Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Bases de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
J Pathol ; 242(2): 178-192, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28299802

RESUMEN

The functional role of bone morphogenetic protein (BMP) signalling in colorectal cancer (CRC) is poorly defined, with contradictory results in cancer cell line models reflecting the inherent difficulties of assessing a signalling pathway that is context-dependent and subject to genetic constraints. By assessing the transcriptional response of a diploid human colonic epithelial cell line to BMP ligand stimulation, we generated a prognostic BMP signalling signature, which was applied to multiple CRC datasets to investigate BMP heterogeneity across CRC molecular subtypes. We linked BMP and Notch signalling pathway activity and function in human colonic epithelial cells, and normal and neoplastic tissue. BMP induced Notch through a γ-secretase-independent interaction, regulated by the SMAD proteins. In homeostasis, BMP/Notch co-localization was restricted to cells at the top of the intestinal crypt, with more widespread interaction in some human CRC samples. BMP signalling was downregulated in the majority of CRCs, but was conserved specifically in mesenchymal-subtype tumours, where it interacts with Notch to induce an epithelial-mesenchymal transition (EMT) phenotype. In intestinal homeostasis, BMP-Notch pathway crosstalk is restricted to differentiating cells through stringent pathway segregation. Conserved BMP activity and loss of signalling stringency in mesenchymal-subtype tumours promotes a synergistic BMP-Notch interaction, and this correlates with poor patient prognosis. BMP signalling heterogeneity across CRC subtypes and cell lines can account for previous experimental contradictions. Crosstalk between the BMP and Notch pathways will render mesenchymal-subtype CRC insensitive to γ-secretase inhibition unless BMP activation is concomitantly addressed. © 2017 The Authors. Journal of Pathology published by John Wiley & Sons Ltd on behalf of Pathological Society of Great Britain and Ireland.


Asunto(s)
Proteínas Morfogenéticas Óseas/genética , Neoplasias Colorrectales/genética , Transición Epitelial-Mesenquimal , Receptores Notch/genética , Transducción de Señal , Secretasas de la Proteína Precursora del Amiloide/genética , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Proteínas Morfogenéticas Óseas/metabolismo , Diferenciación Celular , Estudios de Cohortes , Neoplasias Colorrectales/diagnóstico , Neoplasias Colorrectales/patología , Células Epiteliales/patología , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Estimación de Kaplan-Meier , Modelos Biológicos , Fenotipo , Pronóstico , Receptores Notch/metabolismo , Proteínas Smad/genética , Proteínas Smad/metabolismo
2.
Gut ; 63(5): 792-9, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-23676439

RESUMEN

OBJECTIVE: FBXW7 encodes the substrate recognition component of a ubiquitin ligase that degrades targets such as Notch1, c-Jun, c-Myc and cyclin E. FBXW7 mutations occur in several tumour types, including colorectal cancers. The FBXW7 mutation spectrum in cancers is unusual. Some tumours have biallelic loss of function mutations but most have monoallelic missense mutations involving specific arginine residues at ß-propellor tips involved in substrate recognition. DESIGN: FBXW7 functional studies have generally used null systems. In order to analyse the most common mutations in human tumours, we created a Fbxw7(fl(R482Q))(/+) mouse and conditionally expressed this mutation in the intestines using Vill-Cre. We compared these mice with heterozygous null (Fbxw7(+/-)) mutants. RESULTS: A few sizeable intestinal adenomas occurred in approximately 30% of R482Q/+ and Fbxw7(+/-) mice at age >300 days. Breeding the R482Q allele onto Apc mutant backgrounds led to accelerated morbidity and increased polyp numbers and size. Within the small bowel, polyp distribution was shifted proximally. Elevated levels of two particular Fbxw7 substrates, Klf5 and Tgif1, were found in normal intestine and adenomas of R482Q/+, R482Q/R482Q and Fbxw7(-/-) mice, but not Fbxw7(+/-) animals. On the Apc mutant background, Fbxw7(+/-) mutants had a phenotype intermediate between Fbxw7 wild-type and R482Q/+ mice. CONCLUSIONS: Heterozygous Fbxw7 propellor tip (R482Q) mutations promote intestinal tumorigenesis on an Apc mutant background. Klf5 and Tgif1 are strong candidates for mediating this effect. Although heterozygous null Fbxw7 mutations also promote tumour growth, these have a weaker effect than R482Q. These findings explain the FBXW7 mutation spectrum found in human cancers, and emphasise the need for animal models faithfully to reflect human disease.


Asunto(s)
Pólipos Adenomatosos/genética , Biomarcadores de Tumor/genética , Proteínas de Ciclo Celular/genética , Neoplasias Colorrectales/genética , Proteínas F-Box/genética , Pólipos Intestinales/genética , Mutación Missense , Ubiquitina-Proteína Ligasas/genética , Animales , Western Blotting , Línea Celular Tumoral , Proteína 7 que Contiene Repeticiones F-Box-WD , Marcadores Genéticos , Heterocigoto , Humanos , Inmunohistoquímica , Ratones
3.
PLoS Genet ; 7(6): e1002105, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21655089

RESUMEN

Genome-wide association studies (GWAS) have identified 14 tagging single nucleotide polymorphisms (tagSNPs) that are associated with the risk of colorectal cancer (CRC), and several of these tagSNPs are near bone morphogenetic protein (BMP) pathway loci. The penalty of multiple testing implicit in GWAS increases the attraction of complementary approaches for disease gene discovery, including candidate gene- or pathway-based analyses. The strongest candidate loci for additional predisposition SNPs are arguably those already known both to have functional relevance and to be involved in disease risk. To investigate this proposition, we searched for novel CRC susceptibility variants close to the BMP pathway genes GREM1 (15q13.3), BMP4 (14q22.2), and BMP2 (20p12.3) using sample sets totalling 24,910 CRC cases and 26,275 controls. We identified new, independent CRC predisposition SNPs close to BMP4 (rs1957636, P = 3.93×10(-10)) and BMP2 (rs4813802, P = 4.65×10(-11)). Near GREM1, we found using fine-mapping that the previously-identified association between tagSNP rs4779584 and CRC actually resulted from two independent signals represented by rs16969681 (P = 5.33×10(-8)) and rs11632715 (P = 2.30×10(-10)). As low-penetrance predisposition variants become harder to identify-owing to small effect sizes and/or low risk allele frequencies-approaches based on informed candidate gene selection may become increasingly attractive. Our data emphasise that genetic fine-mapping studies can deconvolute associations that have arisen owing to independent correlation of a tagSNP with more than one functional SNP, thus explaining some of the apparently missing heritability of common diseases.


Asunto(s)
Proteína Morfogenética Ósea 2/genética , Proteína Morfogenética Ósea 4/genética , Neoplasias Colorrectales/genética , Predisposición Genética a la Enfermedad , Péptidos y Proteínas de Señalización Intercelular/genética , Anciano , Proteína Morfogenética Ósea 2/metabolismo , Proteína Morfogenética Ósea 4/metabolismo , Estudios de Casos y Controles , Neoplasias Colorrectales/metabolismo , Frecuencia de los Genes , Variación Genética , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Persona de Mediana Edad , Polimorfismo de Nucleótido Simple , Carácter Cuantitativo Heredable , Transducción de Señal
4.
Gut ; 62(1): 83-93, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22287596

RESUMEN

OBJECTIVE: Wnt signalling is critical for normal intestinal development and homeostasis. Wnt dysregulation occurs in almost all human and murine intestinal tumours and an optimal but not excessive level of Wnt activation is considered favourable for tumourigenesis. The authors assessed effects of pan-intestinal Wnt activation on tissue homeostasis, taking into account underlying physiological Wnt activity and stem-cell number in each region of the bowel. DESIGN: The authors generated mice that expressed temporally controlled, stabilised ß-catenin along the crypt-villus axis throughout the intestines. Physiological Wnt target gene activity was assessed in different regions of normal mouse and human tissue. Human intestinal tumour mutation spectra were analysed. RESULTS: In the mouse, ß-catenin stabilisation resulted in a graduated neoplastic response, ranging from dysplastic transformation of the entire epithelium in the proximal small bowel to slightly enlarged crypts of non-dysplastic morphology in the colorectum. In contrast, stem and proliferating cell numbers were increased in all intestinal regions. In the normal mouse and human intestines, stem-cell and Wnt gradients were non-identical, but higher in the small bowel than large bowel in both species. There was also variation in the expression of some Wnt modulators. Human tumour analysis confirmed that different APC mutation spectra are selected in different regions of the bowel. CONCLUSIONS: There are variable gradients in stem-cell number, physiological Wnt activity and response to pathologically increased Wnt signalling along the crypt-villus axis and throughout the length of the intestinal tract. The authors propose that this variation influences regional mutation spectra, tumour susceptibility and lesion distribution in mice and humans.


Asunto(s)
Biomarcadores de Tumor/metabolismo , Mucosa Intestinal/metabolismo , Neoplasias Intestinales/metabolismo , Células Madre/fisiología , Vía de Señalización Wnt/fisiología , Animales , Biomarcadores de Tumor/genética , Recuento de Células , Genes APC , Marcadores Genéticos , Homeostasis , Humanos , Hibridación in Situ , Mucosa Intestinal/citología , Mucosa Intestinal/patología , Neoplasias Intestinales/genética , Neoplasias Intestinales/patología , Intestinos/citología , Intestinos/patología , Ratones , Ratones Transgénicos , Mutación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Wnt/genética , Proteínas Wnt/metabolismo , beta Catenina/metabolismo
5.
J Pathol ; 226(1): 73-83, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22009253

RESUMEN

Adenomatous polyposis coli (APC ) mutations are found in most colorectal tumours. These mutations are almost always protein-truncating, deleting both central domains that regulate Wnt signalling and C-terminal domains that interact with the cytoskeleton. The importance of Wnt dysregulation for colorectal tumourigenesis is well characterized. It is, however, unclear whether loss of C-terminal functions contributes to tumourigenesis, although this protein region has been implicated in cellular processes--including polarity, migration, mitosis, and chromosomal instability (CIN)­that have been postulated as critical for the development and progression of intestinal tumours. Since almost all APC mutations in human patients disrupt both central and C-terminal regions, we created a mouse model to test the role of the C-terminus of APC in intestinal tumourigenesis. This mouse (Apc(ΔSAMP)) carries an internal deletion within Apc that dysregulates Wnt by removing the beta-catenin-binding and SAMP repeats, but leaves the C-terminus intact. We compared Apc(ΔSAMP) mice with Apc(1322T) animals. The latter allele represented the most commonly found human APC mutation and was identical to Apc(ΔSAMP) except for absence of the entire C-terminus. Apc(ΔSAMP) mice developed numerous intestinal adenomas indistinguishable in number, location, and dysplasia from those seen in Apc(1322T) mice. No carcinomas were found in Apc(ΔSAMP) or Apc(1322T) animals. While similar disruption of the Wnt signalling pathway was observed in tumours from both mice, no evidence of differential C-terminus functions (such as cell migration, CIN, or localization of APC and EB1) was seen. We conclude that the C-terminus of APC does not influence intestinal adenoma development or progression.


Asunto(s)
Adenoma/genética , Proteína de la Poliposis Adenomatosa del Colon/genética , Neoplasias Intestinales/genética , Adenoma/patología , Proteína de la Poliposis Adenomatosa del Colon/química , Animales , Western Blotting , Movimiento Celular/genética , Progresión de la Enfermedad , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Hibridación in Situ , Neoplasias Intestinales/patología , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Análisis de Secuencia por Matrices de Oligonucleótidos , Estructura Terciaria de Proteína/genética , Transducción de Señal/genética , Vía de Señalización Wnt
6.
Nat Genet ; 36(12): 1291-5, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15516931

RESUMEN

Imprinted genes are expressed from only one of the parental chromosomes and are marked epigenetically by DNA methylation and histone modifications. The imprinting center 2 (IC2) on mouse distal chromosome 7 is flanked by several paternally repressed genes, with the more distant ones imprinted exclusively in the placenta. We found that most of these genes lack parent-specific DNA methylation, and genetic ablation of methylation does not lead to loss of their imprinting in the trophoblast (placenta). The silent paternal alleles of the genes are marked in the trophoblast by repressive histone modifications (dimethylation at Lys9 of histone H3 and trimethylation at Lys27 of histone H3), which are disrupted when IC2 is deleted, leading to reactivation of the paternal alleles. Thus, repressive histone methylation is recruited by IC2 (potentially through a noncoding antisense RNA) to the paternal chromosome in a region of at least 700 kb and maintains imprinting in this cluster in the placenta, independently of DNA methylation. We propose that an evolutionarily older imprinting mechanism limited to extraembryonic tissues was based on histone modifications, and that this mechanism was subsequently made more stable for use in embryonic lineages by the recruitment of DNA methylation.


Asunto(s)
Cromosomas de los Mamíferos/genética , Epigénesis Genética/genética , Impresión Genómica/genética , Ratones/genética , Modelos Biológicos , Placenta , Animales , Southern Blotting , Cromatina/genética , Islas de CpG/genética , Metilación de ADN , Femenino , Histonas/metabolismo , Inmunoprecipitación , Metilación , Ratones Mutantes , Reacción en Cadena de la Polimerasa , Polimorfismo Conformacional Retorcido-Simple , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Cancers (Basel) ; 15(14)2023 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-37509364

RESUMEN

PTK6, a non-receptor tyrosine kinase, modulates the pathogenesis of breast and prostate cancers and is recognized as a biomarker of breast cancer prognosis. There are over 30 known substrates of PTK6, including signal transducers, transcription factors, and RNA-binding proteins. Many of these substrates are known drivers of other cancer types, such as colorectal cancer. Colon and rectal tumors also express higher levels of PTK6 than the normal intestine suggesting a potential role in tumorigenesis. However, the importance of PTK6 in colorectal cancer remains unclear. PTK6 inhibitors such as XMU-MP-2 and Tilfrinib have demonstrated potency and selectivity in breast cancer cells when used in combination with chemotherapy, indicating the potential for PTK6 targeted therapy in cancer. However, most of these inhibitors are yet to be tested in other cancer types. Here, we discuss the current understanding of the function of PTK6 in normal intestinal cells compared with colorectal cancer cells. We review existing PTK6 targeting therapeutics and explore the possibility of PTK6 inhibitory therapy for colorectal cancer.

8.
EMBO Mol Med ; 15(10): e17094, 2023 Oct 11.
Artículo en Inglés | MEDLINE | ID: mdl-37589076

RESUMEN

High-risk endometrial cancer has poor prognosis and is increasing in incidence. However, understanding of the molecular mechanisms which drive this disease is limited. We used genetically engineered mouse models (GEMM) to determine the functional consequences of missense and loss of function mutations in Fbxw7, Pten and Tp53, which collectively occur in nearly 90% of high-risk endometrial cancers. We show that Trp53 deletion and missense mutation cause different phenotypes, with the latter a substantially stronger driver of endometrial carcinogenesis. We also show that Fbxw7 missense mutation does not cause endometrial neoplasia on its own, but potently accelerates carcinogenesis caused by Pten loss or Trp53 missense mutation. By transcriptomic analysis, we identify LEF1 signalling as upregulated in Fbxw7/FBXW7-mutant mouse and human endometrial cancers, and in human isogenic cell lines carrying FBXW7 mutation, and validate LEF1 and the additional Wnt pathway effector TCF7L2 as novel FBXW7 substrates. Our study provides new insights into the biology of high-risk endometrial cancer and suggests that targeting LEF1 may be worthy of investigation in this treatment-resistant cancer subgroup.


Asunto(s)
Carcinogénesis , Neoplasias Endometriales , Femenino , Humanos , Ratones , Animales , Proteína 7 que Contiene Repeticiones F-Box-WD/genética , Proteína 7 que Contiene Repeticiones F-Box-WD/metabolismo , Carcinogénesis/genética , Neoplasias Endometriales/genética , Neoplasias Endometriales/metabolismo , Mutación , Mutación Missense
9.
J Pathol ; 224(2): 180-9, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21503901

RESUMEN

FBXW7 is the substrate recognition component of a SCF-type E3 ubiquitin ligase. It has multiple targets such as Notch1, c-Jun, and cyclin E that function in critical developmental and signalling pathways. Mutations in FBXW7 are often found in many types of cancer. In most cases, these mutations do not inactivate the protein, but are mono-allelic missense changes at specific arginine resides involved in substrate binding. We have hypothesized that FBXW7 mutations are selected in cancers for reasons other than haploinsufficiency or full loss-of-function. Given that the existing mutant Fbxw7 mice carry null alleles, we created a mouse model carrying one of the commonly occurring point mutations (Fbxw7(R482Q)) in the WD40 substrate recognition domain of Fbxw7. Mice heterozygous for this mutation apparently developed normally in utero, died perinatally due to a defect in lung development, and in some cases showed cleft palate and eyelid fusion defects. By comparison, Fbxw7(+/-) mice were viable and developed normally. Fbxw7(-/-) animals died of vascular abnormalities at E10.5. We screened known FBXW7 targets for changes in the lungs of the Fbxw7(R482Q/+) mice and found Tgif1 and Klf5 to be up-regulated. Fbxw7(R482Q) alleles are not functionally equivalent to heterozygous or homozygous null alleles, and we propose that they are selected in tumourigenesis because they cause a selective or partial loss of FBXW7 function.


Asunto(s)
Proteínas F-Box/genética , Pulmón/anomalías , Neoplasias/genética , Mutación Puntual , Ubiquitina-Proteína Ligasas/genética , Anomalías Múltiples/genética , Alelos , Secuencia de Aminoácidos , Animales , Arginina/análisis , Proteínas de Ciclo Celular/genética , Modelos Animales de Enfermedad , Proteína 7 que Contiene Repeticiones F-Box-WD , Humanos , Pulmón/embriología , Ratones , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Alveolos Pulmonares/patología , Alineación de Secuencia , Factor de Crecimiento Transformador beta/fisiología
10.
Results Probl Cell Differ ; 70: 71-102, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36348105

RESUMEN

The organisation of the genome in its home, the cell nucleus, is reliant on a number of different aspects to establish, maintain and alter its functional non-random positioning. The genome is dispersed throughout a cell nucleus in specific chromosome territories which are further divided into topologically associated domains (TADs), where regions of the genome from different and the same chromosomes come together. This organisation is both controlled by DNA and chromatin epigenetic modification and the association of the genome with nuclear structures such as the nuclear lamina, the nucleolus and nuclear bodies and speckles. Indeed, sequences that are associated with the first two structures mentioned are termed lamina-associated domains (LADs) and nucleolar-associated domains (NADs), respectively. The modifications and nuclear structures that regulate genome function are altered through a cell's life from stem cell to differentiated cell through to reversible quiescence and irreversible senescence, and hence impacting on genome organisation, altering it to silence specific genes and permit others to be expressed in a controlled way in different cell types and cell cycle statuses. The structures and enzymes and thus the organisation of the genome can also be deleteriously affected, leading to disease and/or premature ageing.


Asunto(s)
Núcleo Celular , Genoma , Cromatina/metabolismo , Cromosomas , Células Madre
11.
Gut ; 59(12): 1680-6, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20926645

RESUMEN

BACKGROUND AND AIMS: Adenomatous polyposis coli (APC) is a tumour suppressor gene mutated in the germline of patients with familial adenomatous polyposis (FAP) and somatically in most colorectal cancers. APC mutations impair ß-catenin degradation, resulting in increased Wnt signalling. The most frequent APC mutation is a codon 1309 truncation that is associated with severe FAP. A previous study compared two mouse models of intestinal tumorigenesis, Apc(R850X) (Min) and Apc(1322T) (1322T), the latter a model of human codon 1309 changes. 1322T mice had more severe polyposis but, surprisingly, these tumours had lower levels of nuclear ß-catenin than Min tumours. The consequences of these different ß-catenin levels were investigated. METHODS: Enterocytes were isolated from 1322T and Min tumours by microdissection and gene expression profiling was performed. Differentially expressed Wnt targets and other stem cell markers were validated using quantitative PCR, in situ hybridisation and immunohistochemistry. RESULTS: As expected, lower nuclear ß-catenin levels in 1322T lesions were associated with generally lower levels of Wnt target expression. However, expression of the Wnt target and stem cell marker Lgr5 was significantly higher in 1322T tumours than in Min tumours. Other stem cell markers (Musashi1, Bmi1 and the Wnt target Cd44) were also at higher levels in 1322T tumours. In addition, expression of the Bmp antagonist Gremlin1 was higher in 1322T tumours, together with lower Bmp2 and Bmp4 expression. CONCLUSIONS: The severe phenotype caused by truncation of Apc at codon 1322 is associated with an increased number of stem cells. Thus, a submaximal level of Wnt signalling favours the stem cell phenotype and this may promote tumorigenesis. A level of Wnt signalling exists that is too high for optimal tumour growth.


Asunto(s)
Poliposis Adenomatosa del Colon/metabolismo , Genes APC , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Wnt/metabolismo , Poliposis Adenomatosa del Colon/genética , Animales , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Ratones , Ratones Endogámicos C57BL , Microdisección/métodos , Proteínas de Neoplasias/metabolismo , Transducción de Señal/fisiología , Proteínas Wnt/genética
12.
Antioxidants (Basel) ; 10(9)2021 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-34573012

RESUMEN

Radiation resistance is a significant clinical problem in rectal cancer treatment, the mechanisms of which are poorly understood. NRF2 signalling is known to contribute to chemo/radioresistance in some cancers, but its role in therapeutic resistance in colorectal cancer (CRC) is unexplored. Using siRNA and CRiSPR/Cas9 isogenic CRC cell lines, we investigated the effect of the knockdown and upregulation of the NRF2 pathway on chemo-radiosensitivity. Poly (A) enriched RNA sequencing and geneset enrichment analysis (GSEA) were carried out on both sensitive and resistant cell models for mechanistic insights. Finally, a cohort of rectal patient samples was profiled to understand the clinical relevance of NRF2 signalling. Radioresistant cell lines were significantly radiosensitised by siRNA knockdown (SW1463, SER10 1.22, ANOVA p < 0.0001; HT55, SER10 1.17, ANOVA p < 0.01), but not the (already) radiosensitive HCT116. The constitutive activation of NRF2 via a CRISPR Cas9 NFE2L2 mutation, E79K, induced radioresistance in HCT116 (SER10 0.71, ANOVA, p < 0.0001). GSEA demonstrated significant opposing metabolic dependencies in NRF2 signalling, specifically, the downregulation of amino acid and protein synthesis with low levels of NRF2 and upregulation with over expression. In a clinical cohort of 127 rectal patients, using a validated mRNA signature, higher baseline NRF2 signalling was associated with incomplete responses to radiation higher final neoadjuvant rectal (NAR) score (OR 1.34, 95% C.I. 1.01-1.80, LRT p-value = 0.023), where high NAR indicates poor radiation response and poor long-term prognosis. This is the first demonstration of NRF2-mediated radiation resistance in colorectal cancer. NRF2 appears to regulate crucial metabolic pathways, which could be exploited for therapeutic interventions.

13.
Gastroenterology ; 136(7): 2204-2213.e1-13, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19248780

RESUMEN

BACKGROUND & AIMS: We previously demonstrated that the 2 APC mutations in human colorectal tumors are coselected, because tumorigenesis requires an optimal level of Wnt signaling. We and others subsequently showed that the truncated APC proteins in colorectal tumors usually retain a total of 1-2 beta-catenin binding/degradation repeats (20AARs); very few intestinal tumors have proteins with no 20AARs. The coselection of the "2 hits" at APC makes it difficult to undertake further mechanistic studies in this area in humans. In mice, however, second hits appear to vary with the strain or genetic background used. This suggested the possibility of creating suboptimal Apc genotypes in the mouse. METHODS: We have constructed a mouse, Apc(1322T), with a mutant protein retaining one 20AAR. After repeated backcrossing to the C57BL/6J background, we compared the 1322T animals with the widely used Min mouse in which the mutant Apc protein has zero 20AARs. RESULTS: In both mice, intestinal adenomas showed copy-neutral loss of heterozygosity, making them homozygous for the mutant Apc allele. 1322T animals had markedly more severe polyposis, with earlier-onset, larger, more numerous, and more severely dysplastic adenomas. 1322T tumors also had more marked Paneth cell differentiation and higher frequencies of crypt fission. Somewhat surprisingly, nuclear beta-catenin expression was lower in 1322T than Min tumors. CONCLUSIONS: We propose that the Apc(1322T) mutation produces submaximal beta-catenin levels that promote early tumor growth more effectively than the Apc(Min) mutation.


Asunto(s)
Poliposis Adenomatosa del Colon/genética , Transformación Celular Neoplásica/genética , Mutación/genética , Transducción de Señal/genética , beta Catenina/metabolismo , Poliposis Adenomatosa del Colon/patología , Alelos , Animales , Western Blotting , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Genes APC , Predisposición Genética a la Enfermedad , Pérdida de Heterocigocidad , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos DBA , Modelos Genéticos , Células de Paneth/citología , Células de Paneth/fisiología , Transducción de Señal/fisiología , Especificidad de la Especie , beta Catenina/genética
14.
Cancer Genet ; 248-249: 1-10, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32871287

RESUMEN

We hypothesise that the NRF2 transcription factor would act a biomarker of poor prognosis in colorectal cancer. We derived and validated an mRNA based metagene signature of NRF2 signalling and validated it in 1360 patients from 4 different datasets as an independent biomarker of poor prognosis. This is a novel insight into the molecular signalling of colorectal cancer. BACKGROUND: NRF2 over activity confers poor prognosis in some cancers but its prognostic role in colorectal cancer (CRC) is unknown. As a transcription factor, we hypothesise a signature of NRF2 regulated genes could act as a prognostic biomarker in CRC and reveal novel biological insights. METHODS: Using known NRF2 regulated genes, differentially expressed in CRC, we defined a signature of NRF2 pathway activity using principal component analysis and Cox proportional hazard models and tested it in four independent mRNA datasets, profiled on three different mRNA platforms. RESULTS: 36 genes comprised the final NRF2 signature. 1360 patients were included in the validation. High NRF2 was associated with worse disease free survival (DFS) and/or overall survival (OS) in all datasets: (GSE14333 HR=1.55, 95% C.I 1.2-2.004, p = 0.0008; GSE39582 HR=1.24, 95% C.I 1.086-1.416, p = 0.001; GSE87211 HR=1.431, 95% C.I 1.06-1.93, p = 0.056; MRC FOCUS trial HR=1.14, 95% C.I 1.04-1.26, p = 0.008). In multivariate analyses, NRF2 remained significant when adjusted for stage and adjuvant chemotherapy in stage I-III disease, and BRAF V600E mutation and sidedness in stage IV disease. NRF2 activity was particularly enriched in Consensus Molecular Subtype (CMS) 4. CONCLUSION: For the first time, NRF2 is shown to be a consistent, robust prognostic biomarker across all stages of colorectal cancer with additional clinical value to current known prognostic biomarkers. High NRF2 signalling in CMS 4 further refines the molecular taxonomy of CRC, a new biological insight, suggesting avenues of further study.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Colorrectales/patología , Perfilación de la Expresión Génica , Factor 2 Relacionado con NF-E2/genética , Transcriptoma , Quimioradioterapia/métodos , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/terapia , Humanos , Pronóstico , Tasa de Supervivencia
15.
Clin Epigenetics ; 12(1): 102, 2020 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-32641106

RESUMEN

Both colorectal (CRC, 15%) and endometrial cancers (EC, 30%) exhibit microsatellite instability (MSI) due to MLH1 hypermethylation and silencing. The MLH1 promoter polymorphism, rs1800734 is associated with MSI CRC risk, increased methylation and reduced MLH1 expression. In EC samples, we investigated rs1800734 risk using MSI and MSS cases and controls. We found no evidence that rs1800734 or other MLH1 SNPs were associated with the risk of MSI EC. We found the rs1800734 risk allele had no effect on MLH1 methylation or expression in ECs. We propose that MLH1 hypermethylation occurs by different mechanisms in CRC and EC.


Asunto(s)
Neoplasias Endometriales/genética , Epigenómica/métodos , Homólogo 1 de la Proteína MutL/genética , Polimorfismo de Nucleótido Simple/genética , Alelos , Metilación de ADN , Neoplasias Endometriales/diagnóstico , Femenino , Silenciador del Gen , Humanos , Metaanálisis como Asunto , Inestabilidad de Microsatélites , Regiones Promotoras Genéticas/genética , ARN Mensajero/genética , Medición de Riesgo
16.
CRISPR J ; 2(6): 406-416, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31742432

RESUMEN

CRISPR-Cas9 has quickly become the method of choice for genome editing, with multiple publications describing technical advances and novel applications. It has been widely adopted as a tool for basic research and has significant translational and clinical potential. However, its usage has outpaced the establishment of essential and rigorous controls for unwanted off-target effects, manifested as small mutations, large deletions of target loci, or large-scale chromosomal rearrangements. A common application of CRISPR-Cas9 is as a tool for creating isogenic cell-line models to study the effects of precise mutations, or variants, on disease traits. Here, we describe the effect of standard CRISPR-Cas9 mutagenesis protocols on well characterized cancer cell lines. We demonstrate that commonly used methods for detecting correctly mutated clones fail to uncover large-scale rearrangements. We show that simple cytogenetic methods can be used to identify clones carrying chromosomal abnormalities and large mutations at target loci. These methods are quick and cost-efficient, and we suggest that such controls should be performed prior to publication of studies based on novel CRISPR-Cas9 mutated cancer cell lines.


Asunto(s)
Inestabilidad Cromosómica/genética , Análisis Citogenético/métodos , Edición Génica/métodos , Sistemas CRISPR-Cas/genética , Línea Celular Tumoral , Inestabilidad Cromosómica/fisiología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Reordenamiento Génico/genética , Humanos , Mutagénesis/genética , Mutación , Neoplasias/genética , ARN Guía de Kinetoplastida/genética , Eliminación de Secuencia/genética
17.
Sci Rep ; 9(1): 13463, 2019 09 17.
Artículo en Inglés | MEDLINE | ID: mdl-31530880

RESUMEN

Expression of the mismatch repair gene MutL homolog 1 (MLH1) is silenced in a clinically important subgroup of sporadic colorectal cancers. These cancers exhibit hypermutability with microsatellite instability (MSI) and differ from microsatellite-stable (MSS) colorectal cancers in both prognosis and response to therapies. Loss of MLH1 is usually due to epigenetic silencing with associated promoter methylation; coding somatic mutations rarely occur. Here we use the presence of a colorectal cancer (CRC) risk variant (rs1800734) within the MLH1 promoter to investigate the poorly understood mechanisms of MLH1 promoter methylation and loss of expression. We confirm the association of rs1800734 with MSI+ but not MSS cancer risk in our own data and by meta-analysis. Using sensitive allele-specific detection methods, we demonstrate that MLH1 is the target gene for rs1800734 mediated cancer risk. In normal colon tissue, small allele-specific differences exist only in MLH1 promoter methylation, but not gene expression. In contrast, allele-specific differences in both MLH1 methylation and expression are present in MSI+ cancers. We show that MLH1 transcriptional repression is dependent on DNA methylation and can be reversed by a methylation inhibitor. The rs1800734 allele influences the rate of methylation loss and amount of re-expression. The transcription factor TFAP4 binds to the rs1800734 region but with much weaker binding to the risk than the protective allele. TFAP4 binding is absent on both alleles when promoter methylation is present. Thus we propose that TFAP4 binding shields the protective rs1800734 allele of the MLH1 promoter from BRAF induced DNA methylation more effectively than the risk allele.


Asunto(s)
Neoplasias Colorrectales/genética , Proteínas de Unión al ADN/metabolismo , Homólogo 1 de la Proteína MutL/genética , Polimorfismo de Nucleótido Simple , Regiones Promotoras Genéticas , Factores de Transcripción/metabolismo , Alelos , Estudios de Casos y Controles , Islas de CpG , Metilación de ADN , Proteínas de Unión al ADN/genética , Bases de Datos Factuales , Regulación Neoplásica de la Expresión Génica , Predisposición Genética a la Enfermedad , Humanos , Inestabilidad de Microsatélites , Homólogo 1 de la Proteína MutL/metabolismo , ARN Mensajero/genética , Factores de Transcripción/genética
18.
Curr Biol ; 15(8): R291-4, 2005 Apr 26.
Artículo en Inglés | MEDLINE | ID: mdl-15854893

RESUMEN

The imprinted Dlk1-Gtl2 region of the mammalian genome - which in sheep encompasses the Callipyge locus, known for its unusual mode of inheritance - encodes a number of maternally expressed miRNAs. Five of these miRNAs, hosted by the antisense transcript antiPeg11, have now been shown to target degradation of the paternally expressed Peg11 mRNA by an RNAi-mediated mechanism.


Asunto(s)
Impresión Genómica/genética , Glicoproteínas/genética , Mamíferos/genética , MicroARNs/genética , Proteínas/genética , Interferencia de ARN , ARN Mensajero/metabolismo , Animales , Componentes del Gen , Humanos , Patrón de Herencia/genética , Mamíferos/crecimiento & desarrollo , MicroARNs/metabolismo , Mutación/genética , ARN Largo no Codificante
19.
Curr Biol ; 14(7): R284-6, 2004 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-15062124

RESUMEN

The DNA-binding protein CTCF, which acts as a chromatin 'insulator', regulates imprinting of the mammalian Igf2 and H19 genes in a methylation-sensitive manner. It has now been shown that CTCF is also required for protection against de novo methylation of the differentially methylated domain of H19 in the female germline.


Asunto(s)
Cromatina/metabolismo , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Impresión Genómica/genética , ARN no Traducido/metabolismo , Proteínas Represoras/metabolismo , Factor de Unión a CCCTC , Proteínas de Unión al ADN/genética , Femenino , Humanos , Masculino , Unión Proteica , ARN Largo no Codificante , Proteínas Represoras/genética
20.
Mol Cell Biol ; 24(13): 5650-6, 2004 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15199123

RESUMEN

Igf2 and H19 are reciprocally imprinted genes on mouse distal chromosome 7. They share several regulatory elements, including a differentially methylated region (DMR) upstream of H19 that is paternally methylated throughout development. The cis-acting sequence requirements for targeting DNA methylation to the DMR remain unknown; however, it has been suggested that direct tandem repeats near DMRs could be involved. Previous studies of the imprinted Rasgrf1 locus demonstrate indeed that a direct repeat element adjacent to a DMR is responsible for establishing paternal allele-specific methylation at the DMR and therefore allelic expression of the Rasgrf1 transcript. We identified a prominent and conserved direct tandem repeat 1 kb upstream of the H19 DMR and proposed that it played a similar role in imprinted regulation of H19. To test our hypothesis, we generated mice harboring a 1.7-kb targeted deletion of the direct repeat element and analyzed fetal growth, allelic expression, and methylation within the Igf2-H19 region. Surprisingly the deletion had no effect on imprinting. These results together with deletions of other repeats close to imprinted genes suggest that direct repeats may not be important for the targeting of methylation at the majority of imprinted loci and that the Rasgrf1 locus may be an exception to this rule.


Asunto(s)
Impresión Genómica , Factor II del Crecimiento Similar a la Insulina/genética , ARN no Traducido/genética , Secuencias Repetidas en Tándem/fisiología , Animales , Células Cultivadas , Mapeo Cromosómico , Secuencia Conservada , Metilación de ADN , Embrión de Mamíferos/citología , Desarrollo Embrionario y Fetal/genética , Ratones , Ratones Noqueados , Proteínas/genética , ARN Largo no Codificante , Secuencias Reguladoras de Ácidos Nucleicos , Eliminación de Secuencia , Células Madre , Secuencias Repetidas en Tándem/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA