Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Blood ; 123(25): 3914-24, 2014 Jun 19.
Artículo en Inglés | MEDLINE | ID: mdl-24802772

RESUMEN

The histone methyltransferase EZH2 is frequently mutated in germinal center-derived diffuse large B-cell lymphoma and follicular lymphoma. To further characterize these EZH2 mutations in lymphomagenesis, we generated a mouse line where EZH2(Y641F) is expressed from a lymphocyte-specific promoter. Spleen cells isolated from the transgenic mice displayed a global increase in trimethylated H3K27, but the mice did not show an increased tendency to develop lymphoma. As EZH2 mutations often coincide with other mutations in lymphoma, we combined the expression of EZH2(Y641F) by crossing these transgenic mice with Eµ-Myc transgenic mice. We observed a dramatic acceleration of lymphoma development in this combination model of Myc and EZH2(Y641F). The lymphomas show histologic features of high-grade disease with a shift toward a more mature B-cell phenotype, increased cycling and gene expression, and epigenetic changes involving important pathways in B-cell regulation and function. Furthermore, they initiate disease in secondary recipients. In summary, EZH2(Y641F) can collaborate with Myc to accelerate lymphomagenesis demonstrating a cooperative role of EZH2 mutations in oncogenesis. This murine lymphoma model provides a new tool to study global changes in the epigenome caused by this frequent mutation and a promising model system for testing novel treatments.


Asunto(s)
Transformación Celular Neoplásica/genética , Linfoma/genética , Mutación , Complejo Represivo Polycomb 2/genética , Proteínas Proto-Oncogénicas c-myc/genética , Animales , Linfocitos B/metabolismo , Linfocitos B/patología , Western Blotting , Células de la Médula Ósea/metabolismo , Proliferación Celular , Transformación Celular Neoplásica/metabolismo , Modelos Animales de Enfermedad , Femenino , Citometría de Flujo , Perfilación de la Expresión Génica , Histonas/metabolismo , Humanos , Estimación de Kaplan-Meier , Linfoma/metabolismo , Linfoma/patología , Linfoma de Células B/genética , Linfoma de Células B/metabolismo , Linfoma de Células B/patología , Lisina/metabolismo , Masculino , Metilación , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Complejo Represivo Polycomb 2/metabolismo , Proteínas Proto-Oncogénicas c-myc/metabolismo , Bazo/metabolismo , Bazo/patología
2.
Can J Infect Dis Med Microbiol ; 2016: 8207418, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27366181

RESUMEN

Background. Clostridium difficile is a major cause of gastrointestinal illness. Epidemic NAP1 strains contain toxins A and B, a deletion in repressor tcdC, and a binary toxin. Objectives. To determine the molecular epidemiology of C. difficile in British Columbia and compare between two time points in one region. Methods. C. difficile isolates from hospital and community laboratories (2008) and one Island Health hospital laboratory (2013) were characterized by pulsed-field gel electrophoresis, PCR-ribotyping, toxin possession, tcdC genotype, and antimicrobial susceptibility. Results. In 2008, 42.7% of isolates had NAP1 designation. Hospital-collected isolates were associated with older patients and more NAP1 types. Unlike other isolates, most NAP1 isolates possessed binary toxin and a 19 bp loss in tcdC. All isolates were susceptible to metronidazole and vancomycin. A 2013 follow-up revealed a 28.9% decrease in NAP1 isolates and 20.0% increase in isolates without NAP designation in one region. Then, community-associated cases were seen in younger patients, while NAP types were evenly distributed. Isolates without NAP designation did not cluster with a PFGE pattern or ribotype. Conclusions. Evaluation of C. difficile infections within British Columbia revealed demographic associations, epidemiological shifts, and characteristics of strain types. Continuous surveillance of C. difficile will enable detection of emerging strains.

3.
Microvasc Res ; 101: 38-47, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26092680

RESUMEN

Pericytes are perivascular support cells, the origin of which in tumor tissue is not clear. Recently, we identified a Tie1(+) precursor cell that differentiates into vascular smooth muscle, in a Notch-dependent manner. To understand the involvement of Notch in the ontogeny of tumor pericytes we used a novel flow immunophenotyping strategy to define CD146(+)/CD45(-)/CD31(-/lo) pericytes in the tumor stroma. This strategy combined with ex vivo co-culture experiments identified a novel pericyte progenitor cell population defined as Sca1(hi)/CD146(-)/CD45(-)/CD31(-). The differentiation of these progenitor cells was stimulated by co-culture with endothelial cells. Overexpression of the Notch ligand Jagged1 in endothelial cells further stimulated the differentiation of Sca1(hi)/CD146(-)/CD45(-)/CD31(-) cells into pericytes, while inhibition of Notch signaling with a γ-secretase inhibitor reduced this differentiation. However, Notch inhibition specifically in Tie1-expressing cells did not change the abundance of pericytes in tumors, suggesting that the pericyte precursor is distinct from the vascular smooth muscle cell precursor. Transplant experiments showed that the bone marrow contributes minimally to tumor pericytes. Immunophenotyping revealed that Sca1(hi)/CD146(-)/CD45(-)/CD31(-) cells have greater potential to differentiate into pericytes and have increased expression of classic mesenchymal stem cell markers (CD13, CD44, Nt5e and Thy-1) compared to Sca1(-/lo)/CD146(-)/CD45(-)/CD31(-) cells. Our results suggest that a local Sca1(hi)/CD146(-)/CD45(-)/CD31(-) pericyte progenitor resides in the tumor microenvironment and requires Notch signaling for differentiation into mature pericytes.


Asunto(s)
Neoplasias/metabolismo , Pericitos/citología , Receptores Notch/metabolismo , Animales , Ataxina-1/metabolismo , Trasplante de Médula Ósea , Antígeno CD146/metabolismo , Carcinoma Pulmonar de Lewis , Diferenciación Celular , Técnicas de Cocultivo , Citometría de Flujo , Células Endoteliales de la Vena Umbilical Humana , Humanos , Antígenos Comunes de Leucocito/metabolismo , Melanoma Experimental , Células Madre Mesenquimatosas/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Trasplante de Neoplasias , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , Receptor TIE-1/metabolismo , Transducción de Señal , Células Madre/citología
4.
Dev Dyn ; 243(7): 894-905, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24633789

RESUMEN

BACKGROUND: Valvuloseptal defects are the most common congenital heart defects. Notch signaling-induced endothelial-to-mesenchymal transition (EMT) in the atrioventricular canal (AVC) cushions at murine embryonic day (E)9.5 is a required step during early valve development. Insights to the transcriptional network that is activated in endocardial cells (EC) during EMT and how these pathways direct valve maturation are lacking. RESULTS: We show that at E11.5, AVC-EC retain the ability to undergo Notch-dependent EMT when explanted on collagen. EC-Notch inhibition at E10.5 blocks expression of known mesenchymal genes in E11.5 AVC-EC. To understand the genetic network and AVC development downstream of Notch signaling beyond E9.5, we constructed Tag-Seq libraries corresponding to different cell types of the E11.5 AVC and atrium in wild-type mice and in EC-Notch inhibited mice. We identified 1,400 potential Notch targets in the AVC-EC, of which 124 are transcription factors (TF). From the 124 TFs, we constructed a transcriptional hierarchy and identify 10 upstream TFs within the network. CONCLUSIONS: We validated 4 of the upstream TFs as Notch targets that are enriched in AVC-EC. Functionally, we show these 4 TFs regulate EMT in AVC explant assays. These novel signaling pathways downstream of Notch are potentially relevant to valve development.


Asunto(s)
Transdiferenciación Celular/genética , Cojinetes Endocárdicos/embriología , Cojinetes Endocárdicos/metabolismo , Regulación del Desarrollo de la Expresión Génica/genética , Redes Reguladoras de Genes/genética , Receptores Notch/metabolismo , Animales , Línea Celular , Transdiferenciación Celular/fisiología , Femenino , Regulación del Desarrollo de la Expresión Génica/fisiología , Redes Reguladoras de Genes/fisiología , Humanos , Masculino , Ratones , Embarazo , Receptores Notch/genética
5.
J Exp Med ; 219(3)2022 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-35089323

RESUMEN

Inflammation is associated with bone marrow failure syndromes, but how specific molecules impact the bone marrow microenvironment is not well elucidated. We report a novel role for the miR-145 target, Toll/interleukin-1 receptor domain containing adaptor protein (TIRAP), in driving bone marrow failure. We show that TIRAP is overexpressed in various types of myelodysplastic syndromes (MDS) and suppresses all three major hematopoietic lineages. TIRAP expression promotes up-regulation of Ifnγ, leading to myelosuppression through Ifnγ-Ifnγr-mediated release of the alarmin, Hmgb1, which disrupts the bone marrow endothelial niche. Deletion of Ifnγ blocks Hmgb1 release and is sufficient to reverse the endothelial defect and restore myelopoiesis. Contrary to current dogma, TIRAP-activated Ifnγ-driven bone marrow suppression is independent of T cell function or pyroptosis. In the absence of Ifnγ, TIRAP drives myeloproliferation, implicating Ifnγ in suppressing the transformation of MDS to acute leukemia. These findings reveal novel, noncanonical roles of TIRAP, Hmgb1, and Ifnγ in the bone marrow microenvironment and provide insight into the pathophysiology of preleukemic syndromes.


Asunto(s)
Trastornos de Fallo de la Médula Ósea/etiología , Trastornos de Fallo de la Médula Ósea/metabolismo , Endotelio/metabolismo , Proteína HMGB1/metabolismo , Interferón gamma/metabolismo , Glicoproteínas de Membrana/genética , Mielopoyesis/genética , Receptores de Interleucina-1/genética , Animales , Biomarcadores , Trastornos de Fallo de la Médula Ósea/patología , Microambiente Celular/genética , Susceptibilidad a Enfermedades , Expresión Génica , Hematopoyesis/genética , Glicoproteínas de Membrana/metabolismo , Células Madre Mesenquimatosas/metabolismo , Ratones , Trastornos Mieloproliferativos/etiología , Trastornos Mieloproliferativos/metabolismo , Trastornos Mieloproliferativos/patología , Receptores de Interleucina-1/metabolismo
6.
Nat Cell Biol ; 22(5): 526-533, 2020 05.
Artículo en Inglés | MEDLINE | ID: mdl-32251398

RESUMEN

Interstitial deletion of the long arm of chromosome 5 (del(5q)) is the most common structural genomic variant in myelodysplastic syndromes (MDS)1. Lenalidomide (LEN) is the treatment of choice for patients with del(5q) MDS, but half of the responding patients become resistant2 within 2 years. TP53 mutations are detected in ~20% of LEN-resistant patients3. Here we show that patients who become resistant to LEN harbour recurrent variants of TP53 or RUNX1. LEN upregulated RUNX1 protein and function in a CRBN- and TP53-dependent manner in del(5q) cells, and mutation or downregulation of RUNX1 rendered cells resistant to LEN. LEN induced megakaryocytic differentiation of del(5q) cells followed by cell death that was dependent on calpain activation and CSNK1A1 degradation4,5. We also identified GATA2 as a LEN-responsive gene that is required for LEN-induced megakaryocyte differentiation. Megakaryocytic gene-promoter analyses suggested that LEN-induced degradation of IKZF1 enables a RUNX1-GATA2 complex to drive megakaryocytic differentiation. Overexpression of GATA2 restored LEN sensitivity in the context of RUNX1 or TP53 mutations by enhancing LEN-induced megakaryocytic differentiation. Screening for mutations that block LEN-induced megakaryocytic differentiation should identify patients who are resistant to LEN.


Asunto(s)
Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Cromosomas Humanos Par 5/genética , Lenalidomida/farmacología , Megacariocitos/efectos de los fármacos , Síndromes Mielodisplásicos/genética , Línea Celular , Cromosomas Humanos Par 5/efectos de los fármacos , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Regulación hacia Abajo/efectos de los fármacos , Regulación hacia Abajo/genética , Factor de Transcripción GATA2/genética , Células HEK293 , Humanos , Mutación/efectos de los fármacos , Mutación/genética , Proteína p53 Supresora de Tumor/genética
7.
Cell Rep ; 27(6): 1769-1780.e4, 2019 05 07.
Artículo en Inglés | MEDLINE | ID: mdl-31067462

RESUMEN

The sterile alpha motif (SAM) and SRC homology 3 (SH3) domain containing protein 1 (Sash1) acts as a scaffold in TLR4 signaling. We generated Sash1-/- mice, which die in the perinatal period due to respiratory distress. Constitutive or endothelial-restricted Sash1 loss leads to a delay in maturation of alveolar epithelial cells causing reduced surfactant-associated protein synthesis. We show that Sash1 interacts with ß-arrestin 1 downstream of the TLR4 pathway to activate Akt and endothelial nitric oxide synthase (eNOS) in microvascular endothelial cells. Generation of nitric oxide downstream of Sash1 in endothelial cells affects alveolar epithelial cells in a cGMP-dependent manner, inducing maturation of alveolar type 1 and 2 cells. Thus, we identify a critical cell nonautonomous function for Sash1 in embryonic development in which endothelial Sash1 regulates alveolar epithelial cell maturation and promotes pulmonary surfactant production through nitric oxide signaling. Lung immaturity is a major cause of respiratory distress and mortality in preterm infants, and these findings identify the endothelium as a potential target for therapy.


Asunto(s)
Células Endoteliales/metabolismo , Pulmón/crecimiento & desarrollo , Óxido Nítrico/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Línea Celular , GMP Cíclico/metabolismo , Pérdida del Embrión/metabolismo , Pérdida del Embrión/patología , Embrión de Mamíferos/metabolismo , Embrión de Mamíferos/patología , Endotelio/metabolismo , Células Epiteliales/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Pulmón/ultraestructura , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo III/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-akt/metabolismo , Alveolos Pulmonares/patología , Proteínas Asociadas a Surfactante Pulmonar/metabolismo , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo , beta-Arrestinas/metabolismo
8.
Nat Commun ; 9(1): 2418, 2018 06 20.
Artículo en Inglés | MEDLINE | ID: mdl-29925839

RESUMEN

Expression of miR-143 and miR-145 is reduced in hematopoietic stem/progenitor cells (HSPCs) of myelodysplastic syndrome patients with a deletion in the long arm of chromosome 5. Here we show that mice lacking miR-143/145 have impaired HSPC activity with depletion of functional hematopoietic stem cells (HSCs), but activation of progenitor cells (HPCs). We identify components of the transforming growth factor ß (TGFß) pathway as key targets of miR-143/145. Enforced expression of the TGFß adaptor protein and miR-145 target, Disabled-2 (DAB2), recapitulates the HSC defect seen in miR-143/145-/- mice. Despite reduced HSC activity, older miR-143/145-/- and DAB2-expressing mice show elevated leukocyte counts associated with increased HPC activity. A subset of mice develop a serially transplantable myeloid malignancy, associated with expansion of HPC. Thus, miR-143/145 play a cell context-dependent role in HSPC function through regulation of TGFß/DAB2 activation, and loss of these miRNAs creates a preleukemic state.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , MicroARNs/metabolismo , Síndromes Mielodisplásicos/genética , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/metabolismo , Proteínas Adaptadoras Transductoras de Señales , Proteínas Adaptadoras del Transporte Vesicular/genética , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis , Médula Ósea/patología , Trasplante de Médula Ósea , Línea Celular Tumoral , Modelos Animales de Enfermedad , Femenino , Regulación Neoplásica de la Expresión Génica , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/genética , Síndromes Mielodisplásicos/patología , Quimera por Trasplante
9.
Cancer Res ; 74(9): 2402-11, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24599126

RESUMEN

Notch signaling is important for tumor angiogenesis induced by vascular endothelial growth factor A. Blockade of the Notch ligand Dll4 inhibits tumor growth in a paradoxical way. Dll4 inhibition increases endothelial cell sprouting, but vessels show reduced perfusion. The reason for this lack of perfusion is not currently understood. Here we report that inhibition of Notch signaling in endothelial cell using an inducible binary transgenic system limits VEGFA-driven tumor growth and causes endothelial dysfunction. Neither excessive endothelial cell sprouting nor defects of pericyte abundance accompanied the inhibition of tumor growth and functional vasculature. However, biochemical and functional analysis revealed that endothelial nitric oxide production is decreased by Notch inhibition. Treatment with the soluble guanylate cyclase activator BAY41-2272, a vasorelaxing agent that acts downstream of endothelial nitric oxide synthase (eNOS) by directly activating its soluble guanylyl cyclase receptor, rescued blood vessel function and tumor growth. We show that reduction in nitric oxide signaling is an early alteration induced by Notch inhibition and suggest that lack of functional vessels observed with Notch inhibition is secondary to inhibition of nitric oxide signaling. Coculture and tumor growth assays reveal that Notch-mediated nitric oxide production in endothelial cell requires VEGFA signaling. Together, our data support that eNOS inhibition is responsible for the tumor growth and vascular function defects induced by endothelial Notch inhibition. This study uncovers a novel mechanism of nitric oxide production in endothelial cells in tumors, with implications for understanding the peculiar character of tumor blood vessels.


Asunto(s)
Melanoma Experimental/enzimología , Neovascularización Patológica/enzimología , Óxido Nítrico Sintasa de Tipo III/fisiología , Receptores Notch/metabolismo , Inhibidores de la Angiogénesis/farmacología , Animales , Línea Celular Tumoral , Técnicas de Cocultivo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/metabolismo , Guanilato Ciclasa/antagonistas & inhibidores , Guanilato Ciclasa/metabolismo , Melanoma Experimental/irrigación sanguínea , Melanoma Experimental/patología , Ratones Endogámicos C57BL , Ratones Transgénicos , Microvasos/efectos de los fármacos , Microvasos/patología , Trasplante de Neoplasias , Óxido Nítrico/metabolismo , Pericitos/metabolismo , Pirazoles/farmacología , Piridinas/farmacología , Transducción de Señal , Carga Tumoral/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA