Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 56
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Development ; 149(19)2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-36205077

RESUMEN

Notch3 promotes mammary luminal cell specification and forced Notch3 activation can induce mammary tumor formation. However, recent studies suggest a tumor-suppressive role for Notch3. Here, we report on Notch3 expression and functional analysis in the mouse mammary gland. Notch3 is expressed in the luminal compartment throughout mammary gland development, but switches to basal cells with initiation of post-lactational involution. Deletion of Notch3 caused a decrease of Notch activation in luminal cells and diminished luminal progenitors at puberty, as well as reduced alveolar progenitors during pregnancy. Parous Notch3-/- mammary glands developed hyperplasia with accumulation of CD24hiCD49flo cells, some of which progressed to invasive tumors with luminal features. Notch3 deletion abolished Notch activation in basal cells during involution, accompanied by altered apoptosis and reduced brown adipocytes, leading to expansion of parity-identified mammary epithelial cells (PI-MECs). Interestingly, the postpartum microenvironment is required for the stem cell activity of Notch3-/- PI-MECs. Finally, high expression of NOTCH3 is associated with prolonged survival in patients with luminal breast cancer. These results highlight an unexpected tumor-suppressive function for Notch3 in the parous mammary gland through restriction of PI-MEC expansion.


Asunto(s)
Células Epiteliales , Glándulas Mamarias Animales , Animales , Células Epiteliales/metabolismo , Femenino , Lactancia , Ratones , Ratones Transgénicos , Embarazo , Células Madre
2.
Breast Cancer Res ; 26(1): 86, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38807216

RESUMEN

Copy number gains in genes coding for Rho activating exchange factors as well as losses affecting genes coding for RhoGAP proteins are common in breast cancer (BC), suggesting that elevated Rho signaling may play an important role. Extra copies and overexpression of RHOC also occur, although a role for RhoC overexpression in driving tumor formation has not been assessed in vivo. To this end, we report on the development of a Rosa26 (R26)-targeted Cre-conditional RhoC overexpression mouse (R26RhoC). This mouse was crossed to two models for ERBB2/NEU+ breast cancer: one based on expression of an oncogenic ErbB2/Neu cDNA downstream of the endogenous ErbB2 promoter (FloxNeoNeuNT), the other, a metastatic model that is based on high-level expression from MMTV regulatory elements (NIC). RhoC overexpression dramatically enhanced mammary tumor formation in FloxNeoNeuNT mice but showed a more subtle effect in the NIC line, which forms multiple mammary tumors after a very short latency. RhoC overexpression also enhanced mammary tumor formation in an activated Pik3ca model for breast cancer (Pik3caH1047R). The transforming effect of RhoC was associated with epithelial/mesenchymal transition (EMT) in ErbB2/NeuNT and Pik3caH1047R systems. Thus, our study reveals the importance of elevated wildtype Rho protein expression as a driver of breast tumor formation and highlights the significance of Copy Number Abberations that affect Rho signalling.


Asunto(s)
Fosfatidilinositol 3-Quinasa Clase I , Regulación Neoplásica de la Expresión Génica , Fosfatidilinositol 3-Quinasas , Receptor ErbB-2 , Proteínas de Unión al GTP rho , Proteína rhoC de Unión a GTP , Animales , Receptor ErbB-2/metabolismo , Receptor ErbB-2/genética , Femenino , Proteína rhoC de Unión a GTP/metabolismo , Proteína rhoC de Unión a GTP/genética , Ratones , Fosfatidilinositol 3-Quinasa Clase I/genética , Fosfatidilinositol 3-Quinasa Clase I/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Proteínas de Unión al GTP rho/metabolismo , Proteínas de Unión al GTP rho/genética , Humanos , Ratones Transgénicos , Neoplasias Mamarias Experimentales/genética , Neoplasias Mamarias Experimentales/patología , Neoplasias Mamarias Experimentales/metabolismo , Transición Epitelial-Mesenquimal/genética , Neoplasias de la Mama/genética , Neoplasias de la Mama/patología , Neoplasias de la Mama/metabolismo , Transducción de Señal
3.
Immunity ; 30(2): 254-63, 2009 Feb 20.
Artículo en Inglés | MEDLINE | ID: mdl-19217325

RESUMEN

Notch2 activation induced by Delta-like-1 (DL1) drives development of splenic marginal zone (MZ) B cells, an innate-like lineage that protects against sepsis. DL1 interacts with Notch2 weakly, but it is not known whether enhancement of DL1-induced Notch2 activation by Fringe glycosyltransferases is important for MZ B cell development. Furthermore, DL1-expressing cells that promote MZ B cell development have not been identified. We show that Lunatic Fringe (Lfng) and Manic Fringe (Mfng) cooperatively enhanced the DL1-Notch2 interaction to promote MZ B cell development. We also identified radio-resistant red pulp endothelial cells in the splenic MZ that express high amounts of DL1 and promoted MZ B generation. Finally, MZ B cell precursor competition for DL1 homeostatically regulated entry into the MZ B cell pool. Our study has revealed that the Fringe-Notch2 interaction has important functions in vivo and provides insights into mechanisms regulating MZ B cell development.


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Células Endoteliales/inmunología , Glicosiltransferasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas/metabolismo , Bazo/inmunología , Animales , Fusión Artificial Génica , Linfocitos B/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Proteínas de Unión al Calcio , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Células Endoteliales/citología , Glucosiltransferasas , Glicosiltransferasas/deficiencia , Glicosiltransferasas/genética , Glicosiltransferasas/inmunología , Proteínas de Homeodominio , Ratones , Ratones Noqueados , Proteínas/genética , Proteínas/inmunología , ARN Mensajero/genética , Receptor Notch2/metabolismo , Bazo/citología , Bazo/metabolismo , Factor de Transcripción HES-1
4.
Neuroimage ; 163: 220-230, 2017 12.
Artículo en Inglés | MEDLINE | ID: mdl-28882630

RESUMEN

MRI is a powerful modality to detect neuroanatomical differences that result from mutations and treatments. Knowing which genes drive these differences is important in understanding etiology, but candidate genes are often difficult to identify. We tested whether spatial gene expression data from the Allen Brain Institute can be used to inform us about genes that cause neuroanatomical differences. For many single-gene-mutation mouse models, we found that affected neuroanatomy was not strongly associated with the spatial expression of the altered gene and there are specific caveats for each model. However, among models with significant neuroanatomical differences from their wildtype controls, the mutated genes had preferential spatial expression in affected neuroanatomy. In mice exposed to environmental enrichment, candidate genes could be identified by a genome-wide search for genes with preferential spatial expression in the altered neuroanatomical regions. These candidates have functions related to learning and plasticity. We demonstrate that spatial gene expression of single-genes is a poor predictor of altered neuroanatomy, but altered neuroanatomy can identify candidate genes responsible for neuroanatomical phenotypes.


Asunto(s)
Encéfalo/anatomía & histología , Animales , Modelos Animales de Enfermedad , Estudios de Asociación Genética , Ratones , Ratones Endogámicos C57BL , Mutación , Fenotipo
5.
Proc Natl Acad Sci U S A ; 111(52): E5716-23, 2014 Dec 30.
Artículo en Inglés | MEDLINE | ID: mdl-25512545

RESUMEN

Inherited photoreceptor degenerations (IPDs), a group of incurable progressive blinding diseases, are caused by mutations in more than 200 genes, but little is known about the molecular pathogenesis of photoreceptor (PR) death. Increased retinal expression of STAT3 has been observed in response to many retinal insults, including IPDs, but the role of this increase in PR death is unknown. Here, we show that the expression of Stat3 is increased in PRs of the Tg(RHO P347S) and Prph2(rds) (/+) mouse models of IPD and is activated by tyrosine phosphorylation. PR-specific deletion of Stat3 substantially accelerated PR degeneration in both mutant strains. In contrast, increased PR-specific expression of ROSA26 (R26) alleles encoding either WT STAT3 (Stat3(wt)) or the gain-of-function variant STAT3(C) (Stat3(C)) improved PR survival in both models. Moreover, PR signaling in Tg(RHO P347S) mice carrying either a R26-Stat3(wt) or R26-Stat3(C) allele demonstrated increased a-wave amplitude of the scotopic electroretinogram. Phosphorylation of STAT3 at tyrosine 705 was required for the prosurvival effect because an R26-Stat3(Y705F) allele was not protective. The prosurvival role of enhanced Stat3 activity was validated using recombinant adenoassociated virus (rAAV) vector-mediated PR Stat3 expression in Tg(RHO P347S) mice. Our findings (i) establish that the increase in endogenous PR Stat3 expression is a protective response in IPDs, (ii) suggest that therapeutic augmentation of PR Stat3 expression has potential as a common neuroprotective therapy for these disorders, and (iii) indicate that prosurvival molecules whose expression is increased in mutant PRs may have promise as novel therapies for IPDs.


Asunto(s)
Enfermedades Genéticas Congénitas/metabolismo , Mutación , Células Fotorreceptoras de Vertebrados/metabolismo , Degeneración Retiniana/metabolismo , Factor de Transcripción STAT3/metabolismo , Animales , Supervivencia Celular/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/genética , Enfermedades Genéticas Congénitas/genética , Enfermedades Genéticas Congénitas/patología , Ratones , Ratones Transgénicos , Células Fotorreceptoras de Vertebrados/patología , ARN no Traducido/biosíntesis , ARN no Traducido/genética , Degeneración Retiniana/genética , Degeneración Retiniana/patología , Factor de Transcripción STAT3/genética
6.
Proc Natl Acad Sci U S A ; 109(15): 5832-7, 2012 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-22460789

RESUMEN

Human Epidermal Growth Factor Receptor 2-positive (HER2(+)) breast cancer (BC) is a highly aggressive disease commonly treated with chemotherapy and anti-HER2 drugs, including trastuzumab. There is currently no way to predict which HER2(+) BC patients will benefit from these treatments. Previous prognostic signatures for HER2(+) BC were developed irrespective of the subtype or the hierarchical organization of cancer in which only a fraction of cells, tumor-initiating cells (TICs), can sustain tumor growth. Here, we used serial dilution and single-cell transplantation assays to identify MMTV-Her2/Neu mouse mammary TICs as CD24(+):JAG1(-) at a frequency of 2-4.5%. A 17-gene Her2-TIC-enriched signature (HTICS), generated on the basis of differentially expressed genes in TIC versus non-TIC fractions and trained on one HER2(+) BC cohort, predicted clinical outcome on multiple independent HER2(+) cohorts. HTICS included up-regulated genes involved in S/G2/M transition and down-regulated genes involved in immune response. Its prognostic power was independent of other predictors, stratified lymph node(+) HER2(+) BC into low and high-risk subgroups, and was specific for HER2(+):estrogen receptor alpha-negative (ERα(-)) patients (10-y overall survival of 83.6% for HTICS(-) and 24.0% for HTICS(+) tumors; hazard ratio = 5.57; P = 0.002). Whereas HTICS was specific to HER2(+):ERα(-) tumors, a previously reported stroma-derived signature was predictive for HER2(+):ERα(+) BC. Retrospective analyses revealed that patients with HTICS(+) HER2(+):ERα(-) tumors resisted chemotherapy but responded to chemotherapy plus trastuzumab. HTICS is, therefore, a powerful prognostic signature for HER2(+):ERα(-) BC that can be used to identify high risk patients that would benefit from anti-HER2 therapy.


Asunto(s)
Neoplasias de la Mama/genética , Receptor alfa de Estrógeno/metabolismo , Perfilación de la Expresión Génica , Genes Relacionados con las Neoplasias/genética , Células Madre Neoplásicas/patología , Receptor ErbB-2/metabolismo , Animales , Anticuerpos Monoclonales Humanizados/farmacología , Anticuerpos Monoclonales Humanizados/uso terapéutico , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Antígeno CD24/metabolismo , Proteínas de Unión al Calcio/metabolismo , Diferenciación Celular/efectos de los fármacos , División Celular/efectos de los fármacos , Femenino , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteína Jagged-1 , Proteínas de la Membrana/metabolismo , Ratones , Terapia Neoadyuvante , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/metabolismo , Pronóstico , Proteínas Serrate-Jagged , Transducción de Señal/efectos de los fármacos , Trastuzumab , Resultado del Tratamiento
7.
J Neurosci ; 33(9): 4055-65, 2013 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-23447614

RESUMEN

Invertebrate studies have highlighted a role for EH and SH3 domain Intersectin (Itsn) proteins in synaptic vesicle recycling and morphology. Mammals have two Itsn genes (Itsn1 and Itsn2), both of which can undergo alternative splicing to include DBL/PH and C2 domains not present in invertebrate Itsn proteins. To probe for specific and redundant functions of vertebrate Itsn genes, we generated Itsn1, Itsn2, and double mutant mice. While invertebrate mutants showed severe synaptic abnormalities, basal synaptic transmission and plasticity were unaffected at Schaffer CA1 synapses in mutant mice. Surprisingly, intercortical tracts-corpus callosum, ventral hippocampal, and anterior commissures-failed to cross the midline in mice lacking Itsn1, but not Itsn2. In contrast, tracts extending within hemispheres and those that decussate to more caudal brain segments appeared normal. Itsn1 mutant mice showed severe deficits in Morris water maze and contextual fear memory tasks, whereas mice lacking Itsn2 showed normal learning and memory. Thus, coincident with the acquisition of additional signaling domains, vertebrate Itsn1 has been functionally repurposed to also facilitate interhemispheric connectivity essential for high order cognitive functions.


Asunto(s)
Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Corteza Cerebral/citología , Corteza Cerebral/crecimiento & desarrollo , Cognición/fisiología , Cuerpo Calloso/fisiología , Lateralidad Funcional/genética , 2-Amino-5-fosfonovalerato/farmacología , Proteínas Adaptadoras del Transporte Vesicular/genética , Análisis de Varianza , Animales , Biofisica , Mapeo Encefálico , Condicionamiento Psicológico/efectos de los fármacos , Condicionamiento Psicológico/fisiología , Señales (Psicología) , Imagen de Difusión Tensora , Embrión de Mamíferos , Antagonistas de Aminoácidos Excitadores/farmacología , Conducta Exploratoria/fisiología , Miedo , Conos de Crecimiento/efectos de los fármacos , Conos de Crecimiento/fisiología , Hipocampo/citología , Imagenología Tridimensional , Técnicas In Vitro , Discapacidades para el Aprendizaje/genética , Imagen por Resonancia Magnética , Aprendizaje por Laberinto/fisiología , Trastornos de la Memoria/genética , Ratones , Ratones Transgénicos , Mutación/genética , Fibras Nerviosas/fisiología , Plasticidad Neuronal/efectos de los fármacos , Plasticidad Neuronal/genética , Neuronas/citología , Neuronas/efectos de los fármacos , Neuronas/fisiología , Desempeño Psicomotor/efectos de los fármacos , Desempeño Psicomotor/fisiología , Ratas , Ratas Wistar , Transmisión Sináptica/efectos de los fármacos , Transmisión Sináptica/genética , Vertebrados/genética , Vertebrados/metabolismo
8.
Am J Physiol Endocrinol Metab ; 307(7): E596-610, 2014 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-25117410

RESUMEN

The multidrug resistance efflux transporter ATP-binding cassette subfamily G member 2 (ABCG2) is not only overexpressed in certain drug-resistant cancers but is also highly expressed in the mammary gland during lactation, carrying xenobiotics and nutrients into milk. We sought to investigate the molecular mechanisms involved in the upregulation of ABCG2 during lactation. Expression profiling of different mouse Abcg2 mRNA isoforms (E1a, E1b, and E1c) revealed that E1b is predominantly expressed and induced in the lactating mouse mammary gland. Despite this induction, analyses of CpG methylation status and published ChIP-seq datasets reveal that E1b promoter sequences in the virgin gland are already hypomethylated and marked with the open chromatin histone mark H3K4me2. Using a forced-weaning model to shut down lactation, we found that within 24 h there was a significant reduction in Abcg2 mRNA expression and a loss of signal transducer and activator of transcription-5 (STAT5) occupancy at the mouse Abcg2 gene. Luciferase reporter assays further showed that some of these STAT5-binding regions that contained interferon-γ-activated sequence (GAS) motifs function as an enhancer after prolactin treatment. We conclude that Abcg2 is already poised for expression in the virgin mammary gland and that STAT5 plays an important role in Abcg2 expression during lactation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/genética , Epigénesis Genética , Células Epiteliales/metabolismo , Lactancia/genética , Glándulas Mamarias Animales/metabolismo , Isoformas de ARN/genética , ARN Mensajero/genética , Factor de Transcripción STAT5/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Islas de CpG , Metilación de ADN , Femenino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos , Regiones Promotoras Genéticas , Transducción de Señal , Regulación hacia Arriba
9.
Blood ; 119(19): 4486-98, 2012 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-22451425

RESUMEN

The miR-17-92 cluster and its 6 encoded miRNAs are frequently amplified and aberrantly expressed in various malignancies. This study demonstrates that retroviral-mediated miR-17-92 overexpression promotes expansion of multipotent hematopoietic progenitors in mice. Cell lines derived from these miR-17-92-overexpressing mice are capable of myeloid and lymphoid lineage differentiation, and recapitulate the normal lymphoid phenotype when transplanted to nonobese diabetic/severe combined immunodeficiency mice. However, overexpression of individual miRNAs from this locus, miR-19a or miR-92a, results in B-cell hyperplasia and erythroleukemia, respectively. Coexpression of another member of this cluster miR-17, with miR-92a, abrogates miR-92a-induced erythroleukemogenesis. Accordingly, we identified several novel miR-92a and miR-17 target genes regulating erythroid survival and proliferation, including p53. Expression of this critical target results in marked growth inhibition of miR-92a erythroleukemic cells. In both murine and human leukemias, p53 inactivation contributed to the selective overexpression of oncogenic miR-92a and miR-19a, and down-regulation of tumor-suppressive miR-17. This miR-17-92 expression signature was also detected in p53- B-cell chronic lymphocytic leukemia patients displaying an aggressive clinical phenotype. These results revealed that imbalanced miR-17-92 expression, also mediated by p53, directly transforms the hematopoietic compartment. Thus examination of such miRNA expression signatures should aid in the diagnosis and treatment of cancers displaying miR-17-92 gene amplification.


Asunto(s)
Células Madre Hematopoyéticas/fisiología , Leucemia/genética , MicroARNs/genética , Animales , Animales Recién Nacidos , Transformación Celular Neoplásica/genética , Células Cultivadas , Regulación Leucémica de la Expresión Génica , Células HL-60 , Células Madre Hematopoyéticas/metabolismo , Humanos , Células K562 , Leucemia/patología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones SCID , Ratones Transgénicos , Familia de Multigenes/genética , Células Madre Multipotentes/metabolismo , Células Madre Multipotentes/fisiología , Células 3T3 NIH , ARN Largo no Codificante
10.
Dev Dyn ; 242(6): 678-86, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23526493

RESUMEN

BACKGROUND: The Notch signaling pathway plays complex roles in developing lungs, including regulation of proximodistal fates, airway cell specification and differentiation. However, the specific Notch-mediated signals involved in lung development remain unclear. RESULTS: Here we report that Jagged1 is expressed in a subset of bronchial and bronchiolar epithelial cells, where it controls proximal airway cell fate and differentiation. In agreement with previous studies involving disruption of all Notch signaling, we found that deletion of Jagged1 in airway epithelium increased the number of ciliated cells at the expense of Clara cells, a phenotype associated with downregulation of Hes1. Deletion of Jagged1 also led to an increased number of pulmonary neuroendocrine cells (PNEC), suggesting that Jagged1/Notch signaling inhibits PNEC cell fate. As expected, Jagged1 deletion did not affect alveolar cell differentiation, although alveolar septation was impaired, likely an indirect effect of proximal airway defects. Finally, in the postnatal lung, Jagged1 deletion induced mucous metaplasia, accompanied by downregulation of Hes1 and Hes5. CONCLUSIONS: Our results demonstrate that Jagged1-mediated Notch signaling regulates multiple cell fate decisions as well as differentiation in the respiratory system to coordinate lung development and to maintain a balance of airway cell types in adult life.


Asunto(s)
Proteínas de Unión al Calcio/fisiología , Linaje de la Célula , Regulación del Desarrollo de la Expresión Génica , Péptidos y Proteínas de Señalización Intercelular/fisiología , Pulmón/embriología , Proteínas de la Membrana/fisiología , Receptores Notch/metabolismo , Animales , Proteínas de Unión al Calcio/genética , Diferenciación Celular , Células Epiteliales/metabolismo , Inmunohistoquímica , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína Jagged-1 , Pulmón/metabolismo , Proteínas de la Membrana/genética , Ratones , Ratones Transgénicos , Células Neuroendocrinas/metabolismo , Fenotipo , Proteínas Serrate-Jagged , Transducción de Señal , Factores de Tiempo
11.
Blood ; 117(4): 1184-95, 2011 Jan 27.
Artículo en Inglés | MEDLINE | ID: mdl-21097675

RESUMEN

Lunatic Fringe (Lfng) enhances Notch1 activation by Delta-like 4 (DL4) to promote Notch1-dependent T-lineage commitment of thymus-seeding progenitors. Subsequently, Notch1 and T-cell receptor-ß (TCRß)-containing pre-TCR complexes signal CD4/CD8 double-negative 3 (DN3) committed T-cell progenitors to survive, proliferate, and differentiate into CD4/CD8 double-positive (DP) αß T-cell precursors. Few DP thymocytes develop without Notch1 or pre-TCR signals, whereas ectopic Notch1 activation causes T-cell leukemia. However, mechanisms of a Notch-pre-TCR collaboration during this "ß-selection" process are poorly understood. We genetically manipulated Lfng to attenuate or enhance Notch1 activation in DN3 thymocytes without inducing leukemogenesis. We show that Lfng temporally sustains DL-induced Notch1 signaling to prolong proliferative self-renewal of pre-DP thymocytes. Pre-TCR signaling greatly augmented Notch trophic functions to promote robust proliferation of pre-DP progenitors. In contrast, in the absence of DL/Notch signaling, pre-TCR-expressing progenitors rapidly atrophied and differentiated into DP thymocytes. Thus, Lfng prolongs Notch1 signaling to promote self-renewal more than differentiation during the early stages of ß-selection. Our data provide novel insights into the Notch-pre-TCR collaboration, and suggest that decreasing Lfng expression during the DN3-DP transition minimizes the potent leukemogenic potential of Notch1 signaling.


Asunto(s)
Proliferación Celular , Glicosiltransferasas/fisiología , Péptidos y Proteínas de Señalización Intracelular/fisiología , Células Progenitoras Linfoides/fisiología , Proteínas de la Membrana/fisiología , Receptor Notch1/fisiología , Receptores de Antígenos de Linfocitos T alfa-beta/metabolismo , Linfocitos T/fisiología , Proteínas Adaptadoras Transductoras de Señales , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/fisiología , Proteínas de Unión al Calcio , Células Cultivadas , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intracelular/genética , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Células Progenitoras Linfoides/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptor Notch1/genética , Receptor Notch1/metabolismo , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/metabolismo
12.
Neurobiol Learn Mem ; 103: 39-49, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23567106

RESUMEN

It is well-known that Notch signaling plays a critical role in brain development and growing evidence implicates this signaling pathway in adult synaptic plasticity and memory formation. The Notch1 receptor is activated by two subclasses of ligands, Delta-like (including Dll1 and Dll4) and Jagged (including Jag1 and Jag2). Ligand-induced Notch1 receptor signaling is modulated by a family of Fringe proteins, including Lunatic fringe (Lfng). Although Dll1, Jag1 and Lfng are critical regulators of Notch signaling, their relative contribution to memory formation in the adult brain is unknown. To investigate the roles of these important components of Notch signaling in memory formation, we examined spatial and fear memory formation in adult mice with reduced expression of Dll1, Jag1, Lfng and Dll1 plus Lfng. We also examined motor activity, anxiety-like behavior and sensorimotor gating using the acoustic startle response in these mice. Of the lines of mutant mice tested, we found that only mice with reduced Jag1 expression (mice heterozygous for a null mutation in Jag1, Jag1(+/-)) showed a selective impairment in spatial memory formation. Importantly, all other behavior including open field activity, conditioned fear memory (both context and discrete cue), acoustic startle response and prepulse inhibition, was normal in this line of mice. These results provide the first in vivo evidence that Jag1-Notch signaling is critical for memory formation in the adult brain.


Asunto(s)
Proteínas de Unión al Calcio/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Proteínas de la Membrana/metabolismo , Memoria/fisiología , Receptores Notch/metabolismo , Transducción de Señal/fisiología , Percepción Espacial/fisiología , Animales , Ansiedad/genética , Ansiedad/metabolismo , Proteínas de Unión al Calcio/genética , Miedo/fisiología , Glicosiltransferasas/genética , Glicosiltransferasas/metabolismo , Péptidos y Proteínas de Señalización Intercelular/genética , Proteína Jagged-1 , Proteína Jagged-2 , Proteínas de la Membrana/genética , Ratones , Actividad Motora/fisiología , Receptores Notch/genética , Reflejo de Sobresalto/genética , Proteínas Serrate-Jagged
13.
Neurobiol Learn Mem ; 105: 20-30, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23850596

RESUMEN

It is well-known that Notch signaling plays a critical role in brain development and growing evidence implicates this signaling pathway in adult synaptic plasticity and memory formation. The Notch1 receptor is activated by two subclasses of ligands, Delta-like (including Dll1 and Dll4) and Jagged (including Jag1 and Jag2). Ligand-induced Notch1 receptor signaling is modulated by a family of Fringe proteins, including Lunatic fringe (Lfng). Although Dll1, Jag1 and Lfng are critical regulators of Notch signaling, their relative contribution to memory formation in the adult brain is unknown. To investigate the roles of these important components of Notch signaling in memory formation, we examined spatial and fear memory formation in adult mice with reduced expression of Dll1, Jag1, Lfng and Dll1 plus Lfng. We also examined motor activity, anxiety-like behavior and sensorimotor gating using the acoustic startle response in these mice. Of the lines of mutant mice tested, we found that only mice with reduced Jag1 expression (mice heterozygous for a null mutation in Jag1, Jag1(+/-)) showed a selective impairment in spatial memory formation. Importantly, all other behavior including open field activity, conditioned fear memory (both context and discrete cue), acoustic startle response and prepulse inhibition, was normal in this line of mice. These results provide the first in vivo evidence that Jag1-Notch signaling is critical for memory formation in the adult brain.

14.
J Am Soc Nephrol ; 23(1): 27-35, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22052054

RESUMEN

Notch signaling in podocytes causes proteinuria and glomerulosclerosis in humans and rodents, but the underlying mechanism remains unknown. Here, we analyzed morphologic, molecular, and cellular events before the onset of proteinuria in newborn transgenic mice that express activated Notch in podocytes. Immunohistochemistry revealed a loss of the slit diaphragm protein nephrin exclusively in podocytes expressing activated Notch. Podocyte-specific deletion of Rbpj, which is essential for canonical Notch signaling, prevented this loss of nephrin. Overexpression of activated Notch decreased cell surface nephrin and increased cytoplasmic nephrin in transfected HEK293T cells; pharmacologic inhibition of dynamin, but not depletion of cholesterol, blocked these effects on nephrin, suggesting that Notch promotes dynamin-dependent, raft-independent endocytosis of nephrin. Supporting an association between Notch signaling and nephrin trafficking, electron microscopy revealed shortened podocyte foot processes and fewer slit diaphragms among the transgenic mice compared with controls. These data suggest that Notch signaling induces endocytosis of nephrin, thereby triggering the onset of proteinuria.


Asunto(s)
Endocitosis , Proteínas de la Membrana/metabolismo , Podocitos/metabolismo , Proteinuria/etiología , Receptores Notch/metabolismo , Animales , Animales Recién Nacidos , Dinaminas/metabolismo , Células HEK293 , Humanos , Ratones , Ratones Transgénicos , Podocitos/ultraestructura , Proteinuria/metabolismo , Proteinuria/patología , Transducción de Señal , beta-Ciclodextrinas
15.
Nat Commun ; 14(1): 4313, 2023 07 18.
Artículo en Inglés | MEDLINE | ID: mdl-37463901

RESUMEN

Metastatic breast-cancer is a major cause of death in women worldwide, yet the relationship between oncogenic drivers that promote metastatic versus primary cancer is still contentious. To elucidate this relationship in treatment-naive animals, we hereby describe mammary-specific transposon-mutagenesis screens in female mice together with loss-of-function Rb, which is frequently inactivated in breast-cancer. We report gene-centric common insertion-sites (gCIS) that are enriched in primary-tumors, in metastases or shared by both compartments. Shared-gCIS comprise a major MET-RAS network, whereas metastasis-gCIS form three additional hubs: Rho-signaling, Ubiquitination and RNA-processing. Pathway analysis of four clinical cohorts with paired primary-tumors and metastases reveals similar organization in human breast-cancer with subtype-specific shared-drivers (e.g. RB1-loss, TP53-loss, high MET, RAS, ER), primary-enriched (EGFR, TGFß and STAT3) and metastasis-enriched (RHO, PI3K) oncogenic signaling. Inhibitors of RB1-deficiency or MET plus RHO-signaling cooperate to block cell migration and drive tumor cell-death. Thus, targeting shared- and metastasis- but not primary-enriched derivers offers a rational avenue to prevent metastatic breast-cancer.


Asunto(s)
Neoplasias de la Mama , Femenino , Humanos , Animales , Ratones , Neoplasias de la Mama/patología , Transducción de Señal , Metástasis de la Neoplasia
16.
Adv Exp Med Biol ; 727: 89-98, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22399341

RESUMEN

Notch signaling plays an essential role in development and homeostasis of multiple organs including the lung. Dysregulation of Notch signaling has been implicated in various lung diseases including lung cancer. Here we review functions of Notch signaling in coordinating events during lung development, such as early proximodistal fate generation and branching, airway epithelial cell fate specification, alveogenesis and pulmonary vascular development. We also discuss roles of Notch in chronic obstructive pulmonary disease, progressive pulmonary fibrosis, pulmonary arterial hypertension, asthma and lung cancer.


Asunto(s)
Enfermedades Pulmonares/metabolismo , Enfermedades Pulmonares/patología , Pulmón/citología , Pulmón/metabolismo , Receptores Notch/metabolismo , Transducción de Señal , Animales , Humanos
17.
Cancer Discov ; 12(12): 2930-2953, 2022 12 02.
Artículo en Inglés | MEDLINE | ID: mdl-36108220

RESUMEN

Systematically investigating the scores of genes mutated in cancer and discerning disease drivers from inconsequential bystanders is a prerequisite for precision medicine but remains challenging. Here, we developed a somatic CRISPR/Cas9 mutagenesis screen to study 215 recurrent "long-tail" breast cancer genes, which revealed epigenetic regulation as a major tumor-suppressive mechanism. We report that components of the BAP1 and COMPASS-like complexes, including KMT2C/D, KDM6A, BAP1, and ASXL1/2 ("EpiDrivers"), cooperate with PIK3CAH1047R to transform mouse and human breast epithelial cells. Mechanistically, we find that activation of PIK3CAH1047R and concomitant EpiDriver loss triggered an alveolar-like lineage conversion of basal mammary epithelial cells and accelerated formation of luminal-like tumors, suggesting a basal origin for luminal tumors. EpiDriver mutations are found in ∼39% of human breast cancers, and ∼50% of ductal carcinoma in situ express casein, suggesting that lineage infidelity and alveogenic mimicry may significantly contribute to early steps of breast cancer etiology. SIGNIFICANCE: Infrequently mutated genes comprise most of the mutational burden in breast tumors but are poorly understood. In vivo CRISPR screening identified functional tumor suppressors that converged on epigenetic regulation. Loss of epigenetic regulators accelerated tumorigenesis and revealed lineage infidelity and aberrant expression of alveogenesis genes as potential early events in tumorigenesis. This article is highlighted in the In This Issue feature, p. 2711.


Asunto(s)
Neoplasias de la Mama , Carcinoma Intraductal no Infiltrante , Humanos , Ratones , Animales , Femenino , Neoplasias de la Mama/patología , Epigénesis Genética , Recurrencia Local de Neoplasia/genética , Carcinoma Intraductal no Infiltrante/genética , Transformación Celular Neoplásica/genética
18.
Cancers (Basel) ; 13(4)2021 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-33578666

RESUMEN

Metastatic dissemination of cancer cells, their colonization at distal sites, and ultimate disruption of tissue physiology are the root causes of most deaths from solid cancers, particularly in tumor types where the primary lesion can be easily dissected and discarded [...].

19.
EMBO Mol Med ; 13(3): e13807, 2021 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-33616312

RESUMEN

Invasive lobular carcinoma (ILC) of the breast is a very common disease. Despite its prevalence, these tumors are relatively understudied. One reason for this is a relative lack of models for ILC. This challenge was addressed by Brisken and colleagues through development of an intraductal injection-based xenograft system for the study of ERα+ breast cancers, including both ILC and more common invasive ductal carcinoma (IDC; Sflomos et al, 2016). In this issue of EMBO Molecular Medicine, the same group have applied intraductal injection-based xenografts to identify novel tumor cell-specific transcriptional signatures in ILC (Sflomos et al, 2021). In doing so they found overexpression of lysyl oxidase-like 1 (LOXL1) to be both responsible for the frequently seen stiff collagen-rich extracellular matrix of lobular breast cancer and essential for their robust growth and metastatic dissemination in vivo, thereby identifying a novel therapeutic target.


Asunto(s)
Neoplasias de la Mama , Carcinoma Ductal de Mama , Carcinoma Lobular , Mama , Femenino , Humanos , Estudios Retrospectivos
20.
Front Cell Dev Biol ; 9: 645386, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33996804

RESUMEN

Epigenetic and chromatin regulation of craniofacial development remains poorly understood. Ankyrin Repeat Domain 11 (ANKRD11) is a chromatin regulator that has previously been shown to control neural stem cell fates via modulation of histone acetylation. ANKRD11 gene variants, or microdeletions of the 16q24.3 chromosomal region encompassing the ANKRD11 gene, cause KBG syndrome, a rare autosomal dominant congenital disorder with variable neurodevelopmental and craniofacial involvement. Craniofacial abnormalities include a distinct facial gestalt, delayed bone age, tooth abnormalities, delayed fontanelle closure, and frequently cleft or submucosal palate. Despite this, the dramatic phenotype and precise role of ANKRD11 in embryonic craniofacial development remain unexplored. Quantitative analysis of 3D images of KBG syndromic subjects shows an overall reduction in the size of the middle and lower face. Here, we report that mice with heterozygous deletion of Ankrd11 in neural crest cells (Ankrd11nchet) display a mild midfacial hypoplasia including reduced midfacial width and a persistent open fontanelle, both of which mirror KBG syndrome patient facial phenotypes. Mice with a homozygous Ankrd11 deletion in neural crest cells (Ankrd11ncko) die at birth. They show increased severity of several clinical manifestations described for KBG syndrome, such as cleft palate, retrognathia, midfacial hypoplasia, and reduced calvarial growth. At E14.5, Ankrd11 expression in the craniofacial complex is closely associated with developing bony structures, while expression at birth is markedly decreased. Conditional deletion of Ankrd11 leads to a reduction in ossification of midfacial bones, with several ossification centers failing to expand and/or fuse. Intramembranous bones show features of delayed maturation, with bone remodeling severely curtailed at birth. Palatal shelves remain hypoplastic at all developmental stages, with a local reduction in proliferation at E13.5. Our study identifies Ankrd11 as a critical regulator of intramembranous ossification and palate development and suggests that Ankrd11nchet and Ankrd11ncko mice may serve as pre-clinical models for KBG syndrome in humans.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA