Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Mol Cell ; 54(6): 960-974, 2014 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-24857548

RESUMO

Many mutant p53 proteins (mutp53s) exert oncogenic gain-of-function (GOF) properties, but the mechanisms mediating these functions remain poorly defined. We show here that GOF mutp53s inhibit AMP-activated protein kinase (AMPK) signaling in head and neck cancer cells. Conversely, downregulation of GOF mutp53s enhances AMPK activation under energy stress, decreasing the activity of the anabolic factors acetyl-CoA carboxylase and ribosomal protein S6 and inhibiting aerobic glycolytic potential and invasive cell growth. Under conditions of energy stress, GOF mutp53s, but not wild-type p53, preferentially bind to the AMPKα subunit and inhibit AMPK activation. Given the importance of AMPK as an energy sensor and tumor suppressor that inhibits anabolic metabolism, our findings reveal that direct inhibition of AMPK activation is an important mechanism through which mutp53s can gain oncogenic function.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Carcinoma de Células Escamosas/genética , Metabolismo Energético/genética , Neoplasias de Cabeça e Pescoço/genética , Proteína Supressora de Tumor p53/metabolismo , Proteínas Quinases Ativadas por AMP/antagonistas & inibidores , Acetil-CoA Carboxilase/metabolismo , Animais , Antimetabólitos Antineoplásicos/farmacologia , Movimento Celular/genética , Proliferação de Células , Ativação Enzimática/genética , Fluoruracila/farmacologia , Humanos , Camundongos , Camundongos Nus , Invasividade Neoplásica/genética , Transplante de Neoplasias , Ligação Proteica/genética , Interferência de RNA , RNA Interferente Pequeno , Proteína S6 Ribossômica/metabolismo , Transdução de Sinais/genética , Esferoides Celulares/citologia , Carcinoma de Células Escamosas de Cabeça e Pescoço , Transplante Heterólogo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
2.
Cancer Prev Res (Phila) ; 14(8): 767-778, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-34021022

RESUMO

Although the principle of systemic treatment to prevent the progression of oral premalignant lesions (OPL) has been demonstrated, there remains a lack of consensus about an optimal approach that balances clinical efficacy with toxicity concerns. Recent advances in cancer therapy using approaches targeting the tumor immune microenvironment (TIME) including immune-checkpoint inhibitors indicate that these agents have significant clinically activity against different types of cancers, including oral cancer, and therefore they may provide an effective oral cancer prevention strategy for patients with OPLs. Our past work showed that systemic delivery of a monoclonal antibody to the programmed death receptor 1 (PD-1) immune checkpoint can inhibit the progression of OPLs to oral cancer in a syngeneic murine oral carcinogenesis model. Here we report a novel approach of local delivery of a PD-1 immune-checkpoint inhibitor loaded using a hydrogel, which significantly reduces the progression of OPLs to carcinomas. In addition, we detected a significant infiltration of regulatory T cells associated with oral lesions with p53 mutation, and a severe loss of expression of STING, which correlated with a decreased infiltration of dendritic cells in the oral lesions. However, a single local dose of PD-1 inhibitor was found to restore stimulator of interferon response cGAMP interactor 1 (STING) and CD11c expression and increase the infiltration of CD8+ T cells into the TIME irrespective of the p53 mutational status. Overall, we provide evidence for the potential clinical value of local delivery of biomaterials loaded with anti-PD-1 antibodies to prevent malignant progression of OPLs. PREVENTION RELEVANCE: Oral cancer is an aggressive disease, with an overall survival rate of 50%. Preinvasive histologic abnormalities such as tongue dysplasia represent an early stage of oral cancer; however, there are no treatments to prevent oral carcinoma progression. Here, we combined biomaterials loaded with an immunotherapeutic agent preventing oral cancer progression.


Assuntos
Inibidores de Checkpoint Imunológico/administração & dosagem , Neoplasias Bucais/prevenção & controle , Lesões Pré-Cancerosas/tratamento farmacológico , Receptor de Morte Celular Programada 1/imunologia , 4-Nitroquinolina-1-Óxido , Animais , Anticorpos Monoclonais/administração & dosagem , Carcinogênese/induzido quimicamente , Carcinogênese/efeitos dos fármacos , Modelos Animais de Doenças , Sistemas de Liberação de Medicamentos , Feminino , Genes p53/genética , Neoplasias de Cabeça e Pescoço/induzido quimicamente , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Neoplasias de Cabeça e Pescoço/prevenção & controle , Humanos , Injeções Intralesionais , Masculino , Camundongos , Camundongos Transgênicos , Neoplasias Bucais/patologia , Lesões Pré-Cancerosas/induzido quimicamente , Lesões Pré-Cancerosas/genética , Lesões Pré-Cancerosas/patologia , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Quinolonas , Carcinoma de Células Escamosas de Cabeça e Pescoço/induzido quimicamente , Carcinoma de Células Escamosas de Cabeça e Pescoço/genética , Carcinoma de Células Escamosas de Cabeça e Pescoço/patologia , Carcinoma de Células Escamosas de Cabeça e Pescoço/prevenção & controle
3.
Oral Oncol ; 97: 56-61, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31421472

RESUMO

OBJECTIVES: Sinonasal undifferentiated carcinoma (SNUC) is a rare, highly aggressive cancer. Despite aggressive multimodal therapy, its prognosis remains poor. Because of its locally advanced nature and high propensity for distant metastasis, we frequently use induction chemotherapy before definitive therapy in patients with SNUC. However, about 30% of patients do not respond to induction chemotherapy, and lack of response is associated with a poor survival rate. Therefore, in this study, we performed gene expression analysis of SNUC samples to identify prognostic markers for induction chemotherapy response. MATERIALS AND METHODS: Formalin-fixed, paraffin-embedded SNUC tumor samples from previously untreated patients harvested before induction chemotherapy were used. Gene expression was performed using an oncology gene expression panel. RESULTS: We identified 34 differentially expressed genes that distinguish the responders from the non-responders. Pathway analysis using these genes revealed alteration of multiple pathways between the two groups. Of these 34 genes, 24 distinguished between these two groups. Additionally, 16 gene pairs were associated with response to induction therapy. CONCLUSION: We identified genes predictive of SNUC response to induction chemotherapy and pathways potentially associated with treatment outcome. This is the first report of identification of predictive biomarkers for response of SNUC to induction chemotherapy, and it may help us develop therapeutic strategies to improve the treatment outcomes of non-responders.


Assuntos
Biomarcadores Tumorais/genética , Carcinoma/tratamento farmacológico , Carcinoma/genética , Neoplasias do Seio Maxilar/tratamento farmacológico , Neoplasias do Seio Maxilar/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma/patologia , Terapia Combinada/métodos , Feminino , Humanos , Quimioterapia de Indução/métodos , Masculino , Neoplasias do Seio Maxilar/patologia , Pessoa de Meia-Idade , Prognóstico , Resultado do Tratamento , Adulto Jovem
4.
Head Neck ; 41(8): 2688-2695, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-30932264

RESUMO

BACKGROUND: Sinonasal undifferentiated carcinoma (SNUC) is a rare, highly aggressive cancer. It is often difficult to determine whether SNUC is a distinct pathologic entity with poorly differentiated neuroendocrine features or it represents an undifferentiated tumor of squamous lineage. Also, reliable histopathologic markers that distinguish SNUC from poorly differentiated sinonasal squamous cell carcinoma (SNSCC) are lacking. Therefore, identification of new diagnostic molecular markers for SNUC is needed. METHODS: Treatment-naïve tumor specimens obtained from 15 SNUC and 6 SNSCC patients were used. Gene expression analysis was performed using an oncology panel. RESULTS: An unsupervised cluster analysis divided the patients into the one with only SNUCs and the one with mainly SNSCCs. Of 132 differentially expressed genes, 7 genes completely distinguished SNUCs from SNSCCs. SNUCs were enriched in sets of genes related to DNA repair, synthesis/replication, and cell division. CONCLUSIONS: Our study identified new diagnostic markers and potential therapeutic targets for SNUC.


Assuntos
Carcinoma de Células Escamosas/diagnóstico , Carcinoma/diagnóstico , Expressão Gênica , Neoplasias do Seio Maxilar/diagnóstico , Neoplasias dos Seios Paranasais/diagnóstico , Biomarcadores Tumorais/metabolismo , Carcinoma/genética , Carcinoma de Células Escamosas/genética , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Neoplasias do Seio Maxilar/genética , Neoplasias dos Seios Paranasais/genética , RNA Neoplásico/metabolismo , Estudos Retrospectivos
5.
Int J Oncol ; 33(1): 5-15, 2008 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-18575745

RESUMO

Focal necrosis is a key pathologic feature that distinguishes glioblastoma from lower grade glioma. The presence of necrosis in a glioblastoma could promote its rapid growth and clinical progression. Focal necrosis of glioblastoma seems to be associated with thrombosis that result from hyper-coagulability. In the present study, we found that glioblastoma cells had a high level of constitutive nuclear factor (NF)-kappaB activity, which was directly correlated with necrosis in glioblastomas. We also found a direct correlation between NF-kappaB activity and the expression of tissue factor (TF), a potent procoagulant factor in gliomas. Inhibition of TF by an inhibitory antibody prevented the procoagulant activity of glioblastoma cells, indicating a TF-dependent mechanism. Blockade of NF-kappaB activation significantly inhibited TF expression and the procoagulant activity of glioblastoma cells in vitro. Blockade of NF-kappaB activation also significantly inhibited in vivo expression of TF, which was directly correlated with decreased necrosis formation and tumor growth of glioblastoma cells in nude mice. Collectively, these results suggest that elevated NF-kappaB activity in glioblastomas cells plays a critical role in necrosis formation of glioblastoma and that inhibition of NF-kappaB activity in glioblastoma can suppress necrosis formation and progressive growth.


Assuntos
Glioblastoma/patologia , NF-kappa B/fisiologia , Tromboplastina/análise , Linhagem Celular Tumoral , Glioblastoma/química , Glioblastoma/metabolismo , Humanos , Proteínas I-kappa B/metabolismo , Imuno-Histoquímica , Interleucina-8/análise , Inibidor de NF-kappaB alfa , NF-kappa B/antagonistas & inibidores , Necrose , Transfecção , Fator A de Crescimento do Endotélio Vascular/análise
6.
Cancer Res ; 66(7): 3593-602, 2006 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-16585184

RESUMO

The transcription factor Forkhead box M1 (FoxM1) is overexpressed in malignant glioma. However, the functional importance of this factor in human glioma is not known. In the present study, we found that FoxM1B was the predominant FoxM1 isoform expressed in human glioma but not in normal brain tissue. The level of FoxM1 protein expression in human glioma tissues was directly correlated with the glioma grade. The level of FoxM1 protein expression in human glioblastoma tissues was inversely correlated with patient survival. Enforced FoxM1B expression caused SW1783 and Hs683 glioma cells, which do not form tumor xenografts, to regain tumorigenicity in nude mouse model systems. Moreover, gliomas that arose from FoxM1B-transfected anaplastic astrocytoma SW1783 cells displayed glioblastoma multiforme phenotypes. Inhibition of FoxM1 expression in glioblastoma U-87MG cells suppressed their anchorage-independent growth in vitro and tumorigenicity in vivo. Furthermore, we found that FoxM1 regulates the expression of Skp2 protein, which is known to promote degradation of the cell cycle regulator p27(Kip1). These results showed that FoxM1 is overexpressed in human glioblastomas and contributes to glioma tumorigenicity. Therefore, FoxM1 might be a new potential target of therapy for human malignant gliomas.


Assuntos
Astrocitoma/metabolismo , Astrocitoma/patologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Fatores de Transcrição Forkhead/biossíntese , Glioblastoma/metabolismo , Glioblastoma/patologia , Animais , Astrocitoma/genética , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Ciclina D1/metabolismo , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Proteína Forkhead Box M1 , Fatores de Transcrição Forkhead/antagonistas & inibidores , Fatores de Transcrição Forkhead/genética , Glioblastoma/genética , Humanos , Camundongos , Camundongos Nus , Isoformas de Proteínas , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Transplante Heterólogo
7.
Cancer Res ; 66(6): 3188-96, 2006 Mar 15.
Artigo em Inglês | MEDLINE | ID: mdl-16540670

RESUMO

Brain metastasis is a major cause of morbidity and mortality in patients with melanoma. The molecular changes that lead to brain metastasis remain poorly understood. In this study, we developed a model to study human melanoma brain metastasis and found that Stat3 activity was increased in human brain metastatic melanoma cells when compared with that in cutaneous melanoma cells. The expression of activated Stat3 is also increased in human brain metastasis specimens when compared with that in the primary melanoma specimens. Increased Stat3 activation by transfection with a constitutively activated Stat3 enhanced brain metastasis, whereas blockade of Stat3 activation by transfection with a dominant-negative Stat3 suppressed brain metastasis of human melanoma cells in animal models. Furthermore, altered Stat3 activity profoundly affected melanoma angiogenesis in vivo and melanoma cell invasion in vitro and significantly affected the expression of basic fibroblast growth factor (bFGF), vascular endothelial growth factor (VEGF), and matrix metalloproteinase-2 (MMP-2) in vivo and in vitro. Finally, Stat3 activity transcriptionally regulated the promoter activity of bFGF in addition to VEGF and MMP-2 in human melanoma cells. These results indicated that Stat3 activation plays an important role in dysregulated expression of bFGF, VEGF, and MMP-2 as well as angiogenesis and invasion of melanoma cells and contributes to brain metastasis of melanoma. Therefore, Stat3 activation might be a new potential target for therapy of human melanoma brain metastases.


Assuntos
Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/secundário , Melanoma/metabolismo , Melanoma/secundário , Fator de Transcrição STAT3/metabolismo , Animais , Neoplasias Encefálicas/irrigação sanguínea , Feminino , Fator 2 de Crescimento de Fibroblastos/metabolismo , Humanos , Metaloproteinase 2 da Matriz/metabolismo , Melanoma/irrigação sanguínea , Melanoma/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Invasividade Neoplásica , Neovascularização Patológica/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Neoplasias Cutâneas/metabolismo , Neoplasias Cutâneas/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
8.
Cancer Res ; 63(19): 6154-7, 2003 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-14559795

RESUMO

Exclusion of the alpha-exon by alternative RNA splicing of the fibroblast growth factor receptor 1 (FGFR1) primary transcript leads to the production of FGFR1beta. Glial cell transformation is associated with a progressive increase in FGFR1beta expression that coincides with a dramatic increase in the expression of the splicing factor polypyrimidine tract-binding protein (PTB). Cell-specific overexpression of PTB increased alpha-exon skipping, and a reduction in PTB increased alpha-exon inclusion. Targeted disruption of PTB interaction with FGFR1 precursor RNA in vivo by an antisense oligonucleotide also increased alpha-exon inclusion. These results suggest that PTB plays a direct role in alpha-exon splicing.


Assuntos
Processamento Alternativo/fisiologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/fisiologia , Receptores Proteína Tirosina Quinases/genética , Receptores de Fatores de Crescimento de Fibroblastos/genética , Processamento Alternativo/efeitos dos fármacos , Processamento Alternativo/genética , Animais , Linhagem Celular Tumoral , Regulação para Baixo , Éxons/efeitos dos fármacos , Éxons/genética , Humanos , Camundongos , Oligonucleotídeos Antissenso/genética , Oligonucleotídeos Antissenso/farmacologia , Proteína de Ligação a Regiões Ricas em Polipirimidinas/biossíntese , Proteína de Ligação a Regiões Ricas em Polipirimidinas/genética , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/genética , Precursores de RNA/efeitos dos fármacos , Precursores de RNA/genética , Receptores Proteína Tirosina Quinases/biossíntese , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos , Receptores de Fatores de Crescimento de Fibroblastos/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa
9.
Oncogene ; 23(20): 3550-60, 2004 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-15116091

RESUMO

The expression of matrix metalloproteinase-2 (MMP-2) has been linked with tumor invasion, angiogenesis, and metastasis. However, the molecular basis for MMP-2 overexpression in tumor cells remains unclear. In this study, by using K-1735 melanoma system, we demonstrated that highly metastatic C4, M2, and X21 tumor cells express elevated MMP-2 mRNA and enzymatic activity, whereas poorly metastatic C10, C19, and C23 tumor cells express much lower levels. Moreover, a concomitant elevated Stat3 activity has been detected in these metastatic tumor cells that overexpress MMP-2. Transfection of constitutively activated Stat3 into poorly metastatic C23 tumor cells directly activated the MMP-2 promoter, whereas the expression of a dominant-negative Stat3 in highly metastatic C4 tumor cells inhibited the MMP-2 promoter. A high-affinity Stat3-binding element was identified in the MMP-2 promoter and Stat3 protein bound directly to the MMP-2 promoter. Blockade of activated Stat3 through expression of a dominant-negative Stat3 significantly suppressed MMP-2 expression in the metastatic tumor cells. Therefore, overexpression of MMP-2 in the metastatic melanoma cells can be attributed to elevated Stat3 activity, and Stat3 upregulates the transcription of MMP-2 through direct interaction with the MMP-2 promoter. Furthermore, blockade of activated Stat3 in highly metastatic C4 cells significantly suppressed the invasiveness of the tumor cells, inhibited tumor growth, and prevented metastasis in nude mice. Collectively, these studies suggest that Stat3 signaling directly regulates MMP-2 expression, tumor invasion, and metastasis, and that Stat3 activation might be a crucial event in the development of metastasis.


Assuntos
Proteínas de Ligação a DNA/metabolismo , Metaloproteinase 2 da Matriz/metabolismo , Invasividade Neoplásica/genética , Metástase Neoplásica/genética , Transativadores/metabolismo , Animais , DNA/metabolismo , Metaloproteinase 2 da Matriz/genética , Melanoma/genética , Melanoma/metabolismo , Camundongos , Regiões Promotoras Genéticas , Ligação Proteica/fisiologia , Fator de Transcrição STAT3
10.
Clin Cancer Res ; 21(21): 4831-44, 2015 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-26124202

RESUMO

PURPOSE: Although the majority of patients with HPV(+) oropharyngeal cancers have a favorable prognosis, there are some patients with tumors that are resistant to aggressive chemoradiotherapy with unusual patterns of locoregional and systemic recurrences. Therefore, more effective therapies are needed. In this study, we investigated the chemosensitizing efficacy of the selective Wee-1 kinase inhibitor, AZD-1775, in HPV(+) head and neck squamous cell carcinoma (HNSCC). EXPERIMENTAL DESIGN: Clonogenic survival assays and an orthotopic mouse model of HPV(+) oral cancer were used to examine the in vitro and in vivo sensitivity of HPV(+) HNSCC cell lines to AZD-1775 in combination with cisplatin, respectively. Cell-cycle analysis, DNA damage (γH2AX), homologous recombination (HR), and apoptosis were examined to dissect molecular mechanisms. RESULTS: We found that AZD-1775 displays single-agent activity and enhances the response of HPV(+) HNSCC cells to cisplatin both in vitro and in vivo. The sensitivity of the HPV(+) HNSCC cells to AZD-1775 alone or in combination with cisplatin was associated with G2 checkpoint abrogation, persistent DNA damage, and apoptosis induction. This finding of AZD-1775 increasing the sensitivity of HPV(+) HNSCC cells to cisplatin through apoptosis was not seen previously in the HPV(-) HNSCC cancer cells and is accompanied by a decreased expression of the antiapoptotic proteins, MCl-1and XIAP, which appear to be cleaved following AZD-1775 treatment. CONCLUSIONS: AZD-1775 selectively sensitizes HPV(+) HNSCC cells and orthotopic oral xenografts to cisplatin through apoptosis and support the clinical investigation of AZD-1775 in combination with cisplatin particularly in patients with advanced and recurrent metastatic HPV(+) HNSCC tumors.


Assuntos
Apoptose/efeitos dos fármacos , Carcinoma de Células Escamosas/metabolismo , Proteínas de Ciclo Celular/antagonistas & inibidores , Neoplasias de Cabeça e Pescoço/metabolismo , Proteína de Sequência 1 de Leucemia de Células Mieloides/metabolismo , Proteínas Nucleares/antagonistas & inibidores , Infecções por Papillomavirus/complicações , Inibidores de Proteínas Quinases/farmacologia , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/metabolismo , Animais , Antineoplásicos/farmacologia , Carcinoma de Células Escamosas/etiologia , Caspases/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Cisplatino/farmacologia , Modelos Animais de Doenças , Resistencia a Medicamentos Antineoplásicos/genética , Sinergismo Farmacológico , Pontos de Checagem da Fase G2 do Ciclo Celular/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Genes p53 , Neoplasias de Cabeça e Pescoço/etiologia , Humanos , Concentração Inibidora 50 , Masculino , Camundongos , Proteína de Sequência 1 de Leucemia de Células Mieloides/genética , Infecções por Papillomavirus/virologia , Pirazóis/farmacologia , Pirimidinas/farmacologia , Pirimidinonas , Carcinoma de Células Escamosas de Cabeça e Pescoço , Carga Tumoral/efeitos dos fármacos , Proteínas Inibidoras de Apoptose Ligadas ao Cromossomo X/genética , Ensaios Antitumorais Modelo de Xenoenxerto
11.
Head Neck ; 36(8): 1094-102, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23836481

RESUMO

BACKGROUND: Little is known about the molecular signature of the rare tumor sinonasal squamous cell carcinoma (SNSCC). The purpose of this study was to comprehensively assess various molecular biomarkers in SNSCC. METHODS: We chose 13 markers for this study, which have been known as prognostic markers in head and neck squamous cell carcinoma. Expression of these markers was examined by either in situ hybridization or immunohistochemical methods on tissue microarrays made from 70 SNSCC specimens and 28 matched-pair normal tissues from patients who underwent surgical resection at our institution. Expression data were correlated with patient clinicopathologic parameters and survival. RESULTS: Of the 13 markers, only epidermal growth factor receptor (EGFR) protein expression was associated with significantly shorter disease-free survival (DFS; p = .01307). EGFR expression was also associated with shorter overall survival (OS), but the difference was not statistically significant. CONCLUSION: Targeted inhibition of tumor EGFR expression may be a new approach to treating SNSCC.


Assuntos
Biomarcadores Tumorais/metabolismo , Carcinoma de Células Escamosas/metabolismo , Neoplasias dos Seios Paranasais/metabolismo , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Carcinoma de Células Escamosas/mortalidade , Intervalo Livre de Doença , Receptores ErbB/metabolismo , Feminino , Humanos , Imuno-Histoquímica , Masculino , Pessoa de Meia-Idade , Neoplasias dos Seios Paranasais/mortalidade , Prognóstico , Estudos Retrospectivos , Análise de Sobrevida , Análise Serial de Tecidos , Adulto Jovem
12.
Laryngoscope ; 123(6): 1416-23, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23625637

RESUMO

OBJECTIVES/HYPOTHESIS: Mutation of the TP53 gene occurs in more than half of cases of head and neck squamous cell carcinoma (HNSCC). However, little is known about how specific TP53 mutations affect tumor progression. The objective of this study is to determine the gain of function of mutant p53 with a proline-to-serine substitution at codon 151. STUDY DESIGN: Laboratory-based study. METHODS: A panel of HNSCC cell lines was determined with anoikis assays, and orthotopic mouse experiments were performed. TP53 was sequenced. The shRNA knockdown and overexpression approaches were used for testing mutant p53 functions. The crystal structure of the p53 protein was analyzed using an in silico approach. RESULTS: An anoikis-resistant cell line, Tu138, was found to have a proline-to-serine substitution at codon 151 of TP53, which results in loss of wild-type p53 transcriptional activity. Moreover, the mutant p53 was shown to promote anoikis resistance and soft agar growth. Using an in silico approach based on the crystal structure of wild-type p53 protein, substitution of proline by serine at position 151 would create a cavity in a hydrophobic pocket, the loss of van der Waals contacts, and the thermodynamically unfavorable placement of a polar group, the hydroxyl oxygen atom of the serine, within a hydrophobic region, all of which likely cause a locally altered structure. CONCLUSIONS: Our data suggest that mutation at position 151 leads to a structural alteration, which results in significant functional changes in the p53 protein that impact tumor progression.


Assuntos
Anoikis/genética , Carcinoma de Células Escamosas/genética , DNA de Neoplasias/genética , Neoplasias de Cabeça e Pescoço/genética , Mutação , Prolina/genética , Serina/genética , Proteína Supressora de Tumor p53/genética , Animais , Western Blotting , Carcinoma de Células Escamosas/metabolismo , Carcinoma de Células Escamosas/patologia , Linhagem Celular Tumoral , Códon , Progressão da Doença , Neoplasias de Cabeça e Pescoço/metabolismo , Neoplasias de Cabeça e Pescoço/patologia , Camundongos , Camundongos Nus , Neoplasias Experimentais , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Carcinoma de Células Escamosas de Cabeça e Pescoço
13.
Cancer Discov ; 3(7): 770-81, 2013 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-23619168

RESUMO

The survival of patients with oral squamous cell carcinoma (OSCC) has not changed significantly in several decades, leading clinicians and investigators to search for promising molecular targets. To this end, we conducted comprehensive genomic analysis of gene expression, copy number, methylation, and point mutations in OSCC. Integrated analysis revealed more somatic events than previously reported, identifying four major driver pathways (mitogenic signaling, Notch, cell cycle, and TP53) and two additional key genes (FAT1, CASP8). The Notch pathway was defective in 66% of patients, and in follow-up studies of mechanism, functional NOTCH1 signaling inhibited proliferation of OSCC cell lines. Frequent mutation of caspase-8 (CASP8) defines a new molecular subtype of OSCC with few copy number changes. Although genomic alterations are dominated by loss of tumor suppressor genes, 80% of patients harbored at least one genomic alteration in a targetable gene, suggesting that novel approaches to treatment may be possible for this debilitating subset of head and neck cancers.


Assuntos
Carcinoma de Células Escamosas/genética , Variações do Número de Cópias de DNA/genética , Metilação de DNA/genética , Regulação Neoplásica da Expressão Gênica/genética , Neoplasias Bucais/genética , Carcinoma de Células Escamosas/patologia , Caspase 8/genética , Caspase 8/metabolismo , Linhagem Celular Tumoral , Genômica , Humanos , Neoplasias Bucais/patologia , Mutação Puntual/genética , Receptores Notch/genética , Receptores Notch/metabolismo
14.
Clin Cancer Res ; 18(22): 6178-87, 2012 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-23032744

RESUMO

PURPOSE: Sinonasal undifferentiated carcinoma (SNUC) is a rare and aggressive cancer. Despite the use of multimodality treatment, the overall prognosis remains poor. To better understand the biologic features of SNUC and help develop new therapies for the disease, we established SNUC cell lines and characterized their biologic behaviors. EXPERIMENTAL DESIGN: Cell lines were established from a patient with a T4N0M0 SNUC of the right maxillary sinus who was treated with surgical resection at our center. Tumor colonies were harvested and were sequentially replated onto larger plates. Two populations were developed and labeled MDA8788-6 and MDA8788-7. These cell lines were characterized with molecular, biomarker, functional, and histologic analyses. RESULTS: Short tandem repeat genotyping revealed that the cell line is isogenic to the parental tumor, and cytogenetic analysis identified 12 chromosomal translocations. The SNUC cell lines do not form colonies in soft agar but are tumorigenic and nonmetastatic in an orthotopic mouse model of sinonasal cancer. Western blot analysis revealed that both MDA8788 cell lines express epithelial markers but do not express mesenchymal markers or the endocrine marker synaptophysin. CONCLUSIONS: This is the first report of the establishment of stable human-derived SNUC cell lines. The lines were highly tumorigenic and maintain the histologic and molecular features of the original tumor. These cell lines should serve as useful tools for the future study of SNUC biology and the development and testing of novel therapies for this deadly disease.


Assuntos
Carcinoma/patologia , Linhagem Celular Tumoral/fisiologia , Neoplasias do Seio Maxilar/patologia , Idoso , Animais , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral/metabolismo , Proliferação de Células , Forma Celular , Transformação Celular Neoplásica/patologia , Bandeamento Cromossômico , Transição Epitelial-Mesenquimal , Feminino , Humanos , Cariótipo , Masculino , Camundongos , Camundongos Nus , Transplante de Neoplasias , Fenótipo
15.
PLoS One ; 6(4): e19412, 2011 Apr 29.
Artigo em Inglês | MEDLINE | ID: mdl-21559372

RESUMO

BACKGROUND: Squamous cell carcinoma of the head and neck (SCCHN) is the seventh most common cancer worldwide. Unfortunately, the survival of patients with SCCHN has not improved in the last 40 years, and thus new targets for therapy are needed. Recently, elevations in serum level of interleukin 6 (IL-6) and expression of Twist in tumor samples were found to be associated with poor clinical outcomes in multiple types of cancer, including SCCHN. Although Twist has been proposed as a master regulator of epithelial-mesenchymal transition and metastasis in cancers, the mechanisms by which Twist levels are regulated post-translationally are not completely understood. Tumor progression is characterized by the involvement of cytokines and growth factors and Twist induction has been connected with a number of these signaling pathways including IL-6. Since many of the effects of IL-6 are mediated through activation of protein phosphorylation cascades, this implies that Twist expression must be under a tight control at the post-translational level in order to respond in a timely manner to external stimuli. METHODOLOGY/PRINCIPAL FINDINGS: Our data show that IL-6 increases Twist expression via a transcription-independent mechanism in many SCCHN cell lines. Further investigation revealed that IL-6 stabilizes Twist in SCCHN cell lines through casein kinase 2 (CK2) phosphorylation of Twist residues S18 and S20, and that this phosphorylation inhibits degradation of Twist. Twist phosphorylation not only increases its stability but also enhances cell motility. Thus, post-translational modulation of Twist contributes to its tumor-promoting properties. CONCLUSIONS/SIGNIFICANCE: Our study shows Twist expression can be regulated at the post-translational level through phosphorylation by CK2, which increases Twist stability in response to IL-6 stimulation. Our findings not only provide novel mechanistic insights into post-translational regulation of Twist but also suggest that CK2 may be a viable therapeutic target in SCCHN.


Assuntos
Carcinoma de Células Escamosas/metabolismo , Caseína Quinase II/metabolismo , Ativação Enzimática , Regulação Neoplásica da Expressão Gênica , Neoplasias de Cabeça e Pescoço/metabolismo , Interleucina-6/metabolismo , Proteínas Nucleares/metabolismo , Proteína 1 Relacionada a Twist/metabolismo , Linhagem Celular Tumoral , Regulação Enzimológica da Expressão Gênica , Humanos , Metástase Neoplásica , Fosforilação , Processamento de Proteína Pós-Traducional , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Cicatrização
16.
Clin Cancer Res ; 17(21): 6658-70, 2011 Nov 01.
Artigo em Inglês | MEDLINE | ID: mdl-21903770

RESUMO

PURPOSE: To characterize tumor growth and metastatic potential in head and neck squamous cell carcinoma (HNSCC) cell lines in an orthotopic murine model of oral tongue cancer and to correlate TP53 mutation status with these findings. EXPERIMENTAL DESIGN: Cells from each of 48 HNSCC cell lines were orthotopically injected into the oral tongues of nude mice. Tumor volume, cervical lymph node metastasis, and mouse survival were recorded. Direct sequencing of the TP53 gene and Western blot analysis for the p53 protein after induction with 5-fluorouracil was conducted. Cell lines were categorized as either mutant TP53 or wild-type TP53, and lines with TP53 mutation were further categorized on the basis of type of mutation (disruptive or nondisruptive) and level of p53 protein expression. The behavior of tumors in these different groups was compared. RESULTS: These 48 HNSCC cell lines showed a wide range of behavior from highly aggressive and metastatic to no tumor formation. Mice injected with cells harboring disruptive TP53 mutations had faster tumor growth, greater incidence of cervical lymph node metastasis, and shorter survival than mice injected with cells lacking these mutations. CONCLUSIONS: HNSCC cell lines display a wide spectrum of behavior in an orthotopic model of oral cancer. Cell lines with disruptive TP53 mutations are more aggressive in this system, corroborating clinical reports that have linked these mutations to poor patient outcome.


Assuntos
Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/patologia , Genes p53 , Neoplasias de Cabeça e Pescoço/genética , Neoplasias de Cabeça e Pescoço/patologia , Mutação , Neoplasias da Língua/genética , Neoplasias da Língua/patologia , Proteína Supressora de Tumor p53/genética , Animais , Linhagem Celular Tumoral , Modelos Animais de Doenças , Humanos , Masculino , Camundongos , Camundongos Nus , Carcinoma de Células Escamosas de Cabeça e Pescoço , Transplante Heterólogo
17.
Science ; 333(6046): 1154-7, 2011 Aug 26.
Artigo em Inglês | MEDLINE | ID: mdl-21798897

RESUMO

Head and neck squamous cell carcinoma (HNSCC) is the sixth most common cancer worldwide. To explore the genetic origins of this cancer, we used whole-exome sequencing and gene copy number analyses to study 32 primary tumors. Tumors from patients with a history of tobacco use had more mutations than did tumors from patients who did not use tobacco, and tumors that were negative for human papillomavirus (HPV) had more mutations than did HPV-positive tumors. Six of the genes that were mutated in multiple tumors were assessed in up to 88 additional HNSCCs. In addition to previously described mutations in TP53, CDKN2A, PIK3CA, and HRAS, we identified mutations in FBXW7 and NOTCH1. Nearly 40% of the 28 mutations identified in NOTCH1 were predicted to truncate the gene product, suggesting that NOTCH1 may function as a tumor suppressor gene rather than an oncogene in this tumor type.


Assuntos
Carcinoma/genética , Proteínas de Ciclo Celular/genética , Proteínas F-Box/genética , Genes Supressores de Tumor , Neoplasias de Cabeça e Pescoço/genética , Mutação , Neoplasias de Células Escamosas/genética , Receptor Notch1/genética , Ubiquitina-Proteína Ligases/genética , Carcinoma/tratamento farmacológico , Carcinoma/virologia , Carcinoma de Células Escamosas , Códon sem Sentido , Éxons , Proteína 7 com Repetições F-Box-WD , Dosagem de Genes , Genes p53 , Neoplasias de Cabeça e Pescoço/tratamento farmacológico , Neoplasias de Cabeça e Pescoço/virologia , Humanos , Mutação INDEL , Mutação de Sentido Incorreto , Neoplasias de Células Escamosas/tratamento farmacológico , Neoplasias de Células Escamosas/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Oncogenes , Papillomaviridae/isolamento & purificação , Infecções por Papillomavirus/virologia , Receptor Notch1/química , Análise de Sequência de DNA , Fumar , Carcinoma de Células Escamosas de Cabeça e Pescoço , Nicotiana
18.
Oncol Rep ; 23(3): 725-32, 2010 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-20127012

RESUMO

Angiogenesis is a key pathologic feature of glioblastoma, which is the most common and most lethal primary brain tumor in adults. The degree of angiogenesis has been shown to be inversely related to patient survival. However, the molecular changes leading to angiogenesis in glioblastoma remain poorly understood. In the present study, we found a direct correlation between nuclear factor (NF)-kappaB activation and angiogenesis in glioblastomas. Blockade of NF-kappaB signaling significantly inhibited glioblastoma growth and angiogenesis in nude mice. These effects were consistent with significant inhibition of the expression of multiple angiogenic molecules, including vascular endothelial growth factor, and interleukin-8, in vitro and in vivo. Furthermore, blockade of NF-kappaB signaling also significantly inhibited the angiogenic potential of glioblastoma cells in vitro and angiogenesis of brain tumors in mouse xenograft models. Collectively, these results suggest that NF-kappaB activation plays a critical role in the growth and progression of glioblastoma and is a potential target for therapy for human glioblastoma.


Assuntos
Regulação Neoplásica da Expressão Gênica , Glioblastoma/irrigação sanguínea , Interleucina-8/genética , NF-kappa B/fisiologia , Neovascularização Patológica/etiologia , Fator A de Crescimento do Endotélio Vascular/genética , Animais , Linhagem Celular Tumoral , Feminino , Humanos , Interleucina-8/análise , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/antagonistas & inibidores , Neovascularização Patológica/prevenção & controle , RNA Mensageiro/análise , Fator A de Crescimento do Endotélio Vascular/análise
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA