Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Gut ; 68(11): 2007-2018, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-30954952

RESUMEN

BACKGROUND AND AIMS: Besides well-defined genetic alterations, the dedifferentiation of mature acinar cells is an important prerequisite for pancreatic carcinogenesis. Acinar-specific genes controlling cell homeostasis are extensively downregulated during cancer development; however, the underlying mechanisms are poorly understood. Now, we devised a novel in vitro strategy to determine genome-wide dynamics in the epigenetic landscape in pancreatic carcinogenesis. DESIGN: With our in vitro carcinogenic sequence, we performed global gene expression analysis and ChIP sequencing for the histone modifications H3K4me3, H3K27me3 and H2AK119ub. Followed by a comprehensive bioinformatic approach, we captured gene clusters with extensive epigenetic and transcriptional remodelling. Relevance of Ring1b-catalysed H2AK119ub in acinar cell reprogramming was studied in an inducible Ring1b knockout mouse model. CRISPR/Cas9-mediated Ring1b ablation as well as drug-induced Ring1b inhibition were functionally characterised in pancreatic cancer cells. RESULTS: The epigenome is vigorously modified during pancreatic carcinogenesis, defining cellular identity. Particularly, regulatory acinar cell transcription factors are epigenetically silenced by the Ring1b-catalysed histone modification H2AK119ub in acinar-to-ductal metaplasia and pancreatic cancer cells. Ring1b knockout mice showed greatly impaired acinar cell dedifferentiation and pancreatic tumour formation due to a retained expression of acinar differentiation genes. Depletion or drug-induced inhibition of Ring1b promoted tumour cell reprogramming towards a less aggressive phenotype. CONCLUSIONS: Our data provide substantial evidence that the epigenetic silencing of acinar cell fate genes is a mandatory event in the development and progression of pancreatic cancer. Targeting the epigenetic repressor Ring1b could offer new therapeutic options.


Asunto(s)
Células Acinares/patología , Epigénesis Genética/fisiología , Neoplasias Pancreáticas/etiología , Neoplasias Pancreáticas/patología , Complejo Represivo Polycomb 1/fisiología , Ubiquitina-Proteína Ligasas/fisiología , Animales , Carcinogénesis , Técnicas de Cultivo de Célula , Modelos Animales de Enfermedad , Ratones , Ratones Noqueados
2.
Gut ; 67(1): 146-156, 2018 01.
Artículo en Inglés | MEDLINE | ID: mdl-27646934

RESUMEN

OBJECTIVE: The initial steps of pancreatic regeneration versus carcinogenesis are insufficiently understood. Although a combination of oncogenic Kras and inflammation has been shown to induce malignancy, molecular networks of early carcinogenesis remain poorly defined. DESIGN: We compared early events during inflammation, regeneration and carcinogenesis on histological and transcriptional levels with a high temporal resolution using a well-established mouse model of pancreatitis and of inflammation-accelerated KrasG12D-driven pancreatic ductal adenocarcinoma. Quantitative expression data were analysed and extensively modelled in silico. RESULTS: We defined three distinctive phases-termed inflammation, regeneration and refinement-following induction of moderate acute pancreatitis in wild-type mice. These corresponded to different waves of proliferation of mesenchymal, progenitor-like and acinar cells. Pancreas regeneration required a coordinated transition of proliferation between progenitor-like and acinar cells. In mice harbouring an oncogenic Kras mutation and challenged with pancreatitis, there was an extended inflammatory phase and a parallel, continuous proliferation of mesenchymal, progenitor-like and acinar cells. Analysis of high-resolution transcriptional data from wild-type animals revealed that organ regeneration relied on a complex interaction of a gene network that normally governs acinar cell homeostasis, exocrine specification and intercellular signalling. In mice with oncogenic Kras, a specific carcinogenic signature was found, which was preserved in full-blown mouse pancreas cancer. CONCLUSIONS: These data define a transcriptional signature of early pancreatic carcinogenesis and a molecular network driving formation of preneoplastic lesions, which allows for more targeted biomarker development in order to detect cancer earlier in patients with pancreatitis.


Asunto(s)
Carcinogénesis/genética , Carcinoma Ductal Pancreático/genética , Neoplasias Pancreáticas/genética , Células Acinares/patología , Enfermedad Aguda , Animales , Carcinogénesis/patología , Carcinoma Ductal Pancreático/patología , Proliferación Celular/genética , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Células Madre Mesenquimatosas/patología , Ratones Transgénicos , Páncreas/fisiología , Neoplasias Pancreáticas/patología , Pancreatitis/genética , Pancreatitis/patología , Lesiones Precancerosas/genética , Lesiones Precancerosas/patología , Proteínas Proto-Oncogénicas p21(ras)/genética , Regeneración/genética
3.
Gut ; 65(4): 647-57, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25601637

RESUMEN

OBJECTIVE: Oncogenic Kras-activated robust Mek/Erk signals phosphorylate to the tuberous sclerosis complex (Tsc) and deactivates mammalian target of rapamycin (mTOR) suppression in pancreatic ductal adenocarcinoma (PDAC); however, Mek and mTOR inhibitors alone have demonstrated minimal clinical antitumor activity. DESIGN: We generated transgenic mouse models in which mTOR was hyperactivated either through the Kras/Mek/Erk cascade, by loss of Pten or through Tsc1 haploinsufficiency. Primary cancer cells were isolated from mouse tumours. Oncogenic signalling was assessed in vitro and in vivo, with and without single or multiple targeted molecule inhibition. Transcriptional profiling was used to identify biomarkers predictive of the underlying pathway alterations and of therapeutic response. Results from the preclinical models were confirmed on human material. RESULTS: Reduction of Tsc1 function facilitated activation of Kras/Mek/Erk-mediated mTOR signalling, which promoted the development of metastatic PDACs. Single inhibition of mTOR or Mek elicited strong feedback activation of Erk or Akt, respectively. Only dual inhibition of Mek and PI3K reduced mTOR activity and effectively induced cancer cell apoptosis. Analysis of downstream targets demonstrated that oncogenic activity of the Mek/Erk/Tsc/mTOR axis relied on Aldh1a3 function. Moreover, in clinical PDAC samples, ALDH1A3 specifically labelled an aggressive subtype. CONCLUSIONS: These results advance our understanding of Mek/Erk-driven mTOR activation and its downstream targets in PDAC, and provide a mechanistic rationale for effective therapeutic matching for Aldh1a3-positive PDACs.


Asunto(s)
Adenocarcinoma/patología , Carcinogénesis/patología , Carcinoma Ductal Pancreático/patología , Sistema de Señalización de MAP Quinasas/fisiología , Neoplasias Pancreáticas/patología , Proteínas Proto-Oncogénicas p21(ras)/fisiología , Serina-Treonina Quinasas TOR/fisiología , Adenocarcinoma/metabolismo , Animales , Biomarcadores de Tumor/análisis , Carcinogénesis/metabolismo , Carcinoma Ductal Pancreático/metabolismo , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Neoplasias Pancreáticas/metabolismo , Fosfatidilinositol 3-Quinasas , Fosforilación , Transducción de Señal , Neoplasias Pancreáticas
4.
Mol Cancer ; 14: 94, 2015 Apr 25.
Artículo en Inglés | MEDLINE | ID: mdl-25910082

RESUMEN

BACKGROUND: Previous studies identified microRNAs (miRNAs) and messenger RNAs with significantly different expression between normal pancreas and pancreatic cancer (PDAC) tissues. Due to technological limitations of microarrays and real-time PCR systems these studies focused on a fixed set of targets. Expression of other RNA classes such as long intergenic non-coding RNAs or sno-derived RNAs has rarely been examined in pancreatic cancer. Here, we analysed the coding and non-coding transcriptome of six PDAC and five control tissues using next-generation sequencing. RESULTS: Besides the confirmation of several deregulated mRNAs and miRNAs, miRNAs without previous implication in PDAC were detected: miR-802, miR-2114 or miR-561. SnoRNA-derived RNAs (e.g. sno-HBII-296B) and piR-017061, a piwi-interacting RNA, were found to be differentially expressed between PDAC and control tissues. In silico target analysis of miR-802 revealed potential binding sites in the 3' UTR of TCF4, encoding a transcription factor that controls Wnt signalling genes. Overexpression of miR-802 in MiaPaCa pancreatic cancer cells reduced TCF4 protein levels. Using Massive Analysis of cDNA Ends (MACE) we identified differential expression of 43 lincRNAs, long intergenic non-coding RNAs, e.g. LINC00261 and LINC00152 as well as several natural antisense transcripts like HNF1A-AS1 and AFAP1-AS1. Differential expression was confirmed by qPCR on the mRNA/miRNA/lincRNA level and by immunohistochemistry on the protein level. CONCLUSIONS: Here, we report a novel lncRNA, sncRNA and mRNA signature of PDAC. In silico prediction of ncRNA targets allowed for assigning potential functions to differentially regulated RNAs.


Asunto(s)
Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , MicroARNs/genética , Neoplasias Pancreáticas/genética , ARN Largo no Codificante/genética , ARN Interferente Pequeño/genética , Células Acinares/metabolismo , Células Acinares/patología , Secuencia de Bases , Estudios de Casos y Controles , Simulación por Computador , Regulación hacia Abajo/genética , Redes Reguladoras de Genes , Humanos , Inmunohistoquímica , MicroARNs/metabolismo , Datos de Secuencia Molecular , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Sistemas de Lectura Abierta/genética , ARN Largo no Codificante/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Reproducibilidad de los Resultados , Regulación hacia Arriba/genética
5.
Adv Exp Med Biol ; 736: 247-60, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22161333

RESUMEN

The process of differentiation of embryonic stem cells (ESCs) is currently becoming the focus of many systems biologists not only due to mechanistic interest but also since it is expected to play an increasingly important role in regenerative medicine, in particular with the advert to induced pluripotent stem cells. These ESCs give rise to the formation of the three germ layers and therefore to the formation of all tissues and organs. Here, we present a computational method for inferring regulatory interactions between the genes involved in ESC differentiation based on time resolved microarray profiles. Fully quantitative methods are commonly unavailable on such large-scale data; on the other hand, purely qualitative methods may fail to capture some of the more detailed regulations. Our method combines the beneficial aspects of qualitative and quantitative (ODE-based) modeling approaches searching for quantitative interaction coefficients in a discrete and qualitative state space. We further optimize on an ensemble of networks to detect essential properties and compare networks with respect to robustness. Applied to a toy model our method is able to reconstruct the original network and outperforms an entire discrete boolean approach. In particular, we show that including prior knowledge leads to more accurate results. Applied to data from differentiating mouse ESCs reveals new regulatory interactions, in particular we confirm the activation of Foxh1 through Oct4, mediating Nodal signaling.


Asunto(s)
Diferenciación Celular/genética , Células Madre Embrionarias/metabolismo , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Modelos Genéticos , Algoritmos , Animales , Blastocisto/metabolismo , Biología Computacional/métodos , Factores de Transcripción Forkhead/genética , Gastrulación/genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Proteína Nodal/genética , Factor 3 de Transcripción de Unión a Octámeros/genética , Reproducibilidad de los Resultados , Biología de Sistemas/métodos , Factores de Tiempo
6.
Leukemia ; 35(7): 2002-2016, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33953289

RESUMEN

B cells have the unique property to somatically alter their immunoglobulin (IG) genes by V(D)J recombination, somatic hypermutation (SHM) and class-switch recombination (CSR). Aberrant targeting of these mechanisms is implicated in lymphomagenesis, but the mutational processes are poorly understood. By performing whole genome and transcriptome sequencing of 181 germinal center derived B-cell lymphomas (gcBCL) we identified distinct mutational signatures linked to SHM and CSR. We show that not only SHM, but presumably also CSR causes off-target mutations in non-IG genes. Kataegis clusters with high mutational density mainly affected early replicating regions and were enriched for SHM- and CSR-mediated off-target mutations. Moreover, they often co-occurred in loci physically interacting in the nucleus, suggesting that mutation hotspots promote increased mutation targeting of spatially co-localized loci (termed hypermutation by proxy). Only around 1% of somatic small variants were in protein coding sequences, but in about half of the driver genes, a contribution of B-cell specific mutational processes to their mutations was found. The B-cell-specific mutational processes contribute to both lymphoma initiation and intratumoral heterogeneity. Overall, we demonstrate that mutational processes involved in the development of gcBCL are more complex than previously appreciated, and that B cell-specific mutational processes contribute via diverse mechanisms to lymphomagenesis.


Asunto(s)
Genoma/genética , Centro Germinal/metabolismo , Linfoma de Células B/genética , Mutación/genética , Adulto , Linfocitos B/metabolismo , Línea Celular , Línea Celular Tumoral , Genes de Inmunoglobulinas/genética , Células HeLa , Células Hep G2 , Células Endoteliales de la Vena Umbilical Humana , Humanos , Cambio de Clase de Inmunoglobulina/genética , Células K562 , Células MCF-7 , Hipermutación Somática de Inmunoglobulina/genética , Recombinación V(D)J/genética
8.
Cell Mol Gastroenterol Hepatol ; 6(4): 429-449, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-30258965

RESUMEN

Background & Aims: Although nearly half of pancreatic ductal adenocarcinoma (PDAC) patients have diabetes mellitus with episodes of hyperglycemia, its tumor microenvironment is hypoglycemic. Thus, it is crucial for PDAC cells to develop adaptive mechanisms dealing with oscillating glucose levels. So far, the biological impact of such glycemic variability on PDAC biology remains unknown. Methods: Murine PDAC cells were cultured in low- and high-glucose medium to investigate the molecular, biochemical, and metabolic influence of glycemic variability on tumor behavior. A set of in vivo functional assays including orthotopic implantation and portal and tail vein injection were used. Results were further confirmed on tissues from PDAC patients. Results: Glycemic variability has no significant effect on PDAC cell proliferation. Hypoglycemia is associated with local invasion and angiogenesis, whereas hyperglycemia promotes metastatic colonization. Increased metastatic colonization under hyperglycemia is due to increased expression of runt related transcription factor 3 (Runx3), which further activates expression of collagen, type VI, alpha 1 (Col6a1), forming a glycemic pro-metastatic pathway. Through epigenetic machinery, retinoic acid receptor beta (Rarb) expression fluctuates according to glycemic variability, acting as a critical sensor relaying the glycemic signal to Runx3/Col6a1. Moreover, the signal axis of Rarb/Runx3/Col6a1 is pharmaceutically accessible to a widely used antidiabetic substance, metformin, and Rar modulator. Finally, PDAC tissues from patients with diabetes show an increased expression of COL6A1. Conclusions: Glycemic variability promotes both local invasion and metastatic colonization of PDAC. A pro-metastatic signal axis Rarb/Runx3/Col6a1 whose activity is controlled by glycemic variability is identified. The therapeutic relevance of this pathway needs to be explored in PDAC patients, especially in those with diabetes.


Asunto(s)
Carcinoma Ductal Pancreático/patología , Hiperglucemia/patología , Hipoglucemia/patología , Neoplasias Pancreáticas/patología , Animales , Carcinoma Ductal Pancreático/irrigación sanguínea , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Colágeno Tipo VI/metabolismo , Subunidad alfa 3 del Factor de Unión al Sitio Principal/metabolismo , Metilación de ADN/efectos de los fármacos , Metilación de ADN/genética , Diabetes Mellitus/patología , Epigénesis Genética/efectos de los fármacos , Ontología de Genes , Histonas/metabolismo , Humanos , Metformina/farmacología , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Neovascularización Patológica/patología , Neoplasias Pancreáticas/irrigación sanguínea , Regiones Promotoras Genéticas/genética , Receptores de Ácido Retinoico/metabolismo , Neoplasias Pancreáticas
9.
Nat Commun ; 7: 11807, 2016 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-27339797

RESUMEN

The circadian clock and the cell cycle are major cellular systems that organize global physiology in temporal fashion. It seems conceivable that the potentially conflicting programs are coordinated. We show here that overexpression of MYC in U2OS cells attenuates the clock and conversely promotes cell proliferation while downregulation of MYC strengthens the clock and reduces proliferation. Inhibition of the circadian clock is crucially dependent on the formation of repressive complexes of MYC with MIZ1 and subsequent downregulation of the core clock genes BMAL1 (ARNTL), CLOCK and NPAS2. We show furthermore that BMAL1 expression levels correlate inversely with MYC levels in 102 human lymphomas. Our data suggest that MYC acts as a master coordinator that inversely modulates the impact of cell cycle and circadian clock on gene expression.


Asunto(s)
Ciclo Celular/fisiología , Proliferación Celular/fisiología , Relojes Circadianos/fisiología , Regulación de la Expresión Génica/fisiología , Factores de Transcripción de Tipo Kruppel/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Línea Celular , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Linfoma/metabolismo , Osteosarcoma/metabolismo , Proteínas Proto-Oncogénicas c-myc/genética
10.
Int J Surg ; 14: 67-74, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25599917

RESUMEN

INTRODUCTION: Surgical site infections (SSI) represent a significant cause of morbidity in abdominal surgery. The objective of this study was to determine the gene expression signature in subcutaneous tissues in relation to SSI. METHODS: To determine differences in gene expression, microarray analysis were performed from bulk tissue mRNA of subcutaneous tissues prospectively collected in 92 patients during open abdominal surgery. 10 patients (11%) developed incisional (superficial and deep) SSI. RESULTS: Preoperative risk factors in patients with SSI were not significantly different from those in patients without wound infections. 1025 genes were differentially expressed between the groups, of which the AZGP1 and ALDH1A3 genes were the highest down- and upregulated ones. Hierarchical clustering demonstrated strong similarity within the respective groups (SSI vs. no-SSI) indicating inter-group distinctness. In a functional classification, genes controlling cell metabolism were mostly down-regulated in subcutaneous tissues of patients that subsequently developed SSI. CONCLUSION: Altered expression of metabolism genes in subcutaneous tissues might constitute a risk factor for postoperative abdominal SSI.


Asunto(s)
Infección de la Herida Quirúrgica/genética , Transcriptoma , Abdomen , Adipoquinas , Adulto , Anciano , Anciano de 80 o más Años , Aldehído Oxidorreductasas/metabolismo , Aminohidrolasas/metabolismo , Proteínas Portadoras/metabolismo , Procedimientos Quirúrgicos del Sistema Digestivo , Femenino , Formiato-Tetrahidrofolato Ligasa/metabolismo , Glicoproteínas/metabolismo , Humanos , Inmunohistoquímica , Masculino , Metilenotetrahidrofolato Deshidrogenasa (NADP)/metabolismo , Persona de Mediana Edad , Complejos Multienzimáticos/metabolismo , Periodo Posoperatorio , Tejido Subcutáneo/metabolismo , Infección de la Herida Quirúrgica/metabolismo , Análisis de Matrices Tisulares , Adulto Joven
11.
Neoplasia ; 17(2): 155-66, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25748234

RESUMEN

Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal cancers principally because of early invasion and metastasis. The epidermal growth factor receptor (EGFR) is essential for PDAC development even in the presence of Kras, but its inhibition with erlotinib gives only a modest clinical response, making the discovery of novel EGFR targets of critical interest. Here, we revealed by mining a human pancreatic gene expression database that the metastasis promoter Na(+)/H(+) exchanger (NHE1) associates with the EGFR in PDAC. In human PDAC cell lines, we confirmed that NHE1 drives both basal and EGF-stimulated three-dimensional growth and early invasion via invadopodial extracellular matrix digestion. EGF promoted the complexing of EGFR with NHE1 via the scaffolding protein Na+/H+ exchanger regulatory factor 1, engaging EGFR in a negative transregulatory loop that controls the extent and duration of EGFR oncogenic signaling and stimulates NHE1. The specificity of NHE1 for growth or invasion depends on the segregation of the transient EGFR/Na+/H+ exchanger regulatory factor 1/NHE1 signaling complex into dimeric subcomplexes in different lipid raftlike membrane domains. This signaling complex was also found in tumors developed in orthotopic mice. Importantly, the specific NHE1 inhibitor cariporide reduced both three-dimensional growth and invasion independently of PDAC subtype and synergistically sensitized these behaviors to low doses of erlotinib.


Asunto(s)
Carcinoma Ductal Pancreático/secundario , Proteínas de Transporte de Catión/metabolismo , Receptores ErbB/metabolismo , Neoplasias Pancreáticas/patología , Intercambiadores de Sodio-Hidrógeno/metabolismo , Animales , Antiarrítmicos/uso terapéutico , Western Blotting , Carcinoma Ductal Pancreático/tratamiento farmacológico , Carcinoma Ductal Pancreático/metabolismo , Línea Celular , Línea Celular Tumoral , Quimioterapia Combinada , Clorhidrato de Erlotinib , Guanidinas/uso terapéutico , Humanos , Ratones , Ratones Desnudos , Ratones SCID , Neoplasias Pancreáticas/tratamiento farmacológico , Neoplasias Pancreáticas/metabolismo , Inhibidores de Proteínas Quinasas/uso terapéutico , Quinazolinas/uso terapéutico , Transducción de Señal , Intercambiador 1 de Sodio-Hidrógeno , Sulfonas/uso terapéutico
12.
World J Gastrointest Oncol ; 5(1): 1-3, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23355925

RESUMEN

Individualized cancer treatment (e.g. targeted therapy) based on molecular alterations has emerged as an important strategy to improve the current standard-of-care chemotherapy. A large number of studies have demonstrated the importance of biomarkers not only in predicting prognosis but more importantly in predicting the response towards therapies. For example, amplification or mutation status of the two biomarkers HER2 (human epidermal growth factor 2) and BRCA (breast cancer) can be used to decide on a specific targeted therapy in breast cancer. However, no biomarkers with a similar clinical impact have been identified in pancreatic ductal adenocarcinoma. Although many genome-wide and proteome-based high-throughput studies have identified candidate genes or proteins as promising biomarkers, none of them were eventually transferred into the clinical setting. Notably, the most reliable markers for predicting prognosis are still the tumor stage and grade and biomarkers for therapy response remain undefined. One reason lies in the lack of systemic approaches to analyze the complexity of dominating cancer pathways and the impact of such signal complexity on prognosis and therapy response.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA