Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Más filtros











Intervalo de año de publicación
1.
Cancer Discov ; 13(12): 2548-2565, 2023 12 12.
Artículo en Inglés | MEDLINE | ID: mdl-37655965

RESUMEN

PML nuclear bodies (NB) are disrupted in PML-RARA-driven acute promyelocytic leukemia (APL). Arsenic trioxide (ATO) cures 70% of patients with APL, driving PML-RARA degradation and NB reformation. In non-APL cells, arsenic binding onto PML also amplifies NB formation. Yet, the actual molecular mechanism(s) involved remain(s) elusive. Here, we establish that PML NBs display some features of liquid-liquid phase separation and that ATO induces a gel-like transition. PML B-box-2 structure reveals an alpha helix driving B2 trimerization and positioning a cysteine trio to form an ideal arsenic-binding pocket. Altering either of the latter impedes ATO-driven NB assembly, PML sumoylation, and PML-RARA degradation, mechanistically explaining clinical ATO resistance. This B2 trimer and the C213 trio create an oxidation-sensitive rheostat that controls PML NB assembly dynamics and downstream signaling in both basal state and during stress response. These findings identify the structural basis for arsenic targeting of PML that could pave the way to novel cancer drugs. SIGNIFICANCE: Arsenic curative effects in APL rely on PML targeting. We report a PML B-box-2 structure that drives trimer assembly, positioning a cysteine trio to form an arsenic-binding pocket, which is disrupted in resistant patients. Identification of this ROS-sensitive triad controlling PML dynamics and functions could yield novel drugs. See related commentary by Salomoni, p. 2505. This article is featured in Selected Articles from This Issue, p. 2489.


Asunto(s)
Arsénico , Arsenicales , Leucemia Promielocítica Aguda , Humanos , Arsénico/farmacología , Cuerpos Nucleares de la Leucemia Promielocítica , Cisteína , Arsenicales/farmacología , Óxidos/farmacología , Trióxido de Arsénico/farmacología , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/genética , Leucemia Promielocítica Aguda/metabolismo , Proteínas Oncogénicas , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo
2.
Nat Commun ; 13(1): 5726, 2022 09 29.
Artículo en Inglés | MEDLINE | ID: mdl-36175410

RESUMEN

Membrane-less organelles are condensates formed by phase separation whose functions often remain enigmatic. Upon oxidative stress, PML scaffolds Nuclear Bodies (NBs) to regulate senescence or metabolic adaptation. PML NBs recruit many partner proteins, but the actual biochemical mechanism underlying their pleiotropic functions remains elusive. Similarly, PML role in embryonic stem cell (ESC) and retro-element biology is unsettled. Here we demonstrate that PML is essential for oxidative stress-driven partner SUMO2/3 conjugation in mouse ESCs (mESCs) or leukemia, a process often followed by their poly-ubiquitination and degradation. Functionally, PML is required for stress responses in mESCs. Differential proteomics unravel the KAP1 complex as a PML NB-dependent SUMO2-target in arsenic-treated APL mice or mESCs. PML-driven KAP1 sumoylation enables activation of this key epigenetic repressor implicated in retro-element silencing. Accordingly, Pml-/- mESCs re-express transposable elements and display 2-Cell-Like features, the latter enforced by PML-controlled SUMO2-conjugation of DPPA2. Thus, PML orchestrates mESC state by coordinating SUMO2-conjugation of different transcriptional regulators, raising new hypotheses about PML roles in cancer.


Asunto(s)
Arsénico , Sumoilación , Animales , Elementos Transponibles de ADN , Células Madre Embrionarias , Ratones , Cuerpos Nucleares , Factores de Transcripción
3.
J Exp Med ; 218(2)2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33075130

RESUMEN

Interferon α (IFNα) is used to treat JAK2V617F-driven myeloproliferative neoplasms (MPNs) but rarely clears the disease. We investigated the IFNα mechanism of action focusing on PML, an interferon target and key senescence gene whose targeting by arsenic trioxide (ATO) drives eradication of acute promyelocytic leukemia. ATO sharply potentiated IFNα-induced growth suppression of JAK2V617F patient or mouse hematopoietic progenitors, which required PML and was associated with features of senescence. In a mouse MPN model, combining ATO with IFNα enhanced and accelerated responses, eradicating MPN in most mice by targeting disease-initiating cells. These results predict potent clinical efficacy of the IFNα+ATO combination in patients and identify PML as a major effector of therapy, even in malignancies with an intact PML gene.


Asunto(s)
Trióxido de Arsénico/farmacología , Interferón-alfa/farmacología , Janus Quinasa 2/metabolismo , Trastornos Mieloproliferativos/tratamiento farmacológico , Proteína de la Leucemia Promielocítica/metabolismo , Animales , Línea Celular , Línea Celular Tumoral , Modelos Animales de Enfermedad , Humanos , Leucemia Promielocítica Aguda/tratamiento farmacológico , Leucemia Promielocítica Aguda/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trastornos Mieloproliferativos/metabolismo
4.
Semin Cell Dev Biol ; 80: 29-34, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29157919

RESUMEN

PML Nuclear bodies (PML NBs) are spherical domains associated with a broad range of activities upon stress responses such as apoptosis, senescence DNA repair, epigenetic control, as well as control of oncogenesis. These bodies are considered as privileged sites for post-translational modifications, where sumoylation plays a key role. Here we summarize recent in vitro and in vivo findings on the link between PML NBs and ROS, in particular PML contributions to oxidative stress response. We discuss how it may regulate switch from cell protection against stress to cell arrest/cell death.


Asunto(s)
Núcleo Celular/metabolismo , Cuerpos de Inclusión Intranucleares/metabolismo , Proteínas Nucleares/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Animales , Humanos , Proteínas Nucleares/genética , Estrés Oxidativo/fisiología , Factores de Transcripción/metabolismo
5.
J Exp Med ; 214(11): 3197-3206, 2017 Nov 06.
Artículo en Inglés | MEDLINE | ID: mdl-28931625

RESUMEN

Promyelocytic leukemia (PML) nuclear bodies (NBs) recruit partner proteins, including p53 and its regulators, thereby controlling their abundance or function. Investigating arsenic sensitivity of acute promyelocytic leukemia, we proposed that PML oxidation promotes NB biogenesis. However, physiological links between PML and oxidative stress response in vivo remain unexplored. Here, we identify PML as a reactive oxygen species (ROS) sensor. Pml-/- cells accumulate ROS, whereas PML expression decreases ROS levels. Unexpectedly, Pml-/- embryos survive acute glutathione depletion. Moreover, Pml-/- animals are resistant to acetaminophen hepatotoxicity or fasting-induced steatosis. Molecularly, Pml-/- animals fail to properly activate oxidative stress-responsive p53 targets, whereas the NRF2 response is amplified and accelerated. Finally, in an oxidative stress-prone background, Pml-/- animals display a longevity phenotype, likely reflecting decreased basal p53 activation. Thus, similar to p53, PML exerts basal antioxidant properties but also drives oxidative stress-induced changes in cell survival/proliferation or metabolism in vivo. Through NB biogenesis, PML therefore couples ROS sensing to p53 responses, shedding a new light on the role of PML in senescence or stem cell biology.


Asunto(s)
Estrés Oxidativo , Proteína de la Leucemia Promielocítica/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Western Blotting , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/metabolismo , Femenino , Cuerpos de Inclusión Intranucleares/metabolismo , Masculino , Ratones de la Cepa 129 , Ratones Endogámicos BALB C , Ratones Noqueados , Microscopía Confocal , Factor 2 Relacionado con NF-E2/metabolismo , Oxidación-Reducción , Proteína de la Leucemia Promielocítica/genética , Proteína p53 Supresora de Tumor/genética
6.
Oncotarget ; 6(32): 32713-22, 2015 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-26418719

RESUMEN

The role of PDGF-B and its receptor in meningeal tumorigenesis is not clear. We investigated the role of PDGF-B in mouse meningioma development by generating autocrine stimulation of the arachnoid through the platelet-derived growth factor receptor (PDGFR) using the RCAStv-a system. To specifically target arachnoid cells, the cells of origin of meningioma, we generated the PGDStv-a mouse (Prostaglandin D synthase). Forced expression of PDGF-B in arachnoid cells in vivo induced the formation of Grade I meningiomas in 27% of mice by 8 months of age. In vitro, PDGF-B overexpression in PGDS-positive arachnoid cells lead to increased proliferation.We found a correlation of PDGFR-B expression and NF2 inactivation in a cohort of human meningiomas, and we showed that, in mice, Nf2 loss and PDGF over-expression in arachnoid cells induced meningioma malignant transformation, with 40% of Grade II meningiomas. In these mice, additional loss of Cdkn2ab resulted in a higher incidence of malignant meningiomas with 60% of Grade II and 30% of Grade III meningiomas. These data suggest that chronic autocrine PDGF signaling can promote proliferation of arachnoid cells and is potentially sufficient to induce meningiomagenesis. Loss of Nf2 and Cdkn2ab have synergistic effects with PDGF-B overexpression promoting meningioma malignant transformation.


Asunto(s)
Aracnoides/metabolismo , Transformación Celular Neoplásica/metabolismo , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/metabolismo , Inhibidor p16 de la Quinasa Dependiente de Ciclina/metabolismo , Oxidorreductasas Intramoleculares/metabolismo , Lipocalinas/metabolismo , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Proteínas Proto-Oncogénicas c-sis/metabolismo , Animales , Aracnoides/patología , Proliferación Celular , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Cultivadas , Inhibidor p15 de las Quinasas Dependientes de la Ciclina/genética , Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Oxidorreductasas Intramoleculares/genética , Lipocalinas/genética , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Meningioma/genética , Meningioma/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Clasificación del Tumor , Proteínas Proto-Oncogénicas c-sis/genética , Receptor beta de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Estudios Retrospectivos , Transducción de Señal , Factores de Tiempo , Transfección
7.
J Cell Biol ; 204(6): 931-45, 2014 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-24637324

RESUMEN

The promyelocytic leukemia (PML) protein organizes PML nuclear bodies (NBs), which are stress-responsive domains where many partner proteins accumulate. Here, we clarify the basis for NB formation and identify stress-induced partner sumoylation as the primary NB function. NB nucleation does not rely primarily on intermolecular interactions between the PML SUMO-interacting motif (SIM) and SUMO, but instead results from oxidation-mediated PML multimerization. Oxidized PML spherical meshes recruit UBC9, which enhances PML sumoylation, allow partner recruitment through SIM interactions, and ultimately enhance partner sumoylation. Intermolecular SUMO-SIM interactions then enforce partner sequestration within the NB inner core. Accordingly, oxidative stress enhances NB formation and global sumoylation in vivo. Some NB-associated sumoylated partners also become polyubiquitinated by RNF4, precipitating their proteasomal degradation. As several partners are protein-modifying enzymes, NBs could act as sensors that facilitate and confer oxidative stress sensitivity not only to sumoylation but also to other post-translational modifications, thereby explaining alterations of stress response upon PML or NB loss.


Asunto(s)
Proteínas Nucleares/metabolismo , Estrés Oxidativo , Sumoilación , Factores de Transcripción/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Animales , Células CHO , Células COS , Núcleo Celular/metabolismo , Senescencia Celular , Chlorocebus aethiops , Cricetinae , Cricetulus , Células HeLa , Humanos , Ratones , Proteína de la Leucemia Promielocítica , Transporte de Proteínas , Especies Reactivas de Oxígeno/metabolismo , Proteínas Modificadoras Pequeñas Relacionadas con Ubiquitina/metabolismo , Enzimas Ubiquitina-Conjugadoras/metabolismo , Ubiquitina-Proteína Ligasas
8.
Clin Cancer Res ; 19(14): 3856-70, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23714726

RESUMEN

PURPOSE: The growth and survival of neurofibromatosis type 2 (NF2)-deficient cells are enhanced by the activation of multiple signaling pathways including ErbBs/IGF-1R/Met, PI3K/Akt, and Ras/Raf/Mek/Erk1/2. The chaperone protein HSP90 is essential for the stabilization of these signaling molecules. The aim of the study was to characterize the effect of HSP90 inhibition in various NF2-deficient models. EXPERIMENTAL DESIGN: We tested efficacy of the small-molecule NXD30001, which has been shown to be a potent HSP90 inhibitor. The antiproliferative activity of NXD30001 was tested in NF2-deficient cell lines and in human primary schwannoma and meningioma cultures in vitro. The antitumor efficacy of HSP90 inhibition in vivo was verified in two allograft models and in one NF2 transgenic model. The underlying molecular alteration was further characterized by a global transcriptome approach. RESULTS: NXD30001 induced degradation of client proteins in and suppressed proliferation of NF2-deficient cells. Differential expression analysis identified subsets of genes implicated in cell proliferation, cell survival, vascularization, and Schwann cell differentiation whose expression was altered by NXD30001 treatment. The results showed that NXD30001 in NF2-deficient schwannoma suppressed multiple pathways necessary for tumorigenesis. CONCLUSIONS: HSP90 inhibition showing significant antitumor activity against NF2-related tumor cells in vitro and in vivo represents a promising option for novel NF2 therapies.


Asunto(s)
Antineoplásicos/farmacología , Proteínas HSP90 de Choque Térmico/metabolismo , Lactonas/farmacología , Neurofibromatosis 2/tratamiento farmacológico , Oximas/farmacología , Animales , Línea Celular Tumoral , Proliferación Celular , Femenino , Proteínas HSP90 de Choque Térmico/antagonistas & inhibidores , Humanos , Ratones , Ratones Desnudos , Ratones Transgénicos , Neurofibromatosis 2/metabolismo , Proteolisis , Transcriptoma/efectos de los fármacos , Ensayos Antitumor por Modelo de Xenoinjerto
9.
Nat Neurosci ; 16(4): 426-33, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23455610

RESUMEN

The autosomal dominant disorder neurofibromatosis type 2 (NF2) is a hereditary tumor syndrome caused by inactivation of the NF2 tumor suppressor gene, encoding merlin. Apart from tumors affecting the peripheral and central nervous systems, most NF2 patients develop peripheral neuropathies. This peripheral nerve disease can occur in the absence of nerve-damaging tumors, suggesting an etiology that is independent of gross tumor burden. We discovered that merlin isoform 2 (merlin-iso2) has a specific function in maintaining axonal integrity and propose that reduced axonal NF2 gene dosage leads to NF2-associated polyneuropathy. We identified a merlin-iso2-dependent complex that promotes activation of the GTPase RhoA, enabling downstream Rho-associated kinase to promote neurofilament heavy chain phosphorylation. Merlin-iso2-deficient mice exhibited impaired locomotor capacities, delayed sensory reactions and electrophysiological signs of axonal neuropathy. Sciatic nerves from these mice and sural nerve biopsies from NF2 patients revealed reduced phosphorylation of the neurofilament H subunit, decreased interfilament spacings and irregularly shaped axons.


Asunto(s)
Neurofibromatosis 2/metabolismo , Neurofibromina 2/fisiología , Polineuropatías/metabolismo , Adulto , Secuencia de Aminoácidos , Animales , Animales Recién Nacidos , Línea Celular Tumoral , Células Cultivadas , Femenino , Ganglios Espinales/metabolismo , Ganglios Espinales/patología , Células HEK293 , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Datos de Secuencia Molecular , Neurofibromatosis 2/genética , Neurofibromatosis 2/patología , Neurofibromina 2/genética , Fosforilación/fisiología , Polineuropatías/genética , Polineuropatías/patología , Embarazo , Isoformas de Proteínas/genética , Isoformas de Proteínas/fisiología
10.
J Clin Invest ; 123(1): 340-7, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23221341

RESUMEN

Neurofibromatosis type 1 (NF1) patients develop benign neurofibromas and malignant peripheral nerve sheath tumors (MPNST). These incurable peripheral nerve tumors result from loss of NF1 tumor suppressor gene function, causing hyperactive Ras signaling. Activated Ras controls numerous downstream effectors, but specific pathways mediating the effects of hyperactive Ras in NF1 tumors are unknown. We performed cross-species transcriptome analyses of mouse and human neurofibromas and MPNSTs and identified global negative feedback of genes that regulate Ras/Raf/MEK/ERK signaling in both species. Nonetheless, ERK activation was sustained in mouse and human neurofibromas and MPNST. We used a highly selective pharmacological inhibitor of MEK, PD0325901, to test whether sustained Ras/Raf/MEK/ERK signaling contributes to neurofibroma growth in a neurofibromatosis mouse model (Nf1(fl/fl);Dhh-Cre) or in NF1 patient MPNST cell xenografts. PD0325901 treatment reduced aberrantly proliferating cells in neurofibroma and MPNST, prolonged survival of mice implanted with human MPNST cells, and shrank neurofibromas in more than 80% of mice tested. Our data demonstrate that deregulated Ras/ERK signaling is critical for the growth of NF1 peripheral nerve tumors and provide a strong rationale for testing MEK inhibitors in NF1 clinical trials.


Asunto(s)
Benzamidas/farmacología , Difenilamina/análogos & derivados , Quinasas de Proteína Quinasa Activadas por Mitógenos/antagonistas & inhibidores , Neurofibromatosis 1/tratamiento farmacológico , Neurofibromatosis 1/enzimología , Neoplasias del Sistema Nervioso Periférico/tratamiento farmacológico , Neoplasias del Sistema Nervioso Periférico/enzimología , Animales , Niño , Preescolar , Difenilamina/farmacología , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Femenino , Humanos , Masculino , Ratones , Ratones Mutantes , Quinasas de Proteína Quinasa Activadas por Mitógenos/genética , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Trasplante de Neoplasias , Neurofibromatosis 1/genética , Neurofibromatosis 1/patología , Proteína Oncogénica p21(ras)/genética , Proteína Oncogénica p21(ras)/metabolismo , Neoplasias del Sistema Nervioso Periférico/genética , Neoplasias del Sistema Nervioso Periférico/patología , Transcriptoma/efectos de los fármacos , Transcriptoma/genética , Trasplante Heterólogo , Ensayos Antitumor por Modelo de Xenoinjerto , Quinasas raf/genética , Quinasas raf/metabolismo
11.
Dis Model Mech ; 2(3-4): 168-77, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19259391

RESUMEN

Cerebral cavernous malformations (CCM) are vascular malformations of the brain that lead to cerebral hemorrhages. In 20% of CCM patients, this results from an autosomal dominant condition caused by loss-of-function mutations in one of the three CCM genes. High expression levels of the CCM genes in the neuroepithelium indicate that CCM lesions might be caused by a loss of function of these genes in neural cells rather than in vascular cells. However, their in vivo function, particularly during cerebral angiogenesis, is totally unknown. We developed mice with constitutive and tissue-specific CCM2 deletions to investigate CCM2 function in vivo. Constitutive deletion of CCM2 leads to early embryonic death. Deletion of CCM2 from neuroglial precursor cells does not lead to cerebrovascular defects, whereas CCM2 is required in endothelial cells for proper vascular development. Deletion of CCM2 from endothelial cells severely affects angiogenesis, leading to morphogenic defects in the major arterial and venous blood vessels and in the heart, and results in embryonic lethality at mid-gestation. These findings establish the essential role of endothelial CCM2 for proper vascular development and strongly suggest that the endothelial cell is the primary target in the cascade of events leading from CCM2 mutations to CCM cerebrovascular lesions.


Asunto(s)
Endotelio Vascular/metabolismo , Hemangioma Cavernoso del Sistema Nervioso Central/genética , Proteínas de Microfilamentos/genética , Neovascularización Patológica , Animales , Vasos Sanguíneos/patología , Células Madre Embrionarias/citología , Eliminación de Gen , Técnicas Genéticas , Genotipo , Hemangioma Cavernoso del Sistema Nervioso Central/fisiopatología , Humanos , Ratones , Ratones Noqueados , Modelos Genéticos , Mutación
12.
J Neurosci ; 28(42): 10472-81, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18923024

RESUMEN

Schwannomin/merlin is the product of a tumor suppressor gene mutated in neurofibromatosis type 2 (NF2). Although the consequences of NF2 mutations on Schwann cell proliferation are well established, the physiological role of schwannomin in differentiated cells is not known. To unravel this role, we studied peripheral nerves in mice overexpressing in Schwann cells schwannomin with a deletion occurring in NF2 patients (P0-SCH-Delta39-121) or a C-terminal deletion. The myelin sheath and nodes of Ranvier were essentially preserved in both lines. In contrast, the ultrastructural and molecular organization of contacts between Schwann cells and axons in paranodal and juxtaparanodal regions were altered, with irregular juxtaposition of normal and abnormal areas of contact. Similar but more severe alterations were observed in mice with conditional deletion of the Nf2 gene in Schwann cells. The number of Schmidt-Lanterman incisures, which are cytoplasmic channels interrupting the compact myelin and characterized by distinct autotypic contacts, was increased in the three mutant lines. P0-SCH-Delta39-121 and conditionally deleted mice displayed exuberant wrapping of nonmyelinated fibers and short internodes, an abnormality possibly related to altered control of Schwann cell proliferation. In support of this hypothesis, Schwann cell number was increased along fibers before myelination in P0-SCH-Delta39-121 mice but not in those with C-terminal deletion. Schwann cell numbers were also more numerous in mice with conditional deletion. Thus, schwannomin plays an important role in the control of Schwann cell number and is necessary for the correct organization and regulation of axoglial heterotypic and glio-glial autotypic contacts.


Asunto(s)
Comunicación Celular/fisiología , Neurofibromina 2/fisiología , Células de Schwann/metabolismo , Células de Schwann/ultraestructura , Proteínas Supresoras de Tumor/fisiología , Animales , Proliferación Celular , Eliminación de Gen , Humanos , Ratones , Ratones Transgénicos , Neurofibromina 2/biosíntesis , Neurofibromina 2/deficiencia , Neurofibromina 2/genética , Nervios Periféricos/metabolismo , Nervios Periféricos/ultraestructura , Proteínas Supresoras de Tumor/genética
13.
PLoS One ; 3(7): e2769, 2008 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-18648508

RESUMEN

Nucleotide oligomerisation domain 2 (NOD2) is a component of the innate immunity known to be involved in the homeostasis of Peyer patches (PPs) in mice. However, little is known about its role during gut infection in vivo. Yersinia pseudotuberculosis is an enteropathogen causing gastroenteritis, adenolymphitis and septicaemia which is able to invade its host through PPs. We investigated the role of Nod2 during Y. pseudotuberculosis infection. Death was delayed in Nod2 deleted and Crohn's disease associated Nod2 mutated mice orogastrically inoculated with Y. pseudotuberculosis. In PPs, the local immune response was characterized by a higher KC level and a more intense infiltration by neutrophils and macrophages. The apoptotic and bacterial cell counts were decreased. Finally, Nod2 deleted mice had a lower systemic bacterial dissemination and less damage of the haematopoeitic organs. This resistance phenotype was lost in case of intraperitoneal infection. We concluded that Nod2 contributes to the susceptibility to Y. pseudotuberculosis in mice.


Asunto(s)
Predisposición Genética a la Enfermedad , Proteína Adaptadora de Señalización NOD2/genética , Proteína Adaptadora de Señalización NOD2/fisiología , Ganglios Linfáticos Agregados/microbiología , Infecciones por Yersinia pseudotuberculosis/genética , Infecciones por Yersinia pseudotuberculosis/microbiología , Yersinia pseudotuberculosis/genética , Animales , Apoptosis , Células de la Médula Ósea/metabolismo , Susceptibilidad a Enfermedades , Eliminación de Gen , Homeostasis , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fenotipo
14.
Nat Genet ; 40(5): 600-8, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18372904

RESUMEN

Kras is commonly mutated in colon cancers, but mutations in Nras are rare. We have used genetically engineered mice to determine whether and how these related oncogenes regulate homeostasis and tumorigenesis in the colon. Expression of K-Ras(G12D) in the colonic epithelium stimulated hyperproliferation in a Mek-dependent manner. N-Ras(G12D) did not alter the growth properties of the epithelium, but was able to confer resistance to apoptosis. In the context of an Apc-mutant colonic tumor, activation of K-Ras led to defects in terminal differentiation and expansion of putative stem cells within the tumor epithelium. This K-Ras tumor phenotype was associated with attenuated signaling through the MAPK pathway, and human colon cancer cells expressing mutant K-Ras were hypersensitive to inhibition of Raf, but not Mek. These studies demonstrate clear phenotypic differences between mutant Kras and Nras, and suggest that the oncogenic phenotype of mutant K-Ras might be mediated by noncanonical signaling through Ras effector pathways.


Asunto(s)
Neoplasias del Colon/genética , Neoplasias del Colon/patología , Genes ras , Animales , Apoptosis/genética , Diferenciación Celular/genética , Línea Celular Tumoral , Proliferación Celular , Epitelio/metabolismo , Epitelio/patología , Homeostasis/genética , Humanos , Ratones , Ratones Transgénicos , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Mutación , Proteínas Proto-Oncogénicas B-raf/metabolismo , Transducción de Señal
15.
Brain Pathol ; 18(1): 62-70, 2008 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17924978

RESUMEN

Meningiomas account for approximately 30% of all primary central nervous system tumors and are found in half of neurofibromatosis type 2 patients often causing significant morbidity. Although most meningiomas are benign, 10% are classified as atypical or anaplastic, displaying aggressive clinical behavior. Biallelic inactivation of the neurofibromatosis 2 (NF2) tumor suppressor is associated with meningioma formation in all NF2 patients and 60% of sporadic meningiomas. Deletion of the p16(INK4a)/p14(ARF) locus is found in both benign and malignant meningiomas, while mutation of the p53 tumor suppressor gene is uncommon. Previously, we inactivated Nf2 in homozygous conditional knockout mice by adenoviral Cre delivery and showed that Nf2 loss in arachnoid cells is rate-limiting for meningioma formation. Here, we report that additional nullizygosity for p16(Ink4a) increases the frequency of meningioma and meningothelial proliferation in these mice without modifying the tumor grade. In addition, by using magnetic resonance imaging (MRI) to screen a large cohort of mutant mice, we were able to detect meningothelial proliferation and meningioma development opening the way to future studies in which therapeutic interventions can be tested as preclinical assessment of their potential clinical application.


Asunto(s)
Inhibidor p16 de la Quinasa Dependiente de Ciclina/genética , Neoplasias Meníngeas/genética , Neoplasias Meníngeas/patología , Meningioma/genética , Meningioma/patología , Neurofibromina 2/genética , Animales , Proliferación Celular , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Ensayos de Selección de Medicamentos Antitumorales/métodos , Femenino , Eliminación de Gen , Predisposición Genética a la Enfermedad/genética , Humanos , Inmunohistoquímica , Imagen por Resonancia Magnética , Masculino , Neoplasias Meníngeas/metabolismo , Meningioma/metabolismo , Ratones , Ratones Noqueados , Ratones Mutantes , Mutación/genética , Invasividad Neoplásica/genética , Neurofibromatosis 2/complicaciones , Neurofibromatosis 2/genética , Neurofibromatosis 2/fisiopatología
16.
PLoS One ; 2(6): e523, 2007 Jun 13.
Artículo en Inglés | MEDLINE | ID: mdl-17565376

RESUMEN

BACKGROUND: CARD15/NOD2 mutations are associated with susceptibility to Crohn's Disease (CD) and Graft Versus Host Disease (GVHD). CD and GVHD are suspected to be related with the dysfunction of Peyer's patches (PP) and isolated lymphoid follicles (LFs). Using a new mouse model invalidated for Card15/Nod2 (KO), we thus analysed the impact of the gene in these lymphoid formations together with the development of experimental colitis. METHODOLOGY/PRINCIPAL FINDINGS: At weeks 4, 12 and 52, the numbers of PPs and LFs were higher in KO mice while no difference was observed at birth. At weeks 4 and 12, the size and cellular composition of PPs were analysed by flow cytometry and immunohistochemistry. PPs of KO mice were larger with an increased proportion of M cells and CD4(+) T-cells. KO mice were also characterised by higher concentrations of TNFalpha, IFNgamma, IL12 and IL4 measured by ELISA. In contrast, little differences were found in the PP-free ileum and the spleen of KO mice. By using chamber experiments, we found that this PP phenotype is associated with an increased of both paracellular permeability and yeast/bacterial translocation. Finally, KO mice were more susceptible to the colitis induced by TNBS. CONCLUSIONS: Card15/Nod2 deficiency induces an abnormal development and function of the PPs characterised by an exaggerated immune response and an increased permeability. These observations provide a comprehensive link between the molecular defect and the Human CARD15/NOD2 associated disorders: CD and GVHD.


Asunto(s)
Colitis/patología , Proteína Adaptadora de Señalización NOD2/fisiología , Ganglios Linfáticos Agregados/metabolismo , Ganglios Linfáticos Agregados/patología , Animales , Western Blotting , Colitis/inducido químicamente , Colitis/metabolismo , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Homeostasis , Técnicas para Inmunoenzimas , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Bazo/citología , Bazo/metabolismo
17.
Development ; 132(6): 1443-51, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15716339

RESUMEN

Loss of Apc appears to be one of the major events initiating colorectal cancer. However, the first events responsible for this initiation process are not well defined and the ways in which different epithelial cell types respond to Apc loss are unknown. We used a conditional gene-ablation approach in transgenic mice expressing tamoxifen-dependent Cre recombinase all along the crypt-villus axis to analyze the immediate effects of Apc loss in the small intestinal epithelium, both in the stem-cell compartment and in postmitotic epithelial cells. Within 4 days, Apc loss induced a dramatic enlargement of the crypt compartment associated with intense cell proliferation, apoptosis and impairment of cell migration. This result confirms the gatekeeper role of Apc in the intestinal epithelium in vivo. Although Apc deletion activated beta-catenin signaling in the villi, we observed neither proliferation nor morphological change in this compartment. This highlights the dramatic difference in the responses of immature and differentiated epithelial cells to aberrant beta-catenin signaling. These distinct biological responses were confirmed by molecular analyses, revealing that Myc and cyclin D1, two canonical beta-catenin target genes, were induced in distinct compartments. We also showed that Apc is a crucial determinant of cell fate in the murine intestinal epithelium. Apc loss perturbs differentiation along the enterocyte, goblet and enteroendocrine lineages, and promotes commitment to the Paneth cell lineage through beta-catenin/Tcf4-mediated transcriptional control of specific markers of Paneth cells, the cryptdin/defensin genes.


Asunto(s)
Diferenciación Celular/fisiología , División Celular/fisiología , Genes APC , Intestinos/fisiología , Células de Paneth/fisiología , Animales , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/genética , Proteínas del Citoesqueleto/fisiología , Defensinas/genética , Defensinas/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/fisiología , Ratones , Ratones Noqueados , Células de Paneth/citología , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Transducción de Señal/fisiología , Transactivadores/fisiología , beta Catenina
18.
Lab Invest ; 84(12): 1619-30, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15502862

RESUMEN

Murine models of familial adenomatous polyposis harbor a germinal heterozygous mutation on Apc tumor suppressor gene. They are valuable tools for studying intestinal carcinogenesis, as most human sporadic cancers contain inactivating mutations of APC. However, Apc(+/-) mice, such as the well-characterized Apc(Min/+) model, develop cancers principally in the small intestine, while humans develop mainly colorectal cancers. We used a Cre-loxP strategy to achieve a new model of germline Apc invalidation in which exon 14 is deleted. We compared the phenotype of these Apc(Delta14/+) mice to that of the classical Apc(Min/+). The main phenotypic difference is the shift of the tumors in the distal colon and rectum, often associated with a rectal prolapse. Thus, the severity of the colorectal phenotype is partly due to the particular mutation Delta14, but also to environmental parameters, as mice raised in conventional conditions developed more colon cancers than those raised in pathogen-free conditions. All lesions, including early lesions, revealed Apc LOH and loss of Apc gene expression. They accumulated beta-catenin, overexpressed the beta-catenin target genes cyclin D1 and c-Myc, and the distribution pattern of glutamine synthetase, a beta-catenin target gene recently identified in the liver, was mosaic in intestinal adenomas. The Apc(Delta14/+) model is thus a useful new tool for studies on the molecular mechanisms of colorectal tumorigenesis.


Asunto(s)
Neoplasias Colorrectales/genética , Neoplasias Colorrectales/fisiopatología , Eliminación de Gen , Genes APC/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Células Clonales , Colon/patología , Neoplasias del Colon/genética , Neoplasias del Colon/patología , Neoplasias Colorrectales/etiología , Neoplasias Colorrectales/patología , ADN de Neoplasias/genética , Modelos Animales de Enfermedad , Ambiente , Exones , Biblioteca de Genes , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , ARN Neoplásico/genética , Neoplasias del Recto/genética , Neoplasias del Recto/patología , Eliminación de Secuencia
19.
Oncogene ; 23(39): 6541-7, 2004 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-15221010

RESUMEN

Previously, we have mimicked human neurofibromatosis type 2 (NF2) in conditional Nf2 mutant (P0Cre;Nf2flox2/flox2) mice. Schwannomas, characteristic for NF2, were found at low frequency in older mice. Here, we report that these mice, upon additional hemizygosity for p53, rapidly develop multiple tumours showing features consistent with malignant peripheral nerve sheath tumours. Thus, p53 hemizygosity promotes tumorigenesis of mutant Nf2 peripheral nerve cells. In contrast, young P0Cre;Nf2flox2/+;p53+/- cis mice mainly succumb to Nf2/p53-related osteogenic tumours. Therefore, Cre-mediated early biallelic loss of Nf2 function in neural crest-derived cells hemizygous for p53 results in resistance to osteogenic tumours and increased susceptibility to peripheral nerve sheath tumours.


Asunto(s)
Genes de la Neurofibromatosis 2 , Genes p53 , Mutación , Neurilemoma/genética , Animales , Genotipo , Inmunohistoquímica , Ratones
20.
Cancer Res ; 64(10): 3718-24, 2004 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-15150133

RESUMEN

Despite the progress made in our understanding of the biology of neurofibromatosis (NF), the long-term clinical outcome for affected patients has not changed significantly in the past decades, and both NF1 and NF2 are still associated with a significant morbidity and a decreased life span. A number of NF1 and NF2 murine models have been generated to aid in the study of NF tumor biology and in the development of targeted therapies for NF patients. A single, universal pathological classification of the lesions generated in these murine models is essential for the validation of the models, for their analysis and comparison with other models, and for their future effective use in preclinical treatment trials. For the formulation of a pathological classification of these lesions, the WHO classification of human tumors was used as a reference. However, it was not adopted for the classification of the GEM lesions because of some important differences between the human and murine lesions. A novel classification scheme for peripheral nerve sheath tumors in murine models was therefore devised.


Asunto(s)
Neoplasias de la Vaina del Nervio/patología , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Neoplasias de la Vaina del Nervio/clasificación , Neurofibromatosis 1/clasificación , Neurofibromatosis 1/patología , Neurofibromatosis 2/clasificación , Neurofibromatosis 2/patología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA