Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Mol Carcinog ; 63(4): 563-576, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38085124

RESUMEN

Oral squamous cell carcinoma is the predominant subtype of head and neck squamous cell carcinoma, characterized by a challenging prognosis. In this study, we established a murine model of oral carcinogenesis using 4-nitroquinoline-1-oxide (4-NQO) induction to investigate the impact of immunotherapy on microenvironmental alterations. Mice in the precancerous condition were randomly divided into two groups: one receiving programmed death-1 (PD1) monoclonal antibody treatment and the other, control immunoglobulin G. Our observations showed that while PD1 blockade effectively delayed the progression of carcinogenesis, it did not completely impede or reverse it. To unravel the underlying reasons for the limited effectiveness of PD1 blockade, we collected tongue lesions and applied mass cytometry (CyTOF) and RNA sequencing (RNA-seq) to characterize the microenvironment. CyTOF analysis revealed an increased macrophage subset (expressing high levels of IFNγ and iNOS) alongside a diminished Th1-like subset (exhibiting low expression of TCF7) and three myeloid-derived suppressor cell subsets (displaying low expression of MHC Class II or IFNγ) following anti-PD1 treatment. Notably, we observed an increased presence of cancer-associated fibroblasts (CAFs) expressing collagen-related genes after PD1 blockade. Furthermore, we found a negative correlation between the infiltration levels of CAFs and CD8+ T cells. These findings were validated in murine tongue tissue slides, and publicly available multi-omics datasets. Our results suggest that CAFs may impair the therapeutic efficacy of PD1 blockade in oral carcinogenesis by the remodeling of the extracellular matrix.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Ratones , Animales , Carcinoma de Células Escamosas/patología , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/genética , Linfocitos T CD8-positivos , Carcinogénesis , Carcinoma de Células Escamosas de Cabeza y Cuello , Perfilación de la Expresión Génica , Microambiente Tumoral
2.
J Oral Pathol Med ; 52(2): 127-135, 2023 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-36285599

RESUMEN

BACKGROUND: Epithelial-mesenchymal transition is one of the main mechanisms for tumor progression and metastasis. Transcription factors such as TWIST1 are key regulators of the epithelial-mesenchymal transition and are regarded as potential therapeutic targets for the treatment of cancer. The purpose of this study was to examine TWIST1 as a possible epithelial-mesenchymal transition-related prognostic biomarker in oral epithelial dysplasia and oral tongue squamous cell carcinomas, as well as the biological behavior of TWIST1-silencing in oral tongue squamous cell carcinomas cell lines. METHODS: Immunohistochemical analysis of TWIST1, E-cadherin, and N-cadherin was carried out in 47 samples representing oral epithelial dysplasia and 41 oral tongue squamous cell carcinomas. The suppression of TWIST1 expression was performed using shRNA-expression vectors in HSC-3 and SCC-9 cells to investigate in vitro the impact of TWIST1 on proliferation, apoptosis, viability, migration, and invasion of SCC-9 and HSC-3 cells. RESULTS: The expression of nuclear TWIST1 was significantly higher in oral tongue squamous cell carcinomas than in oral epithelial dysplasis (p < 0.0001), whereas TWIST1 in the cytoplasm was more expressed in oral epithelial dysplasis (p = 0.012). The high cytoplasmic expression of TWIST1 was significantly associated with shortened overall survival (p < 0.05), and increased nuclear TWIST1 expression was significantly related to high risk of recurrence (p = 0.03). Knockdown of TWIST1 in oral tongue squamous cell carcinomas cells induced the expression of E-cadherin and inhibited N-cadherin, which were followed by decreased proliferation, migration, and invasion. CONCLUSIONS: Our research suggests that TWIST1 is linked to the development of oral tongue carcinogenesis and may be used as a prognostic indicator and therapeutic target for oral tongue squamous cell carcinomas patients.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Lengua , Humanos , Carcinoma de Células Escamosas de Cabeza y Cuello , Carcinoma de Células Escamosas/patología , Pronóstico , Neoplasias de la Lengua/patología , Cadherinas/metabolismo , Transición Epitelial-Mesenquimal/fisiología , Proteína 1 Relacionada con Twist/metabolismo , Proliferación Celular , Línea Celular Tumoral , Movimiento Celular , Proteínas Nucleares
3.
J Oral Pathol Med ; 52(10): 1004-1012, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37817274

RESUMEN

BACKGROUND: Compelling evidence implicates diabetes-associated hyperglycemia as a promoter of tumor progression in oral potentially malignant disorders (OPMD). Yet, information on hyperglycemia-induced cell signaling networks in oral oncology remains limited. Our group recently reported that glucose-rich conditions significantly enhance oral dysplastic keratinocyte viability and migration through epidermal growth factor receptor (EGFR) activation, a pathway strongly linked to oral carcinogenesis. Here, we investigated the basal metabolic phenotype in these cells and whether specific glucose-responsive EGFR ligands mediate these responses. METHODS: Cell energy phenotype and lactate concentration were evaluated via commercially available assays. EGFR ligands in response to normal (5 mM) or high (20 mM) glucose were analyzed by quantitative real-time PCR, ELISA, and western blotting. Cell viability and migration assays were performed in the presence of pharmacological inhibitors or RNA interference. RESULTS: When compared to normal keratinocytes, basal glycolysis in oral dysplastic keratinocytes was significantly elevated. In highly glycolytic cells, high glucose-activated EGFR increasing viability and migration. Notably, we identified amphiregulin (AREG) as the predominant glucose-induced EGFR ligand. Indeed, enhanced cell migration in response to high glucose was blunted by EGFR inhibitor cetuximab and AREG siRNA. Conversely, AREG treatment under normal glucose conditions significantly increased cell viability, migration, lactate levels, and expression of glycolytic marker pyruvate kinase M2. CONCLUSION: These novel findings point to AREG as a potential high glucose-induced EGFR activating ligand in highly glycolytic oral dysplastic keratinocytes. Future studies are warranted to gain more insight into the role of AREG in hyperglycemia-associated OPMD tumor progression.


Asunto(s)
Diabetes Mellitus , Hiperglucemia , Neoplasias , Humanos , Anfirregulina/genética , Anfirregulina/metabolismo , Glucosa/farmacología , Glucosa/metabolismo , Ligandos , Receptores ErbB/metabolismo , Familia de Proteínas EGF/metabolismo , Queratinocitos/metabolismo , Hiperglucemia/complicaciones , Hiperglucemia/metabolismo , Carcinogénesis/metabolismo , Lactatos/metabolismo
4.
Mol Biol Rep ; 49(9): 8369-8380, 2022 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-35713797

RESUMEN

BACKGROUND: On the background of the epidemiological link between diabetes and oral cancer, the present study aimed to analyze the potential involvement of selected glucose transporters (GLUT1/GLUT3/GLUT4), if any, in such putative association. METHODS AND RESULTS: Oral carcinogenesis was induced by 4-nitroquinoline N-oxide in 10 non-diabetic and 10 diabetic rats; 8 non-diabetic and 7 diabetic rats served as controls. Expressions of selected GLUTs at mRNA and protein levels were analyzed in oral tissue (normal/lesion) by quantitative real-time PCR and immunohistochemistry respectively. Premalignant lesions (hyperplasia/dysplasia/carcinoma-in-situ) appeared on tongues of carcinogen-treated animals. Significant increase of GLUT1mRNA level was seen from normal to lesion tongues, along increasing lesion grades (from hyperplasia/mild dysplasia to moderate/severe dysplasia) and in lesions induced under hyperglycemic condition than that induced under normoglycemic one; a similar trend was found in transcript variant-1 of GLUT1, but not in variant-2. GLUT3 and GLUT4 mRNA levels were comparable among lesions irrespective of grades and glycemic status. Concordant to mRNA level, overall expression of GLUT1 protein was higher in tongue lesions in presence of hyperglycemia than in absence of such condition; non-lesion portions of tongues exposed to carcinogen showed a similar trend. Moreover in carcinogen-treated groups, non-lesion and lesion portions of tongues under hyperglycemic condition showed predominantly membranous expression for GLUT1 which was again significantly higher than equivalent portions of tongue under normoglycemic condition. CONCLUSION: Hyperglycemia seemed to favor GLUT1 over-expression and membrane localization of the protein during oral carcinogenesis. GLUT1 transcript variant-1 appeared to be more important than variant-2 in disease pathogenesis.


Asunto(s)
Diabetes Mellitus Experimental , Hiperglucemia , Animales , Carcinogénesis/genética , Carcinógenos/toxicidad , Diabetes Mellitus Experimental/metabolismo , Transportador de Glucosa de Tipo 1/genética , Transportador de Glucosa de Tipo 3 , Hiperglucemia/inducido químicamente , Hiperglucemia/genética , Hiperglucemia/metabolismo , Hiperplasia , ARN Mensajero/genética , Ratas
5.
J Biochem Mol Toxicol ; 36(6): e23029, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35243731

RESUMEN

The objective of this study is to examine the chemopreventive effects of Nerolidol (NER) on hamster buccal pouch carcinogenesis (HBC) induced by 7,12-dimethylbenz(a)anthracene (DMBA) in male golden Syrian hamsters. In this study, oral squamous cell carcinoma was developed in the buccal pouch of an oral painted hamster with 0.5% DMBA in liquid paraffin three times weekly for 12 weeks. To assess DMBA-induced hamster buccal tissue carcinogenesis, biochemical endpoints such as Phase I and II detoxification enzymes, antioxidants, lipid peroxidation (LPO) by-products, and renal function markers, as well as histopathological examinations, were used. Furthermore, the immunohistochemical studies of interleukin-6 were investigated to find the inflammatory link in the HBC carcinogenesis. In our results, DMBA alone exposed hamsters showed 100% tumor growth, altered levels of antioxidants, detoxification agents, LPO, and renal function identifiers as compared to the control hamsters. The outcome in  present biochemical, histopathological, and immunohistochemistry studies has been found a reverse in NER-treated hamsters against the tumor. This study concluded that NER modulated the biochemical profiles (antioxidants, detoxification, LPO, and renal function markers) and inhibited tumor development in DMBA induced oral carcinogenesis.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de la Boca , 9,10-Dimetil-1,2-benzantraceno/toxicidad , Animales , Antioxidantes/efectos adversos , Carcinogénesis , Carcinógenos/toxicidad , Carcinoma de Células Escamosas/patología , Cricetinae , Masculino , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/prevención & control , Sesquiterpenos
6.
Int J Mol Sci ; 23(3)2022 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-35163557

RESUMEN

Betel quid (BQ) is a package of mixed constituents that is chewed by more than 600 million people worldwide, particularly in Asia. The formulation of BQ depends on a variety of factors but typically includes areca nut, betel leaf, and slaked lime and may or may not contain tobacco. BQ chewing is strongly associated with the development of potentially malignant and malignant diseases of the mouth such as oral submucous fibrosis (OSMF) and oral squamous cell carcinoma (OSCC), respectively. We have shown recently that the constituents of BQ vary geographically and that the capacity to induce disease reflects the distinct chemical composition of the BQ. In this review, we examined the diverse chemical constituents of BQ and their putative role in oral carcinogenesis. Four major areca alkaloids-arecoline, arecaidine, guvacoline and guvacine-together with the polyphenols, were identified as being potentially involved in oral carcinogenesis. Further, we propose that fibroblast senescence, which is induced by certain BQ components, may be a key driver of tumour progression in OSMF and OSCC. Our study emphasizes that the characterization of the detrimental or protective effects of specific BQ ingredients may facilitate the development of targeted BQ formulations to prevent and/or treat potentially malignant oral disorders and oral cancer in BQ users.


Asunto(s)
Areca/química , Carcinoma de Células Escamosas/inducido químicamente , Neoplasias de la Boca/inducido químicamente , Fibrosis de la Submucosa Bucal/inducido químicamente , Extractos Vegetales/efectos adversos , Arecolina/efectos adversos , Arecolina/análogos & derivados , Carcinoma de Células Escamosas/patología , Progresión de la Enfermedad , Humanos , Neoplasias de la Boca/patología , Ácidos Nicotínicos/efectos adversos , Fibrosis de la Submucosa Bucal/patología
7.
Mol Carcinog ; 60(11): 758-768, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34432915

RESUMEN

Oral squamous cell carcinoma (OSCC) is the most common type of oral malignancy. Our study uses multipoint materials to explore the heterogeneity and metastasis mechanism of OSCC to find more accurate molecular markers and new therapeutic targets. By using whole-exome capture and sequencing and tumor evolution analysis, we found that most clone-driven mutations were located in the branches of tumor phylogenetic tree, such as COTL1, CASP8, and PROCR. Most clone-driven OSCC mutations occur mainly in tumor suppressor genes, including TP53, SFRP4, and NOTCH1. Our study on intratumor heterogeneity (ITH) and clonal evolution provides an important molecular basis for further understanding of OSCC occurrence and development and metastasis and provides potential targets for the treatment of this disease.


Asunto(s)
Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Secuenciación del Exoma/métodos , Neoplasias de la Boca/genética , Mutación , Evolución Clonal , Evolución Molecular , Genes Supresores de Tumor , Heterogeneidad Genética , Humanos , Proto-Oncogenes
8.
Pathobiology ; 88(5): 327-337, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33965948

RESUMEN

BACKGROUND: The mortality of oral squamous cell carcinoma (OSCC) has remained high for decades; therefore, methods for early detection of OSCC are warranted. However, in the oral cavity, various mucosal diseases may be encountered, including reactive lesions and oral potentially malignant disorders, and it is difficult to differentiate OSCC from these lesions based on both clinical and histopathological findings. It is well known that chronic inflammation contributes to oral cancer development. Macrophages are among the most common inflammatory cells in cancer stromal tissue and have various roles in cancer aggressiveness. Although the roles of macrophages in cancer development have attracted attention, only a few studies have linked macrophages to carcinogenesis, particularly, oral precancerous lesions. SUMMARY: This review article consists of 3 parts: first, we summarize current knowledge on macrophages in human various epithelial precancerous lesions, excluding the oral cavity, to show the importance and gaps in knowledge regarding macrophages in carcinogenesis; second, we review published data related to the role of macrophages in oral carcinogenesis; finally, we present a novel view on oral carcinogenesis, focusing on crosstalk between epithelial cells and macrophages. Key Messages: The biological features of macrophages in oral carcinogenesis differ drastically depending on the anatomical compartment that they infiltrate. Focusing on the alteration of macrophage infiltrating compartment may serve as a useful novel approach for studying the role of the macrophages in oral carcinogenesis and for gaining further insight into cancer prevention and early detection.


Asunto(s)
Carcinogénesis/inmunología , Macrófagos/inmunología , Macrófagos/patología , Neoplasias de la Boca/inmunología , Humanos , Inflamación/complicaciones , Neoplasias de la Boca/mortalidad , Neoplasias de la Boca/patología , Carcinoma de Células Escamosas de Cabeza y Cuello/mortalidad , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Microambiente Tumoral
9.
Anaerobe ; 68: 102300, 2021 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-33246097

RESUMEN

It was estimated that more than 700 bacterial species inhabit the oral cavity of healthy humans. Anaerobes comprise a significant fraction of the oral bacteriome and play an important role in the formation of multi-species biofilms attached to various anatomical sites. Bacterial biofilms are also associated with pathologic laesions of the oral cavity, including oral squamous cell carcinoma (OSCC), and distinct oral taxa could also be detected within the tumors, i.e. in deep biopsy samples. These observations suggested that certain oral bacteria or oral bacterial communities may play a causative role in oral carcinogenesis, in addition to the well characterized risk factors of oral cancer. Alternatively, it was also proposed that a subset of oral bacteria may have a growth advantage in the unique microenvironment of OSCC. Recently, a series of studies analysed the OSCC-associated bacterial communities using metataxonomic, metagenomic and metatranscriptomic approaches. This review outlines the major differences between the community structure of microbiota in tumor biopsy, surface-biofilm and salivary or oral wash samples collected from OSCC patients, compared to corresponding samples from control persons. A special emphasis is given to the anaerobic bacteria Fusobacterium nucleatum and Fusobacterium periodonticum that were characterised repeatedly as "OSCC-associated" in independent studies. Predicted microbial functions and relevant in vivo experimental models of oral carcinogenesis will also be summarized.


Asunto(s)
Bacterias Anaerobias/aislamiento & purificación , Bacterias Anaerobias/fisiología , Microbiota , Neoplasias de la Boca/microbiología , Saliva/microbiología , Anaerobiosis , Animales , Bacterias Anaerobias/clasificación , Bacterias Anaerobias/genética , Biopelículas , Humanos
10.
J Cell Mol Med ; 24(7): 4011-4022, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32180353

RESUMEN

This study aimed to explore new therapeutic targets to improve the survival rate of patients with oral squamous cell carcinoma (OSCC).MiR-210-3p, EphrinA3 and EMT related indices were evaluated in OSCC tissues and cell lines. In addition, the relationship between differential EphrinA3 expression and tumour progression was explored through molecular biology techniques, in vitro functional experiments and tumour xenotransplantation models. The expression of EphrinA3 (rs  = -0.719, P < .05) and E-cadherin (rs  = -0.856, P < .05) was negatively correlated with the pathological grading in OSCC tissues. Protein clustering shows EphrinA3 may be associated with tumour progression. EphrinA3 also can regulate the biological behaviour of oral cancer cells. And it regulates the EMT by the PI3K/AKT signalling pathway. MiR-210-3p targeted the gen EFNA3. Up-regulation of miR-210-3p expression can decrease the expression of EphrinA3 and further to influence the biological behaviour of OSCC. The miR-210-3p-EphrinA3-PI3K/AKT signalling axis plays an important role in the progress of OSCC. EphrinA3 may serve as a novel target for oral cancer treatment.


Asunto(s)
Carcinogénesis/genética , Efrina-A3/genética , MicroARNs/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Anciano , Animales , Línea Celular Tumoral , Proliferación Celular/genética , Progresión de la Enfermedad , Supervivencia sin Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica/genética , Xenoinjertos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Fosfatidilinositol 3-Quinasas/genética , Proteínas Proto-Oncogénicas c-akt/genética , Transducción de Señal , Carcinoma de Células Escamosas de Cabeza y Cuello/patología
11.
Biochim Biophys Acta Mol Cell Res ; 1865(1): 209-219, 2018 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-29104085

RESUMEN

Accumulating evidences indicate that alcohol might play a causative in oral cancer. Unfortunately, in vitro cell systems, uncovering the molecular background of the underlying cell transformation process, are rare. Therefore, this study was conducted, to identify molecular changes and characterize their putative cell behavioral consequences in epitheloid (EPI) and fibroblastoid (FIB) oral keratinocyte phenotypes, arising from chronical alcohol treatment. Concerning adherens junctions (AJs), both EPI and FIB showed membrane-bound ß-catenin, but exhibited differences for E-cadherin and zyxin. While EPI revealed E-cadherin/ß-catenin membrane co-localization, which in parts also applied for zyxin, FIB membranes were devoid of E-cadherin and exhibited marginal zyxin expression. Fetal calf serum (FCS) administration in starved cells promoted proliferation in both keratinocyte phenotypes, whereat EPI and FIB yielded a strikingly modified FCS sensitivity on the temporal scale. Impedance measurement-based cell index detection yielded proliferation stimulation occurring much earlier in FIB (<20h) compared to EPI (>45h). Nuclear preference of the proliferation-associated YAP co-transcription factor in FIB was FCS independent, while it required FCS in EPI. Taken together, the lack of membrane-inherent E-cadherin/ß-catenin co-localization together with low zyxin - reveals perturbation of AJ integrity in FIB. Regarding cell behavior, perturbed AJs in FIB correlate with temporal proliferation sensitivity towards FCS. CYF of 5.6 strongly suggests involvement of chromatin-bound YAP in FIB's proliferation temperosensitivity. These molecular differences detected for EPI and FIB are part of the underlying cell transformation process of alcohol-induced oral carcinogenesis, and indicate FIB being in a more advanced transformation stage.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/fisiología , Uniones Adherentes/patología , Carcinogénesis/inducido químicamente , Proliferación Celular , Etanol/toxicidad , Mucosa Bucal/efectos de los fármacos , Fosfoproteínas/fisiología , Uniones Adherentes/efectos de los fármacos , Uniones Adherentes/genética , Uniones Adherentes/metabolismo , Carcinogénesis/genética , Carcinogénesis/metabolismo , Línea Celular Transformada , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Transformación Celular Neoplásica/inducido químicamente , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Células Epiteliales/patología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Fibroblastos/patología , Humanos , Mucosa Bucal/metabolismo , Mucosa Bucal/patología , Factores de Transcripción , Proteínas Señalizadoras YAP
12.
Genes Cells ; 23(4): 264-273, 2018 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-29441648

RESUMEN

Periodontal disease, an inflammatory disease, is caused by infection with periodontal pathogens. Long-term periodontal disease increases the risk of oral carcinogenesis. Similar to other peptic cancers, oral carcinogenesis also requires multiple genome instabilities; however, the risk factors related to the accumulation of genome instabilities are poorly understood. Here, we suggested that specific periodontal pathogens may increase the risk of genome instability. Accordingly, we screened several periodontal pathogens based on the ability to induce DNA double-strand breaks (DSBs) in host cells. We found that Aggregatibacter actinomycetemcomitans Y4 infection induced DSB formation in host cells. To assess whether DSB formation induced by infection with A. actinomycetemcomitans occurred through apoptotic chromosome fragmentation, cells were treated with a caspase inhibitor, Z-VAD-FMK. DSB accumulation induced by infection with A. actinomycetemcomitans was observed, even in the presence of Z-VAD-FMK, suggesting that this breakage occurred independently of apoptosis. These results suggested that some periodontal pathogens can increase the risk of genome instabilities in host cells and subsequently increase the risk of carcinogenesis.


Asunto(s)
Aggregatibacter actinomycetemcomitans/fisiología , Carcinoma de Células Escamosas/genética , Roturas del ADN de Doble Cadena , Inestabilidad Genómica , Infecciones por Pasteurellaceae/microbiología , Neoplasias de la Lengua/genética , Apoptosis , Carcinoma de Células Escamosas/microbiología , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Células HeLa , Humanos , Infecciones por Pasteurellaceae/patología , Neoplasias de la Lengua/microbiología , Neoplasias de la Lengua/patología
13.
Mol Carcinog ; 58(5): 820-831, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30644602

RESUMEN

Understanding the cellular interactions during oral carcinogenesis has the potential to identify novel prognostic and therapeutic targets. This study aimed at investigating the cancer stem cell (CSC)-fibroblast niche interactions using in-vitro dysplastic cell line models developed from different stages of 4NQO-induced oral carcinogenic mice model. The spontaneously transformed epithelial cells (DysMSCTR6, 14 and 16) were developed from three time points (mild/moderate/severe), while two fibroblast cell lines (FibroMSCTR12, 16) were developed from moderate and severe dysplastic tissue. The epithelial (Epcam+/Ck+) and the fibroblast cell lines (Vimentin+/α-SMA+/Ck-) were authenticated and assessment of cells representing progressive grades of dysplastic severity indicated a significant increase in dysplastic marker profile (P < 0.05). Evaluation of the CSC characteristics showed that an increase in expression of Cd133, Cd44, Aldh1a1, Notch1, and Sox2 was accompanied by an increase in migratory (P > 0.05) and colony formation capacity (P > 0.005). Targeting Notch1 (GSI inhibitor PZ0187; 30 µM), showed a significant reduction in cell proliferation capacity (P < 0.05) and in the dysplastic marker profile. Further, Notch1 inhibition resulted in down regulation of Cd133 and Aldh1a 1 (P < 0.05) and a complete abrogation of colony formation ability (P < 0.0001). The effect of niche interactions evaluated using FibroMSCTR12-conditioned media studies, revealed an enrichment of ALDH1A1+ cells (P < 0.05), induction of spheroid formation ability (P < 0.0001) and increased proliferation capacity (3.7 fold; P < 0.005). Although PZ0187 reduced cell viability by ∼40%, was unable to abrogate the conditioned-media induced increase in proliferation capacity completely. This study reports a Notch-1 dependent enrichment of CSC properties during dysplastic progression and a Notch-1 independent dysplastic cell-fibroblast interaction during oral carcinogenesis.


Asunto(s)
Modelos Animales de Enfermedad , Fibroblastos/patología , Mucosa Bucal/patología , Neoplasias de la Boca/patología , Células Madre Neoplásicas/patología , Lesiones Precancerosas/patología , Animales , Proliferación Celular , Células Cultivadas , Técnicas de Cocultivo , Femenino , Fibroblastos/metabolismo , Técnicas In Vitro , Ratones , Ratones Endogámicos C57BL , Mucosa Bucal/metabolismo , Neoplasias de la Boca/metabolismo , Células Madre Neoplásicas/metabolismo , Lesiones Precancerosas/metabolismo
14.
Cytokine ; 120: 130-143, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31071674

RESUMEN

AIMS: Signal transducer and activator of transcription (STAT)-3 lies at the convergence point of key pathways involved in many malignancies including oral squamous cell carcinoma (OSCC). Endoplasmic reticulum stress (ERS) and the unfolded protein response have been shown to be involved in the pathogenesis and progression of different cancers by influencing key cellular processes such as apoptosis. We investigated the differential expression of STAT3 pathway-related genes and proteins under ERS in OSCC. METHODS: Three normal oral keratinocyte (NOK) and three OSCC cell lines were subjected to tunicamycin to induce ERS for 24 h or to the vehicle medium as control. A pathway-focussed array was used to analyse the modulation of STAT3 pathway gene expression under ERS using qPCR. The expression of key regulated proteins was investigated in the cell lines using immunocytochemistry and in 76 OSCC and 9 normal oral mucosa (NOM) tissue samples using tissue microarray technology and immunohistochemistry. RESULTS: ERS resulted in up-regulation of interleukin-6 receptor (IL6R) gene in NOK cell lines (p = 0.001) and IL5 (p = 0.005) and IL22 (p = 0.024) in OSCC cell lines. Greater STAT3 (p = 0.019) and leukaemia inhibitory factor receptor (p = 0.042) protein expression was observed in treated than untreated NOK cell lines. CONCLUSIONS: The gene and protein regulation patterns show that ERS plays a role in modifying the tumour microenvironment in OSCC by up-regulating tumour-promoting cytokines.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Citocinas/metabolismo , Estrés del Retículo Endoplásmico , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Tunicamicina/efectos adversos , Regulación hacia Arriba , Carcinoma de Células Escamosas/genética , Línea Celular Tumoral , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica , Humanos , Neoplasias de la Boca/genética , Factor de Transcripción STAT3/metabolismo , Regulación hacia Arriba/efectos de los fármacos
15.
J Oral Pathol Med ; 48(7): 546-551, 2019 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-31183906

RESUMEN

The oral microbiome is composed of microorganisms residing in the oral cavity, which are critical components of health and disease. Disruption of the oral microbiome has been proven to influence the course of oral diseases, especially among immunocompromised patients. Oral microbiome is comprised of inter-kingdom microorganisms, including yeasts such as Candida albicans, bacteria, archaea and viruses. These microorganisms can interact synergistically, mutualistically and antagonistically, wherein the sum of these interactions dictates the composition of the oral microbiome. For instance, polymicrobial interactions can improve the ability of C albicans to form biofilm, which subsequently increases the colonisation of oral mucosa by the yeast. Polymicrobial interactions of C albicans with other members of the oral microbiome have been reported to enhance the malignant phenotype of oral cancer cells, such as the attachment to extracellular matrix molecules (ECM) and epithelial-mesenchymal transition (EMT). Polymicrobial interactions may also exacerbate an inflammatory response in oral epithelial cells, which may play a role in carcinogenesis. This review focuses on the role of polymicrobial interactions between C albicans and other oral microorganisms, including its role in promoting oral carcinogenesis.


Asunto(s)
Candida albicans , Biopelículas , Carcinogénesis , Humanos , Interacciones Microbianas , Boca
16.
Adv Dent Res ; 30(1): 16-21, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31538805

RESUMEN

Traditional tobacco products have well-known systemic and local oral effects, including inflammation, vasoconstriction, delayed wound healing, and increased severity of periodontal disease. Specifically in the oral cavity and the lung, cigarette smoking produces cancer, increased infectivity, acute and chronic inflammation, changes in gene expression in epithelial lining cells, and microbiome changes. In recent years, cigarette smoking has greatly decreased in the United States, but the use of new tobacco products has gained tremendous popularity. Without significant knowledge of the oral sequelae of products such as electronic cigarettes, researchers must evaluate current in vitro and in vivo methods to study these agents, as well as develop new tools to adequately study their effects. Some in vitro testing has been performed for electronic cigarettes, including toxicologic models and assays, but these mostly study the effect on the respiratory tract. Recently, direct exposure of the aerosol to in vitro 3-dimensional tissue constructs has been performed, demonstrating changes in cell viability and inflammatory cytokines. For in vivo studies, a universal e-cigarette testing machine or standard vaping regime is needed. A standard research electronic cigarette has recently been developed by the National Institute of Drug Abuse, and other devices delivering aerosols with different nicotine concentrations are becoming available. One of the biggest challenges in this research is keeping up with the new products and the rapidly changing technologies in the industry.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Investigación , Productos de Tabaco , Técnicas In Vitro , Nicotina , Investigación/tendencias , Productos de Tabaco/normas , Estados Unidos , Vapeo
17.
Oral Dis ; 24(1-2): 98-102, 2018 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-29480608

RESUMEN

OBJECTIVES: To examine cytoplasmic cyclin D1 expression levels in oral carcinogenesis and evaluate their possible oncogenic significance and their clinicopathological and prognostic implications. MATERIALS AND METHODS: Immunohistochemical analysis of 69 oral squamous cell carcinomas (OSCCs) was performed, revealing 23 with cytoplasmic cyclin D1 expression. We analyzed the association of the percentage of cyclin D1-positive cells and the intensity of expression with TNM classification, tumor stage, differentiation degree, cell morphology, and Ki-67 expression. RESULTS: Cytoplasmic cyclin D1 expression was associated with advanced tumor stage, poor differentiation, elevated Ki-67 expression, and the presence of invasive cell morphology, indicators of a poor prognosis. An association was observed between nuclear and cytoplasmic expressions of cyclin D1. CONCLUSIONS: Cytoplasmic expression of cyclin D1 appears to possess functions related to increased cell migration and invasion in OSCC.


Asunto(s)
Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/secundario , Ciclina D1/metabolismo , Citoplasma/metabolismo , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Anciano , Anciano de 80 o más Años , Carcinogénesis , Femenino , Humanos , Antígeno Ki-67/metabolismo , Masculino , Persona de Mediana Edad , Clasificación del Tumor , Invasividad Neoplásica , Estadificación de Neoplasias
18.
Oral Dis ; 23(7): 897-912, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-27973759

RESUMEN

Cyclin D1 promotes cell cycle progression during G1 phase, a key event in G1-S transition. The protein is encoded by gene CCND1, located in chromosomal band 11q13. Cyclin D1 plays key roles in cell biology, including cell proliferation and growth regulation, mitochondrial activity modulation, DNA repair, and cell migration control. CCND1 gene and its protein cyclin D1 are frequently altered by different molecular mechanisms, including amplification, chromosomal translocations, mutations, and activation of the pathways involved in cyclin D1 expression, alterations which appear to be essential in the development of human cancers, including oral carcinoma. This is the first published review of the specific features of cyclin D1 overexpression in oral oncogenesis. Starting with the physiological regulation of cyclin D1, there is an evaluation of its functions, overexpression mechanisms, and the implications of the oncogenic activation of CCND1/cyclin D1 in oral squamous cell carcinoma. The potential diagnostic and prognostic value of cyclin D1 is reviewed. The influence of CCND1/cyclin D1 on tumor size and clinical stage is reported, and an update is provided on the utilization of cyclin D1 as therapeutic target and on the combination of cyclin D1 inhibitors with cytotoxic agents. Future research lines in this field are also proposed.


Asunto(s)
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Ciclina D1/genética , Ciclina D1/metabolismo , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Carcinogénesis , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Proliferación Celular , Supervivencia sin Enfermedad , Amplificación de Genes , Expresión Génica , Humanos , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/patología , Polimorfismo Genético , Tasa de Supervivencia , Regulación hacia Arriba
19.
J Formos Med Assoc ; 116(6): 457-463, 2017 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-28292622

RESUMEN

BACKGROUND/PURPOSE: Langerhans cells (LCs) are antigen presenting cells. This study assessed the LC counts in oral epithelial dysplasia (OED) and their correlation to clinicopathological parameters. METHODS: This study examined the LC counts in the epithelia and subepithelial connective tissues of 58 patients with OED (21 mild, 18 moderate, and 19 severe OED lesions) and 10 specimens of normal oral mucosa (NOM) by anti-S-100 protein immunostaining. RESULTS: We found that the mean LC counts in the epithelia or subepithelial connective tissues increased significantly from NOM samples through mild and moderate OED to severe OED samples. In addition, a significant correlation was found between higher mean LC counts in the dysplastic epithelia of OED samples and OED lesions with thicker epithelial layers (p<0.001) or wider inflammatory zones (p<0.001), and between higher mean LC counts in the subepithelial connective tissues of OED samples and OED lesions with wider inflammatory zones (p<0.001). Moreover, the nine OED lesions with malignant transformation had a significantly lower mean LC count than the 49 OED lesions without malignant transformation. CONCLUSION: The significant and gradual elevation in LC count from NOM through mild and moderate OED to severe OED lesions suggests an upregulation of immunosurveillance ability in OED patients during the early oral carcinogenesis process. A low LC count in OED lesions may suggest the partial loss of immunosurveillance ability against dysplastic cells; this in turn favors the malignant transformation of an OED lesion into oral cancer.


Asunto(s)
Epitelio/patología , Células de Langerhans/patología , Mucosa Bucal/patología , Adulto , Anciano , Anciano de 80 o más Años , Estudios de Casos y Controles , Recuento de Células , Transformación Celular Neoplásica/patología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Lesiones Precancerosas/patología , Adulto Joven
20.
J Proteome Res ; 15(9): 2981-97, 2016 09 02.
Artículo en Inglés | MEDLINE | ID: mdl-27432155

RESUMEN

The areca nut is a known carcinogen that causes oral cancer in individuals in Southeast Asia, but the molecular mechanism that leads to this malignancy is still unclear. To mimic the habit of areca nut chewing, our laboratory has established four oral cancer cell sublines (SAS, OECM1, K2, C9), which have been chronically exposed to areca nut extract (ANE). To elucidate the molecular basis of areca nut-induced oral carcinogenesis, the differential proteomes between oral cancer cells and the ANE-treated sublines were determined using isobaric mass tag (iTRAQ) labeling and multidimensional liquid chromatography-mass spectrometry (LC-MS/MS). Over 1000 proteins were identified in four sublines, and 196 proteins were found to be differentially expressed in at least two ANE-treated sublines. A bioinformatic analysis revealed that these proteins participate in several pathways, and one of the most prominent pathways was the regulation of epithelial to mesenchymal transition (EMT). In all, 24 proteins including Krt17 were confirmed to be differentially expressed in the ANE-treated sublines. To reveal additional information on the mechanism of ANE-induced carcinogenesis, Krt17 was further investigated. Krt17 knockdown significantly suppressed ANE-induced cell migration and invasion and modulated the EMT process. Furthermore, in a murine model of carcinogen-induced (arecoline cocktail, an active compound of ANE) oral cancer, Krt17 was significantly up-regulated in all hyperplastic tissues and in carcinoma tissues (p < 0.001). In conclusion, we have identified a proteome of oral cancer cells that is associated with chronic areca nut exposure. Krt17 was demonstrated to contribute to areca nut-induced oral malignancy. The results of this study contribute to risk assessment, disease prevention and other clinical applications associated with areca nut-induced oral cancer.


Asunto(s)
Areca/toxicidad , Queratina-17/metabolismo , Neoplasias de la Boca/etiología , Extractos Vegetales/farmacología , Proteómica/métodos , Animales , Areca/química , Línea Celular , Biología Computacional , Transición Epitelial-Mesenquimal , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Queratina-17/fisiología , Ratones , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda