Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Sci Rep ; 14(1): 12487, 2024 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-38816545

RESUMO

Peritoneal metastases (PM) in colorectal cancer (CRC) is associated with a dismal prognosis. Identifying and exploiting new biomarkers, signatures, and molecular targets for personalised interventions in the treatment of PM in CRC is imperative. We conducted transcriptomic profiling using RNA-seq data generated from the primary tissues of 19 CRC patients with PM. Using our dataset established in a previous study, we identified 1422 differentially expressed genes compared to non-metastatic CRC. The profiling demonstrated no differential expression in liver and lung metastatic CRC. We selected 12 genes based on stringent criteria and evaluated their expression patterns in a validation cohort of 32 PM patients and 84 without PM using real-time reverse transcription-polymerase chain reaction. We selected cartilage intermediate layer protein 2 (CILP2) because of high mRNA expression in PM patients in our validation cohort and its association with a poor prognosis in The Cancer Genome Atlas. Kaplan-Meier survival analysis in our validation cohort demonstrated that CRC patients with high CILP2 expression had significantly poor survival outcomes. Knockdown of CILP2 significantly reduced the proliferation, colony-forming ability, invasiveness, and migratory capacity and downregulated the expression of molecules related to epithelial-mesenchymal transition in HCT116 cells. In an in vivo peritoneal dissemination mouse knockdown of CILP2 also inhibited CRC growth. Therefore, CILP2 is a promising biomarker for the prediction and treatment of PM in CRC.


Assuntos
Biomarcadores Tumorais , Neoplasias Colorretais , Regulação Neoplásica da Expressão Gênica , Neoplasias Peritoneais , Humanos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/mortalidade , Neoplasias Peritoneais/secundário , Neoplasias Peritoneais/genética , Neoplasias Peritoneais/metabolismo , Biomarcadores Tumorais/metabolismo , Biomarcadores Tumorais/genética , Animais , Camundongos , Masculino , Feminino , Prognóstico , Transição Epitelial-Mesenquimal/genética , Proliferação de Células , Células HCT116 , Perfilação da Expressão Gênica , Pessoa de Meia-Idade , Movimento Celular , Idoso
2.
JMIR Med Inform ; 11: e47859, 2023 Nov 24.
Artigo em Inglês | MEDLINE | ID: mdl-37999942

RESUMO

BACKGROUND: Synthetic data generation (SDG) based on generative adversarial networks (GANs) is used in health care, but research on preserving data with logical relationships with synthetic tabular data (STD) remains challenging. Filtering methods for SDG can lead to the loss of important information. OBJECTIVE: This study proposed a divide-and-conquer (DC) method to generate STD based on the GAN algorithm, while preserving data with logical relationships. METHODS: The proposed method was evaluated on data from the Korea Association for Lung Cancer Registry (KALC-R) and 2 benchmark data sets (breast cancer and diabetes). The DC-based SDG strategy comprises 3 steps: (1) We used 2 different partitioning methods (the class-specific criterion distinguished between survival and death groups, while the Cramer V criterion identified the highest correlation between columns in the original data); (2) the entire data set was divided into a number of subsets, which were then used as input for the conditional tabular generative adversarial network and the copula generative adversarial network to generate synthetic data; and (3) the generated synthetic data were consolidated into a single entity. For validation, we compared DC-based SDG and conditional sampling (CS)-based SDG through the performances of machine learning models. In addition, we generated imbalanced and balanced synthetic data for each of the 3 data sets and compared their performance using 4 classifiers: decision tree (DT), random forest (RF), Extreme Gradient Boosting (XGBoost), and light gradient-boosting machine (LGBM) models. RESULTS: The synthetic data of the 3 diseases (non-small cell lung cancer [NSCLC], breast cancer, and diabetes) generated by our proposed model outperformed the 4 classifiers (DT, RF, XGBoost, and LGBM). The CS- versus DC-based model performances were compared using the mean area under the curve (SD) values: 74.87 (SD 0.77) versus 63.87 (SD 2.02) for NSCLC, 73.31 (SD 1.11) versus 67.96 (SD 2.15) for breast cancer, and 61.57 (SD 0.09) versus 60.08 (SD 0.17) for diabetes (DT); 85.61 (SD 0.29) versus 79.01 (SD 1.20) for NSCLC, 78.05 (SD 1.59) versus 73.48 (SD 4.73) for breast cancer, and 59.98 (SD 0.24) versus 58.55 (SD 0.17) for diabetes (RF); 85.20 (SD 0.82) versus 76.42 (SD 0.93) for NSCLC, 77.86 (SD 2.27) versus 68.32 (SD 2.37) for breast cancer, and 60.18 (SD 0.20) versus 58.98 (SD 0.29) for diabetes (XGBoost); and 85.14 (SD 0.77) versus 77.62 (SD 1.85) for NSCLC, 78.16 (SD 1.52) versus 70.02 (SD 2.17) for breast cancer, and 61.75 (SD 0.13) versus 61.12 (SD 0.23) for diabetes (LGBM). In addition, we found that balanced synthetic data performed better. CONCLUSIONS: This study is the first attempt to generate and validate STD based on a DC approach and shows improved performance using STD. The necessity for balanced SDG was also demonstrated.

3.
Cancer Med ; 12(8): 10091-10104, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36748835

RESUMO

BACKGROUND: Early-onset colorectal cancer (EOCRC) has been increasing in incidence worldwide but its genomic pathogenesis is mostly undetermined. This study aimed to identify robust EOCRC-specific gene expression patterns in non-familial adenomatous polyposis (FAP) and non-hereditary nonpolyposis colorectal cancer syndrome (HNPCC) EOCRC. METHOD: We first performed gene expression profiling analysis using RNA sequencing of discovery cohort comprised of 49 EOCRC (age <50) and 50 late-onset colorectal cancer (LOCRC) (age >70) specimens. To obtain robust gene expression data from this analysis, we validated differentially expressed genes (DEGs) through TCGA cohort (EOCRC:59 samples, LOCRC:229 samples) and our validation cohort (EOCRC:72 samples, LOCRC:43 samples) using real-time RT-PCR. After the validation of DEGs, we validated the selected gene at protein levels using Western blotting. To identify whether genomic methylation regulates the expression of a particular gene, we selected methylation sites using The Cancer Genome Atlas (TCGA) datasets and validated them by pyrosequencing in our validation cohort. RESULTS: The EOCRC patients included in this study had significantly more prominent family history of cancer than the LOCRC patients (23 [46.9%] vs. 13 [26%], p = 0.050). Alanyl aminopeptidase (ANPEP) was significantly downregulated in the EOCRC tissues (FC = 1.78, p = 0.0007) and was also commonly downregulated in the TCGA cohort (FC = -1.08, p = 0.0021). Moreover, the ANPEP mRNA and protein expression levels were significantly downregulated in the EOCRC tissues of our validation cohort (p = 0.037 and 0.027). In comparisons of the normal and tumor tissues in public datasets, the ANPEP level was significantly lower in the tumor tissue in the TCGA dataset (p < 2.2 × 10-16 ) and GSE196006 dataset (p = 0.0005). Furthermore, the ANPEP expression level did not show a decreasing tendency at a young age in the normal colon tissue of the GTEx dataset. Lastly, the hypermethylation of cg26222247 in ANPEP was identified to be weakly associated with reduced ANPEP expression in our EOCRC cohort. CONCLUSION: The reduced expression of ANPEP was identified as a novel biomarker of non-FAP and non-HNPCC EOCRC.


Assuntos
Polipose Adenomatosa do Colo , Neoplasias Colorretais Hereditárias sem Polipose , Neoplasias Colorretais , Humanos , Neoplasias Colorretais/patologia , Antígenos CD13 , Polipose Adenomatosa do Colo/diagnóstico , Polipose Adenomatosa do Colo/genética , Neoplasias Colorretais Hereditárias sem Polipose/genética , Biomarcadores
4.
Genes Genomics ; 44(8): 967-979, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35751785

RESUMO

BACKGROUND: Colorectal cancer (CRC) is the third most common type of diagnosed cancer in the world and has the second-highest mortality rate. Meanwhile, South Korea has the second-highest incidence rate for CRC in the world. OBJECTIVE: To assess the possible influence of ethnicity on the molecular profile of colorectal cancer, we compared genomic and transcriptomic features of South Korean CRCs with European CRCs. METHODS: We assembled a genomic and transcriptomic dataset of South Korean CRC patients (KOCRC; n = 126) from previous studies and European cases (EUCRC; n = 245) selected from The Cancer Genome Atlas (TCGA). Then, we compared the two datasets in terms of clinical data, driver genes, mutational signature, gene sets, consensus molecular subtype, and fusion genes. RESULTS: These two cohorts showed similar profiles in driver mutations but differences in the mutation frequencies of some driver genes (including APC, TP53, PABPC1, FAT4, MUC7, HSPG2, GNAS, DENND5B, and BRAF). Analysis of hallmark pathways using genomic data sets revealed further differences between these populations in the WNT, TP53, and NOTCH signaling pathways. In consensus molecular subtype (CMS) analyses of the study cases, no BRAF mutations were found in the CMS1 subtype of KOCRC, which contrasts with previous findings. Fusion gene analysis identified oncogenic fusion of PTPRK-RSPO3 in a subset of KOCRC patients without APC mutations. CONCLUSIONS: This study presents insights into the genomic landscape of KOCRCs and reveals some similarities and differences with EUCRCs at the molecular level.


Assuntos
Neoplasias Colorretais , Transcriptoma , Povo Asiático , Neoplasias Colorretais/genética , Perfilação da Expressão Gênica , Genômica , Humanos
5.
Anticancer Res ; 41(9): 4651-4658, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34475094

RESUMO

BACKGROUND/AIM: We evaluated the predictive value of candidate serum biomarkers for recurrence in stage II and III colorectal cancer (CRC) after curative surgery. PATIENTS AND METHODS: A total of 33 and 120 patients with CRC with or without recurrence at 5 years after curative surgery were included in the training set and the validation set, respectively. Possible serum biomarkers were examined for associations with CRC recurrence using receiver operating characteristics (ROC) curve analysis. RESULTS: In the training set, the expression levels of the 14 biomarkers were compared according to recurrence. Among them, five biomarkers that had significantly different expression levels were validated in 60 patients with recurrence at 5 years after curative surgery and 60 patients without. Multivariate analysis showed that natural log-transformed values of carcinoembryonic antigen (CEA), cyclin-dependent kinase regulatory subunit 2 (CKS2), 2'-5'-oligoadenylate synthetase 2 (OAS2), and autophagy-related gene 5 (ATG5) in preoperative serum were significantly related to recurrence. ROC analysis showed that these biomarkers were able to discriminate patients with recurrence from those without (area under the curve=0.828, 95% confidence interval=0.755-0.990). CONCLUSION: Preoperative serum levels of CEA, CKS2, OAS2 and ATG5 were independent risk factors for recurrence. A combination of serum CEA, CKS2, OAS2 and ATG5 predicted tumor recurrence well in patients with stage II and III CRC.


Assuntos
Biomarcadores Tumorais/sangue , Neoplasias Colorretais/patologia , Neoplasias Colorretais/cirurgia , Recidiva Local de Neoplasia/diagnóstico , 2',5'-Oligoadenilato Sintetase/sangue , Idoso , Proteína 5 Relacionada à Autofagia/sangue , Quinases relacionadas a CDC2 e CDC28/sangue , Antígeno Carcinoembrionário/sangue , Proteínas de Ciclo Celular/sangue , Neoplasias Colorretais/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Recidiva Local de Neoplasia/sangue , Estadiamento de Neoplasias , Curva ROC
6.
Artigo em Inglês | MEDLINE | ID: mdl-33572855

RESUMO

In this cross-sectional study, we investigated the baseline risk factors of diabetes mellitus (DM) in patients with undiagnosed DM (UDM). We utilized the Korean National Health and Nutrition Examination Survey (KNHANES) 2010-2017 data. Data regarding the participants' demographic characteristics, health status, health determinants, healthcare accessibility, and laboratory tests were gathered to explore the differences between the DM, UDM, and without-DM groups. Among the 64,759 individuals who participated in the KNHANES 2010-2017, 32,611 individuals aged ≥20 years with fasting plasma glucose levels of <100 or ≥126 mg/dL were selected. The odds ratios (ORs) regarding family history of diabetes and the performance of national health and cancer screening tests were lower in the UDM group than in the DM group (adjusted OR: 0.54; 95% confidence interval (CI): 0.43, 0.66; adjusted OR: 0.74; 95% CI: 0.62, 0.89; adjusted OR: 0.71; 95% CI: 0.60, 0.85). The ORs of hypertension and obesity were higher in the UDM group than in the DM group (adjusted OR: 1.32; 95% CI: 1.06, 1.64; adjusted OR: 1.80; 95% CI: 1.37, 2.36, respectively). Patients with UDM were more likely to be exposed to DM-related risk factors than those with and without DM. Public health interventions to prevent UDM development are necessary.


Assuntos
Diabetes Mellitus , Adulto , Idoso , Estudos Transversais , Diabetes Mellitus/epidemiologia , Humanos , Inquéritos Nutricionais , Prevalência , República da Coreia/epidemiologia , Fatores de Risco
7.
Asian J Surg ; 44(6): 848-853, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33622598

RESUMO

BACKGROUND: Pseudomyxoma peritonei (PMP) is a rare disease characterized by mucinous ascites and deposits on the peritoneal surfaces. The study aimed to assess PMP patients according to the Peritoneal Surface Oncology Group International (PSOGI) classification, as a part of standardization of this rare disease. METHODS: This retrospective study analyzed PMP patients who underwent surgery between January 2007 and December 2017. All histologic slides were re-evaluated and the clinical data were collected. According to the PSOGI, PMP was retrospectively classified into three categories: low-grade (LG-PMP), high-grade (HG-PMP), and signet-ring cells (SRC-PMP). The extent of peritoneal involvement was quantified by the peritoneal cancer index (PCI). The completeness of cytoreduction (CCR) was dichotomized as complete or incomplete. RESULTS: Fifty-seven patients were included in this study, consisted of 39 patients with LG-PMP (74.0%), 14 with HG-PMP (20.8%), and 4 with SRC-PMP (5.2%). There was no operative mortality and major complications occurred in 24 patients (31.2%). The 5-year overall survival was 56.2% ± 8.1% for LG-PMP, 37.5% ± 12.1% for HG-PMP, and 25.0% ± 21.7% for SRC-PMP. Concerning CCR, the 5-year overall (complete: 59.5% ± 8.4% vs. incomplete: 12.7% ± 8.1%, p = 0.001) and disease-free survival (complete: 38.6% ± 8.9% vs. incomplete: 7.7% ± 6.8%, p = 0.001) were significantly different. In a multivariable analysis, PSOGI classification and CCR independently correlated with survival (p = 0.011 and 0.018, respectively). CONCLUSIONS: The PSOGI classification provides prognostic stratification, hopefully requiring further validation including every single case of PMP established as a standard criteria.


Assuntos
Neoplasias Peritoneais , Pseudomixoma Peritoneal , Procedimentos Cirúrgicos de Citorredução , Humanos , Prognóstico , Pseudomixoma Peritoneal/diagnóstico , Pseudomixoma Peritoneal/cirurgia , Estudos Retrospectivos
8.
J Cancer Res Clin Oncol ; 147(1): 117-128, 2021 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-32960359

RESUMO

PURPOSE: As few genotype-phenotype correlations are available for nonsyndromic hereditary colorectal cancer (CRC), we implemented genomic analysis on the basis of the revised Bethesda guideline (RBG) and extended (12 items) to verify possible subtypes. METHODS: Patients with sporadic CRC (n = 249) were enrolled, stratified according to the revised Bethesda guidelines (RBG+ and RBG- groups) plus additional criteria. Exome/transcriptome analyses (n = 98) and cell-based functional assays were conducted. RESULTS: We detected 469 somatic and 830 germline gene mutations differing significantly between the positive and negative groups, associated with 12 RBG items/additional criteria. Twenty-one genes had significantly higher mutation rates in left, relative to right, colon cancer, while USP40, HCFC1, and HSPG2 mutation rates were higher in rectal than colon cancer. FAT4 mutation rates were lower in early-onset CRC, in contrast to increased rates in microsatellite instability (MSI)-positive tumors, potentially defining an early-onset microsatellite-stable subtype. The mutation rates of COL6A5 and MGAM2 were significantly and SETD5 was assumably, associated CRC pedigree with concurrent gastric cancer (GC). The predicted deleterious/damaging germline variants, SH2D4A rs35647122, was associated with synchronous/metachronous CRC with related tumors, while NUP160 rs381660 and KRTAP27-1 rs2244485 were potentially associated with a GC pedigree and less strictly defined hereditary CRC, respectively. SH2D4A and NUP160 acted as oncogenic facilitators. CONCLUSION: Our limited genomic analysis for RBG and additional items suggested that specific somatic alterations in the respective items may enlighten relevant pathogenesis along with the knowledge of germline mutations. Further validation is needed to indicate appropriate surveillance in suspected individuals.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Testes Genéticos/métodos , Instabilidade de Microssatélites , Mutação , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Humanos , Prognóstico
9.
Artigo em Inglês | MEDLINE | ID: mdl-33266117

RESUMO

A screening model for estimating undiagnosed diabetes mellitus (UDM) is important for early medical care. There is minimal research and a serious lack of screening models for people with a family history of diabetes (FHD), especially one which incorporates gender characteristics. Therefore, the primary objective of our study was to develop a screening model for estimating UDM among people with FHD and enable its validation. We used data from the Korean National Health and Nutrition Examination Survey (KNHANES). KNAHNES (2010-2016) was used as a developmental cohort (n = 5939) and was then evaluated in a validation cohort (n = 1047) KNHANES (2017). We developed the screening model for UDM in male (SMM), female (SMF), and male and female combined (SMP) with FHD using backward stepwise logistic regression analysis. The SMM and SMF showed an appropriate performance (area under curve (AUC) = 76.2% and 77.9%) compared with SMP (AUC = 72.9%) in the validation cohort. Consequently, simple screening models were developed and validated, for the estimation of UDM among patients in the FHD group, which is expected to reduce the burden on the national health care system.


Assuntos
Diabetes Mellitus Tipo 2 , Diabetes Mellitus , Área Sob a Curva , Diabetes Mellitus/diagnóstico , Diabetes Mellitus/epidemiologia , Feminino , Humanos , Masculino , Programas de Rastreamento , Inquéritos Nutricionais , Fatores de Risco
10.
Exp Mol Med ; 51(10): 1-12, 2019 10 02.
Artigo em Inglês | MEDLINE | ID: mdl-31578316

RESUMO

Approximately half of colorectal cancer (CRC) patients experience disease recurrence and metastasis, and these individuals frequently fail to respond to treatment due to their clinical and biological diversity. Here, we aimed to identify a prognostic signature consisting of a small gene group for precisely predicting CRC heterogeneity. We performed transcriptomic profiling using RNA-seq data generated from the primary tissue samples of 130 CRC patients. A prognostic index (PI) based on recurrence-associated genes was developed and validated in two larger independent CRC patient cohorts (n = 795). The association between the PI and prognosis of CRC patients was evaluated using Kaplan-Meier plots, log-rank tests, a Cox regression analysis and a RT-PCR analysis. Transcriptomic profiling in 130 CRC patients identified two distinct subtypes associated with systemic recurrence. Pathway enrichment and RT-PCR analyses revealed an eleven gene signature incorporated into the PI system, which was a significant prognostic indicator of CRC. Multivariate and subset analyses showed that PI was an independent risk factor (HR = 1.812, 95% CI = 1.342-2.448, P < 0.001) with predictive value to identify low-risk stage II patients who responded the worst to adjuvant chemotherapy. Finally, a comparative analysis with previously reported Consensus Molecular Subgroup (CMS), high-risk patients classified by the PI revealed a distinct molecular property similar to CMS4, associated with a poor prognosis. This novel PI predictor based on an eleven gene signature likely represents a surrogate diagnostic tool for identifying high-risk CRC patients and for predicting the worst responding patients for adjuvant chemotherapy.


Assuntos
Neoplasias Colorretais/genética , Recidiva Local de Neoplasia/genética , Prognóstico , Transcriptoma/genética , Idoso , Biomarcadores Tumorais/genética , Neoplasias Colorretais/patologia , Intervalo Livre de Doença , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica/genética , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Recidiva Local de Neoplasia/patologia , Fatores de Risco
11.
Anticancer Res ; 39(9): 4853-4864, 2019 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-31519588

RESUMO

BACKGROUND/AIM: Colorectal cancer (CRC) is the leading cause of cancer mortality worldwide. Its poor prognosis can be ascribed primarily to high recurrence rates. Accordingly, the aim of this study was to identify novel prognostic biomarkers and therapeutic targets for management of CRC. MATERIALS AND METHODS: To develop prognostic biomarkers, we performed RNA-seq analysis and real-time RT-PCR in primary cancer tissues with or without systemic recurrence. To characterize the molecular functions of the encoded proteins, CRC cells underexpressing or overexpressing the candidate genes were established and appropriate cell-based assays were applied. RESULTS: ITGB1 and RHOC mRNA levels were up-regulated in the recurrence group of CRC patients. Overexpression of ITGB1 or RHOC stimulated CRC cell proliferation, invasion and migration, whereas the opposite effects were observed in cells underexpressing either protein. Five-year recurrence-free survival rates were significantly higher in the ITGB1- and RHOC-underexpression groups than those in the overexpression. CONCLUSION: ITGB1 and RHOC are potential predictors of recurrence and therapeutic targets for CRC, possibly predicting a high-risk group of stage II patients.


Assuntos
Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Integrina beta1/metabolismo , Proteína de Ligação a GTP rhoC/metabolismo , Idoso , Biomarcadores Tumorais , Proliferação de Células , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Técnicas de Inativação de Genes , Humanos , Masculino , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo , Pessoa de Meia-Idade , Gradação de Tumores , Estadiamento de Neoplasias , Prognóstico , RNA Interferente Pequeno/genética , Recidiva , Análise de Sobrevida
12.
World J Gastroenterol ; 25(11): 1341-1354, 2019 Mar 21.
Artigo em Inglês | MEDLINE | ID: mdl-30918427

RESUMO

BACKGROUND: Genomic profiling of tumors has contributed to the understanding of colorectal cancer (CRC), facilitating diagnosis, prognosis and selection of treatments, including targeted regimens. A report suggested that a 19-gene-based risk classifier (TCA19) was a prognostic tool for patients with stage III CRC. The survival outcomes in patients with stage IV CRC are still poor and appropriate selection of targeted therapies and immunotherapies is challenging. AIM: To assess clinical implication of TCA19 in patients with stage IV CRC, and to identify TCA19 with involvement in immune-oncology. METHODS: A retrospective review of the medical records of 60 patients with stage IV CRC was conducted, assessing clinicopathological variables and progression-free survival (PFS). TCA19 gene expression was determined by quantitative polymerase chain reaction (qPCR) in matched normal and tumor tissues taken from the study cohort. Expression of potential immune-oncology regulatory proteins and targets was examined by immunohistochemistry (IHC), western blot, immunofluorescence staining in tissues from a validation cohort of 10 patients, and in CRC cell lines co-cultured with monocyte in vitro. RESULTS: In the patients with TCA19 score higher than the median, the PFS rates of eight patients who received the targeted regimens were significantly higher than the PFS rates of four patients who received 5-fluorouracil-based regimen (P = 0.041). In multivariate analysis, expression of signaling lymphocytic activation molecule family, member 7 (SLAMF7) and triggering receptor expressed on myeloid cells 1 (TREM1) was associated with PFS in the 60-patient cohort. After checking another 10 validate set, the expression of the IHC, the level of real-time qPCR, and the level of western blot were lower for SLAMF7 and higher for TREM7 in primary and metastatic tumors than in normal tissues. In CRC cells expressing SLAMF7 that were co-cultured with a monocytic cell line, levels of CD 68 and CD 73 were significantly lower at day 5 of co-culture than at day 0. CONCLUSION: The TCA19 score might be prognostic for target-regimen-specific PFS in stage IV CRC. Down-regulation of SLAMF7 and up-regulation of TREM1 occur in primary and metastatic tumor tissues.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/mortalidade , Família de Moléculas de Sinalização da Ativação Linfocitária/metabolismo , Receptor Gatilho 1 Expresso em Células Mieloides/metabolismo , Idoso , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/imunologia , Linhagem Celular Tumoral , Técnicas de Cocultura , Neoplasias Colorretais/imunologia , Neoplasias Colorretais/patologia , Neoplasias Colorretais/terapia , Regulação para Baixo , Feminino , Perfilação da Expressão Gênica/métodos , Regulação Neoplásica da Expressão Gênica/imunologia , Humanos , Estimativa de Kaplan-Meier , Masculino , Pessoa de Meia-Idade , Terapia de Alvo Molecular/métodos , Estadiamento de Neoplasias , Seleção de Pacientes , Prognóstico , Intervalo Livre de Progressão , Reação em Cadeia da Polimerase em Tempo Real , Estudos Retrospectivos , Medição de Risco/métodos , Fatores de Risco , Família de Moléculas de Sinalização da Ativação Linfocitária/genética , Família de Moléculas de Sinalização da Ativação Linfocitária/imunologia , Receptor Gatilho 1 Expresso em Células Mieloides/genética , Receptor Gatilho 1 Expresso em Células Mieloides/imunologia , Regulação para Cima
13.
PLoS One ; 13(8): e0202856, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30148861

RESUMO

The present study aimed to identify molecules associated with lymphovascular invasion (LVI) and perineural invasion (PNI) and to examine their biological behavior in colorectal cancer (CRC). LVI- and PNI-associated molecules were identified and verified using sequential processes including (1) identification of 117 recurrence-associated genes differentially expressed on RNA-seq analysis using primary cancer tissues from 130 CRC patients with and without systemic recurrence; (2) analysis of molecules associated with LVI and PNI; (3) assessment of biological properties by measuring proliferation, anoikis, invasion/migration, epithelial-mesenchymal transition and autophagy flux; and (4) verification of disease-free survival using public datasets. Gelsolin (GSN) and 2'-5'-oligoadenylate synthetase 2 (OAS2) were associated with PNI and LVI, respectively. Invasion potential was >2-fold greater in GSN-overexpressing LoVo cells than in control cells (p<0.001-0.005), whereas OAS2-overexpressing RKO cells showed reduced invasion (p<0.001-0.005). GSN downregulated E-cadherin, ß-catenin, claudin-1 and snail, and upregulated N-cadherin and ZEB1, whereas OAS2 overexpression had the opposite effects. Several autophagy-related proteins including ATG5-12, ATG6/BECN1, ATG7 and ATG101 were downregulated in GSN-overexpressing LoVo cells, whereas the opposite pattern was observed in OAS2-overexpressing RKO cells. Patients with low GSN expression had significantly higher 5-year recurrence-free survival (RFS) rates than those with GSN overexpression (73.6% vs. 64.7%, p = 0.038), whereas RFS was longer in patients with OAS2 overexpression than in those with underexpression (73.4% vs. 63.7%, p = 0.01). In conclusion, GSN and OAS2 were positively and negatively associated with recurrence, respectively, suggesting their potential value as predictors of recurrence or therapeutic targets in CRC patients.


Assuntos
2',5'-Oligoadenilato Sintetase/metabolismo , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/patologia , Gelsolina/metabolismo , Recidiva Local de Neoplasia/patologia , 2',5'-Oligoadenilato Sintetase/genética , Apoptose , Biomarcadores Tumorais/genética , Movimento Celular , Proliferação de Células , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Gelsolina/genética , Humanos , Invasividade Neoplásica , Metástase Neoplásica , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/metabolismo , Prognóstico , Taxa de Sobrevida , Células Tumorais Cultivadas
14.
Int J Radiat Oncol Biol Phys ; 98(5): 1164-1173, 2017 08 01.
Artigo em Inglês | MEDLINE | ID: mdl-28721901

RESUMO

PURPOSE: The ability to predict individual responsiveness to cancer therapy is urgently needed. This is particularly true for patients with locally advanced rectal cancer (LARC) because a large proportion are resistant to preoperative chemoradiation therapy (CRT). In this study, we sought to identify markers that could predict response by comparing the gene expression profiles of the tumors of patients who received preoperative CRT. METHODS AND MATERIALS: The basal gene expression profiles of tumors from 22 LARC patients who were responders (n=9) and nonresponders (n=13) to preoperative CRT were analyzed using RNA sequencing (RNA-Seq). To validate the RNA-Seq findings, real-time reverse transcriptase polymerase chain reaction (RT-PCR) was performed on tumor samples from an additional 40 LARC patients (n=20 responders; n=20 nonresponders). Candidate genes were stably overexpressed or knocked down in colorectal cancer (CRC) cell lines, and the effect on response to radiation was tested in vitro and also in vivo in a mouse xenograft model. RESULTS: Eight differentially expressed (>16-fold) genes (B3GALT4, HSPA1B, KRBOX1, PPBP, PPP1R18, PSMB8, SLC39A7, and TAP2) associated with the preoperative CRT response were identified (P<.0005). Among these genes, real-time RT-PCR showed that PSMB8 and SLC39A7 were upregulated in the responsive group of the additional 40 LARC patients. In CRC cell lines, PSMB8 overexpression significantly reduced colony formation and increased the apoptosis-inducing molecules cleaved caspase-3 and cleaved PARP after 6-Gy irradiation. PSMB8 knockdown increased colony formation and decreased caspase-3 activation and cleaved PARP levels after irradiation. SLC39A7 overexpression had no significant effects on irradiated CSC cells. After irradiation of the xenografted mice, tumors that arose from CRC cell line HCT116 overexpressing PSMB8 grew more slowly than did those from HCT116 with vector alone. CONCLUSION: These results suggest that PSMB8 is a predictive marker of preoperative radiosensitivity in LARC patients. Clinical validation in a larger cohort is now required.


Assuntos
Biomarcadores Tumorais/genética , Perfilação da Expressão Gênica/métodos , Complexo de Endopeptidases do Proteassoma/genética , Tolerância a Radiação/genética , Neoplasias Retais/genética , Neoplasias Retais/radioterapia , Análise de Sequência de RNA/métodos , Animais , Biomarcadores Tumorais/metabolismo , Caspase 3/genética , Caspase 3/metabolismo , Proteínas de Transporte de Cátions/genética , Proteínas de Transporte de Cátions/metabolismo , Morte Celular , Linhagem Celular Tumoral , Técnicas de Silenciamento de Genes , Inativação Gênica , Marcadores Genéticos , Xenoenxertos , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Poli(ADP-Ribose) Polimerases/genética , Poli(ADP-Ribose) Polimerases/metabolismo , Cuidados Pré-Operatórios/métodos , Complexo de Endopeptidases do Proteassoma/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Neoplasias Retais/patologia , Neoplasias Retais/cirurgia , Ensaio Tumoral de Célula-Tronco , Regulação para Cima
15.
Oncotarget ; 7(40): 65957-65967, 2016 10 04.
Artigo em Inglês | MEDLINE | ID: mdl-27602754

RESUMO

The tumor necrosis factor-related apoptosis inducing ligand (TRAIL) preferentially induces apoptosis in cancer cells. However, many tumors are resistant to TRAIL-induced apoptosis, and resistance mechanisms are not fully understood. To identify novel regulatory molecules of TRAIL resistance, we screened a siRNA library targeting the human kinome, and NEK4 (NIMA-related kinase-4) was identified. Knockdown of NEK4 sensitized TRAIL-resistant cancer cells and in vivo xenografts to cell death. In contrast, over expression of NEK4 suppressed TRAIL-induced cell death in TRAIL-sensitive cancer cells. In addition, loss of NEK4 resulted in decrease of the anti-apoptotic protein survivin, but an increase in apoptotic cell death. Interestingly, NEK4 was highly upregulated in tumor tissues derived from patients with lung cancer and colon cancer. These results suggest that inhibition of NEK4 sensitizes cancer cells to TRAIL-induced apoptosis by regulation of survivin expression.


Assuntos
Apoptose , Biomarcadores Tumorais/metabolismo , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos , Proteínas Inibidoras de Apoptose/metabolismo , Neoplasias Pulmonares/patologia , Quinases Relacionadas a NIMA/metabolismo , Ligante Indutor de Apoptose Relacionado a TNF/farmacologia , Animais , Proliferação de Células , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/metabolismo , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Masculino , Camundongos , Camundongos Nus , Prognóstico , Survivina , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
16.
PLoS One ; 11(5): e0155160, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27152521

RESUMO

Using our data set (GSE50760) previously established by RNA sequencing, the present study aimed to identify upregulated genes associated with colorectal cancer (CRC) liver metastasis (CLM) and verify their biological behavior. The potential roles of candidate genes in tumors were assessed using cell proliferation and invasion assays. Tissue samples were collected from 18 CRC patients with synchronous CLM and two CRC cell lines (SW480 and SW620) were used for transfection and cloning. The roles of the genes identified in CLM were verified using immunohistochemistry in 48 nude mice after intrasplenic transplantation of CRC cells. mRNA and protein expression was determined by quantitative real-time reverse transcription polymerase chain reaction and western blot, respectively. Nine genes were initially selected according to the relevance of their molecular function and biological process and, finally, ALDH1A1 and IGFBP1 were chosen based on differential mRNA expression and a positive correlation with protein expression. The overexpression of ALDH1A1 and IGFBP1 significantly and time-dependently decreased cell proliferation (p ≤ 0.001-0.003) and suppressed invasiveness by ≥3-fold over control cells (p < 0.001) in the SW480 cell line, whereas they had a slight effect on reducing SW620 cell proliferation. The protein expression levels of E-cadherin, N-cadherin, claudin-1, and vimentin were significantly higher in CLM than in primary tumor tissues (p < 0.05). However, the cadherin switch, namely, N-cadherin overexpression with reduced E-cadherin expression, was not observed in CLM tissues and transfected CRC cells. Irrespective of reduced proliferation and invasion found on in vitro cell assays, persistent overexpression of ß-catenin, vimentin, and ZO-1 in IGFBP1-overexpressing SW480 cells possibly contributed to CLM development in mice implanted with IGFBP1-overexpressing SW480 cells (CLM occurrences: SW480/IGFBP1-transfected mice vs. SW480/vector- and SW480/ALDH1A1-transfected mice, 4/8 vs. 0/10, p = 0.023). In conclusion, ALDH1A1 and IGFBP1 are differentially overexpressed in CLM and may play a dual role, functioning as both tumor suppressors and metastasis promoters in CRC.


Assuntos
Aldeído Desidrogenase/genética , Neoplasias Colorretais/patologia , Proteína 1 de Ligação a Fator de Crescimento Semelhante à Insulina/genética , Neoplasias Hepáticas/secundário , Família Aldeído Desidrogenase 1 , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias Colorretais/genética , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Retinal Desidrogenase
17.
Anticancer Res ; 36(5): 2397-406, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-27127149

RESUMO

AIM: Zinc finger with KRAB and SCAN domain 3 (ZKSCAN3) is overexpressed in invasive colorectal cancer (CRC) cells and regulates the expression of several genes favoring tumor progression, including vascular endothelial growth factor (VEGF) and integrin ß4. We evaluated the association of ZKSCAN3 and colorectal cancer liver metastasis (CLM) to determine whether it is related to invasive signaling pathways. MATERIALS AND METHODS: The ratios of expression by primary tumor to normal tissue and metastatic tumor to normal tissue were compared between ZKSCAN3-overexpressing and underexpressing primary tumor groups. RESULTS: In terms of CLM, the ZKSCAN3 overexpression was positively correlated with carcinoembryonic antigen (CEA), VEGF, and AKT expression. The protein-expression analysis showed that ZKSCAN-specific siRNA knockdown reduced CEA expression in LoVo and LS174T CRC cells. Matrigel invasion by ZKSCAN3-overexpressing HCT116 cells was increased when examined on CEA-coated filters compared with phosphate-buffered saline-treated controls. Additionally, matrix metalloproteinase 9 (MMP9) expression was greater in cells with reference allele (GG) than substitution allele (CC) for ZKSCAN3 rs733743 (p=0.032). ZKSCAN3 protein expression of the high serum CEA group was increased in hepatic metastatic tissue compared with the primary tumor tissue, while in the group with normal serum CEA it decreased or was similar. Reference ZKSCAN3 alleles were correlated with male dominance, a family history of malignancy, high serum CEA concentration and stage IV CRC in 450 patients with sporadic CRC. In conclusion, ZKSCAN3 appears to promote colorectal tumor progression and invasion. ZKSCAN3 may facilitate hepatic metastasis of CRC associated with CEA particularly in cases with CEA-producing tumor.


Assuntos
Antígeno Carcinoembrionário/sangue , Neoplasias Colorretais/patologia , Neoplasias Hepáticas/secundário , Fatores de Transcrição/fisiologia , Linhagem Celular Tumoral , Neoplasias Colorretais/imunologia , Humanos , Neoplasias Hepáticas/imunologia , Metaloproteinase 2 da Matriz/metabolismo , Metaloproteinase 9 da Matriz/metabolismo
18.
Anticancer Res ; 31(12): 4329-38, 2011 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-22199298

RESUMO

A chemosensitive single nucleotide polymorphism (SNP) discovery schema is presented that utilizes (i) genome-wide SNP screening, with a human SNP array and an in vitro chemosensitivity assay, in 93 patients with gastric cancer (GC), and (ii) biological utility assessment using cell viability assays of transfected GC cells. Cytotoxicity analysis showed that most of the MKN1 and SNU638 clones transfected with the G allele of Deoxyribonuclease II beta (DNASE2B) rs3738573 were more sensitive to docetaxel than those with the C allele (p≤0.001-0.029) and most of the AGS and SNU638 clones transfected with the T allele of 5-hydroxytryptamine receptor IE (HTRIE) rs3828741 were more sensitive to paclitaxel than those with the C allele (p≤0.001-0.019). Our findings show that the two novel markers, DNASE2B rs3738573 and HTR1E rs3828741, have potential for improving the prediction of chemosensitivity of GC patients.


Assuntos
Estudo de Associação Genômica Ampla , Polimorfismo de Nucleotídeo Único , Neoplasias Gástricas/genética , Adulto , Idoso , Alelos , Sobrevivência Celular , Docetaxel , Ensaios de Seleção de Medicamentos Antitumorais , Feminino , Genótipo , Haplótipos , Humanos , Desequilíbrio de Ligação , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Receptores de Serotonina/genética , Taxoides/farmacologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA