Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 52
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
BMC Cancer ; 23(1): 1062, 2023 Nov 03.
Artículo en Inglés | MEDLINE | ID: mdl-37923984

RESUMEN

BACKGROUND: This study aimed to find out the characteristics in relation to tumor recurrence in diffused-tenosynovial giant cell tumor of temporomandibular joint and to develop and validate the prognostic model for personalized prediction. METHODS: From April 2009 to January 2021, patients with diffused-tenosynovial giant cell tumor of temporomandibular joint at a single center were included in this study. The clinical features and local recurrence-free survival were assessed through the expression of the Ki-67 index and colony-stimulating factor 1 receptor expression. Both univariate and multivariate analyses were performed on the prognostic factors for local recurrence-free survival. An independent predictor nomogram and pertinent tumor characteristics were included. RESULTS: The retrospective study enrolling seventy eligible patients at the Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine. During the follow-up time, eleven patients suffered tumor recurrence. Age was an independent risk factor for local recurrence-free survival (P = 0.032). The Ki-67 index varied significantly in different sites (P = 0.034) and tumor volume (P = 0.017). Multivariate logistic regression was used to develop the prediction model using both statistical significance and prognostic indicators. The C-index of the nomogram based on age, site, Ki-67, and colony-stimulating factor 1 receptor was 0.833. These variates provided good predicted accuracy for a nomogram on local recurrence-free survival. Diffused-tenosynovial giant cell tumor from the temporomandibular joint is extremely uncommon, and certain clinical traits are linked to the tumor proliferation index. CONCLUSIONS: We identified the risk indicators and developed a nomogram in this study to forecast the likelihood of local recurrence-free survival in patients with diffused-tenosynovial giant cell tumor from temporomandibular joint.


Asunto(s)
Tumor de Células Gigantes de las Vainas Tendinosas , Recurrencia Local de Neoplasia , Humanos , Estudios Retrospectivos , Recurrencia Local de Neoplasia/epidemiología , Recurrencia Local de Neoplasia/patología , Factor Estimulante de Colonias de Macrófagos , Antígeno Ki-67 , China , Tumor de Células Gigantes de las Vainas Tendinosas/patología , Articulación Temporomandibular/patología
2.
Cancer Sci ; 113(2): 478-488, 2022 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-34826159

RESUMEN

The aim of this study was to: (a) explore the potential mechanism of cancer cell sensitivity to cisplatin, docetaxel, and 5-fluorouracil (TPF) in oral squamous cell carcinoma (OSCC) patients overexpressing growth differentiation factor 15 (GDF15); and (b) identify potential alternative agents for patients who might not benefit from inductive TPF chemotherapy. The results indicated that OSCC cells overexpressing GDF15 were sensitive to TPF through a caspase-9-dependent pathway both in vitro and in vivo. Immunoprecipitation combined with mass spectrometry revealed that the erbB2 protein was a potential GDF15-binding protein, which was verified by coimmunoprecipitation. Growth differentiation factor 15 overexpression promoted OSCC cell proliferation through erbB2 phosphorylation, as well as downstream AKT and Erk signaling pathways. When GDF15 expression was blocked, the phosphorylation of both the erbB2 and AKT/Erk pathways was downregulated. When OSCC cells with GDF15 overexpression were treated with the erbB2 phosphorylation inhibitor, CI-1033, cell proliferation and xenograft growth colony formation were significantly blocked (P < .05). Thus, GDF15-overexpressing OSCC tumors are sensitive to TPF chemoagents through caspase-9-dependent pathways. Growth differentiation factor 15 overexpression promotes OSCC proliferation through erbB2 phosphorylation. Thus, ErbB2 inhibitors could represent potential targeted drugs or an alternative therapy for OSCC patients with GDF15 overexpression.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Factor 15 de Diferenciación de Crecimiento/metabolismo , Neoplasias de la Boca/metabolismo , Receptor ErbB-2/metabolismo , Carcinoma de Células Escamosas de Cabeza y Cuello/metabolismo , Animales , Apoptosis , Caspasa 9/metabolismo , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transformación Celular Neoplásica/efectos de los fármacos , Cisplatino/farmacología , Fluorouracilo/farmacología , Humanos , Ratones , Morfolinas/farmacología , Fosforilación/efectos de los fármacos , Receptor ErbB-2/antagonistas & inhibidores , Transducción de Señal/efectos de los fármacos , Taxoides/farmacología
3.
BMC Oral Health ; 22(1): 265, 2022 06 29.
Artículo en Inglés | MEDLINE | ID: mdl-35768820

RESUMEN

BACKGROUND: Schwannomas or neurilemmomas are well-encapsulated, benign, solitary, and slow-growing tumors that originate from Schwann cells of the nerve sheath. Extracranial schwannoma is reported to have a relatively high incidence in the tongue while an extremely low incidence in the floor of mouth. In the current study, we presented the first case series of hypoglossal nerve-derived schwannoma in the floor of mouth in Asia. METHODS: A retrospective study of 9 surgical cases of hypoglossal nerve-derived schwannoma in the floor of mouth was performed. The patient and tumor characteristics were evaluated by physical, radiological and pathological examination. Details of operation and complications were also recorded. RESULTS: Hypoglossal nerve-derived schwannoma in the floor of mouth showed a well-defined boundary with a firm texture, smooth surface and good mobility on palpation. The median maximum diameter of the tumors was 4.3 cm (range 2.8-7.0 cm). The median operative time and bleeding volumes were 89.4 min (range 47-180 min) and 99.2 mL (range 15-200 mL), respectively. All cases received complete surgical excision. CONCLUSION: In this study, we presented the diagnosis and management of hypoglossal nerve-derived schwannoma in the floor of mouth for the first time in Asia. The study provided us with a recommendation for consideration of the diagnosis of hypoglossal schwannoma when a patient presents with a mass in the floor of mouth.


Asunto(s)
Neoplasias de los Nervios Craneales , Enfermedades del Nervio Hipogloso , Neurilemoma , Neoplasias de los Nervios Craneales/diagnóstico , Neoplasias de los Nervios Craneales/patología , Neoplasias de los Nervios Craneales/cirugía , Humanos , Nervio Hipogloso/patología , Nervio Hipogloso/cirugía , Enfermedades del Nervio Hipogloso/diagnóstico , Enfermedades del Nervio Hipogloso/etiología , Enfermedades del Nervio Hipogloso/cirugía , Suelo de la Boca/patología , Suelo de la Boca/cirugía , Neurilemoma/diagnóstico por imagen , Neurilemoma/cirugía , Estudios Retrospectivos
4.
J Oral Pathol Med ; 50(8): 795-802, 2021 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-34157171

RESUMEN

BACKGROUND: Annexin A1, a member of the Annexin superfamily, has been shown to play a vital role in a broad range of molecular and cellular processes. This study aims to explore the relationship between the Annexin A1 expression and the clinical response to cisplatin, docetaxel and 5-fluorouracil (TPF) as induction chemotherapy in patients with oral squamous cell carcinoma (OSCC). METHODS: This study recruited two hundred thirty-two patients from a III/IVA OSCC trial. Immunohistochemistry was used to assess the level of Annexin A1 expression. Overexpression and knockdown methods in HB96, HN4 and CAL27 cell lines were used to assess the role of Annexin A1 in the neoplastic cellular response to chemotherapy. RESULTS: We found that reduced expression of Annexin A1 conferred a prognostic benefit from induction chemotherapy based on the TPF drug combination in patients with moderately/poorly differentiated disease. Using an in vitro model, we found that low Annexin A1 enhanced cellular proliferation by activating the EGFR/AKT signalling pathway and inhibiting p27 expression. Furthermore, low Annexin A1 initiated a significant decrease in cell viability after treatment with TPF agents. In addition, downregulation of Annexin A1 promoted apoptosis induced by docetaxel, cisplatin and 5-fluorouracil, and upregulation of Annexin A1 inhibited apoptosis. CONCLUSION: Annexin A1 may be of prognostic value in patients with locally advanced OSCC who are managed with TPF chemotherapy, as low Annexin A1 promotes chemosensitivity to TPF chemotherapy in oral cancer cells via enhanced caspase-dependent apoptosis.


Asunto(s)
Anexina A1 , Carcinoma de Células Escamosas , Neoplasias de Cabeza y Cuello , Neoplasias de la Boca , Anexina A1/genética , Anexina A1/uso terapéutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Cisplatino/farmacología , Cisplatino/uso terapéutico , Docetaxel/farmacología , Docetaxel/uso terapéutico , Fluorouracilo/uso terapéutico , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Quimioterapia de Inducción , Neoplasias de la Boca/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Taxoides/uso terapéutico
5.
J Nanobiotechnology ; 19(1): 157, 2021 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-34039370

RESUMEN

BACKGROUND: Smart nanoscale drug delivery systems that target acidic tumor microenvironments (TME) could offer controlled release of drugs and modulate the hypoxic TME to enhance cancer therapy. The majority of previously reported MnO2 nanostructures are nanoparticles, nanosheets, or nanocomposites incorporated with other types of nanoparticles, which may not offer the most effective method for drug loading or for the controlled release of therapeutic payloads. Previous studies have designed MnO2 nanoshells that achieve tumor-specific and enhanced combination therapy for localized advanced cancer. However, the therapeutic effect of MnO2 nanoshells on metastatic cancer is still uncertain. RESULT: Here, intelligent "theranostic" platforms were synthesized based on hollow mesoporous MnO2 (H-MnO2) nanoshells that were loaded with chemotherapy agents docetaxel and cisplatin (TP) to form H-MnO2-PEG/TP nanoshells, which were designed to alleviate tumor hypoxia, attenuate angiogenesis, trigger the dissolution of Mn2+, and synergize the efficacy of first-class anticancer chemotherapy. The obtained H-MnO2-PEG/TP nanoshells decomposed in the acidic TME, releasing the loaded drugs (TP) and simultaneously attenuated tumor hypoxia and hypoxia-inducible factor-1α (HIF-1α) expression by inducing endogenous tumor hydrogen peroxide (H2O2) decomposition. In vitro experiments showed that compared with the control group, the proliferation, colony formation and migration ability of CAL27 and SCC7 cells were significantly reduced in H-MnO2-PEG/TP group, while cell apoptosis was enhanced, and the expression of hypoxia-inducible factor-1α(HIF-1α) was down-regulated. In vivo experiments showed that tumor to normal organ uptake ratio (T/N ratio) of mice in H-MnO2-PEG/TP group was significantly higher than that in TP group alone (without the nanoparticle), and tumor growth was partially delayed. In the H-MnO2-PEG/TP treatment group, HE staining showed that most of the tumor cells were severely damaged, and TUNEL assay showed cell apoptosis was up-regulated. He staining of renal and liver sections showed no obvious fibrosis, necrosis or hypertrophy, indicating good biosafety. Fluorescence staining showed that HIF-1α expression was decreased, suggesting that the accumulation of MnO2 in the tumor caused the decomposition of H2O2 into O2 and alleviated the hypoxia of the tumor. CONCLUSION: In conclusion, a remarkable in vivo and in vitro synergistic therapeutic effect is achieved through the combination of TP chemotherapy, which simultaneously triggered a series of antiangiogenic and oxidative antitumor reactions.


Asunto(s)
Carcinoma de Células Escamosas/tratamiento farmacológico , Quimioterapia/métodos , Hipoxia/tratamiento farmacológico , Compuestos de Manganeso/química , Neoplasias de la Boca/tratamiento farmacológico , Nanocáscaras/química , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Microambiente Tumoral/efectos de los fármacos , Inhibidores de la Angiogénesis/farmacología , Animales , Antineoplásicos/farmacología , Línea Celular Tumoral , Sistemas de Liberación de Medicamentos , Neoplasias de Cabeza y Cuello/tratamiento farmacológico , Humanos , Peróxido de Hidrógeno/metabolismo , Concentración de Iones de Hidrógeno , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Nanopartículas/química , Óxidos/química , Nanomedicina Teranóstica/métodos , Hipoxia Tumoral/efectos de los fármacos
6.
J Cell Mol Med ; 24(13): 7163-7174, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-32469152

RESUMEN

The up-regulation of EMT regulator Twist1 has been implicated in vasculogenic mimicry (VM) formation in human triple-negative breast cancer (TNBC). Twist1 targets the Claudin15 promoter in hepatocellular carcinoma cells. Claudin family members are related with TNBC. However, the relationship between Claudin15 and VM formation is not clear. In this study, we first found that Claudin15 expression was frequently down-regulated in human TNBC, and Claudin15 down-regulation was significantly associated with VM and Twist1 nuclear expression. Claudin15 down-regulation correlated with shorter survival compared with high levels. Claudin15 silence significantly enhanced cell motility, invasiveness and VM formation in the non-TNBC MCF-7 cells. Conversely, an up-regulation of Claudin15 remarkably reduced TNBC MDA-MB-231 cell migration, invasion and VM formation. We also showed that down-regulation of Claudin15 was Twist1-dependent, and Twist1 repressed Claudin15 promoter activity. Furthermore, GeneChip analyses of mammary glands of Claudin15-deficient mice indicated that Claudin18 and Jun might be downstream factors of Twist1-Claudin15. Our results suggest that Twist1 induced VM through Claudin15 suppression in TNBC, and Twist1 inhibition of Claudin15 might involve Claudin18 and Jun expression.


Asunto(s)
Claudinas , Regulación Neoplásica de la Expresión Génica , Proteínas Nucleares , Neoplasias de la Mama Triple Negativas , Proteína 1 Relacionada con Twist , Animales , Femenino , Humanos , Antígenos CD/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Movimiento Celular/genética , Núcleo Celular/metabolismo , Claudinas/deficiencia , Claudinas/genética , Claudinas/metabolismo , Estimación de Kaplan-Meier , Glándulas Mamarias Animales/patología , Invasividad Neoplásica , Proteínas Nucleares/metabolismo , Fenotipo , Transcripción Genética , Neoplasias de la Mama Triple Negativas/irrigación sanguínea , Neoplasias de la Mama Triple Negativas/genética , Neoplasias de la Mama Triple Negativas/patología , Proteína 1 Relacionada con Twist/metabolismo , Regulación hacia Arriba/genética , Ratones
7.
Cancer Sci ; 111(4): 1303-1313, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31994271

RESUMEN

The survival benefit from docetaxel, cisplatin and 5-fluorouracil (TPF) induction chemotherapy in oral squamous cell carcinoma (OSCC) patients is not satisfactory. Previously, we identified that stathmin, a microtubule-destabilizing protein, is overexpressed in OSCC. Here, we further investigated its role as a biomarker that impacts on OSCC chemosensitivity. We analyzed the predictive value of stathmin on TPF induction chemotherapy and its impact on OSCC cell chemosensitivity. Then, we further investigated the therapeutic effects of the combination therapy of TPF chemotherapy and PI3K-AKT-mTOR inhibitors in vitro and in vivo. We found that OSCC patients with low stathmin expression benefited from TPF induction chemotherapy, while OSCC patients with high stathmin expression could not benefit from TPF induction chemotherapy. Stathmin overexpression promoted cellular proliferation and decreased OSCC cell sensitivity to TPF treatment. In addition, inhibition of the PI3K-AKT-mTOR signaling pathway decreased stathmin expression and phosphorylation. The combination therapy of TPF chemotherapy and PI3K-AKT-mTOR inhibitors exhibited a potent antitumor effect both in vitro and in vivo. Therefore, stathmin can be used as a predictive biomarker for TPF induction chemotherapy and a combination therapy regimen based on stathmin expression might improve the survival of OSCC patients.


Asunto(s)
Protocolos de Quimioterapia Combinada Antineoplásica/administración & dosificación , Carcinoma de Células Escamosas/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Estatmina/genética , Anciano , Animales , Biomarcadores de Tumor/genética , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Cisplatino/administración & dosificación , Supervivencia sin Enfermedad , Docetaxel/administración & dosificación , Femenino , Fluorouracilo/administración & dosificación , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Xenoinjertos , Humanos , Masculino , Ratones , Persona de Mediana Edad , Neoplasias de la Boca/genética , Neoplasias de la Boca/patología , Proteína Oncogénica v-akt/genética , Fosfatidilinositol 3-Quinasas/genética , Medicina de Precisión , Transducción de Señal/efectos de los fármacos , Serina-Treonina Quinasas TOR/genética , Taxoides/administración & dosificación
8.
Biochem Biophys Res Commun ; 531(3): 282-289, 2020 10 20.
Artículo en Inglés | MEDLINE | ID: mdl-32800541

RESUMEN

B7H3 is a member of B7 family of immunoregulatory transmembrane glycoproteins associated with maintaining immune tolerance, tumor cell proliferation, migration, invasion and metabolism, drug resistance, and stem cell differentiation. Neural crest-derived Multipotent Stem Cells (MSCs) from the dental pulp has become a good choice for tissue regeneration because it is easily obtainable and has strong regeneration potentials. Although there have been many studies investigating the role of B7H3 in cancer cells and immune cells, its role in the dental pulp stem cells regeneration is unknown. In this study, we chose SHEDs (stem cells from human exfoliated deciduous teeth) as a research model to analyze the expression and function of B7H3. The result showed that SHEDs were B7H3/CD90, B7H3/CD73, B7H3/CD105 double positive, and the expression of B7H3 is primarily located within the membrane. Downregulation of B7H3 expression significantly accelerated the expansion of SHEDs through the SHP1/AKT signal axis while upregulation of B7H3 expression decreased the proliferation of SHEDs. Hence, this study indicates that B7H3 is a stem cell surface molecule and might be used as a SHEDs marker whereby its downregulation enhances the proliferation of SHEDs via the activation of B7H3/SHP1/AKT signaling pathway.


Asunto(s)
Antígenos B7/metabolismo , Técnicas de Silenciamiento del Gen , Proteína Tirosina Fosfatasa no Receptora Tipo 6/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Membrana Celular/metabolismo , Proliferación Celular , Niño , Preescolar , Humanos , Células Madre , Diente Primario
9.
Cancer Sci ; 109(10): 3197-3208, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-30022558

RESUMEN

Vasculogenic mimicry (VM) refers to the unique capability of aggressive tumor cells to mimic the pattern of embryonic vasculogenic networks. Cancer stem cells (CSC) represent a subpopulation of tumor cells endowed with the capacity for self-renewal and multilineage differentiation. Previous studies have indicated that CSC may participate in the formation of VM. With the advance of high-resolution microarrays and massively parallel sequencing technology, long noncoding RNAs (lncRNAs) are suggested to play a critical role in tumorigenesis and, in particular, the development of human hepatocellular carcinoma (HCC). Currently, no definitive relationship between lncRNA and VM formation has been described. In the current study, we demonstrated that expression of the lncRNA, n339260, is associated with CSC phenotype in HCC, and n339260 level correlated with VM, metastasis, and shorter survival time in an animal model. Overexpression of n339260 in HepG2 cells was associated with a significant increase in CSC. Additionally, the appearance of VM and vascular endothelial (VE)-cadherin, a molecular marker of VM, was also induced by n339260 overexpression. Using a short hairpin RNA approach, n339260 was silenced in tumor cells, and knockdown of n339260 was associated with reduced VM and CSC. The results of this study indicate that n339260 promotes VM, possibly by the development of CSC. The related molecular pathways may be used as novel therapeutic targets for the inhibition of HCC angiogenesis and metastasis.


Asunto(s)
Carcinoma Hepatocelular/genética , Neoplasias Hepáticas/genética , Células Madre Neoplásicas/patología , Neovascularización Patológica/genética , ARN Largo no Codificante/metabolismo , Animales , Carcinoma Hepatocelular/irrigación sanguínea , Carcinoma Hepatocelular/mortalidad , Carcinoma Hepatocelular/patología , Diferenciación Celular/genética , Transición Epitelial-Mesenquimal/genética , Femenino , Regulación Neoplásica de la Expresión Génica , Técnicas de Silenciamiento del Gen , Células Hep G2 , Hepatectomía , Humanos , Hígado/patología , Hígado/cirugía , Neoplasias Hepáticas/irrigación sanguínea , Neoplasias Hepáticas/mortalidad , Neoplasias Hepáticas/patología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Neovascularización Patológica/mortalidad , Neovascularización Patológica/patología , ARN Largo no Codificante/genética , ARN Interferente Pequeño/metabolismo , Esferoides Celulares , Ensayos Antitumor por Modelo de Xenoinjerto
10.
Proc Natl Acad Sci U S A ; 112(4): 1137-42, 2015 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-25583479

RESUMEN

Gaucher disease is caused by mutations of the GBA1 gene, which encodes the lysosomal anchored gluococerebrosidase (GCase). GBA1 mutations commonly result in protein misfolding, abnormal chaperone recognition, and premature degradation, but are less likely to affect catalytic activity. In the present study, we demonstrate that the Hsp90/HOP/Cdc37 complex recruits Hsp27 after recognition of GCase mutants with subsequent targeting of GCase mutant peptides to degradation mechanisms such as VCP and the 26S proteasome. Inhibition of Hsp27 not only increased the quantity of enzyme but also enhanced GCase activity in fibroblasts derived from patients with Gaucher disease. These findings provide insight into a possible therapeutic strategy for protein misfolding diseases by correcting chaperone binding and altering subsequent downstream patterns of protein degradation.


Asunto(s)
Enfermedad de Gaucher/metabolismo , Glucosilceramidasa/metabolismo , Proteínas de Choque Térmico HSP27/metabolismo , Proteínas HSP90 de Choque Térmico/metabolismo , Proteolisis , Deficiencias en la Proteostasis/metabolismo , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo , Chaperoninas/genética , Chaperoninas/metabolismo , Fibroblastos/metabolismo , Enfermedad de Gaucher/genética , Glucosilceramidasa/genética , Proteínas de Choque Térmico HSP27/genética , Proteínas HSP90 de Choque Térmico/genética , Células HeLa , Proteínas de Choque Térmico , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Chaperonas Moleculares , Mutación , Complejo de la Endopetidasa Proteasomal/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Pliegue de Proteína , Deficiencias en la Proteostasis/genética , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
11.
Genes Chromosomes Cancer ; 56(8): 598-607, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28393427

RESUMEN

HnRNPM is an essential splicing factor and its expression is closely correlated with invasion and metastasis of tumor cells. The CD44 cell adhesion molecule is aberrantly expressed in many breast tumors and CD44 splice variants have been implicated in specific oncogenic signaling pathways. To investigate the clinical significance and biological function of hnRNPM, immunohistochemistry, quantitative, and semiquantitative polymerase chain reaction, lentiviral transfection system and transwell invasion assays were performed. We found that hnRNPM expression was significantly upregulated in breast cancer tissues compared with benign breast lesions. Although there was no significant correlation between hnRNPM and total CD44 protein or mRNA level, there was a negative correlation between hnRNPM and CD44v6. HnRNPM and CD44s expression showed positive correlation and in particular, they were dually expressed in breast cancer tissues. Interestingly, cancer stem cells marker, ALDH1+ phenotype was positively associated with overexpression of CD44s or hnRNPM and negatively related to CD44v6. Patients with high hnRNPM tended to have higher levels of CD44s, shorter overall survival (OS) and higher rates of lymph node metastases (LNM). Remarkably, Kaplan-Meier and Cox regression analyses displayed that hnRNPM+ or CD44shigh was a poor prognostic factor for OS of patients with LNM. Upregulation of hnRNPM in MCF-7 cells caused a significant increase in cell invasion, and this effect may occur through the regulation of CD44s expression. In conclusion, overexpression of hnRNPM promotes breast cancer aggressiveness by regulating the level of CD44s, indicates a poor prognosis for patients with LNM, and has potential as therapeutic targets.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias de la Mama/metabolismo , Ribonucleoproteína Heterogénea-Nuclear Grupo M/genética , Receptores de Hialuranos/genética , Aldehído Deshidrogenasa/genética , Aldehído Deshidrogenasa/metabolismo , Biomarcadores de Tumor/metabolismo , Neoplasias de la Mama/patología , Movimiento Celular , Femenino , Ribonucleoproteína Heterogénea-Nuclear Grupo M/metabolismo , Humanos , Receptores de Hialuranos/metabolismo , Metástasis Linfática , Células MCF-7 , ARN Mensajero/genética , ARN Mensajero/metabolismo , Análisis de Supervivencia
12.
Cancer Sci ; 107(8): 1079-91, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27234159

RESUMEN

Sphere formation in conditioned serum-free culture medium supplemented with epidermal growth factor and basic fibroblast growth factor (tumorospheres) is considered useful for the enrichment of cancer stem-like cells, also known as tumor-initiating cells. We used a gene expression microarray to investigate the gene expression profile of melanoma cancer stem-like cells (MCSLCs). The results showed that MCSLCs highly expressed the following Notch signaling pathway molecules: Notch3 (NM_008716), Notch4 (NM_010929), Dtx4 (NM_172442), and JAG2 (NM_010588). Immunofluorescence staining showed tumorosphere cells highly expressed Notch4. Notch4(high) B16F10 cells were isolated by FACS, and Western blotting showed that high Notch4 expression is related to the expression of epithelial-mesenchymal transition (EMT)-associated proteins. Reduced invasive and migratory properties concomitant with the downregulation of the EMT markers Twist1, vimentin, and VE-cadherin and the overexpression of E-cadherin was observed in human melanoma A375 and MUM-2B cells. In these cells, Notch4 was also downregulated, both by Notch4 gene knockdown and by application of the γ-secretase inhibitor, DAPT. Mechanistically, the re-overexpression of Twist1 by the transfection of cells with a Twist1 expression plasmid led to an increase in VE-cadherin expression and a decrease in E-cadherin expression. Immunohistochemical analysis of 120 human melanoma tissues revealed a significant correlation between the high expression of Notch4 and the metastasis of melanoma. Taken together, our findings indicate that Notch4+ MCSLCs trigger EMT and promote the metastasis of melanoma cells.


Asunto(s)
Melanoma/metabolismo , Melanoma/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Proteínas Proto-Oncogénicas/metabolismo , Receptores Notch/metabolismo , Cadherinas/metabolismo , Línea Celular Tumoral , Transición Epitelial-Mesenquimal/genética , Humanos , Melanoma/genética , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/deficiencia , Proteínas Proto-Oncogénicas/genética , Receptor Notch4 , Receptores Notch/deficiencia , Receptores Notch/genética , Proteína 1 Relacionada con Twist/metabolismo
13.
J Neurooncol ; 130(1): 111-122, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27568035

RESUMEN

Transcription factors that induce epithelial-mesenchymal transition (EMT) promote invasion, chemoresistance and a stem-cell phenotype in epithelial tumors, but their roles in central nervous system tumors are not well-understood. We hypothesized these transcription factors have a functional impact in grades II-III gliomas. Using the National Cancer Institute (NCI) Repository for Molecular Brain Neoplasia Data (REMBRANDT) and the Cancer Genome Atlas (TCGA) Lower-Grade Glioma (LGG) data, we determined the impact of EMT-promoting transcription factors (EMT-TFs) on overall survival in grades II-III gliomas, compared their expression across common genetic subtypes and subsequently validated these findings in a set of 31 tumors using quantitative real-time polymerase chain reaction (PCR) and immunohistochemistry. Increased expression of the gene coding for the transcriptional repressor Zinc Finger E box-binding Homeobox 1 (ZEB1) was associated with a significant increase in overall survival (OS) on Kaplan-Meier analysis. Genetic subtype analysis revealed that ZEB1 expression was relatively increased in IDH1/2-mutant gliomas, and IDH1/2-mutant gliomas expressed significantly lower levels of many ZEB1 transcriptional targets. Similarly, IDH1/2-mutant tumors expressed significantly higher levels of targets of microRNA 200C (MIR200C), a key regulator of ZEB1. In a validation study, ZEB1 mRNA was significantly increased in IDH1-mutant grades II-III gliomas, and ZEB1 protein expression was more pronounced in these tumors. Our findings demonstrate a novel relationship between IDH1/2 mutations and expression of ZEB1 and its transcriptional targets. Therapy targeting ZEB1-associated pathways may represent a novel therapeutic avenue for this class of tumors.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Glioma/metabolismo , Isocitrato Deshidrogenasa/genética , Mutación/genética , Homeobox 1 de Unión a la E-Box con Dedos de Zinc/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidad , Bases de Datos Factuales/estadística & datos numéricos , Femenino , Glioma/genética , Glioma/mortalidad , Humanos , Isocitrato Deshidrogenasa/metabolismo , Estimación de Kaplan-Meier , Masculino , ARN Mensajero/metabolismo , Estadística como Asunto
14.
Inflamm Res ; 64(6): 441-51, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25948157

RESUMEN

OBJECTIVE: Toll-like receptors (TLRs) pathway has been demonstrated to play an important role in periodontitis. However, the regulatory mechanism of microRNAs (miRNAs) on TLRs pathway is still unclear. Hence, this study is to explore the function of miRNA-146a in inflammatory reaction induced by Porphyromonas gingivalis lipopolysaccharide (LPS) in human periodontal ligament cells (hPDLCs). METHODS: Cells were treated with 1 or 10 µg/ml P. gingivalis LPS. The expression of TLR2, TLR4 and miRNA-146a were measured by real-time polymerase chain reaction (PCR). Enzyme-linked immunosorbent assay (ELISA) was applied to detect nuclear factor (NF)-κ B p65 nuclear activity, interleukin-1ß (IL-1ß), IL-6, IL-8 and tumor necrosis factor-α (TNF-α). To examine the underlying mechanisms, cells were exposed to anti-TLR2/4 mAb or miRNA-146a inhibitor/mimic and evaluated by real-time PCR and ELISA. RESULTS: 10 µg/ml P. gingivalis LPS increased the expressions of TLR2 (3.79 ± 0.31), TLR4 (2.21 ± 0.31), and miRNA-146a (4.91 ± 0.87), NF-κ B p65 nuclear activity (6.51 ± 0.77 fold) (p < 0.05). 1 µg/ml P. gingivalis LPS induced TLR2 (3.05 ± 0.23), miRNA-146a (3.66 ± 0.83) and NF-κ B p65 nuclear activity (4.06 ± 0.78 fold) (p < 0.05), except TLR4 (1.11 ± 0.30, p > 0.05). Also, cytokines production increased (p < 0.05). The up-regulation of miRNA-146a could be blocked by anti-TLR2/4 mAb (p < 0.05). After the blockage of miRNA-146a, TLR2, TLR4, NF-κ B p65 nuclear activity and proinflammatory cytokines increased. However, after application of miRNA-146a mimic, the levels of these indexes decreased obviously (p < 0.05). CONCLUSION: MiRNA-146a functions as a negative feedback regulator via down-regulating proinflammatory cytokine secretion and blocking TLRs signaling pathway in hPDLCs after P. gingivalis LPS stimulation.


Asunto(s)
Retroalimentación Fisiológica/fisiología , Lipopolisacáridos/farmacología , MicroARNs/genética , Ligamento Periodontal/efectos de los fármacos , Porphyromonas gingivalis/química , Anticuerpos Monoclonales/farmacología , Células Cultivadas , Humanos , Lipopolisacáridos/química , MicroARNs/antagonistas & inhibidores , FN-kappa B/efectos de los fármacos , Ligamento Periodontal/patología , Estimulación Química , Receptor Toll-Like 2/genética , Receptor Toll-Like 4/genética , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/efectos de los fármacos
15.
Exp Mol Pathol ; 98(2): 136-44, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25661888

RESUMEN

The urokinase plasminogen activator receptor (uPAR) is closely associated with poor prognosis in various aggressive cancers including large-cell lung cancer (LCLC). Vasculogenic mimicry (VM) refers to the unique capability of aggressive tumor cells to mimic the pattern of embryonic vasculogenic networks involving the blood supply in early tumor formation. We demonstrate the statistically positive correlation of uPAR expression with VM formation, metastasis, and poor prognosis of LCLC patients. uPAR(+) cells sorted from the LCLC H460 cell line show higher invasion, migration capacity, and tube structure formation capability on Matrigel compared with uPAR(-) cells. uPAR(+) tumor cells highly expressed vimentin and VE-cadherin; the epithelial marker E-cadherin was low expressed. Higher EMT-regulated protein twist and snail expressions were also observed in these cells. uPAR(+) cells injected subcutaneously into nude mice markedly increased tumor growth, induced VM formation and liver metastasis; by contrast, uPAR(-) cells did not. The data suggest that uPAR expression may predict VM formation, tumor metastasis and poorer prognosis of LCLC patients. The uPAR gene may be used as a novel therapeutic target for inhibiting angiogenesis and metastasis in LCLC.


Asunto(s)
Carcinoma de Células Grandes/patología , Neoplasias Hepáticas/secundario , Neoplasias Pulmonares/patología , Neovascularización Patológica/patología , Receptores del Activador de Plasminógeno Tipo Uroquinasa/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Animales , Antígenos CD/biosíntesis , Cadherinas/biosíntesis , Carcinoma de Células Grandes/genética , Carcinoma de Células Grandes/mortalidad , Línea Celular Tumoral , Movimiento Celular , Transición Epitelial-Mesenquimal , Femenino , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/mortalidad , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Persona de Mediana Edad , Invasividad Neoplásica/patología , Trasplante de Neoplasias , Proteínas Nucleares/biosíntesis , Pronóstico , Factores de Transcripción de la Familia Snail , Factores de Transcripción/biosíntesis , Trasplante Heterólogo , Proteína 1 Relacionada con Twist/biosíntesis , Vimentina/biosíntesis
16.
J Oral Pathol Med ; 43(1): 28-34, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23710769

RESUMEN

BACKGROUND: Although molecular mechanism of growth differentiation factor 15 (GDF15) in tumorigenesis of oral squamous cell carcinoma (OSCC) is not clear, the diagnostic and prognostic value of serum GDF15 detection has been noticed. However, serum GDF15 levels in patients with oral leukoplakia and GDF15 as a potential predictive biomarker for response to induction chemotherapy in patients with OSCC have not been reported. METHODS: Pretreatment serum GDF15 concentration was detected using an enzyme-linked immunosorbent assay in 30 healthy persons, 24 patients with oral leukoplakia, and 60 patients with OSCC. RESULTS: Serum GDF15 concentration was significantly higher in patients with oral leukoplakia and OSCC, compared with healthy controls (F = 13.701, df = 2, P < 0.001). From a diagnostic standpoint, a cutoff value of 346.9 ng/l of serum GDF15 concentration was calculated using receiver operating characteristic curve, with a sensitivity of 0.750, specificity of 0.867, Youden's Index of 0.617, and area under curve of 0.863. From a prognostic standpoint, patients with serum GDF15 concentration <346.9 ng/l had an improved 3-year disease-free survival rate (64.3% vs 56.5%) compared with those above 346.9 ng/l, but the difference was not statistically significant. A decreased concentration of GDF15 (<346.9 ng/l) showed a predictive trend toward an improved response to induction chemotherapy compared with elevated concentration with clinical response rates of 100% and 71.4%, respectively, but the difference was not significant. CONCLUSION: Elevated GDF15 level may be not only a diagnostic biomarker for oral leukoplakia, but also a prognostic/predictive biomarker associated with decreased survival and diminished response to induction chemotherapy for patients with OSCC.


Asunto(s)
Carcinoma de Células Escamosas/sangre , Factor 15 de Diferenciación de Crecimiento/sangre , Leucoplasia Bucal/sangre , Neoplasias de la Boca/sangre , Adulto , Anciano , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Área Bajo la Curva , Biomarcadores de Tumor/sangre , Carcinoma de Células Escamosas/secundario , Carcinoma de Células Escamosas/cirugía , Estudios de Casos y Controles , Supervivencia sin Enfermedad , Femenino , Humanos , Quimioterapia de Inducción , Leucoplasia Bucal/cirugía , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/cirugía , Terapia Neoadyuvante , Clasificación del Tumor , Recurrencia Local de Neoplasia/patología , Estadificación de Neoplasias , Pronóstico , Curva ROC , Radioterapia Adyuvante , Inducción de Remisión , Estudios Retrospectivos , Sensibilidad y Especificidad , Resultado del Tratamiento
17.
AME Case Rep ; 8: 7, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38234361

RESUMEN

Background: Giant keratoacanthoma, a rare variant solitary of keratoacanthoma is characterized by a diameter exceeding 20 mm, rapid growth, and destruction of underlying tissue. Traditionally, it has been considered to be a self-resolving or low-grade squamous proliferation. Due to their atypical appearance, Giant keratoacanthomas may present a diagnostic challenge. Histopathological examination remains the reference standard for distinguishing squamous cell carcinoma (SCC) from keratoacanthoma. Surgical management is still the standard treatment for solitary keratoacanthoma. Case Description: We present the case of a 75-year-old man with a giant keratoacanthoma of the cheek, with a transverse length of 25 mm and a longitudinal length of 30 mm located 30 mm below the earlobe that was surgically excised with no obvious recurrence during follow-up. We used the Intraoperative frozen-section examination, which played a crucial role in ensuring clear margins and complete tumor removal. Conclusions: This case demonstrates that keratoacanthoma can be considered surgically curable and should be distinguished from SCC and other crateriform tumors. After surgical resection of giant isolated keratoacanthoma on the face, the high risk of cosmetic problems should be considered. We suggest free-flap transplantation be performed for functional repair, when the invasion range of the tumor is large and a perforation defect is formed.

18.
BMC Cancer ; 13: 301, 2013 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-23786757

RESUMEN

BACKGROUND: The benefit of induction chemotherapy in locally advanced oral squamous cell carcinoma (OSCC) remains to be clearly defined. Induction chemotherapy is likely to be effective for biologically distinct subgroups of patients and biomarker development might lead to identification of the patients whose tumors are to respond to a particular treatment. Annexin A1 may serve as a biomarker for responsiveness to induction chemotherapy. The aim of this study was to investigate Annexin A1 expression in pre-treatment biopsies from a cohort of OSCC patients treated with surgery and post-operative radiotherapy or docetaxel, cisplatin and 5-fluorouracil (TPF) induction chemotherapy followed by surgery and post-operative radiotherapy. Furthermore we sought to assess the utility of Annexin A1 as a prognostic or predictive biomarker. METHODS: Immunohistochemical staining for Annexin A1 was performed in pre-treatment biopsies from 232 of 256 clinical stage III/IVA OSCC patients. Annexin A1 index was estimated as the proportion of tumor cells (low and high, <50% and ≥50% of stained cells, respectively) to Annexin A1 cellular membrane and cytoplasm staining. RESULTS: There was a significant correlation between Annexin A1 expression and pathologic differentiation grade (P=0.015) in OSCC patients. The proportion of patients with low Annexin A1 expression was significantly higher amongst those with moderate/poorly differentiated tumor (78/167) compared to those with well differentiated tumor (18/65). Multivariate Cox model analysis showed clinical stage (P=0.001) and Annexin A1 expression (P=0.038) as independent prognostic risk factors. Furthermore, a low Annexin A1 expression level was predictive of longer disease-free survival (P=0.036, HR=0.620) and locoregional recurrence-free survival (P=0.031, HR=0.607) compared to high Annexin A1 expression. Patients with moderate/poorly differentiated tumor and low Annexin A1 expression benefited from TPF induction chemotherapy as measured by distant metastasis-free survival (P=0.048, HR=0.373) as well as overall survival (P=0.078, HR=0.410). CONCLUSIONS: Annexin A1 can be used as a prognostic biomarker for OSCC. Patients with moderate/poorly differentiated OSCC and low Annexin A1 expression can benefit from the addition of TPF induction chemotherapy to surgery and post-operative radiotherapy. Annexin A1 expression can potentially be used as a predictive biomarker to select OSCC patients with moderate/poorly differentiated tumor who may benefit from TPF induction chemotherapy.


Asunto(s)
Anexina A1/biosíntesis , Biomarcadores de Tumor/análisis , Carcinoma de Células Escamosas/metabolismo , Quimioterapia de Inducción/métodos , Neoplasias de la Boca/metabolismo , Anexina A1/análisis , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Carcinoma de Células Escamosas/tratamiento farmacológico , Carcinoma de Células Escamosas/patología , Diferenciación Celular , Terapia Combinada , Supervivencia sin Enfermedad , Femenino , Humanos , Inmunohistoquímica , Estimación de Kaplan-Meier , Masculino , Persona de Mediana Edad , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/patología , Clasificación del Tumor , Procedimientos Quirúrgicos Orales , Pronóstico , Modelos de Riesgos Proporcionales , Radioterapia
19.
Curr Med Imaging ; 2023 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-36876843

RESUMEN

The objectives study aims to assess the position and route of the alveolar antral artery and the lateral wall thickness of the maxillary sinus using cone-beam computed tomography (CBCT), reducing the risk of complications and improving the success rate of surgery. MATERIALS AND METHODS: This study included CBCT scans from 238 patients. The detection diameter of AAA and distance of the lower border of AAA to the maxillary sinus floor at the first premolar, second premolar, first molar, and second molar locations were evaluated. The route of AAA was observed with novel classification. Furthermore, the distance from the maxillary sinus floor to the alveolar crest at four posterior tooth locations was measured respectively. Moreover, the lateral wall thickness at four locations was assessed. Data were subjected to statistical analysis. RESULTS: AAA was observed in 62.18% of all sinuses. The mean diameter was 0.99±0.21 mm, with significant differences within gender. Half of the route of AAA was intrasinus intraosseous type. The mean distance between the maxillary sinus floor and AAA was 8.00±2.68 mm, with a significant difference between dentate and edentulous status at the first molar location. Distance from the sinus floor to the alveolar ridge crest in edentulous status negatively correlated with the distance from the sinus floor to AAA at the first molar location. The mean thickness of the lateral wall was 2.03±0.91 mm, and the difference in thickness between males and females at the four locations was statistically significant. CONCLUSION: intrasinus-intraosseous type, is the most common route. Special care should be taken at the first molar location during a lateral window sinus floor elevation. CBCT is highly recommended to before lateral wall maxillary sinus floor elevation.

20.
Int J Surg ; 109(8): 2220-2227, 2023 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-37288582

RESUMEN

BACKGROUND: Locally advanced oral squamous cell carcinoma (LAOSCC) is associated with a high rate of recurrence and poor survival. Given the recent successes of neoadjuvant immunochemotherapy (NAICT) in solid tumors, it is promising to use this treatment modality to achieve a better pathological response and improve the survival of LAOSCC, and clinical evidence is needed to assess its safety and efficacy. PATIENTS AND METHODS: A prospective trial of NAICT with toripalimab (PD-1 inhibitor) and albumin paclitaxel/cisplatin (TTP) was conducted in patients with clinical stage III and IVA OSCC. Intravenous albumin paclitaxel (260 mg/m 2 ), cisplatin (75 mg/m 2 ), and toripalimab (240 mg) were given in sequence on day 1 of each 21 day cycle for two cycles, followed by radical surgery and risk-adapted adjuvant (chemo)radiotherapy. The primary endpoints were safety and major pathological response (MPR). Targeted next generation sequencing and multiplex immunofluorescence were performed to assess clinical molecular characteristics and the tumor immune microenvironment in the pre-NAICT and post-NAICT tumor samples. RESULTS: Twenty patients were enrolled. NAICT was well-tolerated with a low incidence of grades 3-4 adverse events in three patients. The completion rates of NAICT and subsequent R0 resection were 100%. The MPR rate was 60%, including a 30% pathological complete response. MPR was achieved in all four patients with a combined positive score of PD-L1>10. The density of tertiary lymphatic structure in post-NAICT tumor samples predicted the pathological response to NAICT. During the median 23-month follow-up, the disease-free survival was 90%, and the overall survival was 95%. CONCLUSIONS: NAICT with the TTP protocol in LAOSCC is feasible and well tolerated, with a promising MPR and no obstruction on subsequent surgery. This trial is supportive of further randomized trials using NAICT in LAOSCC.


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/cirugía , Terapia Neoadyuvante/efectos adversos , Cisplatino , Carcinoma de Células Escamosas de Cabeza y Cuello/inducido químicamente , Carcinoma de Células Escamosas de Cabeza y Cuello/tratamiento farmacológico , Neoplasias de la Boca/tratamiento farmacológico , Neoplasias de la Boca/cirugía , Resultado del Tratamiento , Estudios Prospectivos , Protocolos de Quimioterapia Combinada Antineoplásica , Paclitaxel , Albúminas/uso terapéutico , Microambiente Tumoral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA