Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
Gac Med Mex ; 158(5): 283-288, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36572026

RESUMO

INTRODUCTION: Colorectal cancer (CRC) is a complex disease due to the large number of factors that influence its development, including variants in tumor suppressor genes. OBJECTIVE: To estimate allelic and genotypic frequencies of c.3915G>A and c.5371G>A variants of the TSC2 gene in a Mexican population with CRC, as well as to analyze their association with the development of CRC. METHODS: 126 peripheral blood samples from patients diagnosed with sporadic CRC and 134 from healthy individuals, regarded as the control group, were included. Identification of genotypes was carried out using traditional PCR and enzymatic digestion. All individuals signed an informed consent letter. RESULTS: The A allele of the c.3915G>A variant (OR = 0.31, 95% CI = 0.15-0.69, p = 0.004), as well as A/G haplotype of the c.3915G>A and c.5371G>A variants (OR = 0.28, 95% CI = 0.12-0.68, p = 0.005) showed a possible protective effect against sporadic CRC. In silico analysis indicated that both variants generate modifications in the splicing process. CONCLUSION: The presence of TSC2 gene c.3915G>A variant suggests a possible protective effect against sporadic CRC in the Mexican population; however, no association was observed with the c.5371G>A variant.


INTRODUCCIÓN: El cáncer colorrectal (CCR) es una enfermedad compleja debido al gran número de factores que influyen en su desarrollo, incluyendo variantes en genes supresores de tumores. OBJETIVO: Estimar las frecuencias alélicas y genotípicas de las variantes c.3915G>A y c.5371G>A del gen TSC2 en una población mexicana con CCR, así como analizar la asociación con el desarrollo de CCR. MÉTODOS: Se incluyeron 126 muestras de sangre periférica de pacientes con diagnóstico de CCR esporádico y 134 de individuos sanos, considerados como grupo de control. La identificación de los genotipos se llevó a cabo mediante PCR tradicional y digestión enzimática. Todos los individuos firmaron una carta de consentimiento informado. RESULTADOS: El alelo A de la variante c.3915G>A (RM = 0.31, IC 95 % = 0.15-0.69, p = 0.004), así como el haplotipo A/G de las variantes c.3915G>A y c.5371G>A (RM = 0.28, IC 95 % = 0.12-0.68, p = 0.005) mostraron un posible efecto protector contra CCR esporádico. El análisis in silico indicó que ambas variantes generan modificaciones en el proceso de corte y empalme. CONCLUSIÓN: La presencia de la variante c.3915G>A del gen TSC2 sugiere un posible efecto protector contra CCR esporádico en población mexicana; sin embargo, no se observó esta asociación con la variante c.5371G>A.


Assuntos
Neoplasias Colorretais , Proteína 2 do Complexo Esclerose Tuberosa , Humanos , Neoplasias Colorretais/genética , Neoplasias Colorretais/prevenção & controle , Mutação , Proteína 2 do Complexo Esclerose Tuberosa/genética , Proteínas Supressoras de Tumor/genética
2.
Gac. méd. Méx ; Gac. méd. Méx;158(5): 293-298, sep.-oct. 2022. tab, graf
Artigo em Espanhol | LILACS-Express | LILACS | ID: biblio-1404857

RESUMO

Resumen Introducción: El cáncer colorrectal (CCR) es una enfermedad compleja debido al gran número de factores que influyen en su desarrollo, incluyendo variantes en genes supresores de tumores. Objetivo: Estimar las frecuencias alélicas y genotípicas de las variantes c.3915G>A y c.5371G>A del gen TSC2 en una población mexicana con CCR, así como analizar la asociación con el desarrollo de CCR. Métodos: Se incluyeron 126 muestras de sangre periférica de pacientes con diagnóstico de CCR esporádico y 134 de individuos sanos, considerados como grupo de control. La identificación de los genotipos se llevó a cabo mediante PCR tradicional y digestión enzimática. Todos los individuos firmaron una carta de consentimiento informado. Resultados: El alelo A de la variante c.3915G>A (RM = 0.31, IC 95 % = 0.15-0.69, p = 0.004), así como el haplotipo A/G de las variantes c.3915G>A y c.5371G>A (RM = 0.28, IC 95 % = 0.12-0.68, p = 0.005) mostraron un posible efecto protector contra CCR esporádico. El análisis in silico indicó que ambas variantes generan modificaciones en el proceso de corte y empalme. Conclusión: La presencia de la variante c.3915G>A del gen TSC2 sugiere un posible efecto protector contra CCR esporádico en población mexicana; sin embargo, no se observó esta asociación con la variante c.5371G>A.


Abstract Introduction: Colorectal cancer (CRC) is a complex disease due to the large number of factors that influence its development, including variants in tumor suppressor genes. Objective: To estimate allelic and genotypic frequencies of c.3915G>A and c.5371G>A variants of the TSC2 gene in a Mexican population with CRC, as well as to analyze their association with the development of CRC. Methods: 126 peripheral blood samples from patients diagnosed with sporadic CRC and 134 from healthy individuals, regarded as the control group, were included. Identification of genotypes was carried out using traditional PCR and enzymatic digestion. All individuals signed an informed consent letter. Results: The A allele of the c.3915G>A variant (OR = 0.31, 95% CI = 0.15-0.69, p = 0.004), as well as A/G haplotype of the c.3915G>A and c.5371G>A variants (OR = 0.28, 95% CI = 0.12-0.68, p = 0.005) showed a possible protective effect against sporadic CRC. In silico analysis indicated that both variants generate modifications in the splicing process. Conclusion: The presence of TSC2 gene c.3915G>A variant suggests a possible protective effect against sporadic CRC in the Mexican population; however, no association was observed with the c.5371G>A variant.

3.
Genes (Basel) ; 12(12)2021 11 27.
Artigo em Inglês | MEDLINE | ID: mdl-34946849

RESUMO

Prostate cancers may reactivate a latent embryonic program called the epithelial-mesenchymal transition (EMT) during the development of metastatic disease. Through EMT, tumors can develop a mesenchymal phenotype similar to cancer stem cell traits that contributes to metastasis and variation in therapeutic responses. Some of the recurrent somatic mutations of prostate cancer affect EMT driver genes and effector transcription factors that induce the chromatin- and androgen-dependent epigenetic alterations that characterize castrate-resistant prostate cancer (CRPC). EMT regulators in prostate cancer comprise transcription factors (SNAI1/2, ZEB1, TWIST1, and ETS), tumor suppressor genes (RB1, PTEN, and TP53), and post-transcriptional regulators (miRNAs) that under the selective pressures of antiandrogen therapy can develop an androgen-independent metastatic phenotype. In prostate cancer mouse models of EMT, Slug expression, as well as WNT/ß-Catenin and notch signaling pathways, have been shown to increase stemness potential. Recent single-cell transcriptomic studies also suggest that the stemness phenotype of advanced prostate cancer may be related to EMT. Other evidence correlates EMT and stemness with immune evasion, for example, activation of the polycomb repressor complex I, promoting EMT and stemness and cytokine secretion through RB1, TP53, and PRC1. These findings are helping clinical trials in CRPC that seek to understand how drugs and biomarkers related to the acquisition of EMT can improve drug response.


Assuntos
Biomarcadores Tumorais/genética , Transição Epitelial-Mesenquimal/genética , Células-Tronco Neoplásicas/metabolismo , Neoplasias de Próstata Resistentes à Castração/genética , Neoplasias de Próstata Resistentes à Castração/metabolismo , Transdução de Sinais/genética , Animais , Humanos , Masculino , Medicina de Precisão/métodos
4.
Int J Mol Sci ; 22(17)2021 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-34502458

RESUMO

Immunotherapy has improved patient survival in many types of cancer, but for prostate cancer, initial results with immunotherapy have been disappointing. Prostate cancer is considered an immunologically excluded or cold tumor, unable to generate an effective T-cell response against cancer cells. However, a small but significant percentage of patients do respond to immunotherapy, suggesting that some specific molecular subtypes of this tumor may have a better response to checkpoint inhibitors. Recent findings suggest that, in addition to their function as cancer genes, somatic mutations of PTEN, TP53, RB1, CDK12, and DNA repair, or specific activation of regulatory pathways, such as ETS or MYC, may also facilitate immune evasion of the host response against cancer. This review presents an update of recent discoveries about the role that the common somatic mutations can play in changing the tumor microenvironment and immune response against prostate cancer. We describe how detailed molecular genetic analyses of the tumor microenvironment of prostate cancer using mouse models and human tumors are providing new insights into the cell types and pathways mediating immune responses. These analyses are helping researchers to design drug combinations that are more likely to target the molecular and immunological pathways that underlie treatment failure.


Assuntos
Imunoterapia , Neoplasias da Próstata/genética , Microambiente Tumoral/imunologia , Animais , Ensaios Clínicos como Assunto , Genes Neoplásicos , Humanos , Masculino , Mutação , Neoplasias Experimentais/imunologia , Neoplasias da Próstata/imunologia , Análise de Célula Única , Análise Espacial , Microambiente Tumoral/genética
5.
Rev. cuba. invest. bioméd ; 40(2): e1189, 2021. tab, graf
Artigo em Espanhol | LILACS, CUMED | ID: biblio-1347466

RESUMO

Introducción: El cáncer pulmonar constituye un serio problema de salud mundial por su elevada prevalencia y mortalidad. En la carcinogénesis pulmonar están implicados oncogenes y genes supresores tumorales, que en una compleja interacción con factores ambientales favorecen la transformación cancerosa. Objetivo: Describir los principales genes implicados en el cáncer pulmonar. Métodos: Se buscaron referencias en las bases de datos PubMed Central, Annual Reviews y SciELO. Se revisaron preferentemente los artículos originales, las revisiones bibliográficas, las revisiones sistemáticas y los metaanálisis de los últimos cinco años. Análisis e integración de la información: En la carcinogénesis pulmonar se involucran los oncogenes JUN, FOS, ABL1, BRAF, RAF1, GNAS, KRAS, NRAS, HRAS, CSF 1R, MYC, EGFR, MET, ALK, CCNE1, DDR2, ERBB3, FGFR1, MDM2, ROS1, SOX2 y TP63 y los genes supresores tumorales TP53, CDKN2A, CDKN1A, RB1, CDK2AP1, ATM, ERCC2, BRCA1, CCND1, STK11, PDLIM2, PTEN, ARID1A, ASCL4, CUL3, EP300, KEAP1, KMT2D, NF1, NOTCH1, RASA1, ETD2 y SMARCA4. El conocimiento de la genética molecular del cáncer pulmonar es importante para la identificación de biomarcadores diagnósticos y pronósticos más eficaces y para el diseño de fármacos diana sobre genes específicos(AU)


Introduction: Lung cancer is a serious global health problem due to its high prevalence and mortality. Lung carcinogenesis involves oncogenes and tumor suppressor genes which interact in complex manners with environmental factors, paving the way for the cancerous transformation. Objective: Describe the main genes involved in lung cancer. Methods: References were searched for in the databases PubMed Central, Annual Reviews and SciELO. Particular attention was paid to original papers, bibliographic reviews, systematic reviews and meta-analyses published in the last five years. Data analysis and integration: Lung carcinogenesis involves the oncogenes JUN, FOS, ABL1, BRAF, RAF1, GNAS, KRAS, NRAS, HRAS, CSF 1R, MYC, EGFR, MET, ALK, CCNE1, DDR2, ERBB3, FGFR1, MDM2, ROS1, SOX2 and TP63, and the tumor suppressor genes TP53, CDKN2A, CDKN1A, RB1, CDK2AP1, ATM, ERCC2, BRCA1, CCND1, STK11, PDLIM2, PTEN, ARID1A, ASCL4, CUL3, EP300, KEAP1, KMT2D, NF1, NOTCH1, RASA1, ETD2 and SMARCA4. Knowledge about the molecular genetics of lung cancer is important to identify more efficient diagnostic and prognostic biomarkers and to design targeted drugs for specific genes(AU)


Assuntos
Humanos , Oncogenes , Biomarcadores , Genes Supressores de Tumor
6.
Cell Commun Signal ; 19(1): 5, 2021 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-33430890

RESUMO

BACKGROUND: Androgen receptor (AR) and polycomb repressive complex 2 (PRC2) are known to co-occupy the loci of genes that are downregulated by androgen-stimulus. Long intergenic non-coding RNA (lincRNA) PVT1 is an overexpressed oncogene that is associated with AR in LNCaP prostate cancer cells, and with PRC2 in HeLa and many other types of cancer cells. The possible involvement of PVT1 in mediating androgen-induced gene expression downregulation in prostate cancer has not been explored. METHODS: LNCaP cell line was used. Native RNA-binding-protein immunoprecipitation with anti-AR or anti-EZH2 was followed by RT-qPCR with primers for PVT1. Knockdown of PVT1 with specific GapmeRs (or a control with scrambled GapmeR) was followed by differentially expressed genes (DEGs) determination with Agilent microarrays and with Significance Analysis of Microarrays statistical test. DEGs were tested as a tumor risk classifier with a machine learning Random Forest algorithm run with gene expression data from all TCGA-PRAD (prostate adenocarcinoma) tumors as input. ChIP-qPCR was performed for histone marks at the promoter of one DEG. RESULTS: We show that PVT1 knockdown in androgen-stimulated LNCaP cells caused statistically significant expression upregulation/downregulation of hundreds of genes. Interestingly, PVT1 knockdown caused upregulation of 160 genes that were repressed by androgen, including a significantly enriched set of tumor suppressor genes, and among them FAS, NOV/CCN3, BMF, HRK, IFIT2, AJUBA, DRAIC and TNFRSF21. A 121-gene-set (out of the 160) was able to correctly predict the classification of all 293 intermediate- and high-risk TCGA-PRAD tumors, with a mean ROC area under the curve AUC = 0.89 ± 0.04, pointing to the relevance of these genes in cancer aggressiveness. Native RIP-qPCR in LNCaP showed that PVT1 was associated with EZH2, a component of PRC2. PVT1 knockdown followed by ChIP-qPCR showed significant epigenetic remodeling at the enhancer and promoter regions of tumor suppressor gene NOV, one of the androgen-repressed genes that were upregulated upon PVT1 silencing. CONCLUSIONS: Overall, we provide first evidence that PVT1 was involved in signaling a genome-wide androgen-dependent transcriptional repressive program of tumor suppressor protein-coding genes in prostate cancer cells. Identification of transcriptional inhibition of tumor suppressor genes by PVT1 highlights the pathway to the investigation of mechanisms that lie behind the oncogenic role of PVT1 in cancer. Video Abstract.


Assuntos
Regulação Neoplásica da Expressão Gênica , Neoplasias da Próstata/genética , RNA Longo não Codificante/genética , Androgênios , Linhagem Celular Tumoral , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Humanos , Estimativa de Kaplan-Meier , Masculino , Neoplasias da Próstata/mortalidade , Receptores Androgênicos/genética , Transdução de Sinais , Proteínas Supressoras de Tumor/genética
7.
Cell Commun Signal, v. 19, 5, jan. 2021
Artigo em Inglês | Sec. Est. Saúde SP, SESSP-IBPROD, Sec. Est. Saúde SP | ID: bud-3446

RESUMO

Background: Androgen receptor (AR) and polycomb repressive complex 2 (PRC2) are known to co-occupy the loci of genes that are downregulated by androgen-stimulus. Long intergenic non-coding RNA (lincRNA) PVT1 is an overexpressed oncogene that is associated with AR in LNCaP prostate cancer cells, and with PRC2 in HeLa and many other types of cancer cells. The possible involvement of PVT1 in mediating androgen-induced gene expression downregulation in prostate cancer has not been explored. Methods: LNCaP cell line was used. Native RNA-binding-protein immunoprecipitation with anti-AR or anti-EZH2 was followed by RT-qPCR with primers for PVT1. Knockdown of PVT1 with specifc GapmeRs (or a control with scrambled GapmeR) was followed by diferentially expressed genes (DEGs) determination with Agilent microarrays and with Signifcance Analysis of Microarrays statistical test. DEGs were tested as a tumor risk classifer with a machine learning Random Forest algorithm run with gene expression data from all TCGA-PRAD (prostate adenocarcinoma) tumors as input. ChIP-qPCR was performed for histone marks at the promoter of one DEG. Results: We show that PVT1 knockdown in androgen-stimulated LNCaP cells caused statistically signifcant expression upregulation/downregulation of hundreds of genes. Interestingly, PVT1 knockdown caused upregulation of 160 genes that were repressed by androgen, including a signifcantly enriched set of tumor suppressor genes, and among them FAS, NOV/CCN3, BMF, HRK, IFIT2, AJUBA, DRAIC and TNFRSF21. A 121-gene-set (out of the 160) was able to correctly predict the classifcation of all 293 intermediate- and high-risk TCGA-PRAD tumors, with a mean ROC area under the curve AUC=0.89±0.04, pointing to the relevance of these genes in cancer aggressiveness. Native RIP-qPCR in LNCaP showed that PVT1 was associated with EZH2, a component of PRC2. PVT1 knockdown followed by ChIP-qPCR showed signifcant epigenetic remodeling at the enhancer and promoter regions of tumor suppressor gene NOV, one of the androgen-repressed genes that were upregulated upon PVT1 silencing. Conclusions: Overall, we provide frst evidence that PVT1 was involved in signaling a genome-wide androgendependent transcriptional repressive program of tumor suppressor protein-coding genes in prostate cancer cells. Identifcation of transcriptional inhibition of tumor suppressor genes by PVT1 highlights the pathway to the investiga‑tion of mechanisms that lie behind the oncogenic role of PVT1 in cancer.

8.
Front Oncol ; 10: 1152, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32850327

RESUMO

Despite the recent advances in chemotherapeutic treatments against cancer, some types of highly aggressive and invasive cancer develop drug resistance against conventional therapies, which continues to be a major problem in the fight against cancer. In recent years, studies of alterations of DNA methylome have given us a better understanding of the role of DNA methylation in the development of tumors. DNA methylation (DNAm) is an epigenetic change that promotes the covalent transfer of methyl groups to DNA. This process suppresses gene expression through the modulation of the transcription machinery access to the chromatin or through the recruitment of methyl binding proteins. DNAm is regulated mainly by DNA methyltransferases. Aberrant DNAm contributes to tumor progression, metastasis, and resistance to current anti-tumoral therapies. Aberrant DNAm may occur through hypermethylation in the promoter regions of tumor suppressor genes, which leads to their silencing, while hypomethylation in the promoter regions of oncogenes can activate them. In this review, we discuss the impact of dysregulated methylation in certain genes, which impact signaling pathways associated with apoptosis avoidance, metastasis, and resistance to therapy. The analysis of methylome has revealed patterns of global methylation, which regulate important signaling pathways involved in therapy resistance in different cancer types, such as breast, colon, and lung cancer, among other solid tumors. This analysis has provided gene-expression signatures of methylated region-specific DNA that can be used to predict the treatment outcome in response to anti-cancer therapy. Additionally, changes in cancer methylome have been associated with the acquisition of drug resistance. We also review treatments with demethylating agents that, in combination with standard therapies, seem to be encouraging, as tumors that are in early stages can be successfully treated. On the other hand, tumors that are in advanced stages can be treated with these combination schemes, which could sensitize tumor cells that are resistant to the therapy. We propose that rational strategies, which combine specific demethylating agents with conventional treatment, may improve overall survival in cancer patients.

9.
Pathobiology ; 87(3): 208-216, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32369821

RESUMO

BACKGROUND: Malignant pleural mesothelioma (MPM) is a highly lethal disease comprising a heterogeneous group of tumors with challenging to predict biological behavior. The diagnosis is complex, and the histologic classification includes 2 major subtypes of MPM: epithelioid (∼60% of cases) and sarcomatous (∼20%). Its identification depends upon pathological investigation supported by clinical and radiological evidence and more recently ancillary molecular testing. Treatment options are currently limited, with no known targeted therapies available. OBJECTIVES: To elucidate the mutation profile of driver tumor suppressor and oncogenic genes in a cohort of Brazilian patients. METHODS: We sequenced 16 driver genes in a series of 43 Brazilian malignant mesothelioma (MM) patients from 3 distinct Brazilian centers. Genomic DNA was extracted from formalin-fixed paraffin-embedded tumor tissue blocks, and the TERT promoter region was amplified by PCR followed by direct capillary sequencing. The Illumina TruSight Tumor 15 was used to evaluate 250 amplicons from 15 genes associated with solid tumors (AKT1, GNA11, NRAS, BRAF, GNAQ, PDGFRA, EGFR, KIT, PIK3CA, ERBB2, KRAS, RET, FOXL2, MET,and TP53). Library preparation with the TruSight Tumor 15 was performed before sequencing at the MiSeq platform. Data analysis was performed using Sophia DDM software. RESULTS: Out of 43 MPM patients, 38 (88.4%) were epithelioid subtype and 5 (11.6%) were sarcomatoid histotype. Asbestos exposure was present in 15 (39.5%) patients with epithelioid MPM and 3 (60%) patients with sarcomatoid MPM. We found a TERT promoter mutation in 11.6% of MM, and the c.-146C>T mutation was the most common event. The next-generation sequencing was successful in 33 cases. A total of 18 samples showed at least 1 pathogenic, with a median of 1.8 variants, ranging from 1 to 6. The most mutated genes were TP53 and ERBB2 with 7 variants each, followed by NRAS BRAF, PI3KCA, EGFR and PDGFRA with 2 variants each. KIT, AKT1, and FOXL2 genes exhibited 1 variant each. Interestingly, 2 variants observed in the PDGFRA gene are classic imatinib-sensitive therapy. CONCLUSIONS: We concluded that Brazilian MPM harbor mutation in classic tumor suppressor and oncogenic genes, which might help in the guidance of personalized treatment of MPM.


Assuntos
Genes Supressores de Tumor , Neoplasias Pulmonares/genética , Mesotelioma Maligno/genética , Mutação , Oncogenes , Carcinogênese , Estudos de Coortes , Feminino , Sequenciamento de Nucleotídeos em Larga Escala , Humanos , Masculino , Pessoa de Meia-Idade , Inclusão em Parafina
10.
Natal; s.n; 2020. 86 p. tab, ilus, graf.
Tese em Português | LILACS, BBO - Odontologia | ID: biblio-1537299

RESUMO

As proteínas INGs (inhibitor of growth gene) desempenham papel de supressoras tumorais e podem agir por vias dependentes, ou independentes, da p53 na sinalização do ciclo celular e da apoptose. Este trabalho investigou, por meio de imuno-histoquímica, a correlação entre a expressão das proteínas INGs e a expressão da proteína p53 em ceratocistos odontogênicos (20), TOAs (20) e ameloblastomas sólidos (20). Os espécimes foram submetidos à marcação utilizando os anticorpos anti-Ing3, anti-Ing4, anti-Ing5 e anti-p53. Foi realizada análise quantitativa levando-se em consideração a localização citoplasmática e/ou nuclear para as proteínas INGs e a localização nuclear para a proteína p53. A análise da imunoexpressão das proteínas ING1 e ING2 foi realizada em um estudo prévio e os resultados foram considerados apenas para a análise de correlação com as proteínas estudadas neste estudo. Os dados foram analisados pelo Statistical Package for Social Sciences para Windows (SPSS versão 22.0; IBM, USA). Para a comparação da imunoexpressão entre os grupos de lesões foi utilizado o teste de Kruskal Wallis, e para a investigação das correlações foi utilizado o teste de Spearman. Foram considerados significativos os valores de p ≤ 0.05. O presente estudo evidenciou redução da expressão nuclear e citoplasmática das proteínas ING3, ING4 e ING5 em ceratocistos odontogênicos (COs) e ameloblastomas (AMBs). Além disso, em alguns casos, a perda da expressão nuclear das INGs esteve negativamente correlacionada à expressão da proteína p53. As análises de correlação entre as proteínas INGs indicam a existência de mecanismos compensatórios entre as proteínas INGs em folículos dentários (FDs) e tumores odontogênicos adenomatoides (TOAs), estes mecanismos parecem ser menos evidentes em COs e AMBs. Observou-se redução na expressão da proteína ING3 em AMBs (p=0,003); redução na expressão da proteína ING4, tanto em AMBs (p=0,02) quanto em COs (p=0,001); e uma redução da expressão nuclear da proteína ING5 nos COs (p=0,09) e nos AMBs (p=0,012). Foram evidenciadas correlações positivas entre a expressão nuclear da p53 com a expressão citoplasma/núcleo da proteína ING1 (r=0,603; p=0,05) em COs, e com a expressão citoplasma/ núcleo das proteínas ING3 (r=0,475; p=0,034) e ING4 (r=0,448; p=0,047) em AMBs. Por fim, os resultados deste estudo sugerem que a redução na expressão nuclear das proteínas INGs pode ser um evento envolvido na etiopatogênese de lesões odontogênicas mais agressivas, e que a redução da expressão nuclear/citoplasmática das proteínas INGs não está relacionada ao aumento expressão da p53 em COs e AMBs, o que sugere que a expressão destas proteínas deve resultar em alterações funcionais de maneira independente da p53 em lesões odontogênicas (AU).


INGs (inhibitor of growth gene) proteins play a role of tumor suppressors and can act via p53-dependent or independent pathways in signaling cell cycle and apoptosis. The aim of this study is to evaluate correlation between expression of proteins of ING proteins and expression of protein p53 in dental follicles (DF), odontogenic keratocysts (OK), adenomatoid odontogenic tumors (AOT) and solid ameloblastomas (AMBs). The sample was intentional and non-probabilistic, consisting of 20 cases of solid AMBs, 20 cases of AOT, 20 cases of OKs and 10 samples of DFs. The specimens were subjected to immunohistochemical method, using antibodies anti-Ing3, anti-Ing4, anti-Ing5 and antip53. Quantitative analysis was performed taking into account cytoplasmic and / or nuclear location for ING proteins and nuclear location for the p53 protein. The analysis of ING1 and ING2 immunoexpressions was performed in a previous study and the results were considered only for the correlation analysis. Data were analyzed by Statistical Package for Social Sciences for Windows (SPSS version 22.0; IBM, USA). Kruskal Wallis test was used to compare the immunoexpression between the groups of lesions, and Spearman test was used to investigate correlations. Values of p ≤ 0.05 were considered significant. This study showed a reduction in nuclear and cytoplasmic expression of ING3, ING4 and ING5 in odontogenic keratocysts (OKs) and ameloblastomas (AMBs). In addition, in some cases, loss of INGs nuclear expression was negatively correlated with p53 expression. Correlation analyzes may indicate existence of compensatory mechanisms between all the ING proteins in dental follicles (FDs) and adenomatoid odontogenic tumors (TOAs). These mechanisms seem to be less evident in COs and AMBs. The results of this study showed a reduction in ING3 expression in AMBs (p = 0.003); a reduction in ING4 expression, in OKs (p = 0.02) and in AMBs (p = 0.001); and a reduction in ING5 nuclear expression, also in OK (p = 0.09) and in AMBs (p = 0.012). Positive correlations were found between p53 nuclear expression with ING1 cytoplasm / nucleus expression (r = 0.603; p = 0.05) in OKs, and with ING3 cytoplasm / nucleus expression (r = 0.475; p = 0.034) and also ING4 cytoplasm / nucleus expression (r = 0.448; p = 0.047) in AMBs. Finally, this study suggests that reduction in the expression of INGs proteins seems to be an event that occurred in etiopathogenesis of more aggressive odontogenic lesions. Futhermore, nuclear / cytoplasmic expression of INGs proteins is not related to increase in p53 expression in OKs and AMBs, which indicates that loss of expression of these proteins may results in functional changes independently of p53 (AU).


Assuntos
Tumores Odontogênicos/patologia , Genes Supressores de Tumor , Tumor Adenomatoide/patologia , Proteínas Inibidoras de Apoptose , Imuno-Histoquímica/métodos , Fotomicrografia/instrumentação , Cistos Odontogênicos/patologia , Estatísticas não Paramétricas , Estudos Observacionais como Assunto/métodos
11.
J Oral Pathol Med ; 47(2): 186-191, 2018 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29272070

RESUMO

BACKGROUND: Mutations in the patched 1 (PTCH1) gene are the main genetic alteration reported in sporadic and nevoid basal cell carcinoma-associated odontogenic keratocyst (OKC). Oncogenic mutations, including BRAFV600E, previously considered exclusive of malignant neoplasms have been reported in odontogenic tumors. Recently, a high frequency of BRAFV600E mutation has been reported in OKC. Because of the considerable recurrence rate of OKC, the identification of druggable genetic mutations can be relevant in the management of extensive lesions. METHODS: A set of 28 OKCs was included in this work. Initially, 10 sporadic and eight OKC samples from four NBCCS patients (a pair of lesions from each syndromic patient) were submitted to targeted next-generation sequencing (NGS) of 2800 different mutations in 50 oncogenes and tumor suppressor genes, including BRAF. Ten extra sporadic OKC samples were included to assess BRAFV600E mutation using TaqMan allele-specific qPCR. RESULTS: The following missense mutations occurred in one case each: ATM p.Ser333Phe, SMO p.Gly416Glu, PIK3CA p.Ser326Phe, FBXW7 p.Ser438Phe, JAK2 p.Ser605Phe, PTEN p.Arg173His, ATM p.Cys353Arg, PTEN p.Ser294Arg, MET p.His1112Tyr. None of the 18 samples showed the BRAFV600E (or any other V600) mutation in the NGS. BRAFV600E mutation was detected by qPCR in one of the 10 OKC. Collectively, our results show BRAFV600E mutation in 1 of 28 OKC cases. CONCLUSION: On the basis of our results, OKCs do not present recurrent hotspot mutations in these 50 genes commonly mutated in cancer. In addition, BRAFV600E does not play a central role in OKC pathogenesis.


Assuntos
Síndrome do Nevo Basocelular/genética , Mutação , Cistos Odontogênicos/genética , Receptores de Superfície Celular/genética , Adolescente , Adulto , Idoso , Brasil , Carcinoma/genética , Criança , DNA/genética , DNA/isolamento & purificação , Feminino , Genes Supressores de Tumor , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia , Tumores Odontogênicos , Receptor Patched-1/genética , Adulto Jovem
12.
Rev. colomb. gastroenterol ; 33(1): 32-40, 2018. tab, graf
Artigo em Espanhol | LILACS | ID: biblio-900725

RESUMO

Resumen El cáncer colorrectal (CCR) es una enfermedad con una amplia distribución geográfica, que afecta a millones de personas en el mundo. Esta neoplasia se presenta de manera esporádica en el 80% de los casos, el porcentaje restante tiene una historia familiar. Las alteraciones epigenéticas, como la metilación del ADN, modificación de las histonas y los ácidos ribonucleicos (ARN) no codificantes, están involucradas en el desarrollo de esta enfermedad. En la actualidad, estas alteraciones tienen un potencial valioso como biomarcadores para la detección temprana del CCR y se considera que podrían ser útiles para el diagnóstico y pronóstico de los individuos con CCR. El propósito de esta revisión es describir los principales mecanismos epigenéticos involucrados en el cáncer colorrectal, que tienen una función importante en el desarrollo y progresión de la enfermedad.


Abstract Colorectal cancer (CRC) has broad geographic distribution and affects millions of people throughout the world. It occurs sporadically in 80% of cases, but the rest have family histories. Epigenetic alterations such as DNA methylation and modification of histones and non-coding RNA are involved in the development of this disease. At present, these alterations have valuable potential as biomarkers for early detection of CRC and could be useful for diagnosis and determination of prognosis for individuals with CRC. The purpose of this review is to describe the main epigenetic mechanisms involved in colorectal cancer and the important roles they have in the development and progression of the disease.


Assuntos
Humanos , Neoplasias Colorretais , Neoplasias Colorretais/genética , Metilação de DNA , Epigenômica , Prognóstico , Diagnóstico
13.
Clin Epigenetics ; 9: 74, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28748002

RESUMO

BACKGROUND: Changes in DNA methylation, one of the most studied epigenetic mechanisms, are considered an initial marker for early cancer detection. We evaluated how availability of dietary factors (folates and vitamin B12) involved in one-carbon metabolism may contribute to DNA methylation changes of cancer-related genes in human subjects. METHODS: We studied, by pyrosequencing, the methylation of tumor suppressor gene p16, DNA repair genes MLH1 and MGMT, and the repetitive element LINE-1 (as a surrogate for global DNA methylation), in blood of elderly individuals (n = 249) who had been exposed to folic acid (FA) through FA-fortified wheat flour during the last 12 years. RESULTS: We found that serum folate and to a lesser extent, vitamin B12 concentrations, were significantly correlated with DNA methylation of p16, MLH1, and MGMT, but not with LINE-1. High serum folate concentrations (>45.3 nmol/L) were present in 31.1% of the participants. Although the methylated fraction of CpG sites in p16, MLH1, and MGMT was low (1.17-3.8%), high folate concentrations were significantly associated with methylation at the 3rd tertile of specific CpG sites in all genes with OR between 1.97 and 4.17. CONCLUSIONS: This study shows that a public policy, like food fortification with FA that increases circulating serum folate levels, could affect methylation levels of specific genes linked to cancer risk. Our present results deserve additional studies to clarify the real impact of high FA levels for risk of cancer in a whole population chronically exposed to a fortified food such as wheat flour. TRIAL REGISTRATION: ISRCTN 48153354 and ISRCTN 02694183.


Assuntos
Inibidor p16 de Quinase Dependente de Ciclina/genética , Metilação de DNA , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Ácido Fólico/sangue , Proteína 1 Homóloga a MutL/genética , Proteínas Supressoras de Tumor/genética , Idoso , Idoso de 80 Anos ou mais , Chile , Estudos Transversais , Metilação de DNA/efeitos dos fármacos , Epigênese Genética , Feminino , Ácido Fólico/efeitos adversos , Ácido Fólico/farmacologia , Humanos , Elementos Nucleotídeos Longos e Dispersos , Masculino , Análise de Sequência de DNA , Vitamina B 12/sangue
14.
Exp Ther Med ; 13(5): 2442-2450, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28565861

RESUMO

Tumor suppressor gene promoter CpG island methylation is a well-recognized mechanism in cancer pathogenesis, but its role in multiple myeloma (MM) is controversial. The present study investigated the methylation status and expression of P16, suppressor of cytokine signaling 1 (SOCS-1), P73, E-cadherin and Src homology region 2 domain-containing phosphatase 1 (SHP-1), as well as global methylation in patients with MM during active disease and remission. Bone marrow samples were obtained from 43 patients at the Multiple Myeloma Clinic, Instituto Nacional de Ciencias Médicas y Nutrición Salvador Zubirán (Mexico City, Mexico) during active disease and remission. Methylation-specific polymerase chain reaction and ELISA were performed on bisulfite-treated or untreated DNA to determine promoter-specific or genomic methylation, respectively. Gene expression was measured using reverse-transcription polymerase chain reaction. The results indicated that SOCS-1 methylation occurred more frequently during active disease than remission [29 vs. 3.2% (P=0.021)] and was associated with more advanced forms of the disease [international staging system (ISS) 3, 16.67% vs. ISS 1, 8.3% (P=0.037)]. SHP-1 methylation during active disease was associated with a lower probability of survival at 39-month follow up (median), 52.5 vs. 87.5% (P=0.025). The percentage of methylation was associated with active disease at remission, but this was not significant. Global hypomethylation at remission was a negative predictor factor for overall survival (OS). The results indicated that methylated P16, SOCS-1 and SHP-1 were associated with clinical variables of poor prognosis in MM, likewise the persistence of global hypomethylation at remission. The negative impact on OS of global hypomethylation at remission must be confirmed in a larger sample. Future studies are necessary to investigate whether patients with global hypermethylation at remission should receive more aggressive treatments to improve their OS.

15.
Rev. cuba. estomatol ; 53(3): 128-145, jul.-set. 2016.
Artigo em Espanhol | LILACS | ID: lil-794135

RESUMO

Introducción: el cáncer bucal, una enfermedad con elevada morbilidad y mortalidad, se asocia a factores de riesgo, cuya compleja interrelación está sometida a debate científico. Objetivo: actualizar a los profesionales acerca de los factores de riesgo de cáncer bucal. Métodos: la revisión bibliográfica se realizó en SciELO Regional con los descriptores factores de riesgo y cáncer bucal; se encontraron 31 referencias a texto completo. En Clinical Key con cáncer de labio y cavidad bucal se hallaron 1 746; con los descriptores en inglés genes y oral cancer se obtuvieron 9 822 resultados. En PubMed con el descriptor oral cancer se encontraron 1 207 artículos. En EBSCO con el descriptor oral cancer se encontraron a texto completo de los últimos 5 años, 839 artículos. Se recuperaron 200 artículos en inglés y español, principalmente desde 2010 hasta 2015 y se acotaron 62 artículos. Resultados: en la carcinogénesis bucal se afectan los oncogenes y los genes supresores tumorales. Los factores de riesgo no genéticos son el hábito de fumar, consumo de bebidas alcohólicas, estados carenciales de nutrientes, factores ambientales como las radiaciones y los metales pesados y diferentes infecciones bacterianas, micóticas y virales. Estos factores se relacionan también con inmunodepresión, fricción mecánica por prótesis desajustadas y mala higiene bucal. Conclusiones: se describen los principales factores de riesgo de cáncer bucal. Los genes de susceptibilidad al cáncer interaccionan con factores de riesgo relacionados principalmente con estilos de vida y factores ambientales en una compleja red, cuya identificación y control en la atención primaria de salud es importante en la prevención del cáncer bucal por parte de los estomatólogos(AU)


Introduction: oral cancer is a disease with high morbidity and mortality, associated with risk factors whose complex interplay is subject to scientific debate. Objective: to upgrade to the professionals about the risk factors for oral cancer. Methods: the literature review was conducted in SciELO Regional with the descriptors risk factors and oral cancer, for which 31 full-text references were found. In Clinical Key, with the descriptor cancer of the lip and oral cavity, we found 1746; English descriptors with genes and oral cancer retrieved 9822 results. PubMed, with the descriptor "oral cancer", retrieved 1207 items. EBSCO, with the descriptor "oral cancer", offered full texts of 5 recent years (839 articles). 200 articles in English and Spanish were retrieved, mainly from 2010-2015, and 62 papers were used. Data analisys and integration: in the oral carcinogenesis, tumor suppressor genes and oncogenes are affected. The non-genetic risk factors are smoking, alcohol consumption, and nutrient deficiency states, environmental factors such as radiation and heavy metals and different bacterial, fungal and viral infections. These factors are also related to immunosuppression, mechanical friction by maladjusted dentures and poor oral hygiene. Conclusions: the main risk factors for oral cancer are described. Susceptibility genes to cancer interact with risk factors mainly related to lifestyle and environmental factors in a complex network, whose identification and control in primary health care is important in the prevention of oral cancer by dentists(AU)


Assuntos
Humanos , Carcinoma de Células Escamosas/mortalidade , Bases de Dados Bibliográficas/estatística & dados numéricos , Neoplasias Bucais/prevenção & controle , Fatores de Risco , Consumo de Bebidas Alcoólicas/efeitos adversos , Carcinogênese , Estilo de Vida , Uso de Tabaco/efeitos adversos
16.
CCH, Correo cient. Holguín ; 20(2): 292-304, abr.-jun. 2016. ilus
Artigo em Espanhol | LILACS | ID: lil-787147

RESUMO

El papel de los oncogenes y genes supresores de tumor en el control del ciclo celular es conocido, pero, su efecto directo en el metabolismo de la célula tumoral resulta un tema novedoso en la Oncología actual. El concepto de reprogramación metabólica es retomado como un concepto interesante. Por ello, se realizó la presente búsqueda bibliográfica en la base de datos PubMed usando los descriptores: genes, suppressor and metabolism; oncogenes and metabolism; neoplasms and metabolism. Las mutaciones en oncogenes que codifican para PI3K, AKT, mTORC y Myc inducen un aumento de la expresión de isoenzimas de la vía glucolítica y reprimen la fosforilación oxidativa, lo que garantiza un metabolismo anabólico, además, se relacionan con el aumento del consumo de glucosa y liberación de lactato. En células transformadas se demuestra la importancia del metabolismo anabólico para la progresión del tumor, esta es una alternativa para su tratamiento.


The role of the genes that suppress tumors and oncogenes in cellular cycle control is known but its effect in tumoral metabolism is a novel topic in the modern oncology. Nowadays the concept of metabolic reprograming in tumors has been reexamined as an important process related to carcinogenesis. A literature review was done in PubMed database by using descriptors such as genes, suppressor and metabolism; oncogenes and metabolism; neoplasms and metabolism. Oncogenes mutations of code for PI3K, AKT, mTORC and Myc induce an increase of the expression of isoenzimes of the glycolytic path, while they inhibit oxidative phosphorylation. This makes possible anabolic metabolism. They are also related to an increase in glucose consumption rates and the release of lactase to the tumoral microenvironment. The importance of anabolic metabolism in cancer progression has been proved in tumor cells, which is another option in the treatment of the disease.

17.
Future Oncol ; 11(2): 233-49, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25066711

RESUMO

AIM: The aim of the study was to evaluate the use of global and gene-specific DNA methylation changes as potential biomarkers for gallbladder cancer (GBC) in a cohort from Chile. MATERIAL & METHODS: DNA methylation was analyzed through an ELISA-based technique and quantitative methylation-specific PCR. RESULTS: Global DNA Methylation Index (p = 0.02) and promoter methylation of SSBP2 (p = 0.01) and ESR1 (p = 0.05) were significantly different in GBC when compared with cholecystitis. Receiver curve operator analysis revealed promoter methylation of APC, CDKN2A, ESR1, PGP9.5 and SSBP2, together with the Global DNA Methylation Index, had 71% sensitivity, 95% specificity, a 0.97 area under the curve and a positive predictive value of 90%. CONCLUSION: Global and gene-specific DNA methylation may be useful biomarkers for GBC clinical assessment.


Assuntos
Colecistite/diagnóstico , Metilação de DNA , Neoplasias da Vesícula Biliar/diagnóstico , Adulto , Idoso , Idoso de 80 Anos ou mais , Linhagem Celular Tumoral , Chile , Colecistite/genética , Feminino , Neoplasias da Vesícula Biliar/genética , Humanos , Masculino , Pessoa de Meia-Idade , Técnicas de Diagnóstico Molecular , Regiões Promotoras Genéticas , Curva ROC , Análise de Sequência de DNA
18.
Epigenetics ; 9(7): 1031-46, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-24786473

RESUMO

Tumor suppressor genes (TSGs) are commonly inactivated by somatic mutation and/or promoter methylation; yet, recent high-throughput genomic studies have not identified key TSGs inactivated by both mechanisms. We pursued an integrated molecular analysis based on methylation binding domain sequencing (MBD-seq), 450K Methylation arrays, whole exome sequencing, and whole genome gene expression arrays in primary head and neck squamous cell carcinoma (HNSCC) tumors and matched uvulopalatopharyngoplasty tissue samples (UPPPs). We uncovered 186 downregulated genes harboring cancer specific promoter methylation including PAX1 and PAX5 and we identified 10 key tumor suppressor genes (GABRB3, HOXC12, PARP15, SLCO4C1, CDKN2A, PAX1, PIK3AP1, HOXC6, PLCB1, and ZIC4) inactivated by both promoter methylation and/or somatic mutation. Among the novel tumor suppressor genes discovered with dual mechanisms of inactivation, we found a high frequency of genomic and epigenomic alterations in the PAX gene family of transcription factors, which selectively impact canonical NOTCH and TP53 pathways to determine cell fate, cell survival, and genome maintenance. Our results highlight the importance of assessing TSGs at the genomic and epigenomic level to identify key pathways in HNSCC, deregulated by simultaneous promoter methylation and somatic mutations.


Assuntos
Carcinoma de Células Escamosas/genética , Metilação de DNA , Inativação Gênica , Genes Supressores de Tumor , Neoplasias de Cabeça e Pescoço/genética , Regiões Promotoras Genéticas , Carcinoma de Células Escamosas/metabolismo , Linhagem Celular Tumoral , Estudos de Coortes , Ilhas de CpG , Feminino , Neoplasias de Cabeça e Pescoço/metabolismo , Humanos , Masculino , Mutação , Fator de Transcrição PAX5/genética , Fator de Transcrição PAX5/metabolismo , Fatores de Transcrição Box Pareados/genética , Fatores de Transcrição Box Pareados/metabolismo , Receptor Notch1/genética , Receptor Notch1/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
19.
Rev. cientif. cienc. med ; 17(2): 44-52, 2014. ilus
Artigo em Espanhol | LILACS | ID: lil-738107

RESUMO

La neoplasia es una enfermedad genética que usa mecanismos de progresión e invasión similares a los de los tejidos normales cuando proliferan. Su aparición se debe a la suma de alteraciones genéticas que facilitan la progresión tumoral por fallos en los mecanismos de senescencia celular y apoptosis. Dentro de las principales alteraciones moleculares destacan la expresión aberrante de oncogenes, genes supresores tumorales, enzimas y factores de transcripción que promueven un ciclo celular anómalo. El objetivo de esta revisión es el de conocer algunos de los cambios moleculares implicados en el inicio, promoción y progresión de las neoplasias, con el fin de tener información de los genes útiles para realizar diagnósticos más tempranos del cáncer, que favorezcan el pronóstico de la enfermedad y que sean útiles para la investigación en biotecnología diagnóstica y en terapia génica.


Neoplasia, considered as a genetic disease that uses progression mechanisms similar to the normal tissues' when proliferates. Its presence is due to genetic alterations that facilitate tumor progression by failures in the mechanisms of cellular senescence and apoptosis. Within the main molecular alterations the most important are: aberrant expression of oncogenes, tumor suppressor genes, enzymes and transcription factors that promote abnormal cell cycle. The main objective of this paper is to review some of the molecular changes involved in the initiation, promotion and progression of neoplasms, in order to have genes useful information for earlier diagnosis of cancer, favoring the prognosis of genes disease, for diagnostic research in biotechnology and gene therapy.

20.
Int. j. morphol ; 31(3): 973-979, set. 2013. ilus
Artigo em Espanhol | LILACS | ID: lil-694988

RESUMO

La leucemia linfoblástica aguda (LLA), es la neoplasia mas frecuente en la población infantil. Se manifiesta por una perdida de diferenciación de progenitores linfoides produciendo un aumento de células inmaduras. La hipermetilación en la región promotora de genes supresores de tumores (GST) puede producir un silenciamiento génico que le proporciona a la célula leucémica una ventaja proliferativa o la previene de la apoptosis. Se estudia el estado de hipermetilación de 4 GST involucrados en la apoptosis: APAF1, ASPP1, p73 y FHIT y su asociación con la sobrevida de pacientes menores de 15 años con diagnóstico de LLA. Se analizaron 38 muestras de médula ósea mediante modificación con bisulfito del ADN y reacción en cadena de la polimerasa especifica de metilación (MSP). El rango de edad al diagnóstico fue de 10 meses a 13,8 años. La sobrevida global fue de 69 por ciento a los 5 años. El 81,5 por ciento de los pacientes tuvo al menos un gen hipermetilado. La frecuencia de metilación observada fue: APAF1 68,4 por ciento, FHIT 56,4 por ciento, p73 42 por ciento y ASPP1 18,4 por ciento. La asociación entre hipermetilación y grupo <5 años y 5 años fue: Global p=0,20, APAF1 p=0,03, FHIT p=0,51, p73 p=0,51 y ASPP1 p=0.67. Las curvas de sobrevida se calcularon según frecuencia de hipermetilación de cada gen: APAF1 p=0,05, FHIT p=0,31, p73 p=0,98 y ASPP1 p=0,82. La alta frecuencia de hipermetilación obtenida reafirma la participación de la metilación en la región promotora de GST en la patogénesis de la LLA. La hipermetilación del gen APAF1 fue muy frecuente y se asoció significativamente a la sobrevida del grupo de estudio, mostrando a este gen como un factor predictivo de mal pronostico en pacientes con LLA.


Acute lymphoblastic leukemia (ALL) is the most common malignancy in children. It is manifested by a loss of differentiation of lymphoid progenitors, producing an increase of immature cells. Hypermethylation in promoter region of tumor suppressor genes (GST) may produce a gene silencing that provides a leukemic cell a proliferative advantage or prevent apoptosis. We studied the hypermethylation status of 4 GST involved in apoptosis: APAF1, ASPP1, p73 and FHIT and its association with survival of patients <15 years diagnosed with ALL. We analyzed 38 samples of bone marrow by DNA bisulfite modification and chain reaction methylation-specific polymerase (MSP). The mean age at diagnosis was 10 months to 13.8 years. Overall survival was 69 percent at 5 years. 81.5 percent of patients had at least one hypermethylated gene. The frequency observed was: APAF1 68.4 percent, 56.4 percent FHIT, p73 ASPP1 42 percent and 18.4 percent. The association between hypermethylation and group <5 years and 5 years was: Global p = 0.20, APAF1 p = 0.03, FHIT p = 0.51, p73 p = 0.51, ASPP1 p = 0.67. Survival curves were calculated by frequency of hypermethylation of each gene: APAF1 p = 0.05, p = 0.31 FHIT, p73 p = 0.98 and ASPP1 p = 0.82. The high frequency of hypermethylation obtained confirms enrollment of methylation in the promoter region of GST in the pathogenesis of ALL. APAF1 gene hypermethylation was very frequent and was significantly associated with survival in the study group, showing this gene as a predictor of poor prognosis in patients with ALL.


Assuntos
Humanos , Masculino , Adolescente , Feminino , Recém-Nascido , Lactente , Pré-Escolar , Criança , Metilação de DNA , Genes Supressores de Tumor , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Apoptose , Reação em Cadeia da Polimerase , Análise de Sobrevida
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA