Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
Mais filtros

Base de dados
Tipo de documento
País de afiliação
Intervalo de ano de publicação
1.
J Surg Res ; 254: 183-190, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32450419

RESUMO

BACKGROUND: Hyperplastic polyposis protein 1 (HPP1) encodes a tumor-suppressive transmembrane cleavable epidermal growth factor-like ligand. It is unclear as to whether cleavage and shedding of HPP1 are essential steps in achieving its tumor suppressive properties. ADAM proteins are key players in cellular ectodomain shedding processes with ADAM17 being well characterized and representing the most likely sheddase for HPP1. In this study, we explore the mechanisms and importance of ectodomain shedding in contributing to HPP1-mediated tumor suppression. METHODS: Baseline characterization of HPP1 ectodomain shedding and ADAM family member expression was performed in HCT116 colon cancer cells with forced overexpression of HPP1 and controls. Subsequent impact of attenuation of ADAM expression by short interfering RNA on HPP1 shedding was evaluated. Furthermore, we examined the functional impact of an uncleavable HPP1 mutant construct (HPP1-Δstalk) generated by site-directed mutagenesis. Cellular growth potential functions were analyzed by MTT and soft agar assays. RESULTS: Select proinflammatory cytokines enhanced HPP1 ectodomain shedding, whereas short interfering RNA-mediated knockdown of ADAM17 resulted in abrogation of HPP1 ectodomain shedding. ADAM17 knockdown concomitantly resulted in increased cell proliferation and anchorage-independent growth. HPP1-Δstalk-transfected cells exhibited significantly higher proliferation and reduced STAT1 activation relative to full-length HPP1, further suggesting a critical role for ectodomain shedding in HPP1-mediated tumor suppression. CONCLUSION: The tumor-suppressive properties of HPP1 in colorectal cancer require cleavage and shedding of its ectodomain which in turn are mediated by ADAM17. Further investigations into the regulation of HPP1 may lead to a greater understanding of epidermal growth factor-like ligand family biology and potential novel therapeutic strategies.


Assuntos
Proteína ADAM17/metabolismo , Neoplasias do Colo/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Células HCT116 , Humanos , Fatores de Transcrição STAT/metabolismo
2.
Dis Colon Rectum ; 57(8): 941-57, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-25003289

RESUMO

BACKGROUND: The Radiation Therapy Oncology Group 98-11 clinical trial demonstrated the superiority of standard 5-fluorouracil/mitomycin-C over 5-fluorouracil/cisplatin in combination with radiation in the treatment of anal squamous cell cancer. Tumor size (>5 cm) and lymph node metastases are associated with disease progression. There may be key molecular differences (eg, DNA methylation changes) in tumors at high risk for progression. OBJECTIVE: The objectives of this study were to determine whether there are differences in DNA methylation at individual CpG sites and within genes among locally advanced anal cancers, with large tumor size and/or nodal involvement, compared with those that are less advanced. DESIGN: This was a case-case study among 121 patients defined as high risk (tumor size >5 cm and/or nodal involvement; n = 59) or low risk (≤5 cm, node negative; n = 62) within the mitomycin-C arm of the Radiation Therapy Oncology Group 98-11 trial. DNA methylation was measured using the Illumina HumanMethylation450 Array. SETTINGS: The study was conducted in a tertiary care cancer center in collaboration with a national clinical trials cooperative group. PATIENTS: The patients consisted of 74 women and 47 men with a median age of 54 years (range, 25-79 years). MAIN OUTCOME MEASURES: DNA methylation differences at individual CpG sites and within genes between low- and high-risk patients were compared using the Mann-Whitney test (p < 0.001). RESULTS: A total of 16 CpG loci were differentially methylated (14 increased and 2 decreased) in high- versus low-risk cases. Genes harboring differentially methylated CpG sites included known tumor suppressor genes and novel targets. LIMITATIONS: This study only included patients in the mitomycin-C arm with tumor tissue; however, this sample was representative of the trial. CONCLUSIONS: This is the first study to apply genome-wide methylation analysis to anal cancer. Biologically relevant differences in methylated targets were found to discriminate locally advanced from early anal cancer. Epigenetic events likely play a significant role in the progression of anal cancer and may serve as potential biomarkers.


Assuntos
Neoplasias do Ânus/genética , Neoplasias do Ânus/radioterapia , Carcinoma de Células Escamosas/genética , Carcinoma de Células Escamosas/radioterapia , Epigenômica , Adulto , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias do Ânus/tratamento farmacológico , Carcinoma de Células Escamosas/tratamento farmacológico , Cisplatino/administração & dosagem , Terapia Combinada , Metilação de DNA , Progressão da Doença , Feminino , Fluoruracila/administração & dosagem , Humanos , Metástase Linfática , Masculino , Pessoa de Meia-Idade , Mitomicina/administração & dosagem
3.
Adv Biol Regul ; 88: 100964, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-37004354

RESUMO

Small cell lung cancer (SCLC) often exhibits Rb deficiency, TRß and p130 deletion, and SKP2 amplification, suggesting TRß inactivation and SKP2 activation. It is reported that SKP2 targeted therapy is effective in some cancers in vitro and in vivo, but it is not reported for the treatment of SCLC and retinoblastoma. SKP2 is the synthetic lethal gene in SCLC and retinoblastoma, so SKP2 can be used for targeted therapy in SCLC and retinoblastoma. RB1 knockout mice develop several kinds of tumors, but Rb1 and SKP2 double knockout mice are healthy, suggesting that SKP2 targeted therapy may have significant effects on Rb deficient cancers with less side effects, and if successful in SCLC and retinoblastoma in vitro and in animal model, such compounds may be promising for the clinical treatment of SCLC, retinoblastoma, and variety of Rb deficient cancers. Previously our studies showed that retinoblastomas exhibit retinal cone precursor properties and depend on cone-specific thyroid hormone receptor ß2 (TRß2) and SKP2 signaling. In this study, we sought to suppress SCLC and retinoblastoma cell growth by SKP2 inhibitors as a prelude to targeted therapy in vitro and in vivo. We knocked down TRß2 and SKP2 or over-expressed p27 in SCLC and retinoblastoma cell lines to investigate SKP2 and p27 signaling alterations. The SCLC cell lines H209 as well as retinoblastoma cell lines Y79, WERI, and RB177 were treated with SKP2 inhibitor C1 at different concentrations, following which Western blotting, Immunostaining, and cell cycle kinetics studies were performed to study SKP2 and p27 expression ubiquitination, to determine impact on cell cycle regulation and growth inhibition. TRß2 knockdown in Y79, RB177 and H209 caused SKP2 downregulation and degradation, p27 up-regulation, and S phase arrest, whereas, SKP2 knockdown or p27 over-expression caused p27 accumulation and G1-S phase arrest. In the cell lines Y79, WERI, RB177, and H209 treatment with C1 caused SKP2 ubiquitination and degradation, p27 de-ubiquitination and accumulation, and cell growth arrest. SKP2 inhibitor C1 significantly suppressed retinoblastoma as well as SCLC cell growth by SKP2 degradation and p27 accumulation. In vivo study also showed inhibition of tumor growth with C1 treatment. Potential limitations of the success of such a therapeutic approach and its translational application in human primary tumors, and alternative approaches to overcome such limitations are briefly discussed for the treatment of retinoblastoma, SCLC and other RB-related cancers.


Assuntos
Neoplasias da Retina , Retinoblastoma , Camundongos , Animais , Humanos , Retinoblastoma/tratamento farmacológico , Retinoblastoma/genética , Retinoblastoma/metabolismo , Proteínas Quinases Associadas a Fase S/genética , Proteínas Quinases Associadas a Fase S/metabolismo , Proteína do Retinoblastoma/genética , Proteína do Retinoblastoma/metabolismo , Linhagem Celular Tumoral , Ciclo Celular , Camundongos Knockout , Pulmão/patologia
4.
Cell Oncol (Dordr) ; 44(3): 673-687, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33694102

RESUMO

PURPOSE: The transforming growth factor-beta (TGF-ß) pathway plays a paradoxical, context-dependent role in pancreatic ductal adenocarcinoma (PDAC): a tumor-suppressive role in non-metastatic PDAC and a tumor-promotive role in metastatic PDAC. We hypothesize that non-SMAD-TGF-ß signaling induces PDAC progression. METHODS: We investigated the expression of non-SMAD-TGF-ß signaling proteins (pMAPK14, PD-L1, pAkt and c-Myc) in patient-derived tissues, cell lines and an immunocompetent mouse model. Experimental models were complemented by comparing the signaling proteins in PDAC specimens from patients with various survival intervals. We manipulated models with TGF-ß, gemcitabine (DNA synthesis inhibitor), galunisertib (TGF-ß receptor inhibitor) and MK-2206 (Akt inhibitor) to investigate their effects on NF-κB, ß-catenin, c-Myc and PD-L1 expression. PD-L1 expression was also investigated in cancer cells and tumor associated macrophages (TAMs) in a mouse model. RESULTS: We found that tumors from patients with aggressive PDAC had higher levels of the non-SMAD-TGF-ß signaling proteins pMAPK14, PD-L1, pAkt and c-Myc. In PDAC cells with high baseline ß-catenin expression, TGF-ß increased ß-catenin expression while gemcitabine increased PD-L1 expression. Gemcitabine plus galunisertib decreased c-Myc and NF-κB expression, but induced PD-L1 expression in some cancer models. In mice, gemcitabine plus galunisertib treatment decreased metastases (p = 0.018), whereas galunisertib increased PD-L1 expression (p < 0.0001). In the mice, liver metastases contained more TAMs compared to the primary pancreatic tumors (p = 0.001), and TGF-ß increased TAM PD-L1 expression (p < 0.05). CONCLUSIONS: In PDAC, the non-SMAD-TGF-ß signaling pathway leads to more aggressive phenotypes, TAM-induced immunosuppression and PD-L1 expression. The divergent effects of TGF-ß ligand versus receptor inhibition in tumor cells versus TAMs may explain the TGF-ß paradox. Further evaluation of each mechanism is expected to lead to the development of targeted therapies.


Assuntos
Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/patologia , Neoplasias Pancreáticas/patologia , Fator de Crescimento Transformador beta/metabolismo , Macrófagos Associados a Tumor/metabolismo , Animais , Progressão da Doença , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/fisiologia
5.
Adv Biol Regul ; 78: 100723, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32992231

RESUMO

Cancer as a genetic disease is by now well recognized. Genomic analysis of cancer cells, therefore, has greatly enhanced our ability to identify genetic alterations associated with various cancer types, including both lympho-hematopoietic as well as solid tumors. Chronic myeloid leukemia (CML), based on the specific diagnostic genetic abnormality has served as a prototype disease to clearly demonstrate the significance of the genomic analysis of cancer in identifying targeted therapy. Such a success has provided extra ordinary opportunities to investigate the role of genetic abnormalities and the pathways amenable to targeted therapy, not only in blood cancers but solid tumors such as Lung, Brain, Colon, Renal, Breast cancers as well as other epithelial and mesenchymal tumors. The main focus of this presentation is to illustrate the role of genomic analysis in targeting lung cancer, based on abnormalities or the pathways deregulated in tumor cells from individual patients. Lung cancer is one of the most common epithelial cancers associated with chronic inflammation due to cigarette smoking and other environmental carcinogens, and includes four distinct histologic type; non-small cell lung cancer (NSCLC); small cell lung cancer (SCLC) and squamous cell lung cancer. According to current estimates, 1.3 million cases of lung cancer are expected to be diagnosed worldwide annually, resulting in one million deaths. Since the discovery that patient's tumors with specific mutations in the EGFR may be sensitive to targeted therapeutic approach and the subsequent realization that the such mutations in the gene are not as prevalent, several cancer centers including ours initiated intense efforts to find other mutations or genomic alterations, which may serve as targets of specific therapy. Such efforts have successfully resulted in a battery of genes such as KRAS, ALK, C-MET, HER-2/neu, ROS1, etc., which have helped oncologists to triage the patients for personalized therapies. A significant proportion of patients with lung cancer, however, do not show any of the above genetic abnormalities. Approximately 90% of lung cancers exhibit RB1 mutation/deletion and or KRAS mutations, therefore, the signaling pathways, which regulate multistep tumorigenesis in lung cancer, are important for the treatment of histologic subtypes of lung cancer, which includes NSCLC & SCLC. Equally important was the findings that similar signaling pathways are also shared by other solid tumor types. We have investigated the role of these pathways to target these cancers and develop new strategies to treat lung, brain and related cancers. In addition, our translational studies in other tumor types such as NF2 related malignancies, specifically, Malignant Mesothelioma (MM), in which NF2 related pathway amenable to targeted therapies was identified. Selected examples representing experimental approaches will be discussed to illustrate the critical role of translational research in developing novel therapeutics for the successful and durable responses in some of these cancer types.


Assuntos
Genômica , Neoplasias Pulmonares/genética , Mesotelioma Maligno/genética , Genes ras , Humanos , Mutação , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Transdução de Sinais
6.
Anticancer Res ; 29(4): 1255-62, 2009 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19414372

RESUMO

Malignant peripheral nerve sheath tumors (MPNSTs) develop in patients with underlying NF1, and usually arise as a result of malignant transformation of a pre-existing plexiform neurofibroma. The clonal cytogenetic abnormalities reported in primary MPNST include complex karyotypes with chromosome numbers in the triploid or tetraploid range with recurrent abnormalities of several chromosomes including losses or imbalances. As a prelude to cell biological, pharmacological, and functional studies to investigate pathways and gene(s) associated with multistep tumorigenesis, which includes progression, metastasis and resistance to therapy in MPNST, detailed molecular cytogenetic and genetic analyses of cell lines from primary, metastatic and recurrent MPNST with underlying NF1 disorder have been performed. The clonal cytogenetic abnormalities detected in the primary tumor cell line were similar to those observed in primary cultures of this tumor. Due to the complexity of the rearrangements seen by G-banded karyotype analysis, further characterization of the clonal abnormalities in these three cell lines was performed by molecular cytogenetic techniques, including CGH and SKY. CGH analysis detected recurrent deletions of 9p, 12q21-q32, complete losses of the X-chromosome, and gains of the chromosomal segment 17q25 in all three cell lines. SKY analysis detected extensive clonal abnormalities in these cell lines. The nature and the alterations of the cell cycle regulators, particularly those associated with G1-S checkpoints and known to be deregulated in MPNST, were studied. These cell cycle regulators included those associated with Rb1-cyclin D1 and the p53 pathways. The findings are consistent with the argument that an imbalance between the cyclin activators of CDKs and inhibitory proteins such as p16 result in uncontrollable proliferation in the cell lines, associated with progression of the disease. LOH and expression of the p53 gene in metastatic and recurrent cell lines was observed, as reported by others. The role of biallelic inactivation of p53 gene in MPNST with underlying NF1 mutations, however, needs further study. Overexpression of Rb1-protein observed in metastatic and recurrent cell lines is indicative of its role in the progression of the disease. One of the most important observations of this study is that Nm23-H1 expression is closely associated with advanced or metastatic disease. In summary, MPNST cell lines derived from a patient with metastatic and recurrent disease with NF1 disorder were characterized and a gene associated with metastatic potential which is amenable to therapeutic and chemo-preventative approaches was identified. These cell lines with extensive characterization of genetic abnormalities are likely to provide important reagents for biochemical, molecular and pharmacological studies related to MPNST.


Assuntos
Cromossomos Humanos/genética , Recidiva Local de Neoplasia/genética , Neoplasias de Bainha Neural/genética , Neurofibromatose 1/genética , Adolescente , Northern Blotting , Proliferação de Células , Aberrações Cromossômicas , Deleção Cromossômica , Hibridização Genômica Comparativa , Fibroblastos , Genes da Neurofibromatose 1 , Humanos , Técnicas Imunoenzimáticas , Hibridização in Situ Fluorescente , Cariotipagem , Perda de Heterozigosidade , Recidiva Local de Neoplasia/patologia , Neoplasias de Bainha Neural/patologia , Neurofibromatose 1/patologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Células Tumorais Cultivadas , Proteína Supressora de Tumor p53/genética
7.
Int J Cancer ; 122(7): 1567-72, 2008 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-18059030

RESUMO

HPP1 is a recently discovered gene that is epigenetically silenced in a number of tumor types, suggesting a potential role as a tumor suppressor. However, whether HPP1 has tumor suppressor activity is not clearly known. We have sought to investigate the effects of HPP1 on tumor growth and survival and to identify signaling pathways that mediate HPP1's mechanism of action. Forced expression of HPP1 into HCT116 colon cancer cell lines blocked the ability of HCT116 tumors to grown in vivo in nude mice. In cell culture, ectopic expression of HPP1 induces apoptosis and potently inhibits soft agar colony formation. HPP1 overexpression was also associated with a moderate reduction in in vitro proliferation characterized by an accumulation of cells in the G0/G1 phase of the cell cycle. Microarray analysis revealed that ectopic expression of HPP1 resulted in a dramatic upregulation of STAT1 as well as a large number of associated interferon-inducible genes. RNA interference-mediated abrogation of STAT1 reversed HPP1's antiproliferative effects. We conclude that HPP1 demonstrates tumor suppressive and pro-apoptotic activity, both in vitro and in vivo. Coupled with its inactivation in a number of tumor types, our data provides evidence to support the role of HPP1 as a tumor suppressor gene. Moreover, activation of the STAT1 pathway likely represents the principal mediator of HPP1's tumor suppressive properties.


Assuntos
Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Genes Supressores de Tumor , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Fator de Transcrição STAT1/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Animais , Apoptose , Proliferação de Células , Regulação para Baixo , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Humanos , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/metabolismo , Análise de Sequência com Séries de Oligonucleotídeos , Interferência de RNA , Transdução de Sinais , Regulação para Cima
8.
Sci Rep ; 8(1): 5808, 2018 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-29643359

RESUMO

The precise role of tumor associated macrophages remains unclear in pancreatic ductal adenocarcinoma (PDAC) while TGF-ß has an unclear role in metastases formation. In order to understand the role of IL23, an interleukin associated with macrophage polarization, we investigated IL23 in the context of TGF-ß expression in PDAC. We hypothesized that IL23 expression is associated with metastatic development and survival in PDAC. We investigated IL23 and TGF-ß protein expression on resected PDAC patient tumor sections who were divided into short-term (<12 months) survivors and long-term (>30 months) survivors. Panc-1 cells treated with IL23, TGF-ß, macrophages, or combinations thereof, were orthotopically implanted into NSG mice. Patients in the long-term survivor group had higher IL23 protein expression (P = 0.01). IL23 expression was linearly correlated with TGF-ß expression in patients in the short-term survivor group (P = 0.038). Macrophages induce a higher rate of PDAC metastasis in the mouse model (P = 0.02), which is abrogated by IL23 and TGF-ß treatment (P < 0.001). Macrophages serve a critical role in PDAC tumor growth and metastasis. TGF-ß contributes to a less tumorigenic TME through regulation of macrophages. Macrophages increases PDAC primary tumor growth and metastases formation while combined IL23 and TGF-ß pre-treatment diminishes these processes.


Assuntos
Carcinoma Ductal Pancreático/patologia , Subunidade p19 da Interleucina-23/análise , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Metástase Neoplásica/prevenção & controle , Fator de Crescimento Transformador beta/análise , Idoso , Animais , Modelos Animais de Doenças , Humanos , Imuno-Histoquímica , Camundongos , Pessoa de Meia-Idade , Análise de Sobrevida
9.
DNA Cell Biol ; 34(8): 541-9, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26061044

RESUMO

HPP1, a novel tumor suppressive epidermal growth factor (EGF)-like ligand, mediates its effects through signal transducer and activators of transcription (STAT) activation. We previously demonstrated the importance of STAT1 activation for HPP1 function; however the contribution of STAT2 remains unclear. We sought to delineate the components of JAK-STAT-interferon (IFN) signaling specifically associated with HPP1s biological effects. Using stable HPP1-HCT116 transfectants, expression analyses were performed by polymerase chain reaction (PCR)/western blotting while expression knockdowns were achieved using siRNA. Growth parameters evaluated included proliferation, cell cycle distribution, and anchorage-independent growth. STAT dimerization, translocation, and DNA binding were examined by reporter assays, fluorescent microscopy, and chromatin immunoprecipitation (ChIP), respectively. Forced expression of HPP1 in colon cancer cell lines results in the upregulation of total and activated levels of STAT2. We have also determined that JAK1 and JAK2 are activated in response to HPP1 overexpression, and are necessary for subsequent STAT activation. Overexpression of HPP1 was associated with significant increases in STAT1:STAT1 (p=0.007) and STAT1:STAT2 (p=0.036) dimer formation, as well as subsequent nuclear translocation. By ChIP, binding of activated STAT1 and STAT2 to the interferon-signaling regulatory element promoter sites of the selected genes, protein kinase RNA-activated (PKR), IFI44, and OAS1 was demonstrated. STAT2 knockdown resulted in partial abrogation of HPP1s growth suppressive activity with increased proliferation (p<0.0001), reduced G1/G0 phase cell cycle fraction, and a restoration of growth potential in soft agar (p<0.01). Presumably as a consequence of upregulation of IFN signaling elements, HPP1 overexpression resulted in an acquisition of exogenous IFN sensitivity. Physiologic doses of IFN-α resulted in a significant reduction in proliferation (p<0.001) and increase in G1/G0 cell cycle arrest in HPP1 transfectants. STAT2 is necessary for HPP1-associated growth suppression, and mediates these effects through activation of IFN-α pathways. Given the interest in therapeutic targeting of oncogenic erbB proteins, further understanding of HPP1s role as a tumor suppressive EGF-like ligand is warranted.


Assuntos
Genes Supressores de Tumor , Interferons/fisiologia , Janus Quinases/fisiologia , Proteínas de Membrana/fisiologia , Proteínas de Neoplasias/fisiologia , Fatores de Transcrição STAT/fisiologia , Transporte Ativo do Núcleo Celular , Núcleo Celular/metabolismo , Células HCT116 , Humanos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Multimerização Proteica , Transdução de Sinais/genética , Ativação Transcricional , Transfecção
10.
Pharmacogenetics ; 14(2): 103-9, 2004 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-15077011

RESUMO

OBJECTIVE: The human 8-oxoguanine DNA N-glycosylase 1 gene encodes a DNA glycosylase that is involved in the base excision repair of 8-hydroxy-2-deoxyguanine from oxidatively-damaged DNA and expressed in lung tissue. The codon 326 polymorphism in the hOGG1 gene has been suggested to reduce DNA repair enzyme activity based on in vitro functional analysis. The goal of the present study is to determine whether the codon 326 polymorphism was significantly associated with alterations in individual risk for lung cancer. METHODS: To determine whether hOGG1 plays a role in risk for lung cancer, we measured the prevalence of the Ser326Cys polymorphism in incident lung cancer patients and matched non-cancer controls. hOGG1 genotyping was performed by PCR-restriction fragment length polymorphism analysis of genomic DNA isolated from 179 Caucasian lung cancer cases and 358 controls individually matched in a 1:2 ratio by race-, sex- and age (+/- 5 years). RESULTS: Significantly increased risk for lung cancer was observed for both the hOGG1 326 (odds ratio [OR] = 1.9, 95% confidence interval [CI] = 1.2-2.9) and hOGG1 326 genotypes (OR = 3.8, 95% CI = 1.4-10.6). The increased risk for lung cancer was observed for subjects with both the hOGG1 326 (OR = 1.7, 95% CI = 1.1-2.8) and hOGG1 326 genotypes (OR = 4.9, 95% CI = 1.5-16.1) in ever-smokers. A significant association was found between hOGG1 genotypes and lung cancer risk with a dose-dependent effect with smoking. Significantly increased risk for variant hOGG1 genotypes was observed for all non-small cell lung cancer patients. CONCLUSION: These results suggest that the hOGG1 Ser326Cys polymorphism plays an important role in the risk for lung cancer and is linked to exposure to tobacco smoke.


Assuntos
DNA Glicosilases/genética , Reparo do DNA , Neoplasias Pulmonares/genética , Idoso , Estudos de Casos e Controles , DNA/análise , Feminino , Genótipo , Humanos , Neoplasias Pulmonares/enzimologia , Masculino , Pessoa de Meia-Idade , Razão de Chances , Polimorfismo Genético/genética , Polimorfismo de Fragmento de Restrição , Prevalência , Fatores de Risco , Fumar
11.
J Geriatr Oncol ; 5(1): 40-8, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24484717

RESUMO

OBJECTIVE: Components of metabolic syndrome (MS) have been individually linked to colorectal cancer risk and prognosis; however, an understanding of the dominant mechanisms is lacking. MATERIALS AND METHODS: Twenty-one patients (10 MS; 11 non-MS) with resectable colorectal cancer were prospectively enrolled. Patients were classified for MS by the World Health Organization criteria and tested for circulating vascular endothelial growth factor (VEGF), interleukin-6 (IL-6), insulin-like growth factor-1 (IGF-1), fasting insulin, and tumor expression of IGF-1 receptor (IGF-1R), insulin-receptor (IR) and receptor for advanced glycation end-products (RAGE). Circulating markers were re-tested 6 months after surgery. RESULTS: The MS group had significantly higher baseline and post-operative fasting insulin levels (p < 0.001 and 0.003). No differences were observed in circulating IL-6, VEGF, IGF-1 and free IGF-1. By immunohistochemistry (IHC), IGF-1R expression was significantly higher in tumor vs. normal tissues (p < 0.001) while IR expression showed no difference. Interestingly, 64% of tumors demonstrated high IR positivity in the vessels within or surrounding the tumor stroma, but not in the vessels away from the tumor. By reverse transcription polymerase chain reaction (RT-PCR), tumor IGF-1R over-expression (80%) was confirmed, but there was no difference between MS and non-MS patients. Tumor RAGE over-expression was found in 67% of patients and was equally distributed between the two groups. CONCLUSIONS: Hyperinsulinemia was the only significant factor distinguishing patients with colorectal cancer who have MS. The preferential over-expression of IR in the peri-tumoral microvessels suggests that hyperinsulinemia might contribute to colorectal cancer growth by enhancing angiogenesis.


Assuntos
Neoplasias Colorretais/etiologia , Hiperinsulinismo/complicações , Síndrome Metabólica/complicações , Neovascularização Patológica/etiologia , Idoso , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/irrigação sanguínea , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/cirurgia , Feminino , Humanos , Hiperinsulinismo/metabolismo , Imuno-Histoquímica , Fator de Crescimento Insulin-Like I/metabolismo , Masculino , Síndrome Metabólica/metabolismo , Pessoa de Meia-Idade , Neovascularização Patológica/metabolismo , Cuidados Pós-Operatórios , Cuidados Pré-Operatórios , Estudos Prospectivos , Receptor para Produtos Finais de Glicação Avançada , Receptor IGF Tipo 1/metabolismo , Receptor de Insulina/metabolismo , Receptores Imunológicos/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fatores de Crescimento do Endotélio Vascular/metabolismo
12.
Cancer Biol Ther ; 15(9): 1198-207, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24919179

RESUMO

HPP1 (hyperplastic polyposis protein 1), a tumor suppressor gene, is downregulated by promoter hypermethylation in a number of tumor types including colon cancer. c-Myc is also known to play a role in the suppression of HPP1 expression via binding to a promoter region cognate E-box site. The contribution of histone deacetylation as an additional epigenetic mechanism and its potential interplay with c-Myc in the transcriptional regulation of HPP1 are unknown. We have shown that the treatment of the HPP1-non-expressing colon cancer cell lines, HCT116 and DLD-1 with HDAC inhibitors results in re-expression of HPP1. RNAi-mediated knockdown of c-Myc as well as of HDAC2 and HDAC3 in HCT116 and of HDAC1 and HDAC3 in DLD-1 also resulted in significant re-expression of HPP1. Co-immunoprecipitation (IP), chromatin IP (ChIP), and sequential ChIP experiments demonstrated binding of c-Myc to the HPP1 promoter with recruitment of and direct interaction with HDAC3. In summary, we have demonstrated that c-Myc contributes to the epigenetic regulation of HPP1 via the dominant recruitment of HDAC3. Our findings may lead to a greater biologic understanding for the application of targeted use of HDAC inhibitors for anti-cancer therapy.


Assuntos
Neoplasias do Colo/metabolismo , Histona Desacetilases/metabolismo , Proteínas de Membrana/metabolismo , Proteínas de Neoplasias/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Acetilação , Linhagem Celular Tumoral , Neoplasias do Colo/genética , Neoplasias do Colo/patologia , Regulação para Baixo , Epigênese Genética , Técnicas de Silenciamento de Genes , Inibidores de Histona Desacetilases/farmacologia , Histona Desacetilases/genética , Histonas/metabolismo , Humanos , Proteínas de Membrana/genética , Proteínas de Neoplasias/genética , Regiões Promotoras Genéticas , Proteínas Proto-Oncogênicas c-myc/genética , RNA Interferente Pequeno/genética , Transcrição Gênica
13.
J Gastrointest Surg ; 16(5): 905-12; discussion 912-3, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22362069

RESUMO

BACKGROUND: Altered expression of specific microRNAs (miRNA) is known to occur during colorectal carcinogenesis. However, little is known about the genome-wide alterations in miRNA expression during the neoplastic progression of primary colorectal cancers. METHODS: Using a miRNA array platform, we evaluated the expression of 668 miRNA in primary colonic adenocarcinomas. Biological functions of selected miRNA were evaluated with in vitro invasion assays. RESULTS: RNA was extracted for miRNA analysis from 65 primary colon cancers. We identified a seven-miRNA expression signature that differentiated stage I and stage IV primary colon cancers. We then demonstrated this signature was able to discriminate between stage II and III primary colon cancers. Six differentially expressed miRNA were downregulated in association with the development of metastases, and all 7 miRNA were complementary strand miRNA. We transfected HCT-116, a highly invasive colon cancer cell line, with corresponding downregulated miRNA and demonstrated that overexpression of three miRNA (miR200c*, miR143*, and miR424*) significantly abrogated invasive potential. CONCLUSION: We have identified a seven-miRNA signature that is associated with metastatic potential in the primary tumor. Forced overexpression of three downregulated miRNA resulted in attenuation of in vitro invasion, suggesting direct tumor suppressive function and further supporting the biological importance of complementary strand miRNA.


Assuntos
Adenocarcinoma/genética , Algoritmos , Transformação Celular Neoplásica/genética , Neoplasias do Colo/genética , Regulação Neoplásica da Expressão Gênica , MicroRNAs/análise , Adenocarcinoma/patologia , Adenocarcinoma/cirurgia , Idoso , Biomarcadores Tumorais/genética , Colectomia/métodos , Neoplasias do Colo/patologia , Neoplasias do Colo/cirurgia , Regulação para Baixo , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Estadiamento de Neoplasias , Reação em Cadeia da Polimerase em Tempo Real/métodos , Estudos de Amostragem , Sensibilidade e Especificidade , Técnicas de Cultura de Tecidos
14.
PLoS One ; 7(11): e50533, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23226306

RESUMO

BACKGROUND: Changes in host tumor genome DNA methylation patterns are among the molecular alterations associated with HPV-related carcinogenesis. However, there is little known about the epigenetic changes associated specifically with the development of anal squamous cell cancer (SCC). We sought to characterize broad methylation profiles across the spectrum of anal squamous neoplasia. METHODOLOGY/PRINCIPAL FINDINGS: Twenty-nine formalin-fixed paraffin embedded samples from 24 patients were evaluated and included adjacent histologically normal anal mucosa (NM; n = 3), SCC-in situ (SCC-IS; n = 11) and invasive SCC (n = 15). Thirteen women and 11 men with a median age of 44 years (range 26-81) were included in the study. Using the SFP(10) LiPA HPV-typing system, HPV was detected in at least one tissue from all patients with 93% (27/29) being positive for high-risk HPV types and 14 (93%) of 15 invasive SCC tissues testing positive for HPV 16. Bisulfite-modified DNA was interrogated for methylation at 1,505 CpG loci representing 807 genes using the Illumina GoldenGate Methylation Array. When comparing the progression from normal anal mucosa and SCC-IS to invasive SCC, 22 CpG loci representing 20 genes demonstrated significant differential methylation (p<0.01). The majority of differentially methylated gene targets occurred at or close to specific chromosomal locations such as previously described HPV methylation "hotspots" and viral integration sites. CONCLUSIONS: We have identified a panel of differentially methlylated CpG loci across the spectrum of HPV-associated squamous neoplasia of the anus. To our knowledge, this is the first reported application of large-scale high throughput methylation analysis for the study of anal neoplasia. Our findings support further investigations into the role of host-genome methylation in HPV-associated anal carcinogenesis with implications towards enhanced diagnosis and screening strategies.


Assuntos
Neoplasias do Ânus/genética , Neoplasias do Ânus/virologia , Metilação de DNA , Neoplasias de Células Escamosas/genética , Neoplasias de Células Escamosas/virologia , Análise de Sequência com Séries de Oligonucleotídeos , Papillomaviridae/fisiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Canal Anal/patologia , Canal Anal/virologia , Neoplasias do Ânus/patologia , Ilhas de CpG/genética , Feminino , Loci Gênicos/genética , Técnicas de Genotipagem , Humanos , Masculino , Pessoa de Meia-Idade , Invasividade Neoplásica , Neoplasias de Células Escamosas/patologia , Papillomaviridae/genética , Controle de Qualidade , Análise de Sequência de DNA , Sulfitos/farmacologia
15.
Am J Clin Pathol ; 135(3): 436-41, 2011 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-21350099

RESUMO

The progression of cervical intraepithelial lesions to invasive cancer is associated with corresponding reductions in human papillomavirus (HPV) L1 capsid antigen (L1) expression. We sought to determine whether a similar loss of L1 occurs during anal carcinogenesis using immunohistochemistry on paraffin-embedded sections as well as INNO-LiPA HPV Genotyping (Innogenetics, Gent, Belgium) technology to determine HPV infection status. We analyzed 31 squamous cell carcinomas (SCCs), 26 SCCs in situ (SCC-IS), and 11 normal anal mucosae from 36 patients. High-risk HPV subtypes were detected in all patients. L1 nuclear staining was identified in 38% of SCC-IS; however, there was no detection in normal anal mucosae, SCC, or recurrent SCC. Of those SCC-IS associated with a concomitant invasive SCC, only 15% demonstrated nuclear L1 expression as compared to 62% of isolated SCC-IS (P = .02). Nuclear expression of L1 is lost in the progression of anal SCC-IS to SCC and may serve as a possible prognostic marker of enhanced malignant potential.


Assuntos
Neoplasias do Ânus/metabolismo , Proteínas do Capsídeo/metabolismo , Carcinoma in Situ/metabolismo , Carcinoma de Células Escamosas/metabolismo , Papillomavirus Humano 16/isolamento & purificação , Proteínas Oncogênicas Virais/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Canal Anal/metabolismo , Canal Anal/virologia , Neoplasias do Ânus/virologia , Biomarcadores Tumorais/metabolismo , Proteínas do Capsídeo/imunologia , Carcinoma in Situ/virologia , Carcinoma de Células Escamosas/virologia , Núcleo Celular/metabolismo , Núcleo Celular/patologia , Núcleo Celular/virologia , DNA Viral/análise , Progressão da Doença , Feminino , Papillomavirus Humano 16/imunologia , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/virologia , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Proteínas Oncogênicas Virais/imunologia
16.
Int J Cancer ; 119(4): 801-6, 2006 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-16557577

RESUMO

The recently described gene, RAB32, is a ras proto-oncogene family member that encodes an A-kinase-anchoring protein. RAB32 has been found to be frequently hypermethylated in microsatellite instability-high (MSI-H) colon cancers. We sought to determine the prevalence of RAB32 hypermethylation in gastric and endometrial adenocarcinomas, the 2 other major tumor types in which MSI-H is common. Moreover, we delineated the association of RAB32 hypermethylation with microsatellite instability (MSI) and hMLH1 hypermethylation. MSI status and hypermethylation of the RAB32 and hMLH1 genes were studied in paired primary normal and tumor tissues from 48 patients with gastric cancer. An additional 80 endometrial cancer patients were studied for RAB32 methylation and MSI status. Thirteen (27%) of 48 gastric cancers demonstrated evidence of RAB32 hypermethylation. MSI status was determined in 46 of the tumors, with 7 (100%) of 7 MSI-H tumors, 1 (33%) of 3 MSI-low (MSI-L) tumors and 4 (11%) of 36 microsatellite-stable (MSS) tumors found to harbor RAB32 hypermethylation. RAB32 methylation was significantly associated with intestinal type histology and concomitant hMLH1 hypermethylation in gastric cancer. In contrast, RAB32 methylation occurred in only 1 of 80 endometrial cancers, including 20 MSI-H, 8 MSI-L and 52 MSS tumors. Hypermethylation of hMLH1 was noted in 16 (20%) of 80 endometrial tumors. We conclude that although RAB32 methylation is rare in endometrial cancers, it is strongly associated with hMLH1 hypermethylation and MSI in gastric adenocarcinomas. Given its similar involvement in colon cancer, RAB32 inactivation may represent a component of the oncogenic pathway of microsatellite-unstable gastrointestinal adenocarcinomas.


Assuntos
Adenocarcinoma/genética , Metilação de DNA , Neoplasias do Endométrio/genética , Instabilidade Genômica/genética , Repetições de Microssatélites/genética , Neoplasias Gástricas/genética , Proteínas rab de Ligação ao GTP/genética , Proteínas Adaptadoras de Transdução de Sinal , Adulto , Idoso , Idoso de 80 Anos ou mais , Proteínas de Transporte/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Humanos , Masculino , Pessoa de Meia-Idade , Proteína 1 Homóloga a MutL , Proteínas Nucleares/metabolismo , Proto-Oncogene Mas , Proteínas rab de Ligação ao GTP/metabolismo
17.
J Urol ; 170(6 Pt 1): 2471-4, 2003 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-14634453

RESUMO

PURPOSE: The human oxoguanine glycosylase 1 (hOGG1) gene encodes a DNA glycosylase that is involved in excision repair of 8-OH-dG (8-hydroxy-2-deoxyguanine) from oxidatively damaged DNA. To determine whether hOGG1 has a role in the risk of prostate cancer we screened normal prostate tissue specimens for hOGG1 expression and assessed the role of hOGG1 Ser326Cys polymorphism in the risk of prostate cancer. MATERIALS AND METHODS: In 5 normal prostate tissues hOGG1 expression was determined by reverse transcriptase-polymerase chain reaction. The prevalence of hOGG1 Ser326Cys polymorphism was compared between white patients cases and controls using polymerase chain reaction restriction fragment length polymorphism analysis of genomic DNA isolated from 84 incident patients with primary prostate cancer and 252 individually matched controls (1:3 ratio) by age (+/- 5 years at diagnosis) in white men. RESULTS: In all prostate tissues tested hOGG1 mRNA was detected. A significant association was found between hOGG1 genotypes and prostate cancer with a dose effect relationship (trend test p <0.003). A significantly increased risk of prostate cancer was observed for subjects with hOGG1(326Cys) allele (ORadj 2.1, 95% CI 1.2-3.8). CONCLUSIONS: These results suggest that hOGG1 may have a role in the repair of 8-OH-dG adducts in prostate tissue and hOGG1 Ser326Cys polymorphism is associated with prostate cancer risk.


Assuntos
Adenocarcinoma/genética , DNA Glicosilases/genética , Polimorfismo Genético , Neoplasias da Próstata/genética , Adenocarcinoma/enzimologia , DNA Glicosilases/metabolismo , Reparo do DNA , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Neoplasias da Próstata/enzimologia , Fatores de Risco
18.
Carcinogenesis ; 23(7): 1229-34, 2002 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-12117782

RESUMO

The human OGG1 (hOGG1) gene encodes a DNA glycosylase that is involved in the excision repair of 8-hydroxy-2'-deoxyguanine (8-OH-dG) from oxidatively-damaged DNA. To determine whether hOGG1 plays a role in risk for orolaryngeal cancer, we screened normal orolaryngeal tissue specimens for hOGG1 expression and assessed the role of the hOGG1 Ser326Cys polymorphism in risk for orolaryngeal cancer. hOGG1 expression was determined by reverse transcription-polymerase chain reaction of total RNA from aerodigestive tract tissues, and hOGG1 genotyping was performed by polymerase chain reaction-restriction fragment length polymorphism analysis of buccal cell DNA isolated from 169 Caucasian orolaryngeal cancer cases and 338 race-, sex- and age-matched controls. hOGG1 mRNA was detected in all aerodigestive tract tissues tested including tonsil, tongue, floor of mouth, larynx and esophagus. Significantly increased risk for orolaryngeal cancer was observed for both the hOGG1 326(Ser)/326(Cys) (odds ratio [OR] = 1.6, 95% confidence interval [CI] = 1.04-2.6) and hOGG1 326(Cys)/326(Cys) (OR = 4.1, 95% CI = 1.3-13) genotypes. Although no significant difference in risk for orolaryngeal cancer was observed for hOGG1 genotypes in never-smokers, increased risk for orolaryngeal cancer was observed for subjects with the homozygous polymorphic hOGG1 326(Cys)/326(Cys) genotype in smokers (>100 cigarettes lifetime; OR = 4.8, 95% CI = 1.3-18). Similarly, although no association was observed in never drinkers of alcohol, significantly increased risk was observed for the hOGG1 326(Cys)/326(Cys) genotype in alcohol drinkers (>1 shot/week; OR = 6.9, 95% CI = 1.6-29). These results suggest that hOGG1 may play an important role in the repair of 8-OH-dG adducts in the aerodigestive tract and that the hOGG1 Ser326Cys polymorphism plays an important role in risk for smoking- and alcohol-related orolaryngeal cancer.


Assuntos
Carcinoma de Células Escamosas/enzimologia , Reparo do DNA , Neoplasias Laríngeas/enzimologia , Neoplasias Bucais/enzimologia , N-Glicosil Hidrolases/genética , Adulto , Idoso , Estudos de Casos e Controles , Primers do DNA/química , DNA-Formamidopirimidina Glicosilase , Feminino , Predisposição Genética para Doença , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase , Polimorfismo Genético , RNA Mensageiro/metabolismo , Fatores de Risco
19.
Cancer ; 98(4): 872-80, 2003 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-12910533

RESUMO

BACKGROUND: UGT1A10 exhibits glucuronidating activity against metabolites of the tobacco smoke carcinogen, benzo(a)pyrene, and is expressed highly in numerous target tissues for tobacco-related cancers including the upper aerodigestive tract. The current study was conducted to determine the prevalence of genetic polymorphisms in the UGT1A10-specific region of the UDP-glucuronosyltransferase family 1A locus and their relationship with risk for orolaryngeal carcinoma. METHODS: The authors analyzed UGT1A10-specific sequences in a population of black, white, and Asian individuals. Ten UGT1A10 alleles were identified by direct sequencing of UGT1A10 sequences amplified by polymerase chain reaction (PCR) using DNA purified from buccal cell swabs that were taken from individual subjects. RESULTS: In addition to three silent polymorphisms, three missense polymorphisms were found at codons 139 (Glu > Lys), 240 (Thr > Met), and 244 (Leu > Ile). Using PCR-restriction fragment length polymorphism analysis of buccal cell DNA, the prevalence of the UGT1A10(240Met) variant was less than 0.01% in whites and blacks. Similarly, the prevalence of both the UGT1A10(139Lys) and UGT1A10(244Ile) variants was less than 0.01% in whites but it was significantly higher (0.04 and 0.05, respectively, P < 0.01) in blacks. None of the missense UGT1A10 variants were found in any of the Asian individuals examined. In a case-control study of black individuals, a significant association with orolaryngeal carcinoma risk was found in persons with at least 1 UGT1A10(139Lys) allele (crude odds ratio, 0.29 [95% confidence interval, 0.10-0.81]; adjusted odds ratio, 0.20 [95% confidence interval, 0.05-0.87]). No association was observed for the codon 244 (Leu > Ile) polymorphism. CONCLUSIONS: The data from the current study show that the UGT1A10 gene has several low-frequency missense polymorphisms and that the codon 139 polymorphism is an independent risk factor for orolaryngeal carcinoma in blacks.


Assuntos
Carcinoma de Células Escamosas/genética , Glucuronosiltransferase/genética , Neoplasias Laríngeas/genética , Neoplasias Bucais/genética , Polimorfismo Genético , Consumo de Bebidas Alcoólicas/efeitos adversos , Alelos , Estudos de Casos e Controles , Frequência do Gene , Humanos , Pessoa de Meia-Idade , Mutação de Sentido Incorreto , Razão de Chances , Reação em Cadeia da Polimerase , Polimorfismo de Fragmento de Restrição , Grupos Raciais , Fatores de Risco , Fumar/efeitos adversos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA