression was positively correlated with local adipocytes in OSCC. Survival analysis showed that enrichment of adipocytes and high expression of CD33 were associated with poor prognosis in OSCC patients. Strikingly, depletion of MDSCs significantly ameliorated HFD-promoted carcinogenesis in 4NQO-induced model mice. These findings indicate that obesity is also an important risk factor for OSCC, and cancer immunotherapy, especially targeting MDSCs, may exhibit greater antitumor efficacy in obese patients.


Asunto(s)
Carcinogénesis/patología , Neoplasias de la Boca/etiología , Neoplasias de la Boca/patología , Células Supresoras de Origen Mieloide/patología , Obesidad/complicaciones , 4-Nitroquinolina-1-Óxido , Adipocitos/metabolismo , Animales , Antígenos Ly , Antígeno CD11b/metabolismo , Quimiocinas CC , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Humanos , Terapia de Inmunosupresión , Proteínas Inflamatorias de Macrófagos , Masculino , Ratones Endogámicos C57BL , Modelos Biológicos , Quinolonas , Receptores CCR1/metabolismo , Lectina 3 Similar a Ig de Unión al Ácido Siálico/metabolismo , Transducción de Señal , Análisis de Supervivencia , Lengua/metabolismo , Lengua/patología , Microambiente Tumoral/efectos de los fármacos
9.
Biomed Pharmacother ; 139: 111545, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33873145

RESUMEN

Tacrolimus (TAC, FK506) is a major calcineurin inhibitor and has been commonly used in treatments of patients with organ transplants and immune diseases. Moreover, tacrolimus is recommended by the treatment guidelines for oral potentially malignant disorders (OPMDs) such as oral lichen planus (OLP). However, whether tacrolimus increases the risk of cancer remains controversial. We observed that in a 4-Nitroquinoline N-oxide (4NQO)-induced oral carcinogenesis model, tacrolimus treatment was associated with a significantly lower ratio of cancer formation (52.94% vs. 90%) and a lower proportion of Ki67 and proliferation cell nuclear antigen (PCNA) -positive cells in lesion areas (P < 0.001). Liver, kidney, and lung functions of rats and the tumor immune microenvironment of the tongue were not affected. These observations suggest that tacrolimus blocked oral carcinogenesis through epithelial cell proliferation inhibition, independent of its immunosuppressive effects. As a processing factor, tacrolimus decreased tumor formation and cell proliferation in different stages of oral squamous cell carcinoma (OSCC) progression in vivo and in vitro. Furthermore, we investigated effects on the cell cycle and expression of related proteins. Tacrolimus induced G1/S phase arrest and significantly downregulated the expression of cyclinD1, cyclinE1, and c-Myc. These results suggest that tacrolimus induces G1/S phase arrest via inhibition of cyclinD1, cyclinE1, and c-Myc expression and retards oral cell carcinogenesis in vitro and in vivo. Thus, application of tacrolimus is a safe therapeutic strategy for treating OPMDs.


Asunto(s)
Anticarcinógenos/farmacología , Ciclo Celular/efectos de los fármacos , Neoplasias de la Boca/prevención & control , Tacrolimus/farmacología , 4-Nitroquinolina-1-Óxido , Animales , Carcinógenos , Microambiente Celular/efectos de los fármacos , Ciclinas/antagonistas & inhibidores , Ciclinas/biosíntesis , Genes myc/efectos de los fármacos , Antígeno Ki-67 , Masculino , Ratones , Ratones Endogámicos BALB C , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/patología , Antígeno Nuclear de Célula en Proliferación/análisis , Ratas , Ratas Sprague-Dawley , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/prevención & control , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Cell Death Dis ; 11(10): 925, 2020 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-33116117

RESUMEN

CDK4/6 inhibitors show promising antitumor activity in a variety of solid tumors; however, their role in head and neck squamous cell carcinoma (HNSCC) requires further investigation. The senescence-associated secretory phenotype (SASP) induced by CDK4/6 inhibitors has dual effects on cancer treatment. The need to address the SASP is a serious challenge in the clinical application of CDK4/6 inhibitors. We investigated whether metformin can act as a senostatic drug to modulate the SASP and enhance the anticancer efficacy of CDK4/6 inhibitors in HNSCC. In this study, the efficacy of a combination of the CDK4/6 inhibitor LY2835219 and metformin in HNSCC was investigated in in vitro assays, an HSC6 xenograft model, and a patient-derived xenograft model. Senescence-associated ß-galactosidase staining, antibody array, sphere-forming assay, and in vivo tumorigenesis assay were used to detect the impacts of metformin on the senescence and SASP induced by LY2835219. We found that LY2835219 combined with metformin synergistically inhibited HNSCC by inducing cell cycle arrest in vitro and in vivo. Metformin significantly modulated the profiles of the SASP elicited by LY2835219 by inhibiting the mTOR and stat3 pathways. The LY2835219-induced SASP resulted in upregulation of cancer stemness, while this phenomenon can be attenuated when combined with metformin. Furthermore, results showed that the stemness inhibition by metformin was associated with blockade of the IL6-stat3 axis. Survival analysis demonstrated that overexpression of IL6 and stemness markers was associated with poor survival in HNSCC patients, indicating that including metformin to target these proteins might improve patient prognosis. Collectively, our data suggest that metformin can act as a senostatic drug to enhance the anticancer efficacy of CDK4/6 inhibitors by reprogramming the profiles of the SASP.


Asunto(s)
Antineoplásicos/uso terapéutico , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/metabolismo , Metformina/uso terapéutico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Animales , Antineoplásicos/farmacología , Humanos , Metformina/farmacología , Ratones , Ratones Desnudos , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Cancer Med ; 9(10): 3489-3499, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32162845

RESUMEN

BACKGROUND: Alanine-serine-cysteine transporter 2 (ASCT2), a major glutamine transporter, is essential for cell growth and tumor development in a variety of cancers. However, the clinicopathological significance and pathological role of ASCT2 in OSCC (oral squamous cell carcinoma) lesions remain unclear. METHODS: Sections from 89 OSCC patients and 10 paracancerous tissue controls were stained by immunohistochemistry (IHC) to detect the expression of ASCT2, glutaminase, and Ki-67. Survival analysis was carried out to determine the predictive value of ASCT2 expression using the log-rank test. Moreover, the critical role of ASCT2 in tumor growth was determined by a series of in vitro and in vivo assays. Cell Counting Kit-8 (CCK8), Western Blotting (WB), Reactive Oxygen Species (ROS), and Glutathione (GSH) detection were applied to explore the molecular mechanism of ASCT2 involvement in tumor development. RESULTS: In OSCC lesions, ASCT2 expression was significantly increased and associated with cell proliferation index (Ki-67) and GLS expression. Moreover, survival analysis showed that OSCC patients with high ASCT2 expression had lower overall survival (P = 0.0365). In OSCC cell lines, the high level of ASCT2 was inherent and related to the glutamine addiction of tumor cells. In vitro and in vivo functional experiments revealed that targeted silencing of ASCT2 can effectively inhibit OSCC cell proliferation and tumor growth. Mechanistically, targeting ASCT2 knockdown reduced glutamine uptake and intracellular GSH levels, which contribute to the accumulation of ROS and induce apoptosis in OSCC cells. CONCLUSION: ASCT2 is a significant factor for predicting overall survival in patients with OSCC, and targeting ASCT2 to inhibit glutamine metabolism may be a promising strategy for OSCC treatment.


Asunto(s)
Sistema de Transporte de Aminoácidos ASC/genética , Proliferación Celular/genética , Glutamina/metabolismo , Antígenos de Histocompatibilidad Menor/genética , Neoplasias de la Boca/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Anciano , Sistema de Transporte de Aminoácidos ASC/metabolismo , Animales , Línea Celular Tumoral , Femenino , Técnicas de Silenciamiento del Gen , Glutaminasa/metabolismo , Glutatión/metabolismo , Humanos , Inmunohistoquímica , Masculino , Ratones , Ratones Desnudos , Persona de Mediana Edad , Antígenos de Histocompatibilidad Menor/metabolismo , Neoplasias de la Boca/metabolismo , Trasplante de Neoplasias , Especies Reactivas de Oxígeno/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo
12.
Oral Oncol ; 89: 14-22, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30732952

RESUMEN

OBJECTIVES: Obesity is an important risk factor for several malignancies, but its effect on oral squamous cell carcinoma (OSCC) prognosis is controversial. We aimed to disclose the association between obesity and the OSCC outcome, and explore the potential of some lipid metabolism-related genes as biomarkers for prognostic prediction. MATERIALS AND METHODS: A total of 576 patients diagnosed as T1/2N0M0 OSCC without prediagnosis weight loss was included in this retrospective study. These patients were grouped according to body mass index (BMI). The univariate and multivariate analysis were used to compare the progression-free survival (PFS) and disease specific survival (DSS) between groups. Propensity score matching (PSM) was adopted to minimize confounders. Data from Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) were employed to analyze the potential of some lipid metabolism-related genes for OSCC prognosis prediction. RESULTS: The PFS (P = 0.023) and DSS (P = 0.047) were poorer in obese patients than in normal weight ones. Obesity was an independent risk factor for PFS (Hazard Ratio = 2.016, 95% Confidence Interval 1.101-3.693, P = 0.023) and DSS (Hazard Ratio = 2.022, 95% Confidence Interval 1.040-3.932, P = 0.038). Furthermore, the PSM matched cohort analysis revealed that obesity was associated with poor prognosis of OSCC patients. Finally, 72 dysregulated lipid metabolism-related genes were identified in OSCC, and a combining signature of TGFB1, SPP1, and SERPINE1 was defined as a biomarker for prognostic prediction. CONCLUSIONS: Obesity is an independent risk factor for T1/2N0M0 OSCC, and a combining signature of TGFB1, SPP1, and SERPINE1 may be applied to predict prognosis of OSCC patients.


Asunto(s)
Metabolismo de los Lípidos/genética , Obesidad/genética , Adulto , Anciano , Anciano de 80 o más Años , Carcinoma de Células Escamosas/mortalidad , Carcinoma de Células Escamosas/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/mortalidad , Neoplasias de la Boca/patología , Pronóstico , Estudios Retrospectivos , Tasa de Supervivencia , Adulto Joven
13.
Cancer Lett ; 434: 172-183, 2018 10 10.
Artículo en Inglés | MEDLINE | ID: mdl-30026052

RESUMEN

The dysregulation of glycolysis has been suggested to lead to alteration of cell drug resistance signals, proliferation and metastasis. Emerging evidence indicates that lncRNAs play a key role in the cellular processes of tumor cells, including glycolysis, growth, and movement. However, the role and potential mechanism of lncRNAs in glycolysis-mediated metastasis has not been explored. In this study, we identified a novel lncRNA lnc-p23154 which is associated with OSCC patient metastasis and the promotion of OSCC cell migration and invasion in vitro and in vivo. Furthermore, we found that lnc-p23154 also participates in OSCC glycolysis by facilitating Glut1 expression. Rescue of lnc-p23154 reversed the suppression of OSCC cell migration and invasion induced by Glut1 knockdown. In addition, lnc-p23154 is mainly located in the nucleus and binds to the promoter region of miR-378a-3p, which represses Glut1 expression by targeting to its 3'UTR directly. Therefore, we concluded that lnc-p23154 may play an important role in Glut1-mediated glycolysis by inhibiting miR-378a-3p transcription and accelerate OSCC metastasis.


Asunto(s)
Carcinoma de Células Escamosas/genética , Regulación Neoplásica de la Expresión Génica , Transportador de Glucosa de Tipo 1/genética , Glucólisis/genética , Neoplasias de la Boca/genética , ARN Largo no Codificante/genética , Regiones no Traducidas 3'/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular , Línea Celular Tumoral , Movimiento Celular/genética , Femenino , Transportador de Glucosa de Tipo 1/metabolismo , Células HEK293 , Humanos , Masculino , MicroARNs/genética , Persona de Mediana Edad , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Invasividad Neoplásica , Metástasis de la Neoplasia
14.
Cancer Med ; 7(4): 1056-1069, 2018 04.
Artículo en Inglés | MEDLINE | ID: mdl-29479858

RESUMEN

Radiotherapy for nasopharyngeal carcinoma has been reported to cause second primary oral squamous cell carcinoma (s-OSCC). The prognosis and pathologic characteristic of s-OSCC are largely unknown. Bmi1 was associated with the repair of radiation-induced DNA damage, suggesting its possible involvement in the pathologic process of s-OSCC. Herein, we compared the prognosis between s-OSCC and primary OSCC (p-OSCC) and explored the involvement of Bmi1 in s-OSCC development. In this retrospective study, s-OSCC and p-OSCC patients were matched by propensity scores. Their outcomes were compared by univariate and multivariate analyses. The expression of Bmi1 in s-OSCC and p-OSCC was detected by immunohistochemistry (IHC). Radiation-induced Bmi1 alteration in early-stage was explored in a rat model and HaCaT cells. After matching, 116 pairs of patients with highly balanced characteristics were included. In univariate analysis, the overall survival (OS), disease-specific survival (DSS), and local recurrence-free survival (LRFS) were poorer in s-OSCC than in p-OSCC (P < 0.05), while their regional metastasis-free survival (RMFS) was parallel (P = 0.112). Multivariate analysis further revealed that radiotherapy history was an independent risk factor for OS, DSS, and LRFS (P < 0.05). IHC results showed that the positive rate of Bmi1 was higher in s-OSCC (P = 0.0027). In a rat model of radiotherapy-induced mucositis, Bmi1 upregulation was observed 8 days after irradiation. Consistently, Bmi1 was upregulated in HaCaT cells 1 h after irradiation, and its upregulation was in accord with X-ray exposure duration. In conclusion, the prognosis of s-OSCC is poorer as compared to p-OSCC, which may be attributed to Bmi1 upregulation.


Asunto(s)
Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/mortalidad , Regulación Neoplásica de la Expresión Génica , Neoplasias de la Boca/etiología , Neoplasias de la Boca/mortalidad , Neoplasias Primarias Secundarias/etiología , Neoplasias Primarias Secundarias/mortalidad , Complejo Represivo Polycomb 1/genética , Animales , Biomarcadores de Tumor , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Neoplasias de la Boca/patología , Neoplasias Primarias Secundarias/patología , Complejo Represivo Polycomb 1/metabolismo , Pronóstico , Radioterapia/efectos adversos , Ratas , Factores de Riesgo
15.
Int J Oncol ; 52(3): 733-742, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29328386

RESUMEN

The majority of cases of oral squamous cell carcinoma (OSCC) develop from oral potentially malignant disorders, which have been confirmed to be involved in chronic oxidative stimulation. However, no effective treatment approaches have been used to prevent the development of dysplasia into cancerous lesions thus far. In the present study, a well-established OSCC model was used to detect proteomics profiles at different stages during oral malignant transformation. Of the 15 proteins that were found to be upregulated in both the dysplasia and carcinoma stages, the oxidative stress-associated proteins, thioredoxin-1 (Trx-1), glutaredoxin-1 and peroxiredoxin-2 were note as the proteins with significant changes in expression Trx-1 was identified to be the most significantly upregulated protein in the precancerous stage. Validation experiments confirmed that Trx-1 was overexpressed both in dysplasia and cancerous tissue samples, and the inhibition of Trx-1 was able to promote the apoptosis of OSCC cells under hypoxic conditions. Furthermore, the experimental application of a Trx-1-specific inhibitory agent in an animal model led to a lower cancerization rate and a delay in tumor formation. The possible mechanisms were associated with the increased apoptosis via a reactive oxygen species (ROS)-dependent pathway. Taken together, our findings indicate that Trx-1 may be an important target for delaying oral malignant transformation, which provides a novel therapeutic strategy for the prevention and treatment of OSCC.


Asunto(s)
Carcinoma de Células Escamosas/patología , Transformación Celular Neoplásica/patología , Neoplasias de la Boca/patología , Lesiones Precancerosas/patología , Tiorredoxinas/metabolismo , 4-Nitroquinolina-1-Óxido/toxicidad , Adulto , Anciano , Animales , Apoptosis , Biomarcadores de Tumor/metabolismo , Carcinogénesis/efectos de los fármacos , Carcinogénesis/patología , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/cirugía , Línea Celular Tumoral , Transformación Celular Neoplásica/efectos de los fármacos , Disulfuros/farmacología , Femenino , Humanos , Imidazoles/farmacología , Masculino , Persona de Mediana Edad , Mucosa Bucal/patología , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/cirugía , Neoplasias Experimentales/inducido químicamente , Neoplasias Experimentales/patología , Procedimientos Quirúrgicos Ortognáticos , Estrés Oxidativo , Lesiones Precancerosas/inducido químicamente , Proteómica , Quinolonas/toxicidad , Ratas Sprague-Dawley , Especies Reactivas de Oxígeno/metabolismo , Tiorredoxinas/antagonistas & inhibidores , Lengua/patología
16.
Sci Rep ; 6: 20208, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26831400

RESUMEN

Head and neck squamous cell carcinoma (HNSCC) development is a multistage process includes the normal, dysplasia and squamous cell carcinoma (SCC) stages. Recently, increasing evidence has suggested that the tumor microenvironment (TME) is an integral part of malignant transformation. Exploring certain key node genes in TME for future intervention in dysplasia to interrupt oral carcinogenesis was the primary goal of this research. To achieve this goal, systems biology approaches were first applied to the epithelia and fibroblasts collected at sequential stages in a 4-nitroquinoline-1-oxide (4NQO) -induced rat oral carcinogenesis model. Through bioinformatics network construction, IL-1ß was identified as one of the key node genes in TME during carcinogenesis. Immunohistochemical staining of human and rat samples demonstrated that IL-1ß expression patterns were parallel to the stages of malignant transformation. Silencing IL-1ß with lentivirus-delivered shRNA significantly inhibited oral squamous cell carcinoma cell growth both in vivo and in vitro. Based on these findings, we hypothesized that IL-1ß may be a chemoprevention target in TME during oral carcinogenesis. Therefore, we targeted IL-1 in the TME by oral mucosal injection of an IL-1 receptor antagonist in 4NQO rats. The results demonstrated that targeting IL-1 could interrupt oral carcinogenesis by reprogramming the TME.


Asunto(s)
Transformación Celular Neoplásica/genética , Interleucina-1beta/genética , Neoplasias/etiología , Neoplasias/patología , Microambiente Tumoral/genética , 4-Nitroquinolina-1-Óxido/administración & dosificación , 4-Nitroquinolina-1-Óxido/efectos adversos , Administración Oral , Animales , Carcinoma de Células Escamosas/etiología , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/metabolismo , Análisis por Conglomerados , Modelos Animales de Enfermedad , Transición Epitelial-Mesenquimal , Fibroblastos , Perfilación de la Expresión Génica , Silenciador del Gen , Xenoinjertos , Interleucina-1beta/metabolismo , Masculino , Modelos Biológicos , Mucosa Bucal/efectos de los fármacos , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Ratas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA