Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Proc Natl Acad Sci U S A ; 110(25): 10213-8, 2013 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-23733954

RESUMEN

Tumor suppressors known to date impede cancer growth by arresting the cell cycle or promoting apoptosis. Here we show that unphosphorylated human STAT5A functions as a tumor suppressor capable of repressing multiple oncogenes via heterochromatin formation. Unphosphorylated STAT5A binds to heterochromatin protein 1α (HP1α) and stabilizes heterochromatin. Expressing unphosphorylated STAT5A or HP1α inhibits colon cancer growth in mouse xenograft models. Transcriptome profiling shows that expressing an unphosphorylatable STAT5A has similar effects to overexpressing HP1α in global gene expression. Notably, the majority of the genes commonly repressed by unphosphorylated STAT5A and HP1α have been implicated in cancer development. Finally, down-regulation, somatic mutations, and deletions of STAT5 genes are found in certain human cancers. These results suggest that unphosphorylated STAT5A may epigenetically suppress tumor growth by promoting heterochromatin formation.


Asunto(s)
Neoplasias del Colon/metabolismo , Regulación Neoplásica de la Expresión Génica/fisiología , Heterocromatina/metabolismo , Factor de Transcripción STAT5/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Núcleo Celular/metabolismo , Proliferación Celular/efectos de los fármacos , Homólogo de la Proteína Chromobox 5 , Proteínas Cromosómicas no Histona/genética , Proteínas Cromosómicas no Histona/metabolismo , Proteínas Cromosómicas no Histona/farmacología , Neoplasias del Colon/tratamiento farmacológico , Neoplasias del Colon/patología , Bases de Datos Genéticas , Regulación hacia Abajo/genética , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Células HEK293 , Células HeLa , Humanos , Ratones , Fosforilación/fisiología , ARN Interferente Pequeño/genética , Factor de Transcripción STAT5/genética , Factor de Transcripción STAT5/farmacología , Transducción de Señal/fisiología , Transcriptoma , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/farmacología , Ensayos Antitumor por Modelo de Xenoinjerto
2.
Hum Mol Genet ; 22(2): 284-99, 2013 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-23077212

RESUMEN

Birt-Hogg-Dubé syndrome (BHD) is a human cancer disorder caused by mutations in the tumor suppressor gene Folliculin (FLCN) with unknown biological functions. Here, we show that the Drosophila homolog of FLCN, dFLCN (a.k.a. dBHD) localizes to the nucleolus and physically interacts with the 19S proteasomal ATPase, Rpt4, a nucleolar resident and known regulator of rRNA transcription. Downregulation of dFLCN resulted in an increase in nucleolar volume and upregulation of rRNA synthesis, whereas dFLCN overexpression reduced rRNA transcription and counteracted the effects of Rpt4 on rRNA production by preventing the association of Rpt4 with the rDNA locus. We further show that human FLCN exhibited evolutionarily conserved function and that Rpt4 knockdown inhibits the growth of FLCN-deficient human renal cancer cells in mouse xenografts. Our study suggests that FLCN functions as a tumor suppressor by negatively regulating rRNA synthesis.


Asunto(s)
Síndrome de Birt-Hogg-Dubé/genética , Síndrome de Birt-Hogg-Dubé/metabolismo , Proteínas de Drosophila/metabolismo , Regulación de la Expresión Génica , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , ARN Ribosómico/genética , Proteínas Supresoras de Tumor/fisiología , Adenosina Trifosfatasas/metabolismo , Animales , Animales Modificados Genéticamente , Proteínas Portadoras/metabolismo , Línea Celular , Núcleo Celular/metabolismo , ADN Ribosómico/metabolismo , Drosophila/genética , Drosophila/metabolismo , Proteínas de Drosophila/genética , Femenino , Expresión Génica , Técnicas de Silenciamiento del Gen , Humanos , Ratones , Ratones Desnudos , Unión Proteica , Transporte de Proteínas , Proteínas Proto-Oncogénicas/genética , Precursores del ARN/metabolismo , ARN Ribosómico/biosíntesis , Trasplante Heterólogo , Carga Tumoral/genética , Proteínas ras/genética , Proteínas ras/metabolismo
3.
Bioinformatics ; 30(16): 2310-6, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-24764462

RESUMEN

MOTIVATION: Quantitative real-time PCR (qPCR) is one of the most widely used methods to measure gene expression. Despite extensive research in qPCR laboratory protocols, normalization and statistical analysis, little attention has been given to qPCR non-detects-those reactions failing to produce a minimum amount of signal. RESULTS: We show that the common methods of handling qPCR non-detects lead to biased inference. Furthermore, we show that non-detects do not represent data missing completely at random and likely represent missing data occurring not at random. We propose a model of the missing data mechanism and develop a method to directly model non-detects as missing data. Finally, we show that our approach results in a sizeable reduction in bias when estimating both absolute and differential gene expression. AVAILABILITY AND IMPLEMENTATION: The proposed algorithm is implemented in the R package, nondetects. This package also contains the raw data for the three example datasets used in this manuscript. The package is freely available at http://mnmccall.com/software and as part of the Bioconductor project.


Asunto(s)
Perfilación de la Expresión Génica/métodos , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Algoritmos , Animales , Ratones , Modelos Teóricos , Programas Informáticos
4.
Nature ; 453(7198): 1112-6, 2008 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-18500333

RESUMEN

Understanding the molecular underpinnings of cancer is of critical importance to the development of targeted intervention strategies. Identification of such targets, however, is notoriously difficult and unpredictable. Malignant cell transformation requires the cooperation of a few oncogenic mutations that cause substantial reorganization of many cell features and induce complex changes in gene expression patterns. Genes critical to this multifaceted cellular phenotype have therefore only been identified after signalling pathway analysis or on an ad hoc basis. Our observations that cell transformation by cooperating oncogenic lesions depends on synergistic modulation of downstream signalling circuitry suggest that malignant transformation is a highly cooperative process, involving synergy at multiple levels of regulation, including gene expression. Here we show that a large proportion of genes controlled synergistically by loss-of-function p53 and Ras activation are critical to the malignant state of murine and human colon cells. Notably, 14 out of 24 'cooperation response genes' were found to contribute to tumour formation in gene perturbation experiments. In contrast, only 1 in 14 perturbations of the genes responding in a non-synergistic manner had a similar effect. Synergistic control of gene expression by oncogenic mutations thus emerges as an underlying key to malignancy, and provides an attractive rationale for identifying intervention targets in gene networks downstream of oncogenic gain- and loss-of-function mutations.


Asunto(s)
Transformación Celular Neoplásica/genética , Neoplasias del Colon/genética , Mutación/genética , Oncogenes/genética , Animales , Línea Celular , Colon/citología , Colon/patología , Regulación Neoplásica de la Expresión Génica , Genes p53/genética , Genes ras/genética , Genotipo , Humanos , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Fenotipo
5.
Stat Appl Genet Mol Biol ; 10(1)2011 Oct 05.
Artículo en Inglés | MEDLINE | ID: mdl-23089817

RESUMEN

Gene perturbation experiments are commonly used for the reconstruction of gene regulatory networks. Typical experimental methodology imposes persistent changes on the network. The resulting data must therefore be interpreted as a steady state from an altered gene regulatory network, rather than a direct observation of the original network. In this article an implicit modeling methodology is proposed in which the unperturbed network of interest is scored by first modeling the persistent perturbation, then predicting the steady state, which may then be compared to the observed data. This results in a many-to-one inverse problem, so a computational Bayesian approach is used to assess model uncertainty. The methodology is first demonstrated on a number of synthetic networks. It is shown that the Bayesian approach correctly assigns high posterior probability to the network structure and steady state behavior. Further, it is demonstrated that where uncertainty of model features is indicated, the uncertainty may be accurately resolved with further perturbation experiments. The methodology is then applied to the modeling of a gene regulatory network using perturbation data from nine genes which have been shown to respond synergistically to known oncogenic mutations. A hypothetical model emerges which conforms to reported regulatory properties of these genes. Furthermore, the Bayesian methodology is shown to be consistent in the sense that multiple randomized applications of the fitting algorithm converge to an approximately common posterior density on the space of models. Such consistency is generally not feasible for algorithms which report only single models. We conclude that fully Bayesian methods, coupled with models which accurately account for experimental constraints, are a suitable tool for the inference of gene regulatory networks, in terms of accuracy, estimation of model uncertainty, and experimental design.


Asunto(s)
Teorema de Bayes , Biología Computacional/métodos , Redes Reguladoras de Genes , Algoritmos , Animales , Simulación por Computador , Genes Relacionados con las Neoplasias , Genética de Población/métodos , Humanos , Cadenas de Markov , Modelos Genéticos , Método de Montecarlo , Mutación , Neoplasias/genética , Valor Predictivo de las Pruebas , Reproducibilidad de los Resultados
6.
Nat Struct Mol Biol ; 14(3): 215-23, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17310253

RESUMEN

Many features of the cancer cell phenotype emerge as a result of cooperation between multiple oncogenic mutations. Here we show that activated Ras(V12) and loss of p53 function can cooperate to promote cell motility, a feature closely associated with cancer progression to malignancy. Our analysis indicates that Ras(V12) and loss of p53 synergistically induce RhoA activity, revealing a previously unknown role for p53 in tumor suppression. p53 prevents activation of RhoA and thus induction of cell motility by Ras(V12) through a simple signaling circuit, which integrates multiple inputs that converge on RhoA. Our data suggest that p53 suppresses cancer progression to malignancy by modulating the quality of Ras signaling.


Asunto(s)
Movimiento Celular , Neoplasias/metabolismo , Neoplasias/patología , Proteína p53 Supresora de Tumor/metabolismo , Proteínas ras/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Línea Celular Tumoral , Colon/citología , Colon/patología , Regulación hacia Abajo/genética , Activación Enzimática , Proteínas Activadoras de GTPasa/metabolismo , Humanos , Ratones , Modelos Biológicos , Proteínas Mutantes/metabolismo , Fosfotirosina/metabolismo , Transporte de Proteínas , Proteína p53 Supresora de Tumor/deficiencia , Regulación hacia Arriba/genética , Proteína de Unión al GTP rhoA/genética
7.
STAR Protoc ; 3(4): 101737, 2022 12 16.
Artículo en Inglés | MEDLINE | ID: mdl-36181678

RESUMEN

Inference of gene regulatory networks from gene perturbation experiments is the most reliable approach for investigating interdependence between genes. Here, we describe the initial gene perturbations, expression measurements, and preparation steps, followed by network modeling using TopNet. Summarization and visualization of the estimated networks and optional genetic testing of dependencies revealed by the network model are demonstrated. While developed for gene perturbation experiments, TopNet models data in which nodes are both perturbed and measured. For complete details on the use and execution of this protocol, please refer to McMurray et al. (2021).


Asunto(s)
Redes Reguladoras de Genes , Expresión Génica
8.
Cell Rep ; 40(9): 111253, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36044839

RESUMEN

Activating KRAS mutations and functional loss of members of the SWI/SNF complex, including ARID1A, are found together in the primary liver tumor cholangiocarcinoma (CC). How these mutations cooperate to promote CC has not been established. Using murine models of hepatocyte and biliary-specific lineage tracing, we show that Kras and Arid1a mutations drive the formation of CC and tumor precursors from the biliary compartment, which are accelerated by liver inflammation. Using cultured cells, we find that Arid1a loss causes cellular proliferation, escape from cell-cycle control, senescence, and widespread changes in chromatin structure. Notably, we show that the biliary proliferative response elicited by Kras/Arid1a cooperation and tissue injury in CC is caused by failed engagement of the TGF-ß-Smad4 tumor suppressor pathway. We thus identify an ARID1A-TGF-ß-Smad4 axis as essential in limiting the biliary epithelial response to oncogenic insults, while its loss leads to biliary pre-neoplasia and CC.


Asunto(s)
Neoplasias de los Conductos Biliares , Colangiocarcinoma , Animales , Neoplasias de los Conductos Biliares/genética , Neoplasias de los Conductos Biliares/patología , Conductos Biliares Intrahepáticos/metabolismo , Conductos Biliares Intrahepáticos/patología , Colangiocarcinoma/genética , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Humanos , Ratones , Mutación/genética , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
9.
Cell Rep ; 37(12): 110136, 2021 12 21.
Artículo en Inglés | MEDLINE | ID: mdl-34936873

RESUMEN

Malignant cell transformation and the underlying reprogramming of gene expression require the cooperation of multiple oncogenic mutations. This cooperation is reflected in the synergistic regulation of non-mutant downstream genes, so-called cooperation response genes (CRGs). CRGs affect diverse hallmark features of cancer cells and are not known to be functionally connected. However, they act as critical mediators of the cancer phenotype at an unexpectedly high frequency >50%, as indicated by genetic perturbations. Here, we demonstrate that CRGs function within a network of strong genetic interdependencies that are critical to the malignant state. Our network modeling methodology, TopNet, takes the approach of incorporating uncertainty in the underlying gene perturbation data and can identify non-linear gene interactions. In the dense space of gene connectivity, TopNet reveals a sparse topological gene network architecture, effectively pinpointing functionally relevant gene interactions. Thus, among diverse potential applications, TopNet has utility for identification of non-mutant targets for cancer intervention.


Asunto(s)
Epistasis Genética , Perfilación de la Expresión Génica/métodos , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Neoplasias/genética , Oncogenes , Animales , Femenino , Genes p53 , Genes ras , Genotipo , Humanos , Masculino , Ratones , Modelos Genéticos , Mutación
10.
Oncotarget ; 8(2): 3430-3440, 2017 Jan 10.
Artículo en Inglés | MEDLINE | ID: mdl-27966447

RESUMEN

Prostate cancer is the most common form of non-dermatological cancer among US men, with an increasing incidence due to the aging population. Patients diagnosed with clinically localized disease identified as intermediate or high-risk are often treated by radical prostatectomy. Approximately 33% of these patients will suffer recurrence after surgery. Identifying patients likely to experience recurrence after radical prostatectomy would lead to improved clinical outcomes, as these patients could receive adjuvant radiotherapy. Here, we report a new tool for prediction of prostate cancer recurrence based on the expression pattern of a small set of cooperation response genes (CRGs). CRGs are a group of genes downstream of cooperating oncogenic mutations previously identified in a colon cancer model that are critical to the cancer phenotype. We show that systemic dysregulation of CRGs is also found in prostate cancer, including a 4-gene signature (HBEGF, HOXC13, IGFBP2, and SATB1) capable of differentiating recurrent from non-recurrent prostate cancer. To develop a suitable diagnostic tool to predict disease outcomes in individual patients, multiple algorithms and data handling strategies were evaluated on a training set using leave-one-out cross-validation (LOOCV). The best-performing algorithm, when used in combination with a predictive nomogram based on clinical staging, predicted recurrent and non-recurrent disease outcomes in a blinded validation set with 83% accuracy, outperforming previous methods. Disease-free survival times between the cohort of prostate cancers predicted to recur and predicted not to recur differed significantly (p = 1.38x10-6). Therefore, this test allows us to accurately identify prostate cancer patients likely to experience future recurrent disease immediately following removal of the primary tumor.


Asunto(s)
Biomarcadores de Tumor , Neoplasias de la Próstata/genética , Neoplasias de la Próstata/patología , Transcriptoma , Algoritmos , Perfilación de la Expresión Génica , Humanos , Masculino , Recurrencia Local de Neoplasia , Pronóstico , Prostatectomía/métodos , Neoplasias de la Próstata/mortalidad , Neoplasias de la Próstata/cirugía , Curva ROC , Reproducibilidad de los Resultados , Análisis de Supervivencia
11.
Cell Rep ; 17(3): 821-836, 2016 10 11.
Artículo en Inglés | MEDLINE | ID: mdl-27732857

RESUMEN

Metabolic reprogramming is critical to oncogenesis, but the emergence and function of this profound reorganization remain poorly understood. Here we find that cooperating oncogenic mutations drive large-scale metabolic reprogramming, which is both intrinsic to cancer cells and obligatory for the transition to malignancy. This involves synergistic regulation of several genes encoding metabolic enzymes, including the lactate dehydrogenases LDHA and LDHB and mitochondrial glutamic pyruvate transaminase 2 (GPT2). Notably, GPT2 engages activated glycolysis to drive the utilization of glutamine as a carbon source for TCA cycle anaplerosis in colon cancer cells. Our data indicate that the Warburg effect supports oncogenesis via GPT2-mediated coupling of pyruvate production to glutamine catabolism. Although critical to the cancer phenotype, GPT2 activity is dispensable in cells that are not fully transformed, thus pinpointing a metabolic vulnerability specifically associated with cancer cell progression to malignancy.


Asunto(s)
Glutamina/metabolismo , Glucólisis , Neoplasias/metabolismo , Carcinogénesis/metabolismo , Carcinogénesis/patología , Hipoxia de la Célula , Línea Celular Tumoral , Proliferación Celular , Ciclo del Ácido Cítrico , Genes ras , Humanos , Isoenzimas/metabolismo , L-Lactato Deshidrogenasa/metabolismo , Mutación/genética , Neoplasias/patología , Fenotipo , Transaminasas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo
12.
Oncotarget ; 6(17): 14796-813, 2015 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-26142707

RESUMEN

In searching for small-molecule compounds that inhibit proliferation and survival of diffuse large B-cell lymphoma (DLBCL) cells and may, therefore, be exploited as potential therapeutic agents for this disease, we identified the commonly used and well-tolerated antibiotic doxycycline as a strong candidate. Here, we demonstrate that doxycycline inhibits the growth of DLBCL cells both in vitro and in mouse xenograft models. In addition, we show that doxycycline accumulates in DLBCL cells to high concentrations and affects multiple signaling pathways that are crucial for lymphomagenesis. Our data reveal the deneddylating activity of COP-9 signalosome (CSN) as a novel target of doxycycline and suggest that doxycycline may exert its effects in DLBCL cells in part through a CSN5-HSP90 pathway. Consistently, knockdown of CSN5 exhibited similar effects as doxycycline treatment on DLBCL cell survival and HSP90 chaperone function. In addition to DLBCL cells, doxycycline inhibited growth of several other types of non-Hodgkin lymphoma cells in vitro. Together, our results suggest that doxycycline may represent a promising therapeutic agent for DLBCL and other non-Hodgkin lymphomas subtypes.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Doxiciclina/farmacología , Linfoma de Células B Grandes Difuso/tratamiento farmacológico , Complejos Multiproteicos/metabolismo , Péptido Hidrolasas/metabolismo , Carga Tumoral/efectos de los fármacos , Animales , Antibacterianos/farmacología , Western Blotting , Complejo del Señalosoma COP9 , Supervivencia Celular/efectos de los fármacos , Femenino , Proteínas HSP90 de Choque Térmico/genética , Proteínas HSP90 de Choque Térmico/metabolismo , Humanos , Subunidad gamma Común de Receptores de Interleucina/deficiencia , Subunidad gamma Común de Receptores de Interleucina/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Linfoma de Células B Grandes Difuso/genética , Linfoma de Células B Grandes Difuso/metabolismo , Linfoma no Hodgkin/genética , Linfoma no Hodgkin/metabolismo , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Complejos Multiproteicos/genética , Péptido Hidrolasas/genética , Interferencia de ARN , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
13.
Cell Rep ; 7(4): 1143-55, 2014 May 22.
Artículo en Inglés | MEDLINE | ID: mdl-24794439

RESUMEN

Mutations in p53 and RAS potently cooperate in oncogenic transformation, and correspondingly, these genetic alterations frequently coexist in pancreatic ductal adenocarcinoma (PDA) and other human cancers. Previously, we identified a set of genes synergistically activated by combined RAS and p53 mutations as frequent downstream mediators of tumorigenesis. Here, we show that the synergistically activated gene Plac8 is critical for pancreatic cancer growth. Silencing of Plac8 in cell lines suppresses tumor formation by blocking autophagy, a process essential for maintaining metabolic homeostasis in PDA, and genetic inactivation in an engineered mouse model inhibits PDA progression. We show that Plac8 is a critical regulator of the autophagic machinery, localizing to the lysosomal compartment and facilitating lysosome-autophagosome fusion. Plac8 thus provides a mechanistic link between primary oncogenic mutations and the induction of autophagy, a central mechanism of metabolic reprogramming, during PDA progression.


Asunto(s)
Autofagia/genética , Carcinoma Ductal Pancreático/genética , Mutación , Neoplasias Pancreáticas/genética , Proteínas/genética , Animales , Carcinoma Ductal Pancreático/metabolismo , Carcinoma Ductal Pancreático/patología , Diferenciación Celular/genética , Línea Celular Tumoral , Progresión de la Enfermedad , Xenoinjertos , Humanos , Ratones , Ratones Endogámicos NOD , Ratones Desnudos , Ratones SCID , Neoplasias Pancreáticas/metabolismo , Neoplasias Pancreáticas/patología , Regiones Promotoras Genéticas , Proteínas/metabolismo
14.
Cell Rep ; 2(3): 580-90, 2012 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-22981231

RESUMEN

The ABCA1 protein mediates the transfer of cellular cholesterol across the plasma membrane to apolipoprotein A-I. Loss-of-function mutations in the ABCA1 gene induce Tangier disease and familial hypoalphalipoproteinemia, both cardiovascular conditions characterized by abnormally low levels of serum cholesterol, increased cholesterol in macrophages, and subsequent formation of vascular plaque. Increased intracellular cholesterol levels are also frequently found in cancer cells. Here, we demonstrate anticancer activity of ABCA1 efflux function, which is compromised following inhibition of ABCA1 gene expression by oncogenic mutations or cancer-specific ABCA1 loss-of-function mutations. In concert with elevated cholesterol synthesis found in cancer cells, ABCA1 deficiency allows for increased mitochondrial cholesterol, inhibits release of mitochondrial cell death-promoting molecules, and thus facilitates cancer cell survival, suggesting that elevated mitochondrial cholesterol is essential to the cancer phenotype.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/biosíntesis , Colesterol/metabolismo , Regulación Neoplásica de la Expresión Génica , Mitocondrias/metabolismo , Proteínas Supresoras de Tumor/biosíntesis , Transportador 1 de Casete de Unión a ATP , Transportadoras de Casetes de Unión a ATP/genética , Animales , Línea Celular Tumoral , Supervivencia Celular/genética , Colesterol/genética , Humanos , Ratones , Mitocondrias/genética , Mutación , Proteínas Supresoras de Tumor/genética
15.
Cancer Res ; 72(6): 1557-67, 2012 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-22266220

RESUMEN

Intrahepatic cholangiocarcinoma (IHCC) is a primary cancer of the liver with an increasing incidence and poor prognosis. Preclinical studies of the etiology and treatment of this disease are hampered by the relatively small number of available IHCC cell lines or genetically faithful animal models. Here we report the development of a genetically engineered mouse model of IHCC that incorporates two of the most common mutations in human IHCC, activating mutations of Kras (Kras(G12D)) and deletion of p53. Tissue-specific activation of Kras(G12D) alone resulted in the development of invasive IHCC with low penetrance and long latency. Latency was shortened by combining Kras(G12D) activation with heterozygous or homozygous deletion of p53 (mean survival of 56 weeks vs. 19 weeks, respectively), which also resulted in widespread local and distant metastasis. Serial analysis showed that the murine models closely recapitulated the multistage histopathologic progression of the human disease, including the development of stroma-rich tumors and the premalignant biliary lesions, intraductal papillary biliary neoplasms (IPBN), and Von Meyenburg complexes (VMC; also known as biliary hamartomas). These findings establish a new genetically and histopathologically faithful model of IHCC and lend experimental support to the hypothesis that IPBN and VMC are precursors to invasive cancers.


Asunto(s)
Colangiocarcinoma/genética , Modelos Animales de Enfermedad , Neoplasias Hepáticas/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteína p53 Supresora de Tumor/genética , Animales , Neoplasias de los Conductos Biliares , Conductos Biliares Intrahepáticos , Colangiocarcinoma/patología , Progresión de la Enfermedad , Ingeniería Genética , Humanos , Neoplasias Hepáticas/patología , Ratones , Mutación
16.
Cell Stem Cell ; 11(3): 359-72, 2012 Sep 07.
Artículo en Inglés | MEDLINE | ID: mdl-22863534

RESUMEN

Leukemia stem cells (LSCs) represent a biologically distinct subpopulation of myeloid leukemias, with reduced cell cycle activity and increased resistance to therapeutic challenge. To better characterize key properties of LSCs, we employed a strategy based on identification of genes synergistically dysregulated by cooperating oncogenes. We hypothesized that such genes, termed "cooperation response genes" (CRGs), would represent regulators of LSC growth and survival. Using both a primary mouse model and human leukemia specimens, we show that CRGs comprise genes previously undescribed in leukemia pathogenesis in which multiple pathways modulate the biology of LSCs. In addition, our findings demonstrate that the CRG expression profile can be used as a drug discovery tool for identification of compounds that selectively target the LSC population. We conclude that CRG-based analyses provide a powerful means to characterize the basic biology of LSCs as well as to identify improved methods for therapeutic targeting.


Asunto(s)
Leucemia/genética , Leucemia/patología , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Oncogenes/genética , Animales , Benzotiazoles/farmacología , Crisis Blástica/genética , Crisis Blástica/patología , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Endogámicos C57BL , Células Madre Neoplásicas/efectos de los fármacos , Serpinas/metabolismo , Tirfostinos/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA