Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Gut ; 72(2): 242-255, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35705367

RESUMEN

OBJECTIVE: Cell-cell (CC) and cell-matrix (CM) adhesions are essential for epithelial cell survival, yet dissociation-induced apoptosis is frequently circumvented in malignant cells. DESIGN: We explored CC and CM dependence in 58 gastric cancer (GC) organoids by withdrawing either ROCK inhibitor, matrix or both to evaluate their tumorigenic potential in terms of apoptosis resistance, correlation with oncogenic driver mutations and clinical behaviour. We performed mechanistic studies to determine the role of diffuse-type GC drivers: ARHGAP fusions, RHOA and CDH1, in modulating CC (CCi) or CM (CMi) adhesion independence. RESULTS: 97% of the tumour organoids were CMi, 66% were CCi and 52% were resistant to double withdrawal (CCi/CMi), while normal organoids were neither CMi nor CCi. Clinically, the CCi/CMi phenotype was associated with an infiltrative tumour edge and advanced tumour stage. Moreover, the CCi/CMi transcriptome signature was associated with poor patient survival when applied to three public GC datasets. CCi/CMi and CCi phenotypes were enriched in diffuse-type GC organoids, especially in those with oncogenic driver perturbation of RHO signalling via RHOA mutation or ARHGAP fusions. Inducible knockout of ARHGAP fusions in CCi/CMi tumour organoids led to resensitisation to CC/CM dissociation-induced apoptosis, upregulation of focal adhesion and tight junction genes, partial reversion to a more normal cystic phenotype and inhibited xenograft formation. Normal gastric organoids engineered with CDH1 or RHOA mutations became CMi or CCi, respectively. CONCLUSIONS: The CCi/CMi phenotype has a critical role in malignant transformation and tumour progression, offering new mechanistic information on RHO-ROCK pathway inhibition that contributes to GC pathogenicity.


Asunto(s)
Adhesión Celular , Uniones Célula-Matriz , Neoplasias Gástricas , Humanos , Uniones Célula-Matriz/metabolismo , Uniones Célula-Matriz/patología , Progresión de la Enfermedad , Organoides/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patología
2.
Gut ; 69(12): 2165-2179, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32217638

RESUMEN

OBJECTIVE: Sporadic early-onset colorectal cancer (EOCRC) has bad prognosis, yet is poorly represented by cell line models. We examine the key mutational and transcriptomic alterations in an organoid biobank enriched in EOCRCs. DESIGN: We established paired cancer (n=32) and normal organoids (n=18) from 20 patients enriched in microsatellite-stable EOCRC. Exome and transcriptome analysis was performed. RESULTS: We observed a striking diversity of molecular phenotypes, including PTPRK-RSPO3 fusions. Transcriptionally, RSPO fusion organoids resembled normal colon organoids and were distinct from APC mutant organoids, with high BMP2 and low PTK7 expression. Single cell transcriptome analysis confirmed the similarity between RSPO fusion organoids and normal organoids, with a propensity for maturation on Wnt withdrawal, whereas the APC mutant organoids were locked in progenitor stages. CRISPR/Cas9 engineered mutation of APC in normal human colon organoids led to upregulation of PTK7 protein and suppression of BMP2, but less so with an engineered RNF43 mutation. The frequent co-occurrence of RSPO fusions with SMAD4 or BMPR1A mutation was confirmed in TCGA database searches. RNF43 mutation was found in organoid from a leukaemia survivor with a novel mutational signature; and organoids with POLE proofreading mutation displayed ultramutation. The cancer organoid genomes were stable over long culture periods, while normal human colon organoids tended to be subject to clonal dominance over time. CONCLUSIONS: These organoid models enriched in EOCRCs with linked genomic data fill a gap in existing CRC models and reveal distinct genetic profiles and novel pathway cooperativity.


Asunto(s)
Neoplasias Colorrectales/genética , Perfil Genético , Organoides/patología , Proteína de la Poliposis Adenomatosa del Colon/genética , Proteína Morfogenética Ósea 2/genética , Receptores de Proteínas Morfogenéticas Óseas de Tipo 1/genética , Sistemas CRISPR-Cas , Moléculas de Adhesión Celular/genética , Perfilación de la Expresión Génica , Fusión Génica , Humanos , Modelos Genéticos , Mutación , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Fosfatasas Clase 2 Similares a Receptores/genética , Proteína Smad4/genética , Trombospondinas/genética , Bancos de Tejidos , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba , Secuenciación del Exoma
3.
Gut ; 66(9): 1645-1656, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-27329244

RESUMEN

OBJECTIVE: Serrated polyps (hyperplastic polyps, sessile or traditional serrated adenomas), which can arise in a sporadic or polyposis setting, predispose to colorectal cancer (CRC), especially those with microsatellite instability (MSI) due to MLH1 promoter methylation (MLH1me+). We investigate genetic alterations in the serrated polyposis pathway. DESIGN: We used a combination of exome sequencing and target gene Sanger sequencing to study serrated polyposis families, sporadic serrated polyps and CRCs, with validation by analysis of The Cancer Genome Atlas (TCGA) cohort, followed by organoid-based functional studies. RESULTS: In one out of four serrated polyposis families, we identified a germline RNF43 mutation that displayed autosomal dominant cosegregation with the serrated polyposis phenotype, along with second-hit inactivation through loss of heterozygosity or somatic mutations in all serrated polyps (16), adenomas (5) and cancer (1) examined, as well as coincidental BRAF mutation in 62.5% of the serrated polyps. Concurrently, somatic RNF43 mutations were identified in 34% of sporadic sessile/traditional serrated adenomas, but 0% of hyperplastic polyps (p=0.013). Lastly, in MSI CRCs, we found significantly more frequent RNF43 mutations in the MLH1me+ (85%) versus MLH1me- (33.3%) group (p<0.001). These findings were validated in the TCGA MSI CRCs (p=0.005), which further delineated a significant differential involvement of three Wnt pathway genes between these two groups (RNF43 in MLH1me+; APC and CTNNB1 in MLH1me-); and identified significant co-occurrence of BRAF and RNF43 mutations in the MSI (p<0.001), microsatellite stable (MSS) (p=0.002) and MLH1me+ MSI CRCs (p=0.042). Functionally, organoid culture of serrated adenoma or mouse colon with CRISPR-induced RNF43 mutations had reduced dependency on R-spondin1. CONCLUSIONS: These results illustrate the importance of RNF43, along with BRAF mutation in the serrated neoplasia pathway (both the sporadic and familial forms), inform genetic diagnosis protocol and raise therapeutic opportunities through Wnt inhibition in different stages of evolution of serrated polyps.


Asunto(s)
Adenoma/genética , Pólipos del Colon/genética , Neoplasias Colorrectales/genética , Proteínas de Unión al ADN/genética , Homólogo 1 de la Proteína MutL/metabolismo , Proteínas Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Adenoma/patología , Adulto , Pólipos del Colon/patología , Neoplasias Colorrectales/patología , Familia , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas/métodos , Hong Kong , Humanos , Masculino , Inestabilidad de Microsatélites , Persona de Mediana Edad , Mutación , Ubiquitina-Proteína Ligasas , Vía de Señalización Wnt/fisiología
4.
Nat Genet ; 38(10): 1178-83, 2006 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16951683

RESUMEN

Epimutations in the germline, such as methylation of the MLH1 gene, may contribute to hereditary cancer syndrome in human, but their transmission to offspring has never been documented. Here we report a family with inheritance, in three successive generations, of germline allele-specific and mosaic hypermethylation of the MSH2 gene, without evidence of DNA mismatch repair gene mutation. Three siblings carrying the germline methylation developed early-onset colorectal or endometrial cancers, all with microsatellite instability and MSH2 protein loss. Clonal bisulfite sequencing and pyrosequencing showed different methylation levels in different somatic tissues, with the highest level recorded in rectal mucosa and colon cancer tissue, and the lowest in blood leukocytes. This mosaic state of germline methylation with different tissue distribution could act as the first hit and provide a mechanism for genetic disease inheritance that may deviate from the mendelian pattern and be overlooked in conventional leukocyte-based genetic diagnosis strategy.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Proteína 2 Homóloga a MutS/genética , Adulto , Neoplasias Colorrectales Hereditarias sin Poliposis/patología , Metilación de ADN , Femenino , Mutación de Línea Germinal , Humanos , Masculino , Linaje
5.
Nat Commun ; 13(1): 2710, 2022 05 17.
Artículo en Inglés | MEDLINE | ID: mdl-35581206

RESUMEN

Lynch Syndrome (LS) is an autosomal dominant disease conferring a high risk of colorectal cancer due to germline heterozygous mutations in a DNA mismatch repair (MMR) gene. Although cancers in LS patients show elevated somatic mutation burdens, information on mutation rates in normal tissues and understanding of the trajectory from normal to cancer cell is limited. Here we whole genome sequence 152 crypts from normal and neoplastic epithelial tissues from 10 LS patients. In normal tissues the repertoire of mutational processes and mutation rates is similar to that found in wild type individuals. A morphologically normal colonic crypt with an increased mutation burden and MMR deficiency-associated mutational signatures is identified, which may represent a very early stage of LS pathogenesis. Phylogenetic trees of tumour crypts indicate that the most recent ancestor cell of each tumour is already MMR deficient and has experienced multiple cycles of clonal evolution. This study demonstrates the genomic stability of epithelial cells with heterozygous germline MMR gene mutations and highlights important differences in the pathogenesis of LS from other colorectal cancer predisposition syndromes.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Reparación de la Incompatibilidad de ADN/genética , Células Epiteliales/patología , Mutación de Línea Germinal , Humanos , Mutación , Filogenia
6.
Gastroenterology ; 137(1): 176-87, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19303019

RESUMEN

BACKGROUND & AIMS: Repulsive guidance molecule member A (RGMA) is a glycosylphosphatidylinositol-anchored glycoprotein and axon guidance molecule that signals through its receptor, neogenin (NEO1), a homologue of the deleted-in-colorectal cancer (DCC) gene. RGMA also functions as a bone morphogenetic protein (BMP) coreceptor. We studied the potential roles of RGMA and NEO1 in colorectal cancer (CRC) pathogenesis. METHODS: We analyzed expression of RGMA and NEO1, as well as their epigenetic and genetic changes, in a large series of CRC samples, normal colon tissues, adenomas, and cell lines. These studies were accompanied by in vitro functional assay. RESULTS: RGMA and NEO1 expression were significantly down-regulated in most CRCs, adenomas, and cell lines. RGMA was frequently silenced by promoter methylation in CRCs (86.7%), adenomas (90.9%), and CRC cell lines (92.3%) but not in normal colon tissues; allelic imbalance of RGMA and NEO1 was observed in 40% and 49% of CRCs, respectively. In CRC samples, reduced RGMA levels were significantly associated with mismatch repair deficiency or mutations in KRAS or BRAF. Exposure to 5-aza-2'-deoxycytidine restored RGMA expression in CRC cell lines. Transfection of RGMA into CRC cells suppressed cell proliferation, migration, and invasion and also increased apoptosis in response to DNA-damaging agent. CONCLUSIONS: The frequent genetic and epigenetic inactivation of RGMA in CRCs and adenomas along with its in vitro function collectively support its role as a tumor suppressor in colon cells. These findings add to the expanding list of axon guidance molecules with disrupted function during colon carcinogenesis and create new opportunities for early detection and drug development.


Asunto(s)
Adenoma/genética , Neoplasias del Colon/genética , Metilación de ADN , Regulación Neoplásica de la Expresión Génica , Silenciador del Gen , Genes Supresores de Tumor , Proteínas de la Membrana/genética , Proteínas del Tejido Nervioso/genética , Adenoma/metabolismo , Adenoma/patología , Desequilibrio Alélico , Línea Celular Tumoral , Movimiento Celular/genética , Proliferación Celular , Neoplasias del Colon/metabolismo , Neoplasias del Colon/patología , Reparación de la Incompatibilidad de ADN/genética , Proteínas Ligadas a GPI , Humanos , Proteínas de la Membrana/metabolismo , Mutación , Invasividad Neoplásica , Proteínas del Tejido Nervioso/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas p21(ras) , Transfección , Proteínas ras/genética
7.
Clin Cancer Res ; 14(9): 2560-9, 2008 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-18451217

RESUMEN

PURPOSE: Early events in colorectal tumorigenesis include mutation of the adenomatous polyposis coli (APC) gene and epigenetic hypermethylation with transcriptional silencing of the O(6)-methylguanine DNA methyltransferase (MGMT), human mut L homologue 1 (hMLH1), and P16/CDKN2A genes. Epigenetic alterations affect genetic events: Loss of MGMT via hypermethylation reportedly predisposes to guanine-to-adenine or cytosine-to-thymine (G:C-->A:T) transition mutations in KRAS and P53, and silencing of hMLH1 leads to high levels of microsatellite instability (MSI-H)/mutator phenotype, suggesting that epigenetic-genetic subtypes exist. EXPERIMENTAL DESIGN: We evaluated the relationships of aberrant methylation of APC, MGMT, hMLH1, P16, N33, and five MINTs to mutations in APC, KRAS, BRAF, and P53 in 208 colorectal carcinomas. RESULTS: We found that APC hypermethylation was age related (P = 0.04), in contrast to the other genes, and did not cluster with CpG island methylator phenotype (CIMP) markers. Hypermethylation of APC concurrently with either MGMT or hMLH1 was strongly associated with occurrence of G-to-A transitions in APC [odds ratio (OR), 26.8; P < 0.0002 from multivariable logic regression model], but C-to-T transitions had no associations. There was no relationship of hypermethylation of any gene, including MGMT, with G-to-A or C-to-T transitions in KRAS or P53, although APC hypermethylation was associated with P53 mutation (P < 0.0002). CIMP with MSI-H due to hMLH1 hypermethylation, or CIMP with loss of MGMT expression in non-MSI-H tumors, was associated with BRAF mutation (OR, 4.5; P < 0.0002). CIMP was also associated with BRAF V600E T-to-A transversion (OR, 48.5; P < 0.0002). CONCLUSIONS: Our findings suggest that the heterogeneous epigenetic dysregulation of promoter methylation in various genes is interrelated with the occurrence of mutations, as manifested in epigenetic-genetic subgroups of tumors.


Asunto(s)
Neoplasias Colorrectales/genética , Islas de CpG/genética , Metilación de ADN , Metilasas de Modificación del ADN/genética , Enzimas Reparadoras del ADN/genética , Epigénesis Genética , Genes APC , Proteínas Supresoras de Tumor/genética , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Neoplasias Colorrectales/metabolismo , Metilasas de Modificación del ADN/metabolismo , Enzimas Reparadoras del ADN/metabolismo , Femenino , Genes p53 , Genes ras , Humanos , Masculino , Persona de Mediana Edad , Homólogo 1 de la Proteína MutL , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Proteínas Proto-Oncogénicas p21(ras) , Proteínas Supresoras de Tumor/metabolismo , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , Proteínas ras/genética , Proteínas ras/metabolismo
8.
Cell Stem Cell ; 23(6): 882-897.e11, 2018 12 06.
Artículo en Inglés | MEDLINE | ID: mdl-30344100

RESUMEN

Gastric cancer displays marked molecular heterogeneity with aggressive behavior and treatment resistance. Therefore, good in vitro models that encompass unique subtypes are urgently needed for precision medicine development. Here, we have established a primary gastric cancer organoid (GCO) biobank that comprises normal, dysplastic, cancer, and lymph node metastases (n = 63) from 34 patients, including detailed whole-exome and transcriptome analysis. The cohort encompasses most known molecular subtypes (including EBV, MSI, intestinal/CIN, and diffuse/GS, with CLDN18-ARHGAP6 or CTNND1-ARHGAP26 fusions or RHOA mutations), capturing regional heterogeneity and subclonal architecture, while their morphology, transcriptome, and genomic profiles remain closely similar to in vivo tumors, even after long-term culture. Large-scale drug screening revealed sensitivity to unexpected drugs that were recently approved or in clinical trials, including Napabucasin, Abemaciclib, and the ATR inhibitor VE-822. Overall, this new GCO biobank, with linked genomic data, provides a useful resource for studying both cancer cell biology and precision cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Bancos de Muestras Biológicas , Ensayos de Selección de Medicamentos Antitumorales , Organoides/efectos de los fármacos , Organoides/patología , Neoplasias Gástricas/tratamiento farmacológico , Neoplasias Gástricas/patología , Aminopiridinas/farmacología , Bencimidazoles/farmacología , Benzofuranos/farmacología , Proliferación Celular/efectos de los fármacos , Femenino , Humanos , Isoxazoles/farmacología , Masculino , Naftoquinonas/farmacología , Medicina de Precisión , Pirazinas/farmacología , Neoplasias Gástricas/clasificación , Neoplasias Gástricas/genética
9.
Mol Biol Cell ; 14(8): 3208-15, 2003 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-12925757

RESUMEN

Gastric cancer is the world's second most common cause of cancer death. We analyzed gene expression patterns in 90 primary gastric cancers, 14 metastatic gastric cancers, and 22 nonneoplastic gastric tissues, using cDNA microarrays representing approximately 30,300 genes. Gastric cancers were distinguished from nonneoplastic gastric tissues by characteristic differences in their gene expression patterns. We found a diversity of gene expression patterns in gastric cancer, reflecting variation in intrinsic properties of tumor and normal cells and variation in the cellular composition of these complex tissues. We identified several genes whose expression levels were significantly correlated with patient survival. The variations in gene expression patterns among cancers in different patients suggest differences in pathogenetic pathways and potential therapeutic strategies.


Asunto(s)
Adenocarcinoma/genética , Perfilación de la Expresión Génica , Neoplasias Gástricas/genética , Adenocarcinoma/etiología , Adenocarcinoma/metabolismo , Secuencia de Bases , Mucosa Gástrica/metabolismo , Infecciones por Helicobacter/complicaciones , Helicobacter pylori , Humanos , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Gástricas/etiología , Neoplasias Gástricas/metabolismo
10.
Oncogene ; 21(49): 7585-92, 2002 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-12386821

RESUMEN

High-frequency microsatellite instability (MSI-H) results from deficiency in nucleotide mismatch repair. It contributes significantly to carcinogenesis in the human colorectal mucosa. Here we study 41 colorectal and three other HNPCC-related cancers with MSI-H to provide comprehensive information on the mechanisms of inactivation of the two major proteins involved, hMLH1 and hMSH2. Seventeen of the patients had family histories meeting the criteria for Bethesda grades 1, 2 or 3. Of these familial cases, 14 (83%) had early-onset disease, defined on the basis of diagnosis prior to the age of 50, but in three the disease was of late onset (>50 years). A second subset of 20 patients had early onset disease without family history. The remaining seven patients were selected to allow comparisons with sporadic, late-onset disease, the molecular basis of which has been extensively reported elsewhere. We stratified the tumours initially on the basis of hMLH1 or hMSH2 protein deficiency, detected by immunohistochemistry, and then by analysis of germline and somatic mutation, mRNA transcription, loss of heterozygosity (LOH) at the hMLH1 and hMSH2 loci, and methylation status in two regions of the hMLH1 promoter. The functional significance of several of these changes in the MSI-H tumours was confirmed by comparisons with 16 tumours with low-frequency microsatellite instability and 56 tumours with stable microsatellites. As anticipated, patients with family histories usually showed germline mutation of hMSH2 or hMLH1. In many cases the residual normal allele was silenced in their tumours by loss of heterozygosity (LOH). The small subset of late-onset, sporadic cases confirmed the preponderance in this group of biallelic hMLH1 promoter methylation. In the early-onset, apparently sporadic subset there were 11 tumours with hMLH1 deficiency, five with hMSH2 deficiency and four with no detectable abnormality in expression of either protein. These showed a complex mixture of lesions, including germline and somatic mutations, promoter methylation, LOH, suppression of wild-type RNA by as yet undiscovered mechanisms, or no detectable abnormality in any of these parameters. Evidence is presented to indicate that methylation in proximal region of the hMLH1 promoter is a more reliable correlate of transcriptional silencing in colorectal cancers than methylation in upstream region. These observations have significant implications for management of patients with MSI-H tumours.


Asunto(s)
Disparidad de Par Base , Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Neoplasias Colorrectales/genética , Proteínas de Unión al ADN , Mutación de Línea Germinal , Proteínas Adaptadoras Transductoras de Señales , Secuencia de Bases , Proteínas Portadoras , Metilación de ADN , Cartilla de ADN , Humanos , Inmunohistoquímica , Pérdida de Heterocigocidad , Homólogo 1 de la Proteína MutL , Proteína 2 Homóloga a MutS , Proteínas de Neoplasias/genética , Proteínas Nucleares , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas/genética , Transcripción Genética
11.
Oncogene ; 21(42): 6549-56, 2002 Sep 19.
Artículo en Inglés | MEDLINE | ID: mdl-12226758

RESUMEN

Gastric adenocarcinoma is one of the major malignancies worldwide. Gastric cell lines have been widely used as the model to study the genetics, pharmacology and biochemistry of gastric cancers. Here we describe a comprehensive survey of the gene expression profiles of 12 gastric carcinoma cell lines, using cDNA microarray with 43 000 clones. For comparison, we also explored the gene expression patterns of 15 cell lines derived from lymphoid, endothelial, stromal and other epithelial cancers. Expression levels of specific genes were validated through comparison to protein expression by immunohistochemistry using cell block arrays. We found sets of genes whose expression corresponds to the molecular signature of each cell type. In the gastric cancer cell lines, apart from genes that are highly expressed corresponding to their common epithelial origin from the gastrointestinal tract, we found marked heterogeneity among the gene expression patterns of these cell lines. Some of the heterogeneity may reflect their underlying molecular characteristics or specific differentiation program. Two putative gastric carcinoma cell lines were found to be B-cell lymphoma, and another one had no epithelial specific gene expression and hence was of doubtful epithelial origin. These cell lines should no longer be used in gastric carcinoma research. In conclusion, our gene expression database can serve as a powerful resource for the study of gastric cancer using these cell lines.


Asunto(s)
Mucosa Gástrica/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Neoplasias Gástricas/genética , Linaje de la Célula , Células Epiteliales/metabolismo , Mucosa Gástrica/patología , Humanos , Técnicas para Inmunoenzimas , Linfoma de Células B/química , Linfoma de Células B/patología , Proteínas de Neoplasias/metabolismo , Neoplasias Gástricas/metabolismo , Células Tumorales Cultivadas
12.
Nat Genet ; 46(6): 573-82, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24816253

RESUMEN

Gastric cancer is a heterogeneous disease with diverse molecular and histological subtypes. We performed whole-genome sequencing in 100 tumor-normal pairs, along with DNA copy number, gene expression and methylation profiling, for integrative genomic analysis. We found subtype-specific genetic and epigenetic perturbations and unique mutational signatures. We identified previously known (TP53, ARID1A and CDH1) and new (MUC6, CTNNA2, GLI3, RNF43 and others) significantly mutated driver genes. Specifically, we found RHOA mutations in 14.3% of diffuse-type tumors but not in intestinal-type tumors (P < 0.001). The mutations clustered in recurrent hotspots affecting functional domains and caused defective RHOA signaling, promoting escape from anoikis in organoid cultures. The top perturbed pathways in gastric cancer included adherens junction and focal adhesion, in which RHOA and other mutated genes we identified participate as key players. These findings illustrate a multidimensional and comprehensive genomic landscape that highlights the molecular complexity of gastric cancer and provides a road map to facilitate genome-guided personalized therapy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Mutación , Neoplasias Gástricas/genética , Uniones Adherentes , Algoritmos , Animales , Metilación de ADN , Análisis Mutacional de ADN , Epigénesis Genética , Femenino , Dosificación de Gen , Perfilación de la Expresión Génica , Variación Genética , Genoma Humano , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína de Unión al GTP rhoA/genética
13.
Nat Genet ; 43(12): 1219-23, 2011 Oct 30.
Artículo en Inglés | MEDLINE | ID: mdl-22037554

RESUMEN

Gastric cancer is a heterogeneous disease with multiple environmental etiologies and alternative pathways of carcinogenesis. Beyond mutations in TP53, alterations in other genes or pathways account for only small subsets of the disease. We performed exome sequencing of 22 gastric cancer samples and identified previously unreported mutated genes and pathway alterations; in particular, we found genes involved in chromatin modification to be commonly mutated. A downstream validation study confirmed frequent inactivating mutations or protein deficiency of ARID1A, which encodes a member of the SWI-SNF chromatin remodeling family, in 83% of gastric cancers with microsatellite instability (MSI), 73% of those with Epstein-Barr virus (EBV) infection and 11% of those that were not infected with EBV and microsatellite stable (MSS). The mutation spectrum for ARID1A differs between molecular subtypes of gastric cancer, and mutation prevalence is negatively associated with mutations in TP53. Clinically, ARID1A alterations were associated with better prognosis in a stage-independent manner. These results reveal the genomic landscape, and highlight the importance of chromatin remodeling, in the molecular taxonomy of gastric cancer.


Asunto(s)
Exoma , Mutación , Proteínas Nucleares/genética , Neoplasias Gástricas/genética , Factores de Transcripción/genética , Adulto , Anciano , Anciano de 80 o más Años , Proteínas de Ciclo Celular/genética , Ensamble y Desensamble de Cromatina , Proteínas de Unión al ADN , Femenino , Genes Relacionados con las Neoplasias , Estudios de Asociación Genética , Humanos , Uniones Intercelulares , Masculino , Inestabilidad de Microsatélites , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico , Análisis de Secuencia de ADN , Transducción de Señal , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/mortalidad , Adulto Joven
14.
Proc Natl Acad Sci U S A ; 104(39): 15418-23, 2007 Sep 25.
Artículo en Inglés | MEDLINE | ID: mdl-17881565

RESUMEN

Human colonic epithelial cell renewal, proliferation, and differentiation are stringently controlled by numerous regulatory pathways. To identify genetic programs of human colonic epithelial cell differentiation in vivo as well as candidate marker genes that define colonic epithelial stem/progenitor cells and the stem cell niche, we applied gene expression analysis of normal human colon tops and basal crypts by using expression microarrays with 30,000 genes. Nine hundred and sixty-nine cDNA clones were found to be differentially expressed between human colon crypts and tops. Pathway analysis revealed the differential expression of genes involved in cell cycle maintenance and apoptosis, as well as genes in bone morphogenetic protein (BMP), Notch, Wnt, EPH, and MYC signaling pathways. BMP antagonists gremlin 1, gremlin 2, and chordin-like 1 were found to be expressed by colon crypts. In situ hybridization and RT-PCR confirmed that these BMP antagonists are expressed by intestinal cryptal myofibroblasts and smooth muscle cells at the colon crypt. In vitro analysis demonstrated that gremlin 1 partially inhibits Caco-2 cell differentiation upon confluence and activates Wnt signaling in normal rat intestinal epithelial cells. Collectively, the expression data set provides a comprehensive picture of human colonic epithelial cell differentiation. Our study also suggests that BMP antagonists are candidate signaling components that make up the intestinal epithelial stem cell niche.


Asunto(s)
Proteínas Morfogenéticas Óseas/metabolismo , Colon/metabolismo , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Mucosa Intestinal/metabolismo , Células Madre/metabolismo , Apoptosis , Células CACO-2 , Citocinas , Células Epiteliales/metabolismo , Proteínas del Ojo/biosíntesis , Humanos , Péptidos y Proteínas de Señalización Intercelular/biosíntesis , Proteínas del Tejido Nervioso/biosíntesis , Receptores Notch/metabolismo , Transducción de Señal
15.
Carcinogenesis ; 27(3): 454-64, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16272170

RESUMEN

EphB2, a receptor tyrosine kinase regulated by the beta-catenin/Tcf4 complex, is expressed in the proliferative compartment of mouse intestine and regulates bidirectional migration of intestinal precursor cells in the crypt-villus axis through repulsive interaction with Ephrin-B ligands. Recently, it has been shown that reduction of EphB activity accelerates colon tumour progression in the Apc(Min/+) mice. In this study, we examined the expression of EphB2 in normal colon, adenomas, primary colorectal cancers (CRCs), lymph node metastases and liver metastases using immunohistochemistry on tissue microarrays. In addition, EphB2 was overexpressed in SW480 colon cancer cells to study its effect in vitro. We found that EphB2 was expressed in 100% of normal colon crypt base cells, 78% of adenomas, 55.4% of primary CRCs, 37.8% of lymph node metastases and 32.9% of liver metastases (all differences were statistically significant at P < 0.001 compared with primary CRCs). Patients with CRCs that lose EphB2 expression had more advanced tumour stage (P = 0.005), poor differentiation (P < 0.001), poor overall survival (P = 0.005) and disease-free survival (P = 0.001), with the latter being independent of tumour stage. In vitro studies showed that overexpression of EphB2 inhibited colon cancer cell growth in colony formation assay and activation of EphB2 receptor inhibited colon cancer cell adhesion and migration. Our data demonstrated a progressive loss of EphB2 expression in each critical step of colon carcinogenesis, including the onset of invasion, dedifferentiation and metastasis which are paralleled by adverse patient outcome. EphB2 may achieve its tumour suppressor function through regulation of cell survival, adhesion and migration.


Asunto(s)
Adenoma/genética , Adenoma/patología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Invasividad Neoplásica , Metástasis de la Neoplasia , Receptor EphB2/biosíntesis , Adulto , Anciano , Anciano de 80 o más Años , Supervivencia Celular , Transformación Celular Neoplásica , Colon/enzimología , Supervivencia sin Enfermedad , Regulación hacia Abajo , Femenino , Perfilación de la Expresión Génica , Humanos , Neoplasias Hepáticas/secundario , Metástasis Linfática , Masculino , Persona de Mediana Edad , Estadificación de Neoplasias , Pronóstico
16.
Gastroenterology ; 127(2): 457-69, 2004 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-15300578

RESUMEN

BACKGROUND & AIMS: Gastric cancer is one of the major cancers worldwide. Expression profiling has proven useful in delineating novel prognostic markers in various cancer types. We previously analyzed gene-expression patterns in 90 gastric adenocarcinomas by using complementary DNA microarrays and prioritized a list of genes whose expression levels predict patient outcome. METHODS: We identified a specific gene of interest, chemokine (C-C motif) ligand 18 (CCL18), on the basis of a high absolute standardized log Cox hazard ratio, a high variance in expression among all tumor samples, and putative biologic function. Detailed analysis of CCL18 expression with clinicopathologic and survival data was performed (n = 89). Quantitative reverse-transcription polymerase chain reaction was used to verify the microarray expression data and was further applied to analyze an independent cohort of tumor samples (n = 59). The cellular source of CCL18 was determined with immunohistochemistry and in situ hybridization. RESULTS: High CCL18 expression levels were associated with prolonged overall (P = 0.001; hazard ratio, 0.586) and disease-free (P = 0.002; hazard ratio, 0.416) patient survival in the array-based data set by univariate analysis. The observations were confirmed in an independent set of 59 patients by using quantitative reverse-transcription polymerase chain reaction. In multivariate analysis, tumor stage and CCL18 levels were independent prognostic factors for predicting both overall and disease-free survival. We found that CCL18 was expressed by a subpopulation of tumor-associated macrophages that were preferentially located at the tumor invasion front. CONCLUSIONS: Macrophage-derived CCL18 may function as a local antitumor immunomodulator that affects patient outcome. Our study suggests CCL18 as a novel candidate for antitumor therapeutics and risk stratification in gastric cancer patients.


Asunto(s)
Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Quimiocinas CC/genética , Perfilación de la Expresión Génica , Neoplasias Gástricas/diagnóstico , Neoplasias Gástricas/genética , Adenocarcinoma/mortalidad , Algoritmos , Anticuerpos Biespecíficos , Biomarcadores de Tumor/genética , Quimiocinas CC/inmunología , Mucosa Gástrica/fisiología , Regulación Neoplásica de la Expresión Génica/inmunología , Humanos , Macrófagos/fisiología , Pronóstico , Neoplasias Gástricas/mortalidad , Análisis de Supervivencia , Linfocitos T/inmunología
17.
Am J Hum Genet ; 74(5): 1035-42, 2004 May.
Artículo en Inglés | MEDLINE | ID: mdl-15042510

RESUMEN

Hereditary nonpolyposis colorectal cancer (HNPCC) accounts for approximately 2% of all colorectal cancer (CRC) cases and is the most common hereditary CRC syndrome. We have previously reported a high incidence of microsatellite instability (MSI) and germline mismatch repair (MMR) gene mutations in young Hong Kong Chinese with CRC. Ongoing studies at the Hereditary Gastrointestinal Cancer Registry in Hong Kong have revealed a unique germline MSH2 c.1452-1455delAATG mutation that has not been reported in other ethnic groups. Detailed analysis showed that this specific MSH2 mutation constituted 21% of all germline MMR gene mutations and 36% of all MSH2 germline mutations identified. We designed a specific PCR-based diagnostic test on paraffin-embedded tissues and identified this germline mutation in 2 (1.5%) of 138 consecutive patients with early-onset CRC (<46 years of age at diagnosis). Haplotype analysis was performed using 11 microsatellite markers located between D2S391 and D2S123. All 10 families had the same disease haplotype, suggesting a founder effect. These 10 families all originated from the Chinese province of Guangdong, which historically included Hong Kong. It is the most populous of the Chinese provinces, with a population of >93 million. Further analysis suggested that this founder mutation may date back to between 22 and 103 generations ago. The identification of this MSH2 founder mutation has important implications for the design of mutation-detection strategies for the southern Chinese population. Since there were major emigrations from Hong Kong and Guangdong province during the 19th and 20th centuries, this finding is also significant for Chinese communities worldwide.


Asunto(s)
Neoplasias Colorrectales Hereditarias sin Poliposis/genética , Proteínas de Unión al ADN/genética , Efecto Fundador , Mutación de Línea Germinal , Proteínas Proto-Oncogénicas/genética , China/epidemiología , Neoplasias Colorrectales Hereditarias sin Poliposis/etnología , Reparación del ADN , Femenino , Predisposición Genética a la Enfermedad , Pruebas Genéticas , Haplotipos , Humanos , Desequilibrio de Ligamiento , Masculino , Repeticiones de Microsatélite , Persona de Mediana Edad , Proteína 2 Homóloga a MutS , Reacción en Cadena de la Polimerasa
18.
Proc Natl Acad Sci U S A ; 99(25): 16203-8, 2002 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-12456890

RESUMEN

We analyzed gene expression patterns in human gastric cancers by using cDNA microarrays representing approximately equal 30,300 genes. Expression of PLA2G2A, a gene previously implicated as a modifier of the Apc(Min/+) (multiple intestinal neoplasia 1) mutant phenotype in the mouse, was significantly correlated with patient survival. We confirmed this observation in an independent set of patient samples by using quantitative RT-PCR. Beyond its potential diagnostic and prognostic significance, this result suggests the intriguing possibility that the activity of PLA2G2A may suppress progression or metastasis of human gastric cancer.


Asunto(s)
Adenocarcinoma/enzimología , Proteínas de Neoplasias/fisiología , Fosfolipasas A/fisiología , Neoplasias Gástricas/enzimología , Adenocarcinoma/genética , Adenocarcinoma/mortalidad , ADN Complementario/genética , Progresión de la Enfermedad , Inducción Enzimática , Mucosa Gástrica/enzimología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Fosfolipasas A2 Grupo II , Humanos , Hibridación in Situ , Tablas de Vida , Proteínas de Neoplasias/biosíntesis , Proteínas de Neoplasias/genética , Análisis de Secuencia por Matrices de Oligonucleótidos , Fosfolipasas A/biosíntesis , Fosfolipasas A/genética , Fosfolipasas A2 , Pronóstico , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Neoplasias Gástricas/genética , Neoplasias Gástricas/mortalidad , Análisis de Supervivencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA