Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 35
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
Nature ; 621(7977): 171-178, 2023 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-37648867

RESUMEN

Triacylglycerols (TAGs) are the main source of stored energy in the body, providing an important substrate pool for mitochondrial beta-oxidation. Imbalances in the amount of TAGs are associated with obesity, cardiac disease and various other pathologies1,2. In humans, TAGs are synthesized from excess, coenzyme A-conjugated fatty acids by diacylglycerol O-acyltransferases (DGAT1 and DGAT2)3. In other organisms, this activity is complemented by additional enzymes4, but whether such alternative pathways exist in humans remains unknown. Here we disrupt the DGAT pathway in haploid human cells and use iterative genetics to reveal an unrelated TAG-synthesizing system composed of a protein we called DIESL (also known as TMEM68, an acyltransferase of previously unknown function) and its regulator TMX1. Mechanistically, TMX1 binds to and controls DIESL at the endoplasmic reticulum, and loss of TMX1 leads to the unconstrained formation of DIESL-dependent lipid droplets. DIESL is an autonomous TAG synthase, and expression of human DIESL in Escherichia coli endows this organism with the ability to synthesize TAG. Although both DIESL and the DGATs function as diacylglycerol acyltransferases, they contribute to the cellular TAG pool under specific conditions. Functionally, DIESL synthesizes TAG at the expense of membrane phospholipids and maintains mitochondrial function during periods of extracellular lipid starvation. In mice, DIESL deficiency impedes rapid postnatal growth and affects energy homeostasis during changes in nutrient availability. We have therefore identified an alternative TAG biosynthetic pathway driven by DIESL under potent control by TMX1.


Asunto(s)
Aciltransferasas , Triglicéridos , Animales , Humanos , Ratones , Aciltransferasas/metabolismo , Coenzima A/metabolismo , Diacilglicerol O-Acetiltransferasa/metabolismo , Escherichia coli/metabolismo , Homeostasis , Triglicéridos/biosíntesis , Metabolismo Energético , Nutrientes/metabolismo , Ácidos Grasos/química , Ácidos Grasos/metabolismo
2.
Nature ; 620(7973): 374-380, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37532932

RESUMEN

Low-grade inflammation is a hallmark of old age and a central driver of ageing-associated impairment and disease1. Multiple factors can contribute to ageing-associated inflammation2; however, the molecular pathways that transduce aberrant inflammatory signalling and their impact in natural ageing remain unclear. Here we show that the cGAS-STING signalling pathway, which mediates immune sensing of DNA3, is a critical driver of chronic inflammation and functional decline during ageing. Blockade of STING suppresses the inflammatory phenotypes of senescent human cells and tissues, attenuates ageing-related inflammation in multiple peripheral organs and the brain in mice, and leads to an improvement in tissue function. Focusing on the ageing brain, we reveal that activation of STING triggers reactive microglial transcriptional states, neurodegeneration and cognitive decline. Cytosolic DNA released from perturbed mitochondria elicits cGAS activity in old microglia, defining a mechanism by which cGAS-STING signalling is engaged in the ageing brain. Single-nucleus RNA-sequencing analysis of microglia and hippocampi of a cGAS gain-of-function mouse model demonstrates that engagement of cGAS in microglia is sufficient to direct ageing-associated transcriptional microglial states leading to bystander cell inflammation, neurotoxicity and impaired memory capacity. Our findings establish the cGAS-STING pathway as a driver of ageing-related inflammation in peripheral organs and the brain, and reveal blockade of cGAS-STING signalling as a potential strategy to halt neurodegenerative processes during old age.


Asunto(s)
Envejecimiento , Encéfalo , Disfunción Cognitiva , Inflamación , Proteínas de la Membrana , Enfermedades Neurodegenerativas , Nucleotidiltransferasas , Animales , Humanos , Ratones , Envejecimiento/metabolismo , Envejecimiento/patología , Encéfalo/metabolismo , Encéfalo/patología , Efecto Espectador , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , ADN/inmunología , Inflamación/enzimología , Inflamación/metabolismo , Proteínas de la Membrana/metabolismo , Trastornos de la Memoria/enzimología , Trastornos de la Memoria/metabolismo , Microglía/metabolismo , Mitocondrias/metabolismo , Enfermedades Neurodegenerativas/enzimología , Enfermedades Neurodegenerativas/metabolismo , Nucleotidiltransferasas/metabolismo , Especificidad de Órganos , Transducción de Señal , Hipocampo/metabolismo , Hipocampo/patología
3.
Sci Transl Med ; 14(674): eabj4375, 2022 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-36475903

RESUMEN

Liver transplantation is the only curative option for patients with end-stage liver disease. Despite improvements in surgical techniques, nonanastomotic strictures (characterized by the progressive loss of biliary tract architecture) continue to occur after liver transplantation, negatively affecting liver function and frequently leading to graft loss and retransplantation. To study the biological effects of organ preservation before liver transplantation, we generated murine models that recapitulate liver procurement and static cold storage. In these models, we explored the response of cholangiocytes and hepatocytes to cold storage, focusing on responses that affect liver regeneration, including DNA damage, apoptosis, and cellular senescence. We show that biliary senescence was induced during organ retrieval and exacerbated during static cold storage, resulting in impaired biliary regeneration. We identified decoy receptor 2 (DCR2)-dependent responses in cholangiocytes and hepatocytes, which differentially affected the outcome of those populations during cold storage. Moreover, CRISPR-mediated DCR2 knockdown in vitro increased cholangiocyte proliferation and decreased cellular senescence but had the opposite effect in hepatocytes. Using the p21KO model to inhibit senescence onset, we showed that biliary tract architecture was better preserved during cold storage. Similar results were achieved by administering senolytic ABT737 to mice before procurement. Last, we perfused senolytics into discarded human donor livers and showed that biliary architecture and regenerative capacities were better preserved. Our results indicate that cholangiocytes are susceptible to senescence and identify the use of senolytics and the combination of senotherapies and machine-perfusion preservation to prevent this phenotype and reduce the incidence of biliary injury after transplantation.


Asunto(s)
Sistema Biliar , Humanos , Ratones , Animales , Constricción Patológica , Senescencia Celular
4.
Nat Commun ; 13(1): 6442, 2022 10 28.
Artículo en Inglés | MEDLINE | ID: mdl-36307419

RESUMEN

The experimental need to engineer the genome both in time and space, has led to the development of several photoactivatable Cre recombinase systems. However, the combination of inefficient and non-intentional background recombination has prevented thus far the wide application of these systems in biological and biomedical research. Here, we engineer an optimized photoactivatable Cre recombinase system that we refer to as doxycycline- and light-inducible Cre recombinase (DiLiCre). Following extensive characterization in cancer cell and organoid systems, we generate a DiLiCre mouse line, and illustrated the biological applicability of DiLiCre for light-induced mutagenesis in vivo and positional cell-tracing by intravital microscopy. These experiments illustrate how newly formed HrasV12 mutant cells follow an unnatural movement towards the interfollicular dermis. Together, we develop an efficient photoactivatable Cre recombinase mouse model and illustrate how this model is a powerful genome-editing tool for biological and biomedical research.


Asunto(s)
Doxiciclina , Optogenética , Ratones , Animales , Doxiciclina/farmacología , Ratones Transgénicos , Edición Génica , Integrasas/genética , Integrasas/metabolismo , Ratones Endogámicos
5.
Science ; 377(6614): 1533-1537, 2022 09 30.
Artículo en Inglés | MEDLINE | ID: mdl-36173861

RESUMEN

Protein synthesis generally starts with a methionine that is removed during translation. However, cytoplasmic actin defies this rule because its synthesis involves noncanonical excision of the acetylated methionine by an unidentified enzyme after translation. Here, we identified C19orf54, named ACTMAP (actin maturation protease), as this enzyme. Its ablation resulted in viable mice in which the cytoskeleton was composed of immature actin molecules across all tissues. However, in skeletal muscle, the lengths of sarcomeric actin filaments were shorter, muscle function was decreased, and centralized nuclei, a common hallmark of myopathies, progressively accumulated. Thus, ACTMAP encodes the missing factor required for the synthesis of mature actin and regulates specific actin-dependent traits in vivo.


Asunto(s)
Actinas , Metionina , Péptido Hidrolasas , Citoesqueleto de Actina/química , Citoesqueleto de Actina/metabolismo , Actinas/biosíntesis , Actinas/genética , Animales , Endopeptidasas , Metionina/genética , Metionina/metabolismo , Ratones , Péptido Hidrolasas/genética , Péptido Hidrolasas/metabolismo
6.
Lab Invest ; 102(4): 391-400, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-34921235

RESUMEN

Osteosarcoma is a high-grade bone-forming neoplasm, with a complex genome. Tumours frequently show chromothripsis, many deletions, translocations and copy number alterations. Alterations in the p53 or Rb pathway are the most common genetic alterations identified in osteosarcoma. Using spontaneously transformed murine mesenchymal stem cells (MSCs) which formed sarcoma after subcutaneous injection into mice, it was previously demonstrated that p53 is most often involved in the transformation towards sarcomas with complex genomics, including osteosarcoma. In the current study, not only loss of p53 but also loss of p16Ink4a is shown to be a driver of osteosarcomagenesis: murine MSCs with deficient p15Ink4b, p16Ink4a, or p19Arf transform earlier compared to wild-type murine MSCs. Furthermore, in a panel of nine spontaneously transformed murine MSCs, alterations in p15Ink4b, p16Ink4a, or p19Arf were observed in eight out of nine cases. Alterations in the Rb/p16 pathway could indicate that osteosarcoma cells are vulnerable to CDK4/CDK6 inhibitor treatment. Indeed, using two-dimensional (n = 7) and three-dimensional (n = 3) cultures of human osteosarcoma cell lines, it was shown that osteosarcoma cells with defective p16INK4A are sensitive to the CDK4/CDK6 inhibitor palbociclib after 72-hour treatment. A tissue microarray analysis of 109 primary tumour biopsies revealed a subset of patients (20-23%) with intact Rb, but defective p16 or overexpression of CDK4 and/or CDK6. These patients might benefit from CDK4/CDK6 inhibition, therefore our results are promising and might be translated to the clinic.


Asunto(s)
Neoplasias Óseas , Células Madre Mesenquimatosas , Osteosarcoma , Animales , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/genética , Quinasa 4 Dependiente de la Ciclina/genética , Quinasa 4 Dependiente de la Ciclina/metabolismo , Quinasa 6 Dependiente de la Ciclina/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones , Osteosarcoma/tratamiento farmacológico , Proteína p14ARF Supresora de Tumor/genética , Proteína p14ARF Supresora de Tumor/metabolismo , Proteína p53 Supresora de Tumor/genética
7.
Sci Transl Med ; 13(594)2021 05 19.
Artículo en Inglés | MEDLINE | ID: mdl-34011625

RESUMEN

The ability of the kidney to regenerate successfully after injury is lost with advancing age, chronic kidney disease, and after irradiation. The factors responsible for this reduced regenerative capacity remain incompletely understood, with increasing interest in a potential role for cellular senescence in determining outcomes after injury. Here, we demonstrated correlations between senescent cell load and functional loss in human aging and chronic kidney diseases including radiation nephropathy. We dissected the causative role of senescence in the augmented fibrosis occurring after injury in aged and irradiated murine kidneys. In vitro studies on human proximal tubular epithelial cells and in vivo mouse studies demonstrated that senescent renal epithelial cells produced multiple components of the senescence-associated secretory phenotype including transforming growth factor ß1, induced fibrosis, and inhibited tubular proliferative capacity after injury. Treatment of aged and irradiated mice with the B cell lymphoma 2/w/xL inhibitor ABT-263 reduced senescent cell numbers and restored a regenerative phenotype in the kidneys with increased tubular proliferation, improved function, and reduced fibrosis after subsequent ischemia-reperfusion injury. Senescent cells are key determinants of renal regenerative capacity in mice and represent emerging treatment targets to protect aging and vulnerable kidneys in man.


Asunto(s)
Senescencia Celular , Daño por Reperfusión , Animales , Fibrosis , Riñón/patología , Ratones , Ratones Endogámicos C57BL , Regeneración , Daño por Reperfusión/patología
8.
J Exp Med ; 217(6)2020 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-32271879

RESUMEN

We have generated mouse models of malignant mesothelioma (MM) based upon disruption of the Bap1, Nf2, and Cdkn2ab tumor suppressor loci in various combinations as also frequently observed in human MM. Inactivation of all three loci in the mesothelial lining of the thoracic cavity leads to a highly aggressive MM that recapitulates the histological features and gene expression profile observed in human patients. The tumors also show a similar inflammatory phenotype. Bap1 deletion alone does not cause MM but dramatically accelerates MM development when combined with Nf2 and Cdkn2ab (hereafter BNC) disruption. The accelerated tumor development is accompanied by increased Polycomb repression and EZH2-mediated redistribution of H3K27me3 toward promoter sites with concomitant activation of PI3K and MAPK pathways. Treatment of BNC tumor-bearing mice with cisplatin and pemetrexed, the current frontline treatment, prolongs survival. This makes the autochthonous mouse model described here very well suited to explore the pathogenesis of MM and validate new treatment regimens for MM, including immunotherapy.


Asunto(s)
Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Eliminación de Gen , Mesotelioma Maligno/metabolismo , Neurofibromina 2/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Ubiquitina Tiolesterasa/metabolismo , Animales , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Humanos , Inmunofenotipificación , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Mesotelioma Maligno/genética , Mesotelioma Maligno/patología , Ratones , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de Poli(ADP-Ribosa) Polimerasas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Transcripción Genética/efectos de los fármacos , Microambiente Tumoral/efectos de los fármacos
9.
Nat Commun ; 10(1): 1425, 2019 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-30926782

RESUMEN

Cdkn2ab knockout mice, generated from 129P2 ES cells develop skin carcinomas. Here we show that the incidence of these carcinomas drops gradually in the course of backcrossing to the FVB/N background. Microsatellite analyses indicate that this cancer phenotype is linked to a 20 Mb region of 129P2 chromosome 15 harboring the Wnt7b gene, which is preferentially expressed from the 129P2 allele in skin carcinomas and derived cell lines. ChIPseq analysis shows enrichment of H3K27-Ac, a mark for active enhancers, in the 5' region of the Wnt7b 129P2 gene. The Wnt7b 129P2 allele appears sufficient to cause in vitro transformation of Cdkn2ab-deficient cell lines primarily through CDK6 activation. These results point to a critical role of the Cdkn2ab locus in keeping the oncogenic potential of physiological levels of WNT signaling in check and illustrate that GWAS-based searches for cancer predisposing allelic variants can be enhanced by including defined somatically acquired lesions as an additional input.


Asunto(s)
Carcinogénesis/genética , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/deficiencia , Inhibidor p16 de la Quinasa Dependiente de Ciclina/deficiencia , Variación Genética , Neoplasias Cutáneas/genética , Vía de Señalización Wnt/genética , Alelos , Animales , Emparejamiento Base/genética , Línea Celular Tumoral , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Cromosomas de los Mamíferos/genética , Quinasa 6 Dependiente de la Ciclina/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Fibroblastos/metabolismo , Ligamiento Genético , Pulmón/patología , Metaplasia , Ratones Noqueados , Factor de Crecimiento Derivado de Plaquetas/metabolismo
10.
Nat Med ; 24(7): 961-967, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29808006

RESUMEN

RAS mutations are frequent in human cancer, especially in pancreatic, colorectal and non-small-cell lung cancers (NSCLCs)1-3. Inhibition of the RAS oncoproteins has proven difficult4, and attempts to target downstream effectors5-7 have been hampered by the activation of compensatory resistance mechanisms8. It is also well established that KRAS-mutant tumors are insensitive to inhibition of upstream growth factor receptor signaling. Thus, epidermal growth factor receptor antibody therapy is only effective in KRAS wild-type colon cancers9,10. Consistently, inhibition of SHP2 (also known as PTPN11), which links receptor tyrosine kinase signaling to the RAS-RAF-MEK-ERK pathway11,12, was shown to be ineffective in KRAS-mutant or BRAF-mutant cancer cell lines13. Our data also indicate that SHP2 inhibition in KRAS-mutant NSCLC cells under normal cell culture conditions has little effect. By contrast, SHP2 inhibition under growth factor-limiting conditions in vitro results in a senescence response. In vivo, inhibition of SHP2 in KRAS-mutant NSCLC also provokes a senescence response, which is exacerbated by MEK inhibition. Our data identify SHP2 inhibition as an unexpected vulnerability of KRAS-mutant NSCLC cells that remains undetected in cell culture and can be exploited therapeutically.


Asunto(s)
Carcinoma de Pulmón de Células no Pequeñas/genética , Neoplasias Pulmonares/genética , Mutación/genética , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Proteínas Proto-Oncogénicas p21(ras)/genética , Animales , Carcinoma de Pulmón de Células no Pequeñas/patología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Senescencia Celular/efectos de los fármacos , Guanosina Trifosfato/metabolismo , Humanos , Neoplasias Pulmonares/patología , Ratones , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Ensayos Antitumor por Modelo de Xenoinjerto
11.
Biol Psychiatry ; 83(2): 181-192, 2018 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-28720317

RESUMEN

BACKGROUND: Dopaminergic input to the prefrontal cortex (PFC) increases throughout adolescence and, by establishing precisely localized synapses, calibrates cognitive function. However, why and how mesocortical dopamine axon density increases across adolescence remains unknown. METHODS: We used a developmental application of axon-initiated recombination to label and track the growth of dopamine axons across adolescence in mice. We then paired this recombination with cell-specific knockdown of the netrin-1 receptor DCC to determine its role in adolescent dopamine axon growth. We then assessed how altering adolescent PFC dopamine axon growth changes the structural and functional development of the PFC by quantifying pyramidal neuron morphology and cognitive performance. RESULTS: We show, for the first time, that dopamine axons continue to grow from the striatum to the PFC during adolescence. Importantly, we discover that DCC, a guidance cue receptor, controls the extent of this protracted growth by determining where and when dopamine axons recognize their final target. When DCC-dependent adolescent targeting events are disrupted, dopamine axons continue to grow ectopically from the nucleus accumbens to the PFC and profoundly change PFC structural and functional development. This leads to alterations in cognitive processes known to be impaired across psychiatric conditions. CONCLUSIONS: The prolonged growth of dopamine axons represents an extraordinary period for experience to influence their adolescent trajectory and predispose to or protect against psychopathology. DCC receptor signaling in dopamine neurons is a molecular link where genetic and environmental factors may interact in adolescence to influence the development and function of the prefrontal cortex.


Asunto(s)
Axones/metabolismo , Receptor DCC/metabolismo , Neuronas Dopaminérgicas/metabolismo , Núcleo Accumbens/metabolismo , Corteza Prefrontal/metabolismo , Animales , Atención/fisiología , Conducta Animal/fisiología , Receptor DCC/genética , Técnicas de Silenciamiento del Gen , Inhibición Psicológica , Masculino , Aprendizaje por Laberinto/fisiología , Ratones , Núcleo Accumbens/crecimiento & desarrollo , Corteza Prefrontal/crecimiento & desarrollo , Disposición en Psicología
12.
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
13.
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
14.
J Neurosci ; 34(29): 9768-78, 2014 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-25031414

RESUMEN

Paranodal axoglial junctions are critical for maintaining the segregation of axonal domains along myelinated axons; however, the proteins required to organize and maintain this structure are not fully understood. Netrin-1 and its receptor Deleted in Colorectal Cancer (DCC) are proteins enriched at paranodes that are expressed by neurons and oligodendrocytes. To identify the specific function of DCC expressed by oligodendrocytes in vivo, we selectively eliminated DCC from mature myelinating oligodendrocytes using an inducible cre regulated by the proteolipid protein promoter. We demonstrate that DCC deletion results in progressive disruption of the organization of axonal domains, myelin ultrastructure, and myelin protein composition. Conditional DCC knock-out mice develop balance and coordination deficits and exhibit decreased conduction velocity. We conclude that DCC expression by oligodendrocytes is required for the maintenance and stability of myelin in vivo, which is essential for proper signal conduction in the CNS.


Asunto(s)
Uniones Comunicantes/fisiología , Regulación del Desarrollo de la Expresión Génica , Vaina de Mielina/fisiología , Oligodendroglía/metabolismo , Receptores de Superficie Celular/deficiencia , Proteínas Supresoras de Tumor/deficiencia , Animales , Axones/fisiología , Recuento de Células , Receptor DCC , Embrión de Mamíferos , Antagonistas de Estrógenos/farmacología , Conducta Exploratoria/fisiología , Uniones Comunicantes/ultraestructura , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Regulación del Desarrollo de la Expresión Génica/genética , Integrasas/genética , Ratones , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Proteína Proteolipídica de la Mielina/genética , Proteína Proteolipídica de la Mielina/metabolismo , Vaina de Mielina/ultraestructura , Conducción Nerviosa/efectos de los fármacos , Conducción Nerviosa/genética , Oligodendroglía/ultraestructura , Trastornos Psicomotores/genética , Nódulos de Ranvier/metabolismo , Nódulos de Ranvier/ultraestructura , Receptores de Superficie Celular/genética , Tamoxifeno/farmacología , Proteínas Supresoras de Tumor/genética
15.
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
16.
EMBO Mol Med ; 6(2): 212-25, 2014 02.
Artículo en Inglés | MEDLINE | ID: mdl-24401838

RESUMEN

Human cancers modeled in Genetically Engineered Mouse Models (GEMMs) can provide important mechanistic insights into the molecular basis of tumor development and enable testing of new intervention strategies. The inherent complexity of these models, with often multiple modified tumor suppressor genes and oncogenes, has hampered their use as preclinical models for validating cancer genes and drug targets. In our newly developed approach for the fast generation of tumor cohorts we have overcome this obstacle, as exemplified for three GEMMs; two lung cancer models and one mesothelioma model. Three elements are central for this system; (i) The efficient derivation of authentic Embryonic Stem Cells (ESCs) from established GEMMs, (ii) the routine introduction of transgenes of choice in these GEMM-ESCs by Flp recombinase-mediated integration and (iii) the direct use of the chimeric animals in tumor cohorts. By applying stringent quality controls, the GEMM-ESC approach proofs to be a reliable and effective method to speed up cancer gene assessment and target validation. As proof-of-principle, we demonstrate that MycL1 is a key driver gene in Small Cell Lung Cancer.


Asunto(s)
Células Madre Embrionarias/citología , Técnicas de Transferencia de Gen , Neoplasias Pulmonares/patología , Animales , Carcinogénesis/metabolismo , Carcinogénesis/patología , Proliferación Celular , Células Cultivadas , Quimera , Células Clonales , ADN Nucleotidiltransferasas/metabolismo , Modelos Animales de Enfermedad , Células Madre Embrionarias/metabolismo , Genes Reporteros , Inestabilidad Genómica , Genotipo , Células Germinativas/metabolismo , Humanos , Luciferasas/metabolismo , Neoplasias Pulmonares/metabolismo , Ratones , Ratones Endogámicos C57BL , Oncogenes , Fenotipo , Células Madre Pluripotentes/metabolismo , Control de Calidad , Reproducibilidad de los Resultados , Carcinoma Pulmonar de Células Pequeñas/metabolismo , Carcinoma Pulmonar de Células Pequeñas/patología
17.
Cell Rep ; 3(1): 173-85, 2013 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-23291093

RESUMEN

The transmembrane protein deleted in colorectal cancer (DCC) and its ligand, netrin-1, regulate synaptogenesis during development, but their function in the mature central nervous system is unknown. Given that DCC promotes cell-cell adhesion, is expressed by neurons, and activates proteins that signal at synapses, we hypothesized that DCC expression by neurons regulates synaptic function and plasticity in the adult brain. We report that DCC is enriched in dendritic spines of pyramidal neurons in wild-type mice, and we demonstrate that selective deletion of DCC from neurons in the adult forebrain results in the loss of long-term potentiation (LTP), intact long-term depression, shorter dendritic spines, and impaired spatial and recognition memory. LTP induction requires Src activation of NMDA receptor (NMDAR) function. DCC deletion severely reduced Src activation. We demonstrate that enhancing NMDAR function or activating Src rescues LTP in the absence of DCC. We conclude that DCC activation of Src is required for NMDAR-dependent LTP and certain forms of learning and memory.


Asunto(s)
Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Plasticidad Neuronal , Neuronas/metabolismo , Receptores de Superficie Celular/metabolismo , Sinapsis/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Envejecimiento/metabolismo , Animales , Receptor DCC , Espinas Dendríticas/metabolismo , Espinas Dendríticas/ultraestructura , Activación Enzimática , Eliminación de Gen , Hipocampo/metabolismo , Hipocampo/patología , Hipocampo/fisiopatología , Potenciación a Largo Plazo , Aprendizaje por Laberinto , Memoria , Ratones , Factores de Crecimiento Nervioso/metabolismo , Netrina-1 , Neuronas/patología , Neuronas/ultraestructura , Fosfolipasa C gamma/metabolismo , Fosforilación , Prosencéfalo/metabolismo , Prosencéfalo/patología , Prosencéfalo/fisiopatología , Ratas , Receptores de Superficie Celular/deficiencia , Receptores de N-Metil-D-Aspartato/metabolismo , Fracciones Subcelulares/metabolismo , Sinapsis/patología , Sinapsis/ultraestructura , Proteínas Supresoras de Tumor/deficiencia , Familia-src Quinasas/metabolismo
18.
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
19.
Bioessays ; 33(9): 701-10, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21735458

RESUMEN

Recent technological advances have opened the door for the fast and cost-effective generation of genetically engineered mouse models (GEMMs) to study cancer. We describe here a conceptually novel approach for the generation of chimeric GEMMs based on the controlled introduction of various genetic elements in embryonic stem cells (ESCs) that are derived from existing mouse strains with a predisposition for cancer. The isolation of GEMM-derived ESC lines is greatly facilitated by the availability of the newly defined culture media containing inhibitors that effectively preserve ESC pluripotency. The feasibility of the GEMM-ESC approach is discussed in light of current literature and placed into the context of existing models. This approach will allow for fast and flexible validation of candidate cancer genes and drug targets and will result in a repository of GEMM-ESC lines and corresponding vector collections that enable easy distribution and use of preclinical models to the wider scientific community.


Asunto(s)
Quimera/genética , Modelos Animales de Enfermedad , Genes Relacionados con las Neoplasias , Ratones , Neoplasias/genética , Células Madre Pluripotentes/metabolismo , Animales , Animales Modificados Genéticamente , Quimera/metabolismo , Medios de Cultivo , Sistemas de Liberación de Medicamentos , Células Madre Embrionarias/metabolismo , Humanos
20.
Dis Model Mech ; 4(3): 347-58, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21282721

RESUMEN

Breast cancer is the most common malignancy in women of the Western world. Even though a large percentage of breast cancer patients show pathological complete remission after standard treatment regimes, approximately 30-40% are non-responsive and ultimately develop metastatic disease. To generate a good preclinical model of invasive breast cancer, we have taken a tissue-specific approach to somatically inactivate p53 and E-cadherin, the cardinal cell-cell adhesion receptor that is strongly associated with tumor invasiveness. In breast cancer, E-cadherin is found mutated or otherwise functionally silenced in invasive lobular carcinoma (ILC), which accounts for 10-15% of all breast cancers. We show that mammary-specific stochastic inactivation of conditional E-cadherin and p53 results in impaired mammary gland function during pregnancy through the induction of anoikis resistance of mammary epithelium, resulting in loss of epithelial organization and a dysfunctional mammary gland. Moreover, combined inactivation of E-cadherin and p53 induced lactation-independent development of invasive and metastatic mammary carcinomas, which showed strong resemblance to human pleomorphic ILC. Dissemination patterns of mouse ILC mimic the human malignancy, showing metastasis to the gastrointestinal tract, peritoneum, lung, lymph nodes and bone. Our results confirm that loss of E-cadherin contributes to both mammary tumor initiation and metastasis, and establish a preclinical mouse model of human ILC that can be used for the development of novel intervention strategies to treat invasive breast cancer.


Asunto(s)
Cadherinas/genética , Carcinoma Lobular/patología , Silenciador del Gen , Glándulas Mamarias Animales/crecimiento & desarrollo , Glándulas Mamarias Animales/patología , Neoplasias Mamarias Animales/patología , Proteína p53 Supresora de Tumor/genética , Adenocarcinoma/patología , Animales , Cadherinas/metabolismo , Femenino , Humanos , Integrasas/metabolismo , Lactancia , Neoplasias Mamarias Animales/metabolismo , Ratones , Invasividad Neoplásica , Metástasis de la Neoplasia , Especificidad de Órganos/genética , Lesiones Precancerosas/metabolismo , Lesiones Precancerosas/patología , Embarazo , Proteína p53 Supresora de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...