Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Int J Mol Sci ; 23(14)2022 Jul 19.
Artículo en Inglés | MEDLINE | ID: mdl-35887295

RESUMEN

We recently described cell-projection pumping as a mechanism transferring cytoplasm between cells. The uptake of fibroblast cytoplasm by co-cultured SAOS-2 osteosarcoma cells changes SAOS-2 morphology and increases cell migration and proliferation, as seen by single-cell tracking and in FACS separated SAOS-2 from co-cultures. Morphological changes in SAOS-2 seen by single cell tracking are consistent with previous observations in fixed monolayers of SAOS-2 co-cultures. Notably, earlier studies with fixed co-cultures were limited by the absence of a quantitative method for identifying sub-populations of co-cultured cells, or for quantitating transfer relative to control populations of SAOS-2 or fibroblasts cultured alone. We now overcome that limitation by a novel Cartesian plot analysis that identifies individual co-cultured cells as belonging to one of five distinct cell populations, and also gives numerical measure of similarity to control cell populations. We verified the utility of the method by first confirming the previously established relationship between SAOS-2 morphology and uptake of fibroblast contents, and also demonstrated similar effects in other cancer cell lines including from melanomas, and cancers of the ovary and colon. The method was extended to examine global DNA methylation, and while there was no clear effect on SAOS-2 DNA methylation, co-cultured fibroblasts had greatly reduced DNA methylation, similar to cancer associated fibroblasts.


Asunto(s)
Neoplasias Óseas , Osteosarcoma , Neoplasias Óseas/metabolismo , Línea Celular Tumoral , Femenino , Fibroblastos/metabolismo , Humanos , Osteosarcoma/metabolismo , Fenotipo
2.
Biophys J ; 118(6): 1248-1260, 2020 03 24.
Artículo en Inglés | MEDLINE | ID: mdl-32087096

RESUMEN

We earlier reported cytoplasmic fluorescence exchange between cultured human fibroblasts (Fibs) and malignant cells (MCs). Others report similar transfer via either tunneling nanotubes (TNTs) or shed membrane vesicles, and this changes the phenotype of recipient cells. Our time-lapse microscopy showed most exchange was from Fibs into MCs, with less in the reverse direction. Although TNTs were seen, we were surprised transfer was not via TNTs but was instead via fine and often branching cell projections that defied direct visual resolution because of their size and rapid movement. Their structure was revealed nonetheless by their organellar cargo and the grooves they formed indenting MCs, which was consistent with holotomography. Discrete, rapid, and highly localized transfer events evidenced against a role for shed vesicles. Transfer coincided with rapid retraction of the cell projections, suggesting a hydrodynamic mechanism. Increased hydrodynamic pressure in retracting cell projections normally returns cytoplasm to the cell body. We hypothesize "cell-projection pumping" (CPP), in which cytoplasm in retracting cell projections partially equilibrates into adjacent recipient cells via microfusions that form temporary intercellular cytoplasmic continuities. We tested plausibility for CPP by combined mathematical modeling, comparison of predictions from the model with experimental results, and then computer simulations based on experimental data. The mathematical model predicted preferential CPP into cells with lower cell stiffness, expected from equilibration of pressure toward least resistance. Predictions from the model were satisfied when Fibs were cocultured with MCs and fluorescence exchange was related to cell stiffness by atomic force microscopy. When transfer into 5000 simulated recipient MCs or Fibs was studied in computer simulations, inputting experimental cell stiffness and donor cell fluorescence values generated transfers to simulated recipient cells similar to those seen by experiment. We propose CPP as a potentially novel mechanism in mammalian intercellular cytoplasmic transfer and communication.


Asunto(s)
Comunicación Celular , Nanotubos , Animales , Técnicas de Cocultivo , Citoplasma , Citosol , Humanos , Hidrodinámica
3.
Proc Natl Acad Sci U S A ; 114(40): E8448-E8457, 2017 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-28923937

RESUMEN

Gastrointestinal stromal tumors (GISTs) predominantly harbor activating mutations in the receptor tyrosine kinase KIT. To genetically dissect in vivo the requirement of different signal transduction pathways emanating from KIT for tumorigenesis, the oncogenic KitV558Δ mutation was combined with point mutations abrogating specific phosphorylation sites on KIT. Compared with single-mutant KitV558Δ/+ mice, double-mutant KitV558Δ;Y567F/Y567F knock-in mice lacking the SRC family kinase-binding site on KIT (pY567) exhibited attenuated MAPK signaling and tumor growth. Surprisingly, abrogation of the PI3K-binding site (pY719) in KitV558Δ;Y719F/Y719F mice prevented GIST development, although the interstitial cells of Cajal (ICC), the cells of origin of GIST, were normal. Pharmacologic inhibition of the PI3K pathway in tumor-bearing KitV558Δ/+ mice with the dual PI3K/mTOR inhibitor voxtalisib, the pan-PI3K inhibitor pilaralisib, and the PI3K-alpha-restricted inhibitor alpelisib each diminished tumor proliferation. The addition of the MEK inhibitor PD-325901 or binimetinib further decreased downstream KIT signaling. Moreover, combining PI3K and MEK inhibition was effective against imatinib-resistant KitV558Δ;T669I/+ tumors.


Asunto(s)
Carcinogénesis/patología , Resistencia a Antineoplásicos/genética , Tumores del Estroma Gastrointestinal/patología , Mesilato de Imatinib/farmacología , Mutación , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/metabolismo , Animales , Carcinogénesis/genética , Carcinogénesis/metabolismo , Femenino , Neoplasias Gastrointestinales/genética , Neoplasias Gastrointestinales/metabolismo , Neoplasias Gastrointestinales/patología , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/metabolismo , Humanos , Masculino , Ratones , Fosfatidilinositol 3-Quinasas/genética , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/genética , Transducción de Señal , Células Tumorales Cultivadas
4.
J Biol Chem ; 292(37): 15254-15265, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28620047

RESUMEN

Squamous cell carcinoma-related oncogene (SCCRO)/DCUN1D1, a component of the neddylation E3 complex, regulates the activity of the cullin-RING-ligase type of ubiquitination E3s by promoting neddylation of cullin family members. Studies have shown that SCCRO regulates proliferation in vitro and in vivo Here we show that inactivation of SCCRO results in prolonged mitotic time because of delayed and/or failed abscission. The effects of SCCRO on abscission involve its role in neddylation and localization of Cul3 to the midbody. The Cul3 adaptor KLHL21 mediates the effects of SCCRO on abscission, as it fails to localize to the midbody in SCCRO-deficient cells during abscission, and its inactivation resulted in phenotypic changes identical to SCCRO inactivation. Ubiquitination-promoted turnover of Aurora B at the midbody was deficient in SCCRO- and KLHL21-deficient cells, suggesting that it is the target of Cul3KLHL21 at the midbody. Correction of abscission delays in SCCRO-deficient cells with addition of an Aurora B inhibitor at the midbody stage suggests that Aurora B is the target of SCCRO-promoted Cul3KLHL21 activity. The activity of other Cul3-anchored complexes, including Cul3KLHL9/KLHL13, was intact in SCCRO-deficient cells, suggesting that SCCRO selectively, rather than collectively, neddylates cullins in vivo Combined, these findings support a model in which the SCCRO, substrate, and substrate adaptors cooperatively provide tight control of neddylation and cullin-RING-ligase activity in vivo.


Asunto(s)
Proteínas Cullin/metabolismo , Proteínas de Microfilamentos/metabolismo , Procesamiento Proteico-Postraduccional , Proteínas Proto-Oncogénicas/metabolismo , Ubiquitinas/metabolismo , Sustitución de Aminoácidos , Animales , Aurora Quinasa B/genética , Aurora Quinasa B/metabolismo , Biomarcadores/metabolismo , Proteínas de Ciclo Celular , Células Cultivadas , Proteínas Cullin/química , Proteínas Cullin/genética , Proteínas del Citoesqueleto , Embrión de Mamíferos/citología , Péptidos y Proteínas de Señalización Intracelular , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Ratones , Ratones Noqueados , Proteínas de Microfilamentos/química , Proteínas de Microfilamentos/genética , Microscopía Confocal , Mutación , Proteína NEDD8 , Multimerización de Proteína , Transporte de Proteínas , Proteínas Proto-Oncogénicas/genética , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Telofase , Imagen de Lapso de Tiempo
5.
PLoS Pathog ; 11(9): e1005132, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26334306

RESUMEN

Antibiotic resistance among enterococci and γ-proteobacteria is an increasing problem in healthcare settings. Dense colonization of the gut by antibiotic-resistant bacteria facilitates their spread between patients and also leads to bloodstream and other systemic infections. Antibiotic-mediated destruction of the intestinal microbiota and consequent loss of colonization resistance are critical factors leading to persistence and spread of antibiotic-resistant bacteria. The mechanisms underlying microbiota-mediated colonization resistance remain incompletely defined and are likely distinct for different antibiotic-resistant bacterial species. It is unclear whether enterococci or γ-proteobacteria, upon expanding to high density in the gut, confer colonization resistance against competing bacterial species. Herein, we demonstrate that dense intestinal colonization with vancomycin-resistant Enterococcus faecium (VRE) does not reduce in vivo growth of carbapenem-resistant Klebsiella pneumoniae. Reciprocally, K. pneumoniae does not impair intestinal colonization by VRE. In contrast, transplantation of a diverse fecal microbiota eliminates both VRE and K. pneumoniae from the gut. Fluorescence in situ hybridization demonstrates that VRE and K. pneumoniae localize to the same regions in the colon but differ with respect to stimulation and invasion of the colonic mucus layer. While VRE and K. pneumoniae occupy the same three-dimensional space within the gut lumen, their independent growth and persistence in the gut suggests that they reside in distinct niches that satisfy their specific in vivo metabolic needs.


Asunto(s)
Enteritis/microbiología , Enterococcus faecium/fisiología , Infecciones por Bacterias Grampositivas/microbiología , Mucosa Intestinal/microbiología , Infecciones por Klebsiella/microbiología , Klebsiella pneumoniae/fisiología , Enterococos Resistentes a la Vancomicina/fisiología , Ampicilina/efectos adversos , Animales , Antibacterianos/efectos adversos , Antibacterianos/farmacología , Carbapenémicos/farmacología , Recuento de Colonia Microbiana , Farmacorresistencia Bacteriana , Enteritis/patología , Enteritis/prevención & control , Enterococcus faecium/efectos de los fármacos , Enterococcus faecium/crecimiento & desarrollo , Enterococcus faecium/aislamiento & purificación , Trasplante de Microbiota Fecal , Heces/microbiología , Femenino , Microbioma Gastrointestinal/efectos de los fármacos , Infecciones por Bacterias Grampositivas/patología , Infecciones por Bacterias Grampositivas/prevención & control , Interacciones Huésped-Patógeno , Hibridación Fluorescente in Situ , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Infecciones por Klebsiella/patología , Infecciones por Klebsiella/prevención & control , Klebsiella pneumoniae/efectos de los fármacos , Klebsiella pneumoniae/crecimiento & desarrollo , Klebsiella pneumoniae/aislamiento & purificación , Ratones Endogámicos C57BL , Interacciones Microbianas , Organismos Libres de Patógenos Específicos , Enterococos Resistentes a la Vancomicina/efectos de los fármacos , Enterococos Resistentes a la Vancomicina/crecimiento & desarrollo , Enterococos Resistentes a la Vancomicina/aislamiento & purificación
7.
Histochem Cell Biol ; 144(6): 533-42, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26357955

RESUMEN

Differing stimuli affect cell stiffness while cancer metastasis is associated with reduced cell stiffness. Cell stiffness determined by atomic force microscopy has been limited by measurement over nuclei to avoid spurious substratum effects in thin cytoplasmic domains, and we sought to develop a more complete approach including cytoplasmic areas. Ninety µm square fields were recorded from ten separate sites of cultured human dermal fibroblasts (HDF) and three sites each for melanoma (MM39, WM175, and MeIRMu), osteosarcoma (SAOS-2 and U2OS), and ovarian carcinoma (COLO316 and PEO4) cell lines, each site providing 1024 measurements as 32 × 32 square grids. Stiffness recorded below 0.8 µm height was occasionally influenced by substratum, so only stiffness recorded above 0.8 µm was analysed, but all sites were included for height and volume analysis. COLO316 had the lowest cell height and volume, followed by HDF (p < 0.0001) and then PEO4, SAOS-2, MeIRMu, WM175, U2OS, and MM39. HDF were more stiff than all other cells (p < 0.0001), while in descending order of stiffness were PEO4, COLO316, WM175, SAOS-2, U2OS, MM39, and MeIRMu (p < 0.02). Stiffness fingerprints comprised scattergrams of stiffness values plotted against the height at which each stiffness value was recorded and appeared unique for each cell type studied, although in most cases the overall form of fingerprints was similar, with maximum stiffness at low height measurements and a second lower peak occurring at high-height levels. We suggest that our stiffness-fingerprint analytical method provides a more nuanced description than previously reported and will facilitate study of the stiffness response to cell stimulation.


Asunto(s)
Fibroblastos/citología , Microscopía de Fuerza Atómica/métodos , Neoplasias/patología , Células Cultivadas , Humanos
8.
Blood ; 118(4): 1163-74, 2011 Jul 28.
Artículo en Inglés | MEDLINE | ID: mdl-21652679

RESUMEN

During vertebrate angiogenesis, Notch regulates the cell-fate decision between vascular tip cells versus stalk cells. Canonical Notch signaling depends on sequential proteolytic events, whereby interaction of Notch with membrane-anchored ligands triggers proteolytic processing, first by Adam10 and then presenilins. This liberates the Notch intracellular domain, allowing it to enter the nucleus and activate Notch-dependent genes. Here we report that conditional inactivation of Adam10 in endothelial cells (A10ΔEC) recapitulates the increased branching and density of the retinal vasculature that is also caused by interfering with Notch signaling. Moreover, A10ΔEC mice have additional vascular abnormalities, including aberrant subcapsular hepatic veins, enlarged glomeruli, intestinal polyps containing endothelial cell masses, abnormal endochondral ossification, leading to stunted long bone growth and increased pathologic neovascularization following oxygen-induced retinopathy. Our findings support a model in which Adam10 is a crucial regulator of endothelial cell-fate decisions, most likely because of its essential role in canonical Notch signaling.


Asunto(s)
Proteínas ADAM/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Vasos Sanguíneos/embriología , Vasos Sanguíneos/crecimiento & desarrollo , Células Endoteliales/metabolismo , Proteínas de la Membrana/metabolismo , Neovascularización Fisiológica/fisiología , Transducción de Señal/fisiología , Proteína ADAM10 , Animales , Western Blotting , Embrión de Mamíferos , Ratones , Ratones Transgénicos , Receptores Notch/metabolismo , Retina/crecimiento & desarrollo , Vasos Retinianos/crecimiento & desarrollo
9.
Nat Genet ; 36(6): 653-9, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15156143

RESUMEN

Little is known of the molecular mechanisms whereby spermatogonia, mitotic germ cells of the testis, self-renew and differentiate into sperm. Here we show that Zfp145, encoding the transcriptional repressor Plzf, has a crucial role in spermatogenesis. Zfp145 expression was restricted to gonocytes and undifferentiated spermatogonia and was absent in tubules of W/W(v) mutants that lack these cells. Mice lacking Zfp145 underwent a progressive loss of spermatogonia with age, associated with increases in apoptosis and subsequent loss of tubule structure but without overt differentiation defects or loss of the supporting Sertoli cells. Spermatogonial transplantation experiments revealed a depletion of spermatogonial stem cells in the adult. Microarray analysis of isolated spermatogonia from Zfp145-null mice before testis degeneration showed alterations in the expression profile of genes associated with spermatogenesis. These results identify Plzf as a spermatogonia-specific transcription factor in the testis that is required to regulate self-renewal and maintenance of the stem cell pool.


Asunto(s)
Proteínas de Unión al ADN/genética , Espermatogonias/citología , Células Madre/citología , Factores de Transcripción/genética , Animales , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Epigénesis Genética , Expresión Génica , Factores de Transcripción de Tipo Kruppel , Masculino , Ratones , Ratones Noqueados , Proteína de la Leucemia Promielocítica con Dedos de Zinc , Espermatogénesis/genética , Espermatogonias/metabolismo , Espermatogonias/trasplante , Células Madre/metabolismo , Testículo/citología , Testículo/metabolismo , Factores de Transcripción/metabolismo
10.
Nat Cell Biol ; 7(7): 675-85, 2005 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-15965469

RESUMEN

Nijmegen breakage syndrome (NBS) is a chromosomal fragility disorder that shares clinical and cellular features with ataxia telangiectasia. Here we demonstrate that Nbs1-null B cells are defective in the activation of ataxia-telangiectasia-mutated (Atm) in response to ionizing radiation, whereas ataxia-telangiectasia- and Rad3-related (Atr)-dependent signalling and Atm activation in response to ultraviolet light, inhibitors of DNA replication, or hypotonic stress are intact. Expression of the main human NBS allele rescues the lethality of Nbs1-/- mice, but leads to immunodeficiency, cancer predisposition, a defect in meiotic progression in females and cell-cycle checkpoint defects that are associated with a partial reduction in Atm activity. The Mre11 interaction domain of Nbs1 is essential for viability, whereas the Forkhead-associated (FHA) domain is required for T-cell and oocyte development and efficient DNA damage signalling. Reconstitution of Nbs1 knockout mice with various mutant isoforms demonstrates the biological impact of impaired Nbs1 function at the cellular and organismal level.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Proteínas de Ciclo Celular/fisiología , Proteínas de Unión al ADN/metabolismo , Modelos Animales de Enfermedad , Proteínas Nucleares/fisiología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Ácido Anhídrido Hidrolasas , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/inmunología , Linfocitos B/patología , Sitios de Unión/genética , Ciclo Celular/genética , Proteínas de Ciclo Celular/genética , Aberraciones Cromosómicas , Rotura Cromosómica , Trastornos de los Cromosomas/genética , Trastornos de los Cromosomas/metabolismo , Trastornos de los Cromosomas/patología , Daño del ADN/genética , Daño del ADN/fisiología , Enzimas Reparadoras del ADN , Replicación del ADN/genética , Femenino , Predisposición Genética a la Enfermedad/genética , Gónadas/anomalías , Humanos , Cambio de Clase de Inmunoglobulina/genética , Linfoma no Hodgkin/etiología , Linfoma no Hodgkin/genética , Proteína Homóloga de MRE11 , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Mutación , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Fosforilación , Síndrome , Linfocitos T/inmunología , Linfocitos T/patología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo
11.
Cancer Cell ; 4(4): 277-89, 2003 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-14585355

RESUMEN

Angiogenic defects in Id mutant mice inhibit the growth of tumor xenografts, providing a genetic model for antiangiogenic stress. Our work tests the consequences of such stress on progression of more physiological Pten+/- tumors. While tumor growth occurs despite impaired angiogenesis, disruption of vasculature by Id loss causes tumor cells to experience hypoxia and necrosis, the extent of which is tumor dependent. We show that bone-marrow-derived endothelial precursors contribute functionally to neovasculature of some but not all Pten+/- tumors, partially rescuing Id mutant phenotype. We demonstrate that loss of Id1 in tumor endothelial cells results in downregulation of several proangiogenic genes, including alpha6 and beta4 integrins, matrix metalloprotease-2, and fibroblast growth factor receptor-1. Inhibition of these factors phenocopies loss of Id in in vivo angiogenesis assays.


Asunto(s)
Médula Ósea/metabolismo , Células Endoteliales/metabolismo , Neovascularización Patológica/metabolismo , Proteínas Represoras , Factores de Transcripción/metabolismo , Animales , Hipoxia de la Célula , Células Cultivadas , Endotelio Vascular/fisiopatología , Femenino , Proteínas de Peces , Proteína 1 Inhibidora de la Diferenciación , Proteínas Inhibidoras de la Diferenciación , Integrina alfa6/metabolismo , Integrina beta4/metabolismo , Ganglios Linfáticos/fisiopatología , Metaloproteinasa 2 de la Matriz/metabolismo , Ratones , Ratones Noqueados , Proteínas de Neoplasias/metabolismo , Neoplasias Experimentales/fisiopatología , Fosfohidrolasa PTEN , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Proteínas Tirosina Fosfatasas/genética , Proteínas Tirosina Fosfatasas/metabolismo , Proteínas Tirosina Quinasas Receptoras/metabolismo , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos , Receptores de Factores de Crecimiento de Fibroblastos/metabolismo , Trombospondina 1/metabolismo , Factores de Transcripción/genética , Trasplante Heterólogo , Neoplasias Uterinas/fisiopatología
12.
Neoplasia ; 28: 100790, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35398668

RESUMEN

Mutations in IDH1 and IDH2 drive the development of gliomas. These genetic alterations promote tumor cell renewal, disrupt differentiation states, and induce stem-like properties. Understanding how this phenotypic reprogramming occurs remains an area of high interest in glioma research. Previously, we showed that IDH mutation results in the development of a CD24-positive cell population in gliomas. Here, we demonstrate that this CD24-positive population possesses striking stem-like properties at the molecular and phenotypic levels. We found that CD24 expression is associated with stem-like features in IDH-mutant tumors, a patient-derived gliomasphere model, and a neural stem cell model of IDH1-mutant glioma. In orthotopic models, CD24-positive cells display enhanced tumor initiating potency compared to CD24-negative cells. Furthermore, CD24 knockdown results in changes in cell viability, proliferation rate, and gene expression that closely resemble a CD24-negative phenotype. Our data demonstrate that induction of a CD24-positive population is one mechanism by which IDH-mutant tumors acquire stem-like properties. These findings have significant implications for our understanding of the molecular underpinnings of IDH-mutant gliomas.


Asunto(s)
Neoplasias Encefálicas , Glioma , Isocitrato Deshidrogenasa , Células Madre Neoplásicas , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patología , Antígeno CD24/genética , Antígeno CD24/metabolismo , Glioma/genética , Glioma/metabolismo , Glioma/patología , Humanos , Isocitrato Deshidrogenasa/genética , Isocitrato Deshidrogenasa/metabolismo , Mutación , Células Madre Neoplásicas/metabolismo , Células Madre Neoplásicas/patología , Fenotipo
13.
Dev Biol ; 337(2): 199-210, 2010 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-19874813

RESUMEN

Juxtamembrane signaling via the membrane growth factor KitL is critical for Kit mediated functions. KitL has a conserved cytoplasmic domain and has been shown to possess a monomeric leucine-dependent basolateral targeting signal. To investigate the consequences in vivo of impaired basolateral KitL targeting in polarized epithelial cells, we have mutated this critical leucine to alanine using a knock-in strategy. KitL(L263A/L263A) mutant mice are pigmented normally and steady-state hematopoiesis is unaffected although peritoneal and skin mast cell numbers are significantly increased. KitL localization is affected in the Sertoli cells of the KitL(L263A/L263A) testis and testis size is reduced in these mice due to aberrant spermatogonial proliferation. Furthermore, the effect of the KitL L263A mutation on the testicular phenotype is dosage dependent. The tubules of hemizygous KitL(L263A/Sl) mice completely lack germ cells in contrast to the weaker testicular phenotype of KitL(L263A/L263A) mice. The onset of the testis phenotype coincides with the formation of tight junctions between Sertoli cells during postnatal development. Thus, the altered sorting of KitL is dispensable for hematopoietic and melanogenic lineages, yet is crucial in the testicular environment, where the basal membranes of adjacent polarized Sertoli cells form a niche for the proliferating spermatogonia.


Asunto(s)
Polaridad Celular , Hematopoyesis/fisiología , Espermatogénesis/fisiología , Factor de Células Madre/química , Factor de Células Madre/metabolismo , Secuencia de Aminoácidos , Animales , Apoptosis , Recuento de Células , Proliferación Celular , Exones/genética , Marcación de Gen , Linfopoyesis , Masculino , Mastocitos/citología , Ratones , Datos de Secuencia Molecular , Mutación/genética , Estructura Terciaria de Proteína , Transporte de Proteínas , Eliminación de Secuencia , Relación Estructura-Actividad , Testículo/metabolismo , Testículo/patología
14.
Nature ; 437(7055): 147-53, 2005 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-16007073

RESUMEN

Nucleophosmin (also known as NPM, B23, NO38) is a nucleolar protein directly implicated in cancer pathogenesis, as the NPM1 gene is found mutated and rearranged in a number of haematological disorders. Furthermore, the region of chromosome 5 to which NPM1 maps is deleted in a proportion of de novo human myelodysplastic syndromes (MDS), and loss of chromosome 5 is extremely frequent in therapy-related MDS. NPM is a multifunctional protein, and its role in oncogenesis is controversial as NPM has been attributed with both oncogenic and tumour suppressive functions. To study the function of Npm in vivo, we generated a hypomorphic Npm1 mutant series (Npm1+/- < Npm1(hy/hy) < Npm1-/-) in mouse. Here we report that Npm is essential for embryonic development and the maintenance of genomic stability. Npm1-/- and Npm1(hy/hy) mutants have aberrant organogenesis and die between embryonic day E11.5 and E16.5 owing to severe anaemia resulting from defects in primitive haematopoiesis. We show that Npm1 inactivation leads to unrestricted centrosome duplication and genomic instability. We demonstrate that Npm is haploinsufficient in the control of genetic stability and that Npm1 heterozygosity accelerates oncogenesis both in vitro and in vivo. Notably, Npm1+/- mice develop a haematological syndrome with features of human MDS. Our findings uncover an essential developmental role for Npm and implicate its functional loss in tumorigenesis and MDS pathogenesis.


Asunto(s)
Transformación Celular Neoplásica , Desarrollo Embrionario , Neoplasias/metabolismo , Neoplasias/patología , Proteínas Nucleares/metabolismo , Animales , Apoptosis/genética , Ciclo Celular/genética , Transformación Celular Neoplásica/genética , Transformación Celular Neoplásica/patología , Células Cultivadas , Centrosoma/metabolismo , Pérdida del Embrión/genética , Fibroblastos , Eliminación de Gen , Inestabilidad Genómica/genética , Hematopoyesis/genética , Hibridación Fluorescente in Situ , Ratones , Ratones Noqueados , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Neoplasias/genética , Proteínas Nucleares/deficiencia , Proteínas Nucleares/genética , Nucleofosmina
15.
J Exp Med ; 199(6): 867-78, 2004 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-15024050

RESUMEN

The Kit receptor functions in hematopoiesis, lymphocyte development, gastrointestinal tract motility, melanogenesis, and gametogenesis. To investigate the roles of different Kit signaling pathways in vivo, we have generated knock-in mice in which docking sites for PI 3-kinase (KitY719) or Src kinase (KitY567) have been mutated. Whereas steady-state hematopoiesis is normal in KitY719F/Y719F and KitY567F/Y567F mice, lymphopoiesis is affected differentially. The KitY567F mutation, but not the KitY719F mutation, blocks pro T cell and pro B cell development in an age-dependent manner. Thus, the Src family kinase, but not the PI 3-kinase docking site in Kit, mediates a critical signal for lymphocyte development. In agreement with these results, treatment of normal mice with the Kit tyrosine kinase inhibitor imatinib (Gleevec) leads to deficits in pro T and pro B cell development, similar to those seen in KitY567F/Y567F and KitW/W mice. The two mutations do not affect embryonic gametogenesis but the KitY719F mutation blocks spermatogenesis at the spermatogonial stages and in contrast the KitY567F mutation does not affect this process. Therefore, Kit-mediated PI 3-kinase signaling and Src kinase family signaling is highly specific for different cellular contexts in vivo.


Asunto(s)
Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-kit/genética , Proteínas Proto-Oncogénicas c-kit/metabolismo , Transducción de Señal/fisiología , Familia-src Quinasas/metabolismo , Factores de Edad , Animales , Linfocitos B/efectos de los fármacos , Linfocitos B/fisiología , Benzamidas , Western Blotting , Cartilla de ADN , Citometría de Flujo , Técnicas Histológicas , Mesilato de Imatinib , Linfopoyesis/efectos de los fármacos , Linfopoyesis/genética , Masculino , Mastocitos/citología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Mutagénesis Insercional , Mutagénesis Sitio-Dirigida , Mutación/genética , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Piperazinas/farmacología , Reacción en Cadena de la Polimerasa , Pruebas de Precipitina , Pirimidinas/farmacología , Espermatogénesis/genética , Linfocitos T/efectos de los fármacos , Linfocitos T/fisiología , Testículo/anatomía & histología , Familia-src Quinasas/genética
16.
Dev Cell ; 8(3): 353-64, 2005 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-15737931

RESUMEN

The murine septin4 gene (Sept4) has been implicated in diverse cellular functions, including cytokinesis, apoptosis, and tumor suppression. Here, we investigated the function of Sept4 proteins during mouse development by creating a targeted deletion of the Sept4 genomic locus. Sept4 mutant mice are viable but male sterile due to immotile and structurally defective sperm. During spermatogenesis, Sept4 proteins were essential for proper mitochondrial architecture and establishment of the annulus, a ring-like structure in the tail region of sperm. In addition, Sept4 mutant sperm showed defects in the elimination of residual cytoplasm during sperm maturation and had increased staining for the caspase inhibitor XIAP. This is consistent with a role of the proapoptotic Sept4 protein ARTS in promoting caspase-mediated removal of cytoplasm via inhibition of XIAP. Our results indicate that Sept4 proteins play distinct but evolutionarily conserved functions in different cellular compartments.


Asunto(s)
Diferenciación Celular/fisiología , Proteínas del Citoesqueleto/metabolismo , GTP Fosfohidrolasas/metabolismo , Infertilidad Masculina/metabolismo , Espermatogénesis/fisiología , Espermatozoides/citología , Animales , Apoptosis/fisiología , Caspasas/metabolismo , Movimiento Celular/fisiología , Citoplasma/metabolismo , Citoplasma/ultraestructura , Proteínas del Citoesqueleto/genética , GTP Fosfohidrolasas/genética , Masculino , Ratones , Microscopía Electrónica de Transmisión , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , Mutación/genética , Proteínas/metabolismo , Septinas , Espermatozoides/metabolismo , Espermatozoides/ultraestructura , Proteína Inhibidora de la Apoptosis Ligada a X
17.
Blood ; 111(11): 5350-8, 2008 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-18292288

RESUMEN

Overexpression of antiapoptotic members of the Bcl-2 family are observed in approximately 80% of B-cell lymphomas, contributing to intrinsic and acquired drug resistance. Nullifying antiapoptotic function can potentially overcome this in-trinsic and acquired drug resistance. AT-101 is a BH3 mimetic known to be a potent inhibitor of antiapoptotic Bcl-2 family members including Bcl-2, Bcl-X(L), and Mcl-1. In vitro, AT-101 exhibits concentration- and time-dependent cytotoxicity against lymphoma and multiple myeloma cell lines, enhancing the activity of cytotoxic agents. The IC(50) for AT-101 is between 1 and 10 microM for a diverse panel of B-cell lymphomas. AT-101 was synergistic with carfilzomib (C), etoposide (E), doxorubicin (D), and 4-hydroxycyclophosphamide (4-HC) in mantle cell lymphoma (MCL) lines. In a transformed large B-cell lymphoma line (RL), AT-101 was synergistic when sequentially combined with 4-HC, but not when both drugs were added simultaneously. AT-101 also induced potent mitochondrial membrane depolarization (Delta Psi m) and apoptosis when combined with carfilzomib, but not with bortezomib in MCL. In severe combined immunodeficient (SCID) beige mouse models of drug-resistant B-cell lymphoma, 35 mg/kg per day of AT-101 was safe and efficacious. The addition of AT-101 to cyclophosphamide (Cy) and rituximab (R) in a schedule-dependent manner enhanced the efficacy of the conventional therapy.


Asunto(s)
Antineoplásicos Fitogénicos/farmacología , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Gosipol/análogos & derivados , Linfoma de Células B/tratamiento farmacológico , Proteínas Proto-Oncogénicas c-bcl-2/efectos de los fármacos , Animales , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales de Origen Murino , Apoptosis/efectos de los fármacos , Proteína Proapoptótica que Interacciona Mediante Dominios BH3 , Biomimética , Western Blotting , Línea Celular Tumoral , Ciclofosfamida/administración & dosificación , Modelos Animales de Enfermedad , Citometría de Flujo , Gosipol/farmacología , Humanos , Técnicas In Vitro , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Microscopía Confocal , Rituximab , Ensayos Antitumor por Modelo de Xenoinjerto
18.
Blood ; 112(7): 2906-16, 2008 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-18591385

RESUMEN

Overexpression of antiapoptotic members of the Bcl-2 family is observed in approximately 80% of B-cell lymphomas, contributing to intrinsic and acquired drug resistance. Nullifying the antiapoptotic influence of these proteins can potentially overcome this resistance, and may complement conventional chemotherapy. ABT-737 is a BH3-only mimetic and potent inhibitor of the antiapoptotic Bcl-2 family members Bcl-2, Bcl-X(L), and Bcl-w. In vitro, ABT-737 exhibited concentration-dependent cytotoxicity against a broad panel of lymphoma cell lines including mantle cell lymphoma (MCL) and diffuse large B-cell lymphoma (DLBCL). ABT-737 showed synergism when combined with the proteasome inhibitors bortezomib or carfilzomib in select lymphoma cell lines and induced potent mitochondrial membrane depolarization and apoptosis when combined with either. ABT-737 plus bortezomib also induced significant apoptosis in primary samples of MCL, DLBCL, and chronic lymphocytic leukemia (CLL) but no significant cytotoxic effect was observed in peripheral blood mononuclear cells from healthy donors. In severe combined immunodeficient beige mouse models of MCL, the addition of ABT-737 to bortezomib enhanced efficacy compared with either drug alone and with the control. Collectively, these data suggest that ABT-737 alone or in combination with a proteasome inhibitor represents a novel and potentially important platform for the treatment of B-cell malignancies.


Asunto(s)
Antineoplásicos/farmacología , Compuestos de Bifenilo/farmacología , Inhibidores Enzimáticos/farmacología , Linfoma/enzimología , Linfoma/patología , Imitación Molecular/efectos de los fármacos , Nitrofenoles/farmacología , Inhibidores de Proteasoma , Sulfonamidas/farmacología , Animales , Ácidos Borónicos/farmacología , Bortezomib , Muerte Celular/efectos de los fármacos , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Resistencia a Antineoplásicos/efectos de los fármacos , Sinergismo Farmacológico , Salud , Humanos , Leucemia Linfocítica Crónica de Células B/patología , Leucocitos Mononucleares/efectos de los fármacos , Linfoma de Células B Grandes Difuso/patología , Linfoma de Células del Manto/patología , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Ratones , Microscopía Confocal , Piperazinas/farmacología , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Pirazinas/farmacología , Donantes de Tejidos , Ensayos Antitumor por Modelo de Xenoinjerto
19.
Proc Natl Acad Sci U S A ; 104(49): 19506-11, 2007 Dec 04.
Artículo en Inglés | MEDLINE | ID: mdl-18048329

RESUMEN

The establishment of distant metastases depends on the capacity of small numbers of cancer cells to regenerate a tumor after entering a target tissue. The mechanisms that confer this capacity remain to be defined. Here we identify a role for the transcriptional inhibitors of differentiation Id1 and Id3 as selective mediators of lung metastatic colonization in the triple negative [TN, i.e., lacking expression of estrogen receptor and progesterone receptor, and lacking Her2 (human epidermal growth factor receptor 2) amplification] subgroup of human breast cancer. Although broad expression of Id1 has recently been documented in tumors of the rare metaplastic subtype, here we report that rare Id1-expressing cells are also present in the more common TN subset of human breast tumors but not in other subtypes. We also provide evidence that Id1 expression is enriched in clinically obtained hormone receptor negative lung metastases. Functional studies demonstrate that Id1 and its closely related family member Id3 are required for tumor initiating functions, both in the context of primary tumor formation and during metastatic colonization of the lung microenvironment. In vivo characterization of lung metastatic progression reveals that Id1 and Id3 facilitate sustained proliferation during the early stages of metastatic colonization, subsequent to extravasation into the lung parenchyma. These results shed light on the proliferative mechanisms that initiate metastatic colonization, and they implicate Id1 and Id3 as mediators of this malignant function in the TN subgroup of breast cancers.


Asunto(s)
Neoplasias de la Mama/patología , Proteína 1 Inhibidora de la Diferenciación/fisiología , Proteínas Inhibidoras de la Diferenciación/fisiología , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/secundario , Proteínas de Neoplasias/fisiología , Animales , Proliferación Celular , Femenino , Humanos , Proteína 1 Inhibidora de la Diferenciación/análisis , Proteína 1 Inhibidora de la Diferenciación/genética , Proteínas Inhibidoras de la Diferenciación/análisis , Proteínas Inhibidoras de la Diferenciación/genética , Neoplasias Pulmonares/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/análisis , Proteínas de Neoplasias/genética , Trasplante Heterólogo
20.
Science ; 368(6490): 497-505, 2020 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-32355025

RESUMEN

Androgen deprivation is the cornerstone of prostate cancer treatment. It results in involution of the normal gland to ~90% of its original size because of the loss of luminal cells. The prostate regenerates when androgen is restored, a process postulated to involve stem cells. Using single-cell RNA sequencing, we identified a rare luminal population in the mouse prostate that expresses stemlike genes (Sca1 + and Psca +) and a large population of differentiated cells (Nkx3.1 +, Pbsn +). In organoids and in mice, both populations contribute equally to prostate regeneration, partly through androgen-driven expression of growth factors (Nrg2, Rspo3) by mesenchymal cells acting in a paracrine fashion on luminal cells. Analysis of human prostate tissue revealed similar differentiated and stemlike luminal subpopulations that likewise acquire enhanced regenerative potential after androgen ablation. We propose that prostate regeneration is driven by nearly all persisting luminal cells, not just by rare stem cells.


Asunto(s)
Andrógenos/metabolismo , Próstata/fisiología , Próstata/cirugía , Neoplasias de la Próstata/cirugía , Regeneración , Antagonistas de Andrógenos/uso terapéutico , Proteína de Unión a Andrógenos/genética , Animales , Antígenos de Neoplasias/genética , Ataxina-1/genética , Diferenciación Celular/genética , Proteínas Ligadas a GPI/genética , Expresión Génica , Proteínas de Homeodominio/genética , Humanos , Masculino , Células Madre Mesenquimatosas/fisiología , Ratones , Proteínas de Neoplasias/genética , Factores de Crecimiento Nervioso/genética , Tamaño de los Órganos , Organoides/metabolismo , Organoides/fisiología , Próstata/metabolismo , Neoplasias de la Próstata/tratamiento farmacológico , Neoplasias de la Próstata/metabolismo , Regeneración/genética , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Trombospondinas/genética , Factores de Transcripción/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA