Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 124
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 169(1): 3-5, 2017 03 23.
Artículo en Inglés | MEDLINE | ID: mdl-28340347

RESUMEN

In this issue of Cell, Baar et al. show how FOXO4 protects senescent cell viability by keeping p53 sequestered in nuclear bodies, preventing it from inducing apoptosis. Disrupting this interaction with an all-D amino acid peptide (FOXO4-DRI) restores p53's apoptotic role and ameliorates the consequences of senescence-associated loss of tissue homeostasis.


Asunto(s)
Apoptosis , Rejuvenecimiento , Supervivencia Celular , Senescencia Celular , Humanos , Proteína p53 Supresora de Tumor/química
2.
Cell ; 161(7): 1494-6, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26091030

RESUMEN

In mouse intestinal tumors induced by the inhibition of APC, the restoration of APC function causes complete tumor regression with normal differentiation and return of stem cell function irrespective of whether tumors also carried mutations in Kras and p53. These findings by Dow et al. validate the Wnt pathway as an exquisite target for intervention.


Asunto(s)
Proteína de la Poliposis Adenomatosa del Colon/metabolismo , Neoplasias Colorrectales/genética , Modelos Animales de Enfermedad , Intestino Grueso/patología , Intestino Delgado/patología , Animales
3.
Genes Dev ; 35(21-22): 1403-1430, 2021 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-34725129

RESUMEN

Chromatin is highly dynamic, undergoing continuous global changes in its structure and type of histone and DNA modifications governed by processes such as transcription, repair, replication, and recombination. Members of the chromodomain helicase DNA-binding (CHD) family of enzymes are ATP-dependent chromatin remodelers that are intimately involved in the regulation of chromatin dynamics, altering nucleosomal structure and DNA accessibility. Genetic studies in yeast, fruit flies, zebrafish, and mice underscore essential roles of CHD enzymes in regulating cellular fate and identity, as well as proper embryonic development. With the advent of next-generation sequencing, evidence is emerging that these enzymes are subjected to frequent DNA copy number alterations or mutations and show aberrant expression in malignancies and other human diseases. As such, they might prove to be valuable biomarkers or targets for therapeutic intervention.


Asunto(s)
Cromatina , Proteínas de Unión al ADN , Animales , Ensamble y Desensamble de Cromatina/genética , ADN Helicasas/genética , ADN Helicasas/metabolismo , Proteínas de Unión al ADN/metabolismo
4.
Cell ; 154(4): 914-27, 2013 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-23953119

RESUMEN

Reporter genes integrated into the genome are a powerful tool to reveal effects of regulatory elements and local chromatin context on gene expression. However, so far such reporter assays have been of low throughput. Here, we describe a multiplexing approach for the parallel monitoring of transcriptional activity of thousands of randomly integrated reporters. More than 27,000 distinct reporter integrations in mouse embryonic stem cells, obtained with two different promoters, show ∼1,000-fold variation in expression levels. Data analysis indicates that lamina-associated domains act as attenuators of transcription, likely by reducing access of transcription factors to binding sites. Furthermore, chromatin compaction is predictive of reporter activity. We also found evidence for crosstalk between neighboring genes and estimate that enhancers can influence gene expression on average over ∼20 kb. The multiplexed reporter assay is highly flexible in design and can be modified to query a wide range of aspects of gene regulation.


Asunto(s)
Efectos de la Posición Cromosómica , Técnicas Genéticas , Animales , Cromatina/metabolismo , Células Madre Embrionarias/metabolismo , Genes Reporteros , Secuenciación de Nucleótidos de Alto Rendimiento , Ratones , Regiones Promotoras Genéticas
5.
Genes Dev ; 34(15-16): 1017-1032, 2020 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-32747478

RESUMEN

As one of the most common forms of cancer, lung cancers present as a collection of different histological subtypes. These subtypes are characterized by distinct sets of driver mutations and phenotypic appearance, and they often show varying degrees of heterogenicity, aggressiveness, and response/resistance to therapy. Intriguingly, lung cancers are also capable of showing features of multiple subtypes or converting from one subtype to another. The intertumoral and intratumoral heterogeneity of lung cancers as well as incidences of subtype transdifferentiation raise the question of to what extent the tumor characteristics are dictated by the cell of origin rather than the acquired driver lesions. We provide here an overview of the studies in experimental mouse models that try to address this question. These studies convincingly show that both the cell of origin and the genetic driver lesions play a critical role in shaping the phenotypes of lung tumors. However, they also illustrate that there is far from a direct one-to-one relationship between the cell of origin and the cancer subtype, as most epithelial cells can be reprogrammed toward diverse lung cancer fates when exposed to the appropriate set of driver mutations.


Asunto(s)
Neoplasias Pulmonares/etiología , Adenocarcinoma/etiología , Animales , Carcinoma de Células Escamosas/etiología , Modelos Animales de Enfermedad , Células Epiteliales , Neoplasias Pulmonares/genética , Ratones , Mucosa Respiratoria/citología , Carcinoma Pulmonar de Células Pequeñas/etiología
6.
Cell ; 141(1): 18-20, 2010 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-20371339

RESUMEN

Resistance to anticancer drugs is widely observed in vitro and in cancer patients, but its prevalence is too high to be solely explained by the acquisition of mutations. Sharma et al. (2010) now report that dynamic chromatin modifications may be an independent route to drug resistance in cancer cells that can be reversed by epigenetic drugs.

7.
Cell ; 133(1): 29-30, 2008 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-18394985

RESUMEN

Overexpression of Brf1, a transcription factor of the RNA polymerase III apparatus, can transform cells in vitro and cause tumor formation in vivo. Marshall et al. (2008) now show that one of the transcriptional products of RNA polymerase III, the initiator tRNA(Met), mediates this effect, revealing an unexpected role for this tRNA in tumorigenesis.


Asunto(s)
Transformación Celular Neoplásica , ARN de Transferencia de Metionina/metabolismo , Animales , Línea Celular , Humanos , Modelos Biológicos , ARN Polimerasa III/metabolismo , ARN de Transferencia de Metionina/genética , Factor de Transcripción TFIIIB/metabolismo , Transcripción Genética
8.
Cell ; 133(4): 727-41, 2008 May 16.
Artículo en Inglés | MEDLINE | ID: mdl-18485879

RESUMEN

p53 and p19(ARF) are tumor suppressors frequently mutated in human tumors. In a high-throughput screen in mice for mutations collaborating with either p53 or p19(ARF) deficiency, we identified 10,806 retroviral insertion sites, implicating over 300 loci in tumorigenesis. This dataset reveals 20 genes that are specifically mutated in either p19(ARF)-deficient, p53-deficient or wild-type mice (including Flt3, mmu-mir-106a-363, Smg6, and Ccnd3), as well as networks of significant collaborative and mutually exclusive interactions between cancer genes. Furthermore, we found candidate tumor suppressor genes, as well as distinct clusters of insertions within genes like Flt3 and Notch1 that induce mutants with different spectra of genetic interactions. Cross species comparative analysis with aCGH data of human cancer cell lines revealed known and candidate oncogenes (Mmp13, Slamf6, and Rreb1) and tumor suppressors (Wwox and Arfrp2). This dataset should prove to be a rich resource for the study of genetic interactions that underlie tumorigenesis.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Redes Reguladoras de Genes , Genes Supresores de Tumor , Neoplasias/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular Tumoral , Clonación Molecular , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Genes p53 , Genómica/métodos , Humanos , Ratones , Ratones Noqueados , Mutagénesis Insercional , Neoplasias/metabolismo , Análisis de Secuencia de ADN
9.
Genes Dev ; 29(14): 1447-62, 2015 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-26220992

RESUMEN

Lung cancer is the leading cause of cancer deaths, with small cell lung cancer (SCLC) representing the most aggressive subtype. Standard treatments have not changed in decades, and the 5-year survival rate has remained <7%. Genomic analyses have identified key driver mutations of SCLC that were subsequently validated in animal models of SCLC. To provide better treatment options, a deeper understanding of the cellular and molecular mechanisms underlying SCLC initiation, progression, metastasis, and acquisition of resistance is required. In this review, we describe the genetic landscape of SCLC, features of the cell of origin, and targeted therapeutic approaches.


Asunto(s)
Neoplasias Pulmonares/genética , Neoplasias Pulmonares/terapia , Carcinoma Pulmonar de Células Pequeñas/genética , Carcinoma Pulmonar de Células Pequeñas/terapia , Animales , Antineoplásicos/uso terapéutico , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Humanos , Neoplasias Pulmonares/patología , Mutación , Carcinoma Pulmonar de Células Pequeñas/mortalidad , Carcinoma Pulmonar de Células Pequeñas/patología
10.
Genes Dev ; 29(15): 1587-92, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26215568

RESUMEN

Tumor heterogeneity can create a unique symbiotic tumor microenvironment. Earlier, we showed that clonal evolution in mouse small cell lung cancer (SCLC) can result in subclones that, upon cografting, endow the neuroendocrine tumor cells with metastatic potential. We now show that paracrine signaling between SCLC subclones is a critical requirement in the early steps of the metastatic process, such as local invasion and intravasation. We further show evidence that paracrine signaling via fibroblast growth factor 2 (Fgf2) and Mapk between these diverged tumor subclones causes enhanced expression of the Pea3 (polyomavirus enhancer activator 3) transcription factor, resulting in metastatic dissemination of the neuroendocrine tumor subclones. Our data reveal for the first time paracrine signaling between tumor cell subclones in SCLC that results in metastatic spread of SCLC.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Metástasis de la Neoplasia/fisiopatología , Comunicación Paracrina/fisiología , Carcinoma Pulmonar de Células Pequeñas/fisiopatología , Factores de Transcripción/metabolismo , Animales , Línea Celular Tumoral , Medios de Cultivo Condicionados , Factor 2 de Crecimiento de Fibroblastos/metabolismo , Regulación Neoplásica de la Expresión Génica/genética , Técnicas de Silenciamiento del Gen , Ratones , Ratones Endogámicos BALB C , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Invasividad Neoplásica/genética , Metástasis de la Neoplasia/genética , Proteínas Proto-Oncogénicas c-ets/genética , Proteínas Proto-Oncogénicas c-ets/metabolismo , Factores de Transcripción/genética
11.
EMBO Rep ; 17(11): 1516-1531, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27702988

RESUMEN

Historically, cancers have been treated with chemotherapeutics aimed to have profound effects on tumor cells with only limited effects on normal tissue. This approach was followed by the development of small-molecule inhibitors that can target oncogenic pathways critical for the survival of tumor cells. The clinical targeting of these so-called oncogene addictions, however, is in many instances hampered by the outgrowth of resistant clones. More recently, the proper functioning of non-mutated genes has been shown to enhance the survival of many cancers, a phenomenon called non-oncogene addiction. In the current review, we will focus on the distinct non-oncogenic addictions found in cancer cells, including synthetic lethal interactions, the underlying stress phenotypes, and arising therapeutic opportunities.


Asunto(s)
Mutación , Neoplasias/terapia , Animales , Apoptosis , Línea Celular Tumoral , Humanos , Ratones , Terapia Molecular Dirigida , Neoplasias/genética , Neoplasias/metabolismo , Oncogenes , Estrés Fisiológico , Mutaciones Letales Sintéticas
12.
Nature ; 482(7386): 538-41, 2012 Feb 22.
Artículo en Inglés | MEDLINE | ID: mdl-22358843

RESUMEN

Since its discovery in the early 1990s the deleted in colorectal cancer (DCC) gene, located on chromosome 18q21, has been proposed as a tumour suppressor gene as its loss is implicated in the majority of advanced colorectal and many other cancers. DCC belongs to the family of netrin 1 receptors, which function as dependence receptors as they control survival or apoptosis depending on ligand binding. However, the role of DCC as a tumour suppressor remains controversial because of the rarity of DCC-specific mutations and the presence of other tumour suppressor genes in the same chromosomal region. Here we show that in a mouse model of mammary carcinoma based on somatic inactivation of p53, additional loss of DCC promotes metastasis formation without affecting the primary tumour phenotype. Furthermore, we demonstrate that in cell cultures derived from p53-deficient mouse mammary tumours DCC expression controls netrin-1-dependent cell survival, providing a mechanistic basis for the enhanced metastatic capacity of tumour cells lacking DCC. Consistent with this idea, in vivo tumour-cell survival is enhanced by DCC loss. Together, our data support the function of DCC as a context-dependent tumour suppressor that limits survival of disseminated tumour cells.


Asunto(s)
Genes p53/genética , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Metástasis de la Neoplasia/genética , Metástasis de la Neoplasia/patología , Receptores de Superficie Celular/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Apoptosis/genética , Línea Celular Tumoral , Supervivencia Celular/genética , Receptor DCC , Modelos Animales de Enfermedad , Femenino , Neoplasias Mamarias Experimentales/metabolismo , Ratones , Ratones Endogámicos C57BL , Factores de Crecimiento Nervioso/deficiencia , Factores de Crecimiento Nervioso/genética , Factores de Crecimiento Nervioso/metabolismo , Netrina-1 , Receptores de Superficie Celular/deficiencia , Receptores de Superficie Celular/genética , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética
13.
Proc Natl Acad Sci U S A ; 112(27): 8409-14, 2015 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-26100884

RESUMEN

Metaplastic breast carcinoma (MBC) is a rare histological breast cancer subtype characterized by mesenchymal elements and poor clinical outcome. A large fraction of MBCs harbor defects in breast cancer 1 (BRCA1). As BRCA1 deficiency sensitizes tumors to DNA cross-linking agents and poly(ADP-ribose) polymerase (PARP) inhibitors, we sought to investigate the response of BRCA1-deficient MBCs to the PARP inhibitor olaparib. To this end, we established a genetically engineered mouse model (GEMM) for BRCA1-deficient MBC by introducing the MET proto-oncogene into a BRCA1-associated breast cancer model, using our novel female GEMM ES cell (ESC) pipeline. In contrast to carcinomas, BRCA1-deficient mouse carcinosarcomas resembling MBC show intrinsic resistance to olaparib caused by increased P-glycoprotein (Pgp) drug efflux transporter expression. Indeed, resistance could be circumvented by using another PARP inhibitor, AZD2461, which is a poor Pgp substrate. These preclinical findings suggest that patients with BRCA1-associated MBC may show poor response to olaparib and illustrate the value of GEMM-ESC models of human cancer for evaluation of novel therapeutics.


Asunto(s)
Proteína BRCA1/deficiencia , Neoplasias Mamarias Experimentales/tratamiento farmacológico , Ftalazinas/farmacología , Piperazinas/farmacología , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Subfamilia B de Transportador de Casetes de Unión a ATP/genética , Subfamilia B de Transportador de Casetes de Unión a ATP/metabolismo , Animales , Proteína BRCA1/genética , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/genética , Neoplasias de la Mama/metabolismo , Carcinosarcoma/tratamiento farmacológico , Carcinosarcoma/genética , Carcinosarcoma/metabolismo , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos/genética , Inhibidores Enzimáticos/farmacología , Femenino , Humanos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/metabolismo , Metaplasia , Ratones Endogámicos C57BL , Ratones Noqueados , Poli(ADP-Ribosa) Polimerasas/metabolismo , Proto-Oncogenes Mas , Análisis de Supervivencia
14.
Proc Natl Acad Sci U S A ; 111(13): 4952-7, 2014 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-24586047

RESUMEN

Much controversy surrounds the cell-of-origin of mutant K-Ras (K-RasG12D)-induced lung adenocarcinoma. To shed light on this issue, we have used technology that enables us to conditionally target K-RasG12D expression in Surfactant Protein C (SPC)(+) alveolar type 2 cells and in Clara cell antigen 10 (CC10)(+) Clara cells by use of cell-type-restricted recombinant Adeno-Cre viruses. Experiments were performed both in the presence and absence of the tumor suppressor gene p53, enabling us to assess what effect the cell-of-origin and the introduced genetic lesions have on the phenotypic characteristics of the resulting adenocarcinomas. We conclude that both SPC-expressing alveolar type 2 cells and CC10-expressing Clara cells have the ability to initiate malignant transformation following the introduction of these genetic alterations. The lungs of K-Ras(lox-Stop-lox-G12D/+) and K-Ras(lox-Stop-lox-G12D/+);tumor suppressor gene Trp53(F/F) mice infected with Adeno5-SPC-Cre and Adeno5-CC10-Cre viruses displayed differences in their tumor spectrum, indicating distinct cellular routes of tumor initiation. Moreover, using a multicolor Cre reporter line, we demonstrate that the resulting tumors arise from a clonal expansion of switched cells. Taken together, these results indicate that there are multiple cellular paths to K-RasG12D-induced adenocarcinoma and that the initiating cell influences the histopathological phenotype of the tumors that arise.


Asunto(s)
Adenocarcinoma/genética , Adenocarcinoma/patología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patología , Mutación/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Adenocarcinoma del Pulmón , Adenoviridae/metabolismo , Animales , Proliferación Celular , Células Clonales , Progresión de la Enfermedad , Vectores Genéticos , Inmunohistoquímica , Integrasas/metabolismo , Ratones , Ratones Endogámicos C57BL , Modelos Biológicos , Proteínas Mutantes/metabolismo , Lesiones Precancerosas/patología , Recombinación Genética/genética , Proteína p53 Supresora de Tumor/metabolismo , Uteroglobina/metabolismo
15.
PLoS Genet ; 10(4): e1004250, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24721906

RESUMEN

The ability of retroviruses and transposons to insert their genetic material into host DNA makes them widely used tools in molecular biology, cancer research and gene therapy. However, these systems have biases that may strongly affect research outcomes. To address this issue, we generated very large datasets consisting of ~ 120,000 to ~ 180,000 unselected integrations in the mouse genome for the Sleeping Beauty (SB) and piggyBac (PB) transposons, and the Mouse Mammary Tumor Virus (MMTV). We analyzed ~ 80 (epi)genomic features to generate bias maps at both local and genome-wide scales. MMTV showed a remarkably uniform distribution of integrations across the genome. More distinct preferences were observed for the two transposons, with PB showing remarkable resemblance to bias profiles of the Murine Leukemia Virus. Furthermore, we present a model where target site selection is directed at multiple scales. At a large scale, target site selection is similar across systems, and defined by domain-oriented features, namely expression of proximal genes, proximity to CpG islands and to genic features, chromatin compaction and replication timing. Notable differences between the systems are mainly observed at smaller scales, and are directed by a diverse range of features. To study the effect of these biases on integration sites occupied under selective pressure, we turned to insertional mutagenesis (IM) screens. In IM screens, putative cancer genes are identified by finding frequently targeted genomic regions, or Common Integration Sites (CISs). Within three recently completed IM screens, we identified 7%-33% putative false positive CISs, which are likely not the result of the oncogenic selection process. Moreover, results indicate that PB, compared to SB, is more suited to tag oncogenes.


Asunto(s)
Cromatina/genética , Elementos Transponibles de ADN/genética , Retroviridae/genética , Animales , Islas de CpG/genética , Genoma/genética , Ratones , Mutagénesis Insercional/métodos , Oncogenes/genética
16.
Cancer Cell ; 13(1): 5-7, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-18167334

RESUMEN

In contrast to conventional chemotherapeutic agents, modern anticancer therapies are aimed at attacking specific targets in a tumor. While these therapies show promising clinical effects, their success is limited by the development of resistance to the antitumor agent, a phenomenon that is well known in regular cancer therapies. As illustrated in a novel study by Debies and colleagues in The Journal of Clinical Investigation, mouse models for cancer serve as promising tools for advancing our understanding of the tumor response to targeted therapy. However, the experimental setup and selected model system may evoke unexpected escape mechanisms. Here, we discuss the promises and pitfalls of these approaches.


Asunto(s)
Antineoplásicos/uso terapéutico , Escape del Tumor , Animales , Modelos Animales de Enfermedad , Resistencia a Antineoplásicos , Humanos , Ratones , Recurrencia Local de Neoplasia/tratamiento farmacológico , Proteínas Wnt/metabolismo
17.
Blood ; 120(24): 4819-28, 2012 Dec 06.
Artículo en Inglés | MEDLINE | ID: mdl-23074275

RESUMEN

Mixed lineage leukemia (MLL) fusion genes arise from chromosomal translocations and induce acute myeloid leukemia through a mechanism involving transcriptional deregulation of differentiation and self-renewal programs. Progression of MLL-rearranged acute myeloid leukemia is associated with increased activation of Rac GTPases. Here, we demonstrate that MLL fusion oncogenes maintain leukemia-associated Rac activity by regulating Frat gene expression, specifically Frat2. Modulation of FRAT2 leads to concomitant changes in Rac activity, and transformation of Frat knockout hematopoietic progenitor cells by MLL fusions results in leukemias displaying reduced Rac activation and increased sensitivity to chemotherapeutic drugs. FRAT2 activates Rac through a signaling mechanism that requires glycogen synthase kinase 3 and DVL. Disruption of this pathway abrogates the leukemogenic activity of MLL fusions. This suggests a rationale for the paradoxical requirement of canonical Wnt signaling and glycogen synthase kinase 3 activity for MLL fusion oncogenicity and identifies novel therapeutic targets for this disease.


Asunto(s)
Proteínas Portadoras/metabolismo , Proteína de la Leucemia Mieloide-Linfoide/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Proteínas de Unión al GTP rac/metabolismo , Enfermedad Aguda , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Western Blotting , Proteínas Portadoras/genética , Transformación Celular Neoplásica/genética , Células Cultivadas , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteínas Dishevelled , Femenino , Glucógeno Sintasa Quinasa 3/genética , Glucógeno Sintasa Quinasa 3/metabolismo , Células HEK293 , Células Madre Hematopoyéticas/metabolismo , N-Metiltransferasa de Histona-Lisina , Humanos , Leucemia Mieloide/genética , Leucemia Mieloide/metabolismo , Leucemia Mieloide/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteína de la Leucemia Mieloide-Linfoide/genética , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fusión de Oncogenes , Proteínas de Fusión Oncogénica/genética , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Proteínas Proto-Oncogénicas , Interferencia de ARN , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Proteínas de Unión al GTP rac/genética
18.
Transgenic Res ; 23(4): 691-5, 2014 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-24798251

RESUMEN

Nonsurgical embryo transfer (NSET) of blastocysts to pseudopregnant female recipients provides many benefits over surgical implantation with less distress for the mice, no anesthesia or analgesia required and a considerable reduction in implantation time per mouse. Although a disposable device to perform NSET is on the market since 2009, it is not generally used in transgenic facilities, most likely because surgical implantation is efficient and inexpensive. Here, we report that with several refinements to the original protocol, the NSET method becomes very attractive and outperforms the traditional surgical transfer on basis of pregnancy rate, birth rate and implantation-related discomfort. Furthermore, repeated use of the same NSET device on several recipient females reduces the costs to a reasonable level. The data presented covers all embryo transfers over the last 5 years at the transgenic facility of the Netherlands Cancer Institute, of which the last 2 years were performed exclusively with NSET.


Asunto(s)
Tasa de Natalidad , Implantación del Embrión , Transferencia de Embrión/métodos , Transferencia de Embrión/veterinaria , Embarazo/estadística & datos numéricos , Animales , Blastocisto , Femenino , Ratones
19.
Cancer Cell ; 10(5): 437-49, 2006 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-17097565

RESUMEN

Metastatic disease is the primary cause of death in breast cancer, the most common malignancy in Western women. Loss of E-cadherin is associated with tumor metastasis, as well as with invasive lobular carcinoma (ILC), which accounts for 10%-15% of all breast cancers. To study the role of E-cadherin in breast oncogenesis, we have introduced conditional E-cadherin mutations into a mouse tumor model based on epithelium-specific knockout of p53. Combined loss of E-cadherin and p53 resulted in accelerated development of invasive and metastatic mammary carcinomas, which show strong resemblance to human ILC. Moreover, loss of E-cadherin induced anoikis resistance and facilitated angiogenesis, thus promoting metastatic disease. Our results suggest that loss of E-cadherin contributes to both mammary tumor initiation and metastasis.


Asunto(s)
Anoicis/fisiología , Neoplasias de la Mama/metabolismo , Cadherinas/metabolismo , Carcinoma Lobular/metabolismo , Silenciador del Gen , Proteína p53 Supresora de Tumor/metabolismo , Animales , Neoplasias de la Mama/patología , Neoplasias de la Mama/fisiopatología , Cadherinas/genética , Carcinoma Lobular/patología , Carcinoma Lobular/fisiopatología , Modelos Animales de Enfermedad , Femenino , Humanos , Glándulas Mamarias Humanas/anatomía & histología , Glándulas Mamarias Humanas/metabolismo , Glándulas Mamarias Humanas/patología , Ratones , Ratones Endogámicos BALB C , Metástasis de la Neoplasia , Neovascularización Patológica , Neoplasias Cutáneas/metabolismo , Neoplasias Cutáneas/patología , Tasa de Supervivencia , Células Tumorales Cultivadas , Proteína p53 Supresora de Tumor/genética
20.
Mol Oncol ; 18(6): 1351-1354, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38634213

RESUMEN

Discovery research is the starting point for the development of more effective anti-cancer treatments. It requires an interdisciplinary research environment with first-class infrastructural support in which curiosity-driven research can lead to new concepts for treating cancer. Translating such research findings to clinical practice requires complementary skills and infrastructures, including high-quality clinical facilities, access to patient cohorts and participation of pharma. This complex ecosystem has yielded many new but also "me too" treatment regimens, especially in immuno-oncology resulting in an extremely high pricing of anti-cancer agents. The costs of antibodies, vaccines, and cell therapies charged by pharma stand out although the concepts and methodologies have been largely developed in academia, financed from public funds. Comprehensive Cancer Centres (CCCs) covering a coherent stretch of the cancer research continuum are well-positioned to make these personalized treatments more affordable, but this will require restructuring of the way the translational cancer research continuum is funded.


Asunto(s)
Neoplasias , Humanos , Neoplasias/terapia , Neoplasias/economía , Antineoplásicos/uso terapéutico , Antineoplásicos/economía , Investigación Biomédica Traslacional/economía , Academia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA