Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 115
Filtrar
Más filtros

Intervalo de año de publicación
1.
Cancer Causes Control ; 35(3): 429-435, 2024 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-37815646

RESUMEN

PURPOSE: Mounting evidence suggests a possible link between gut microbiome and oral cancer, pointing to some potential modifiable targets for disease prevention. In the present study, Mendelian randomization (MR) was used to explore whether there was a causal link between gut microbiome and oral cancer. METHODS: The single nucleotide polymorphisms (SNPs) significantly associated with gut microbiome were served as instrumental variables. MR analyses were performed using genetic approaches such as inverse variance weighting (IVW), MR Egger and weighted median, with IVW as the primary approach, supplemented by MR Egger and weighted median. Mendelian randomization pleiotropy residual sum and outlier (MR-PRESSO) and MR-Egger regression were used to detect the presence of horizontal pleiotropy and identify outlier SNPs. RESULTS: Causal effect estimates indicated that genetically predicted abundance of Prevotellaceae was associated with higher risk of oral cancer (odds ratio (OR) 1.80, 95% confidence interval (CI) 1.16-2.81, p = 0.009). There was no evidence of notable heterogeneity and horizontal pleiotropy. CONCLUSION: Genetically derived estimates suggest that Prevotellaceae may be associated with the risk of oral cancer. Such robust evidence should be given priority in future studies and explore the underlying mechanisms.


Asunto(s)
Microbioma Gastrointestinal , Neoplasias de la Boca , Humanos , Microbioma Gastrointestinal/genética , Neoplasias de la Boca/genética , Suplementos Dietéticos , Análisis de la Aleatorización Mendeliana , Oportunidad Relativa , Estudio de Asociación del Genoma Completo
2.
Adv Sci (Weinh) ; 10(25): e2207549, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37401236

RESUMEN

LncRNAs play a critical role in oral squamous cell carcinoma (OSCC) progression. However, the function and detailed molecular mechanism of most lncRNAs in OSCC are not fully understood. Here, a novel nuclear-localized lncRNA, DUXAP9 (DUXAP9), that is highly expressed in OSCC is identified. A high level of DUXAP9 is positively associated with lymph node metastasis, poor pathological differentiation, advanced clinical stage, worse overall survival, and worse disease-specific survival in OSCC patients. Overexpression of DUXAP9 significantly promotes OSCC cell proliferation, migration, invasion, and xenograft tumor growth and metastasis, and upregulates N-cadherin, Vimentin, Ki67, PCNA, and EZH2 expression and downregulates E-cadherin in vitro and in vivo, whereas knockdown of DUXAP9 remarkably suppresses OSCC cell proliferation, migration, invasion, and xenograft tumor growth in vitro and in vivo in an EZH2-dependent manner. Yin Yang 1 (YY1) is found to activate the transcriptional expression of DUXAP9 in OSCC. Furthermore, DUXAP9 physically interacts with EZH2 and inhibits EZH2 degradation via the suppression of EZH2 phosphorylation, thereby blocking EZH2 translocation from the nucleus to the cytoplasm. Thus, DUXAP9 can serve as a promising target for OSCC therapy.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , ARN Largo no Codificante , Humanos , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , ARN Largo no Codificante/genética , ARN Largo no Codificante/metabolismo , Yin-Yang , Línea Celular Tumoral , Proliferación Celular/genética , Neoplasias de la Boca/genética , Proteína Potenciadora del Homólogo Zeste 2/genética , Proteína Quinasa CDC2
3.
Cells ; 12(13)2023 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-37443802

RESUMEN

Angelica keiskei Koidzumi (A. keiskei) is used as a traditional medicine, anti-aging agent, and health food, as well as to restore vitality. Xanthoangelol (xanol), a prenylated chalcone, is the predominant constituent of A. keiskei. Oral squamous cell carcinoma (OSCC), the most common malignancy, has a high proliferation rate and frequent metastasis. However, it is unknown whether xanol has anti-OSCC effects on apoptosis, autophagy, and necroptosis. In the present study, we purified xanol from A. keiskei and demonstrated that it suppressed cell proliferation and induced cytotoxicity in human OSCC. Xanol triggered apoptotic cell death by regulating apoptotic machinery molecules but inhibited necroptotic cell death by dephosphorylating the necroptotic machinery molecules RIP1, RIP3, and MLKL in human OSCC. We also found that xanol inhibited the PI3K/AKT/mTOR/p70S6K pathway and induced autophagosome formation by enhancing beclin-1 and LC3 expression levels and reducing p62 expression levels. Furthermore, we showed that xanol prevented the metastatic phenotypes of human OSCC by inhibiting migration and invasion via the reduction of MMP13 and VEGF. Finally, we demonstrated that xanol exerted anticancer effects on tumorigenicity associated with its transformed properties. Taken together, these findings demonstrate the anticancer effects and biological mechanism of action of xanol as an effective phytomedicine for human OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas/patología , Carcinoma de Células Escamosas de Cabeza y Cuello , Proteínas Proto-Oncogénicas c-akt/metabolismo , Fosfatidilinositol 3-Quinasas , Necroptosis , Neoplasias de la Boca/genética , Apoptosis , Autofagia
4.
Am J Chin Med ; 51(3): 741-760, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36869017

RESUMEN

Dysfunction of epidermal growth factor receptor (EGFR) signaling plays a critical role in the tumorigenesis of oral squamous cell carcinoma (OSCC). In the present study, the data analysis results of immunohistochemistry and the TCGA database verified that the expression of EGFR is significantly upregulated in OSCC tumor tissues, and depletion of EGFR inhibits the growth of OSCC cells in vitro and in vivo. Moreover, these results showed that the natural compound, curcumol, exhibited a profound antitumor effect on OSCC cells. Western blotting, MTS, and immunofluorescent staining assays indicated that curcumol inhibited cell proliferation and induced intrinsic apoptosis in OSCC cells via downregulating myeloid cell leukemia 1 (Mcl-1). A mechanistic study revealed that curcumol inhibited the EGFR-Akt signal pathway, which activated GSK-3[Formula: see text]-mediated Mcl-1 phosphorylation. Further research showed that curcumol-induced Mcl-1 Ser159 phosphorylation is required to disrupt the interaction between deubiquitinase JOSD1 and Mcl-1 and eventually induce Mcl-1 ubiquitination and degradation. In addition, curcumol administration can effectively inhibit CAL27 and SCC25 xenograft tumor growth and is well-tolerated in vivo. Finally, we demonstrated that Mcl-1 is upregulated and positively correlates with p-EGFR and p-Akt in OSCC tumor tissues. Collectively, the present results provide new insights into the antitumor mechanism of curcumol, identifying it as an attractive therapeutic agent that reduces Mcl-1 expression and inhibits OSCC growth. Targeting EGFR/Akt/Mcl-1 signaling could be a promising option in the clinical treatment of OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Proteínas Proto-Oncogénicas c-akt , Glucógeno Sintasa Quinasa 3 , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/genética , Receptores ErbB/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , Proliferación Celular , Transducción de Señal , Línea Celular Tumoral , Apoptosis
5.
J Immunol Res ; 2023: 5293677, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36969496

RESUMEN

The morbidity of oral cancer is high in the world. Oridonin is a traditional Chinese medicine that can effectively inhibit oral squamous cell carcinoma (OSCC) growth, but its mechanism remains unclear. Our previous data showed that oridonin inhibited CAL-27 cell proliferation and promoted apoptosis. Herein, we explored the mechanism and target of oridonin in human OSCC through RNA sequencing and integration of multiple bioinformatics analysis strategies. Differences in gene expression can be analyzed with RNA sequencing. Kyoto Encyclopedia of Genes and Genomes (KEGG), Gene Ontology (GO), gene set enrichment analysis (GSEA), Disease Ontology (DO), and other enrichment analyses were used to evaluate differentially expressed genes (DEGs). Protein-protein interaction (PPI) networks were built via the STRING database. It was found that tumor necrosis factor (TNF) signaling pathway, cytokine-cytokine receptor interaction, and nuclear factor-kappa B (NF-kappaB) signaling pathway were associated with the therapeutic effects of oridonin in OSCC. Three key genes (BIRC3, TNFSF10, and BCL6) were found to associate with cell apoptosis in OSCC cells treated with oridonin. Quantitative PCR assays verified the expression of apoptosis-related DEGs: TNFSF10, BIRC3, AIFM2, BCL6, BCL2L2, and Bax. Western blots were employed for verifying proteins expression associated with DEGs: cleaved caspase 3, Bax, Bcl-w, anti-cIAP2, and anti-TRAIL. In conclusion, our findings reveal the molecular pathways and targets by which oridonin can treat and induce cytotoxic effects in OSCC: by affecting the signaling including TNF, NF-κB, and cytokine-cytokine receptor interaction and by regulating the key gene BIRC3, TNFSF10, and BCL6. It should be noted that further clinical trial validation is very necessary. Combined with current research trends, our existing research may provide innovative research drugs for the treatment of OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Transcriptoma , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , ARN , FN-kappa B/metabolismo , Proteína X Asociada a bcl-2 , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Apoptosis , Citocinas/genética , Biología Computacional/métodos
6.
J Gene Med ; 25(6): e3490, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36843559

RESUMEN

BACKGROUND AND AIMS: This study performed a network pharmacology analysis to explore the potential anticancer activity of baicalein against oral squamous cell carcinoma (OSCC). METHODS: The differentially expressed genes in OSCC were identified by the OSCC, and the key module related to OSCC was acquired by the weighted gene co-expression network analysis from GSE30784. Baicalein targets were obtained from GeneCards, SwissTargetPrediction and TCMSP (the traditional Chinese medicine systems pharmacology database and analysis platform) databases. Apart from GSE30784, OSCC-related genes were acquired from the Comparative Toxicogenomics Database and DisGeNET platform. The baicalein targets and OSCC-related genes were overlapped to acquire the drug-disease interaction genes. We input the candidate drug-disease interaction genes into the STRING database and created the protein-protein interaction network. The hub genes in the network were identified by cytoHubba. The expression levels of hub genes between cancer tissues and normal tissues were evaluated based on The Cancer Genome Atlas. Moreover, we performed the cancer immune infiltration analysis and evaluated the association between the expression of HIF1A and the abundance of infiltrating immune cells. The molecular docking between HIFA and baicalein was also performed and visualized. Additionally, the enrichment analysis was performed based on Gene Ontology and Kyoto Encyclopedia of Genes and Genomes database. RESULTS: After overlapping the candidate baicalein targets with OSCC-related genes, 34 candidate drug-disease interaction genes were obtained. cytoHubba identified the top 10 genes with high centrality measures from the network, including AKT1, CD44, EGFR, HIF1A, IGF1, MMP2, MYC, PTGS2, STAT3 and TP53. The enriched biological process terms included regulation of apoptotic signaling pathway, regulation of cysteine-type endopeptidase activity and cellular response to chemical stress. The top five enriched pathways comprised Kaposi sarcoma-associated herpesvirus infection, hepatitis C, p53 signaling pathway, Epstein-Barr virus infection and proteoglycans in cancer. The expression of HIF1A was positively associated with the infiltration levels of CD4 + T cells (cor = 0.131, p = 0.004) and dendritic cells (cor = 0.098, p = 0.03), whereas negatively associated with B cells (cor = -0.098, p = 0.03). CONCLUSION: Our results suggested that baicalein might be a candidate drug against OSCC. More studies are necessary to validate its anticancer effect and explore the underlying mechanisms.


Asunto(s)
Carcinoma de Células Escamosas , Infecciones por Virus de Epstein-Barr , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Humanos , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas de Cabeza y Cuello , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/genética , Farmacología en Red , Simulación del Acoplamiento Molecular , Herpesvirus Humano 4 , Biología Computacional
7.
J Ovarian Res ; 15(1): 77, 2022 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-35778739

RESUMEN

OBJECTIVE: Ying Yang1 (YY1) has already been discussed in oral squamous cell carcinoma (OSCC), but the knowledge about its mediation on long non-coding RNA KCNQ1 overlapping transcript 1/microRNA-506-3p/synaptophysin like 1 (Kcnq1ot/miR-506-3p/SYPL1) axis in OSCC is still in its infancy. Hence, this article aims to explain the mechanism of YY1/Kcnq1ot1/miR-506-3p/SYPL1 axis in OSCC development. METHODS: YY1, Kcnq1ot1, miR-506-3p and SYPL1 expression levels were determined in OSCC tissues. The potential relation among YY1, Kcnq1ot1, miR-506-3p and SYPL1 was explored. Cell progression was observed to figure out the actions of depleted YY1, Kcnq1ot1 and SYPL1 and restored miR-506-3p in OSCC. OSCC tumorigenic ability in mice was examined. RESULTS: Elevated YY1, Kcnq1ot1 and SYPL1 and reduced miR-506-3p were manifested in OSCC. YY1 promoted Kcnq1ot1 transcription and up-regulated Kcnq1ot1 expression, thereby promoting OSCC cell procession. Silencing Kcnq1ot1 or elevating miR-506-3p delayed OSCC cell progression and silencing Kcnq1ot1 impeded tumorigenic ability of OSCC cells in mice. YY1-mediated Kcnq1ot1 sponged miR-506-3p to target SYPL1. CONCLUSION: YY1 promotes OSCC cell progression via up-regulating Kcnq1ot1 to sponge miR-506-3p to elevate SYPL1, guiding a novel way to treat OSCC.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , MicroARNs , Neoplasias de la Boca , ARN Largo no Codificante , Factor de Transcripción YY1 , Animales , Carcinogénesis , Carcinoma de Células Escamosas/genética , Humanos , Ratones , MicroARNs/genética , Neoplasias de la Boca/genética , Canales de Potasio con Entrada de Voltaje , ARN Largo no Codificante/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Sinaptofisina , Factor de Transcripción YY1/genética , Factor de Transcripción YY1/metabolismo
8.
Int J Mol Sci ; 24(1)2022 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-36614140

RESUMEN

Orento is a traditional Japanese medicinal kampo preparation that is also prescribed in oral care. In oral squamous cell carcinoma cell line CAL27, orento significantly inhibited periodontopathogenic bacterium Porphyromonas gingivalis lipopolysaccharide (LPS) and lipoproteins (PAMP)-stimulated production of interleukin (IL)-6. This suggests that orento negatively regulates PAMP-mediated toll-like receptor (TLR) signaling. Orento significantly suppressed PAMP-stimulated activation of the IL-6 promoter, indicating that orento may suppress the production of IL-6 by PAMP at the transcriptional level. Orento also suppressed TLR-mediated activation of transcription factor nuclear factor-kappa B (NF-kB) that was stimulated by PAMP. This finding indicates that orento may suppress the function and activation of factors involved in TLR signaling, thereby suppressing NF-kB-dependent expression of various genes. Orento suppressed IL-1 receptor-associated kinase (IRAK4), IRAK1, and c-Jun N-terminal kinase (JNK) phosphorylation in PAMP-stimulated CAL27 cells. This result indicates that orento is involved in the initiation of TLR signaling by PAMP and suppresses the downstream signaling pathways of myeloid differentiation primary response gene 88 (MyD88) such as mitogen-activated protein kinase (MAPK) and NF-kB cascades. These findings suggest that orento has an inhibitory effect on the production of inflammatory cytokines.


Asunto(s)
Neoplasias de la Boca , Carcinoma de Células Escamosas de Cabeza y Cuello , Humanos , Interleucina-6/genética , Lipopolisacáridos/farmacología , Neoplasias de la Boca/genética , FN-kappa B/metabolismo , Moléculas de Patrón Molecular Asociado a Patógenos , Porphyromonas gingivalis/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Receptores Toll-Like , Línea Celular Tumoral
9.
Med Oncol ; 38(9): 110, 2021 Aug 06.
Artículo en Inglés | MEDLINE | ID: mdl-34357463

RESUMEN

EGFR-targeted therapies are reported to yield modest effect in OSCC. Activation of NFκB signaling is considered as molecular driver of EGFR inhibitor resistance in various cancers. In this scenario, present study focused on the molecular crosstalk between EGFR and NFκB signaling pathways and its therapeutic importance in OSCC. The EGFR- NFκB p65 co-expressed human OSCC cell lines UPCI:SCC066, UPCI:SCC040 and UM-SCC083B were used for in vitro studies. Recombinant human EGF, siRNAs, Western blot and qRT-PCR were used to dissect the molecular crosstalk between EGFR-NFκB signaling pathways in OSCCs. The effect of NFκB p65 knockdown on cancer hallmarks was studied by respective functional assays and RNA-Seq analysis was performed to identify the differentially expressed genes upon NFκB p65 knockdown. Gefitinib and Bay 11-7085 combination treatment was done to study the chemotherapeutic potential of EGFR- NFκB axis. Significant positive correlation between EGFR and NFκB p65 expression was observed in Head and Neck TCGA data set. EGFR induction or knockdown respectively stimulate or impair the NFκB signaling in EGFR- NFκB p65 co-expressed OSCC cell lines. NFκB p65 knockdown causes apoptosis and suppresses the viability, colony formation, migration, invasion, and spheroid formation. Using RNA-seq analysis, we identified PIK3CD as the NFκB target gene, which is commonly involved in these functions. Gefitinib and Bay 11-7085 combination treatment was found to be useful in chemosensitizing the Gefitinib-resistant OSCC cells by capitulating the EGFR- NFκB signaling axis. Combination treatment using Gefitinib and Bay 11-7085 enhanced the apoptosis and reduced cell viability and colony formation in a synergistic way. Our data demonstrated that EGFR-NFκB signaling axis plays a key role in the pathogenesis of OSCCs. Therefore, simultaneous therapeutic intervention of these pathways may be a good alternative approach for the management of OSCCs.


Asunto(s)
Resistencia a Antineoplásicos/efectos de los fármacos , Gefitinib/farmacología , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Boca/tratamiento farmacológico , FN-kappa B/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Apoptosis , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Movimiento Celular , Proliferación Celular , Quimioterapia Combinada , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , FN-kappa B/genética , RNA-Seq , Células Tumorales Cultivadas
10.
Biomed Pharmacother ; 141: 111809, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34144454

RESUMEN

Oral mucocutaneous cancers (OMCs) are cancers that affect both the oral mucosa and perioral cutaneous structures. Common OMCs are squamous cell carcinoma (SCC), basal cell carcinoma (BCC) and malignant melanoma (MM). Anatomical similarities and conventions which categorizes these lesions blur the magnitude of OMCs in diverse populations. The burden of OMC is high in the sub-Saharan Africa and Indian subcontinents, and the cost of management is prohibitive in the resource-limited, developing world. Hence, there is a pressing demand for the use of cost-effective in silico approaches to identify diagnostic tools and treatment targets for diseases with high burdens in these regions. Due to their ubiquitousness and accessibility, the use of therapeutic efficacy of plant bioactive compounds in the management of OMC is both appropriate and plausible. Furthermore, screening known mechanistic disease targets with well annotated plant bioactive compound libraries is poised to improve the routine management of OMCs provided that the requisite access to database resources are available and accessible. Using natural products minimizes the side effects and morbidities associated with conventional therapies. The development of innovative treatments approaches would tremendously benefit the African and Indian populace and reduce the mortalities associated with OMCs in the developing world. Hence, we discuss herein, the potential benefits, opportunities and challenges of using bioactive compound libraries in the management of OMCs.


Asunto(s)
Antineoplásicos Fitogénicos/uso terapéutico , Neoplasias de la Boca/tratamiento farmacológico , Fitoterapia/métodos , Animales , Simulación por Computador , Humanos , Mucosa Bucal , Neoplasias de la Boca/genética
11.
Br J Cancer ; 124(12): 2004-2016, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33762722

RESUMEN

BACKGROUND: Chemoresistance is one of the major factors for treatment failure in OSCC. Identifying key resistance triggering molecules will be useful strategy for developing novel treatment methods. METHODS: To identify the causative factors of chemoresistance, we performed RNA sequencing and global proteomic profiling of human OSCC lines presenting with sensitive, early and late cisplatin-resistance patterns. RESULTS: From the common set of dysregulated genes from both the analysis, RRBP1 was identified to be upregulated in both early and late cisplatin-resistant cells with respect to the sensitive counterpart. Analysis of OSCC patient sample indicates that RRBP1 expression is upregulated in chemotherapy-non-responder tumours as compared to chemotherapy-responder tumours. Genetic (knockout) or pharmacological (Radezolid, represses expression of RRBP1) inhibition of RRBP1 restores cisplatin-mediated cell death in chemo-resistant OSCC. Mechanistically, RRBP1 regulates Yes-associated protein1 (YAP1), a key protein in the Hippo pathway to induce chemoresistance. The PDC xenograft data suggests that knockout of RRBP1 induces cisplatin-mediated cell death and facilitates a significant reduction of tumour burden. CONCLUSION: Overall, our data suggests that (I) RRBP1 is a major driver of cisplatin-resistance in OSCC, (II) RRBP1 regulates YAP1 expression to mediate cisplatin-resistance, (III) Radezolid represses RRBP1 expression and (IV) targeting RRBP1 reverses cisplatin-induced chemoresistance in advanced OSCC.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Proteínas Portadoras/fisiología , Cisplatino/uso terapéutico , Resistencia a Antineoplásicos/genética , Neoplasias de la Boca/tratamiento farmacológico , Animales , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Proteínas Portadoras/genética , Línea Celular Tumoral , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Inactivación de Genes , Células HEK293 , Vía de Señalización Hippo/efectos de los fármacos , Vía de Señalización Hippo/genética , Humanos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 46(12): 1315-1324, 2021 Dec 28.
Artículo en Inglés, Chino | MEDLINE | ID: mdl-35232899

RESUMEN

OBJECTIVES: To screen genes related to the prognosis of oral squamous cell carcinoma (OSCC), and to explore its role and mechanism in the occurrence and development of OSCC. METHODS: The data and the biological information in 330 OSCC tumor samples with head and neck squamous cell carcinoma (HNSCC) (OSCC group) and 37 normal samples (normal sample group) were screened and included, which came from the cancer genome atlas (TCGA) database. The differentially expressed genes were screened out by biological information analysis between the 2 groups. Furthermore, according to the tumor T grade (T1+T2 group: 114 cases, T3+T4 group: 216 cases), metastasis (positive group: 163 cases, negative group: 167 cases) and pathological grade (G1+G2 group: 244 cases, G3 +G4 group: 86 cases), the samples were divided into different groups respectively, and the differential genes were obtained separately, then the intersections of the differential expressed genes related to the prognosis of OSCC were screened. The different gene with the largest different multiples [hyaluronan mediated motility receptor (HMMR)] was selected for the next step in order to explore the relationship between HMMR and clinical grading (Stage I+II group: 69 cases, Stage III +IV group: 261 cases), as well as the relationship between T grade, metastasis and pathological grade. According to the median value of HMMR expression, the samples were divided into a high expression group and a low expression group (high expression group: 165 cases, low expression group: 165 cases); Kaplan-Meier survival analysis was used to explore the relationship between HMMR expression and prognosis. Tumor tissue specimens and corresponding normal oral mucosal tissue specimens in 50 OSCC patients, who underwent surgery in the First Affiliated Hospital of Hunan University of Traditional Chinese Medicine from January 2014 to January 2016, were collected. Real-time RT-PCR and Western blotting and immunohistochemistry were used to verify the bioinformatics analysis results. Kaplan-Meier survival analysis was used to examine the relationship between the positive and negative expression of HMMR immunohistochemical staining (positive group: 32 cases, negative group: 18 cases) and prognostic related factors, and Cox regression analysis model was used to explore the prognostic risk factors of OSCC. The cell proliferation experiment and the cell scratch experiment were used to evaluate the effect of down-regulation of HMMR on the proliferation and migration of OSCC cells. RESULTS: HMMR was highly expressed in OSCC tissues. Compared with the low HMMR expression group, the prognostic related factors in the HMMR high expression group was significantly lower, with significant difference (all P<0.05); the high expression of HMMR was significantly related with the T grade (RR=1.33, P<0.05), lymphonodus metastasis (RR=1.74, P<0.05), the clinical stage (RR=1.49, P<0.05), and it was an independent prognostic risk factor for OSCC (RR=1.45, P<0.05). Down-regulation of HMMR can inhibit the proliferation and migration of OSCC cells, with significant difference (P<0.05 or P<0.01). CONCLUSIONS: HMMR, as a proto-oncogene of OSCC, can promote the occurrence and development of OSCC, and it may be used as a potential early diagnostic marker and a new target for therapy.


Asunto(s)
Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Regulación Neoplásica de la Expresión Génica , Neoplasias de Cabeza y Cuello/genética , Humanos , Ácido Hialurónico , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética
13.
Protein Pept Lett ; 28(7): 735-749, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33302827

RESUMEN

BACKGROUND: Oral cancer is a significant health problem worldwide. Oral squamous cell carcinoma (OSCC) is a malignant neoplasm of epithelial cells that mostly affects different anatomical sites in the head and neck and derives from the squamous epithelium or displays similar morphological characteristics. Generally, OSCC is often the end stage of several changes in the stratified squamous epithelium, which begin as epithelial dysplasia and progress by breaking the basement membrane and invading adjacent tissues. Several plant-based drugs with potent anti-cancer effects are considered inexpensive treatments with limited side effects for cancer and other diseases. OBJECTIVE: The aim of this review is to explore whether some Brazilian plant extracts or constituents exhibit anti-tumorigenic activity or have a cytotoxic effect on human oral carcinoma cells. METHODS: Briefly, OSCC and several metabolites derived from Brazilian plants (i.e., flavonoids, vinblastine, irinotecan, etoposide and paclitaxel) were used as keywords to search the literature on PubMed, GenBank and GeneCards. RESULTS: The results showed that these five chemical compounds found in Cerrado Biome plants exhibit anti-neoplastic effects. Evaluating the compounds revealed that they play a main role in the regulation of cell proliferation. CONCLUSION: Preserving and utilising the biodiversity of our planet, especially in unique ecosystems, such as the Cerrado Biome, may prove essential to preserving and promoting human health in modern contexts.


Asunto(s)
Anticarcinógenos/farmacología , Antineoplásicos Fitogénicos/farmacología , Carcinogénesis/efectos de los fármacos , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Proteínas de Neoplasias/genética , Anticarcinógenos/química , Anticarcinógenos/aislamiento & purificación , Antineoplásicos Fitogénicos/química , Antineoplásicos Fitogénicos/aislamiento & purificación , Brasil , Carcinogénesis/genética , Carcinogénesis/metabolismo , Carcinogénesis/patología , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Proliferación Celular/efectos de los fármacos , Biología Computacional/métodos , Etopósido/química , Etopósido/aislamiento & purificación , Etopósido/farmacología , Flavonoides/química , Flavonoides/aislamiento & purificación , Flavonoides/farmacología , Regulación Neoplásica de la Expresión Génica , Humanos , Irinotecán/química , Irinotecán/aislamiento & purificación , Irinotecán/farmacología , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Proteínas de Neoplasias/antagonistas & inhibidores , Proteínas de Neoplasias/metabolismo , Paclitaxel/química , Paclitaxel/aislamiento & purificación , Paclitaxel/farmacología , Extractos Vegetales/química , Plantas Medicinales , Vinblastina/química , Vinblastina/aislamiento & purificación , Vinblastina/farmacología
14.
Phytomedicine ; 80: 153386, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-33113500

RESUMEN

BACKGROUND: Overexpression of polycomb protein contributes to epigenetic repression in oral squamous cell carcinoma (OSCC) ensuing in poor prognosis and aggressive phenotype. Several plant-based compounds could help prevent epigenome alteration and cancer progression, but their low bioavailability limits their therapeutic activity. HYPOTHESIS: In this study, we have synthesized genistein nanoformulation (GLNPs) and evaluated its epigenetic regulation mechanism for selective apoptosis induction in OSCC. METHODS: Lactalbumin was used to prepare nanoformulation of Genistein. The mechanism of epigenetic regulation and selective apoptosis by Genistein loaded nanoparticles was studied in OSCC cell line JHU011 and fibroblast cell line L929 using immunofluorescence, Western blotting and ChIP-qPCR assay. RESULTS: We have found that GLNPs treatment selectively induced apoptosis in OSCC compared to the normal fibroblast cells. This selective effect in OSCC is achieved through enhanced reactive oxygen species (ROS) generation followed by Bax mitochondrial translocation and caspase 3 activation. Further, GLNPs induced withdrawal of epigenetic transcription repression through concurrent downregulation of the polycomb group proteins (PcG) Bmi 1 and EZH2 along with their successive targets, UbH2AK119 and H3K27me3, which have immense therapeutic implications in the treatment of OSCC. Last, we have established that GLNPs regulate EZH2expression through proteasomal mediated degradation and 3PK inhibition; 3PK protein was found physically linked with EZH2 protein and its promoter region (-1107 to -1002). This event indicates that 3PK might play some crucial role in EZH2 expression and epigenetic control of OSCC. Moreover, the formulation showed improved biodistribution, aqueous dispersibility and enhanced biocompatibility In-vivo. CONCLUSIONS: These results provide evidence that GLNPs may withdraw epigenetic transcriptional repression and selectively induce apoptosis in human oral squamous cell carcinoma.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Carcinoma de Células Escamosas/tratamiento farmacológico , Proteína Potenciadora del Homólogo Zeste 2/metabolismo , Genisteína/farmacología , Neoplasias de la Boca/tratamiento farmacológico , Animales , Antineoplásicos Fitogénicos/farmacocinética , Apoptosis/efectos de los fármacos , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Regulación hacia Abajo/efectos de los fármacos , Sistemas de Liberación de Medicamentos , Epigénesis Genética/efectos de los fármacos , Genisteína/administración & dosificación , Genisteína/farmacocinética , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Masculino , Ratones Endogámicos BALB C , Neoplasias de la Boca/genética , Neoplasias de la Boca/metabolismo , Neoplasias de la Boca/patología , Nanopartículas/administración & dosificación , Nanopartículas/química , Proteínas Serina-Treonina Quinasas/metabolismo , Transducción de Señal/efectos de los fármacos , Distribución Tisular , Ensayos Antitumor por Modelo de Xenoinjerto
15.
J Egypt Natl Canc Inst ; 32(1): 12, 2020 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-32372215

RESUMEN

BACKGROUND: Squamous cell carcinoma of the oral cavity (OSCC) is the sixth most common malignancy. Surgery is mainstay treatment for oral cancers. Surgery in locally advanced OSCC presents many challenges primarily because the head and neck have critical structures that can be damaged by tumor or treatment. It is thought that neoadjuvant chemotherapy (NC) in locally advanced OSCC is able to shrink tumor size. Chemoresistancy is a problem due to hypoxic microenvironment characterized by increased expression of HIF-1α. It is also regulated by miR-210 as well as increased expression of CD44 and CD133. Melatonin has a powerful antioxidant and oncostatic effects that are expected to improve tumor hypoxia and clinical response. Fifty patients with OSCC were included and randomized. miR-210 and CD44 expression were measured before and after intervention using qRT-PCR absolute quantification, and clinical response was evaluated according to RECIST 1.1 criteria. This study aims to determine the effect of melatonin in improving the clinical response of patients with locally advanced oral squamous cell carcinoma (OSCC) after neoadjuvant chemotherapy to miR-210 and CD44 expression. RESULTS: Melatonin administration reduced miR-210 levels but not significant (p = 0.767). CD44 expression also decreased in the melatonin group compared with placebo yet was not significant (p = 0.103). There was a decrease in the expression of miR-210 and CD44 followed by a decrease in the percentage of residual tumor but not significant (p = 0.114). CONCLUSION: In OSCC, the addition of 20-mg melatonin to neoadjuvant chemotherapy (NC) reduced the expression of miR-210 and CD44 and decreased the percentage of tumor residue; however, no statistically significant result was observed. TRIAL REGISTRATION: This study is registered to ClinicalTrials.gov under trial registration number: NCT04137627 with date of registration on October 22, 2019-retrospectively registered, accessible from: https://clinicaltrials.gov/ct2/show/NCT04137627.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Receptores de Hialuranos/metabolismo , Melatonina/administración & dosificación , MicroARNs/metabolismo , Neoplasias de la Boca/terapia , Carcinoma de Células Escamosas de Cabeza y Cuello/terapia , Adolescente , Adulto , Hipoxia de la Célula/efectos de los fármacos , Hipoxia de la Célula/genética , Línea Celular Tumoral , Quimioterapia Adyuvante/métodos , Niño , Método Doble Ciego , Resistencia a Antineoplásicos/efectos de los fármacos , Resistencia a Antineoplásicos/genética , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Receptores de Hialuranos/análisis , Masculino , MicroARNs/análisis , Persona de Mediana Edad , Mucosa Bucal/patología , Mucosa Bucal/cirugía , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Terapia Neoadyuvante/métodos , Estadificación de Neoplasias , Criterios de Evaluación de Respuesta en Tumores Sólidos , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Carga Tumoral/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos , Microambiente Tumoral/genética , Adulto Joven
16.
Cell Biol Int ; 44(7): 1447-1457, 2020 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-32159250

RESUMEN

Esophageal squamous cell carcinoma (ESCC) belongs to one of the most common malignant tumors worldwide and possesses high mortality. Long non-coding RNAs (lncRNAs) have been demonstrated to be essential biological participants in the progression of ESCC. On the basis of bio-informatics prediction, forkhead box P4 antisense RNA 1 (FOXP4-AS1) and forkhead box P4 (FOXP4) were upregulated in esophageal carcinoma samples and were positively correlated with each other. The present study aimed to explore the function of FOXP4-AS1 and FOXP4 in ESCC cells. Function assays disclosed that knockdown of FOXP4-AS1 or FOXP4 efficiently suppressed cell proliferation and induced cell apoptosis. Moreover, FOXP4-AS1 positively regulated FOXP4 by interacting with insulin-like growth factor 2 mRNA-binding protein 2 (IGF2BP2) to stabilize FOXP4 messenger RNA. In addition, FOXP4-AS1 could upregulate the expression of FOXP4 by sponging miR-3184-5p. Finally, we found that Yin Yang 1 (YY1) is a transcription factor that can transcriptionally activate both FOXP4-AS1 and FOXP4 in ESCC cells. In a word, YY1-induced upregulation of FOXP4-AS1 and FOXP4 promote the proliferation of ESCC cells.


Asunto(s)
Proliferación Celular/genética , Neoplasias Esofágicas/patología , Factores de Transcripción Forkhead/genética , Regulación Neoplásica de la Expresión Génica/genética , Factor de Transcripción YY1/genética , Línea Celular Tumoral , Neoplasias Esofágicas/genética , Carcinoma de Células Escamosas de Esófago/genética , Carcinoma de Células Escamosas de Esófago/patología , Neoplasias de Cabeza y Cuello/genética , Humanos , Neoplasias de la Boca/genética , ARN sin Sentido/genética , ARN Largo no Codificante/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Regulación hacia Arriba , Factor de Transcripción YY1/metabolismo
17.
Cancer Prev Res (Phila) ; 13(4): 357-366, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31969344

RESUMEN

We previously reported that the environmental pollutant and tobacco smoke constituent dibenzo[def,p]chrysene (DBP) induced DNA damage, altered DNA methylation and induced oral squamous cell carcinoma (OSCC) in mice. In the present study, we showed that 5% dietary black raspberry (BRB) significantly reduced (P < 0.05) the levels of DBP-DNA adducts in the mouse oral cavity with comparable effect to those of its constitutes. Thus, only BRB was selected to examine if aberrant DNA methylation induced by DBP can be altered by BRB. Using comparative genome-wide DNA methylation analysis, we identified 479 hypermethylated and 481 hypomethylated sites (q < 0.01, methylation difference >25%) between the oral tissues of mice treated with DBP and fed control diet or diet containing BRB. Among the 30 differential methylated sites (DMS) induced by DBP, we found DMS mapped to Fgf3, Qrich2, Rmdn2, and Cbarp were hypermethylated by BRB whereas hypomethylated by DBP at either the exact position or proximal sites; DMS mapped to Vamp3, Ppp1rB1, Pkm, and Zfp316 were hypomethylated by BRB but hypermethylated by DBP at proximal sites. In addition to Fgf3, 2 DMS mapped to Fgf4 and Fgf13 were hypermethylated by BRB; these fibroblast growth factors are involved in regulation of the epithelial-mesenchymal transition (EMT) pathway as identified by IPA. Moreover, BRB significantly reduced (P < 0.05) the tumor incidence from 70% to 46.7%. Taken together, the inhibitory effects of BRB on DNA damage combined with its effects on epigenetic alterations may account for BRB inhibition of oral tumorigenesis induced by DBP. SIGNIFICANCE: We provided mechanistic insights that can account for the inhibition of oral tumors by BRB, which could serve as the framework for future chemopreventive trials for addicted smokers as well as non- or former smokers who are exposed to environmental carcinogens.


Asunto(s)
Benzopirenos/toxicidad , Epigénesis Genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Neoplasias de la Boca/tratamiento farmacológico , Extractos Vegetales/farmacología , Rubus/química , Contaminación por Humo de Tabaco/prevención & control , Animales , Apoptosis , Biomarcadores de Tumor/genética , Carcinógenos/toxicidad , Carcinoma de Células Escamosas/inducido químicamente , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Proliferación Celular , Metilación de ADN , Femenino , Humanos , Ratones , Ratones Endogámicos C57BL , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Células Tumorales Cultivadas , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Nanomedicine (Lond) ; 15(1): 55-75, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31868110

RESUMEN

Background: Oral cancer is highly aggressive due to difficult diagnosis, therapy resistance and increasing frequency; thus finding prevention therapies is very important. Aim: This study evaluates the use of gold and silver nanoparticles (NPs), phyto-synthesized with Cornus mas extract against oral dysplastic lesions. Methods: NPs were characterized by UV-Vis, Fourier-transform infrared spectroscopy, transmission electron microscopy, x-ray diffraction and laser Doppler microelectrophoresis. Biological testing employed two human oral cell lines: gingival fibroblasts and dysplastic keratinocytes and evaluated viability, cell death mechanisms and cellular uptake. Results: NPs induced selective toxic effects against dysplastic cells. p53/BAX/BCL2 activation and PI3K/AKT inhibition led to cell death through necrosis and apoptosis. NPs also induced antioxidant and anti-inflammatory effects. Conclusion: NPs of gold and silver showed promising beneficial effects in the therapy of oral dysplasia.


Asunto(s)
Cornus/química , Nanopartículas del Metal/química , Neoplasias de la Boca/tratamiento farmacológico , Extractos Vegetales/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Fibroblastos/efectos de los fármacos , Encía/efectos de los fármacos , Encía/patología , Oro/química , Humanos , Queratinocitos/efectos de los fármacos , Queratinocitos/patología , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Extractos Vegetales/química , Proteínas Proto-Oncogénicas c-bcl-2/genética , Plata/química , Proteína p53 Supresora de Tumor/genética , Rayos Ultravioleta , Proteína X Asociada a bcl-2/genética
19.
Sci Rep ; 9(1): 16985, 2019 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-31740710

RESUMEN

People of north-eastern states of India consume raw areca-nut (RAN) and lime which could lead to oral, esophageal and gastric cancers. However, the incidence of these cancers are significantly lesser in those who consume pieces of Potentilla fulgens root along with RAN. Since evaluation of anticancer role, if any, of P. fulgens on RAN-mediated genetic alterations in human is difficult because of other compounding factors, this study was undertaken in mice to focus on gastric carcinogenesis since ad libitum administration of RAN extract with lime in drinking water induced stomach cancer due to greater exposure of its lining. A total of 160 mice were used at different time points and either methanol extract of P. fulgens roots (PRE) or mixture of four compounds of ethyl-acetate fraction (EA-mixture) was mixed with mice feed. Histological studies revealed that RAN + lime induced cancer in all the mice and interestingly only 20% developed cancer when PRE/EA-mixture was provided along with RAN + lime. Higher frequency of precocious anaphase and over expression of p53 and Securin genes were significantly reduced by PRE/EA-mixture. Thus PRE/EA-mixture mitigates the RAN-induced tumor-initiating process in stomach by maintaining expression of tumor suppressor and check-point genes under control.


Asunto(s)
Neoplasias/prevención & control , Extractos Vegetales/farmacología , Raíces de Plantas/química , Potentilla/química , Acetatos/química , Animales , Areca/química , Carcinógenos , Neoplasias Esofágicas/inducido químicamente , Neoplasias Esofágicas/genética , Neoplasias Esofágicas/prevención & control , Humanos , India , Metanol/química , Ratones , Neoplasias de la Boca/inducido químicamente , Neoplasias de la Boca/genética , Neoplasias de la Boca/prevención & control , Neoplasias/inducido químicamente , Neoplasias/genética , Nueces/química , Fitoterapia/métodos , Extractos Vegetales/química , Extractos Vegetales/aislamiento & purificación , Securina/genética , Neoplasias Gástricas/inducido químicamente , Neoplasias Gástricas/genética , Neoplasias Gástricas/prevención & control , Proteína p53 Supresora de Tumor/genética
20.
Environ Toxicol ; 34(10): 1085-1093, 2019 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-31184425

RESUMEN

Geraniin has been reported to have numerous biological activities, including antiviral, antihypertensive, antihyperglycaemic, liver protective, antidiabetic, and apoptotic activities. However, the anti-migration effects of geraniin on oral cancer remain elusive. In this study, we revealed the potential antitumor mechanisms of geraniin through the inhibition of the migration and invasion of human oral cancer cell lines SCC-9 and SCC-14. The results of gelatin zymography and Western blot assays revealed that geraniin significantly reduced the activity and expression of matrix metalloproteinase-2 (MMP-2) of oral cancer cells in a concentration-dependent manner. Furthermore, geraniin potently suppressed the phosphorylation of focal adhesion kinase (FAK), Src, and extracellular signal-regulated kinase (ERK)1/2 but did not affect the phosphorylation of p38 mitogen-activated protein kinase (MAPK) and c-Jun N-terminal kinase 1/2. Moreover, blocking the MAPK/ERK1/2 pathway significantly enhanced the anti-migration ability of geraniin in oral cancer cells. In conclusion, we demonstrated that geraniin inhibits the motility of SCC-9 and SCC-14 cells in vitro through a molecular mechanism that involves the attenuation of MMP-2 expression and activity mediated by decreased FAK/Src and ERK1/2 pathways.


Asunto(s)
Antineoplásicos/farmacología , Medicamentos Herbarios Chinos/farmacología , Proteína-Tirosina Quinasas de Adhesión Focal/metabolismo , Glucósidos/farmacología , Taninos Hidrolizables/farmacología , Metaloproteinasa 2 de la Matriz/metabolismo , Neoplasias de la Boca/metabolismo , Familia-src Quinasas/metabolismo , Línea Celular Tumoral , Movimiento Celular/efectos de los fármacos , Proteína-Tirosina Quinasas de Adhesión Focal/genética , Geranium/química , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Metaloproteinasa 2 de la Matriz/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Proteínas Quinasas Activadas por Mitógenos/genética , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Neoplasias de la Boca/genética , Neoplasias de la Boca/fisiopatología , Familia-src Quinasas/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA