Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 42
Filtrar
1.
Proc Natl Acad Sci U S A ; 118(31)2021 08 03.
Artículo en Inglés | MEDLINE | ID: mdl-34330825

RESUMEN

A network of molecular factors drives the development, differentiation, and maintenance of endothelial cells. Friend leukemia integration 1 transcription factor (FLI1) is a bona fide marker of endothelial cells during early development. In zebrafish Tg(fli1:EGFP)y1 , we identified two endothelial cell populations, high-fli1+ and low-fli1+, by the intensity of green fluorescent protein signal. By comparing RNA-sequencing analysis of non-fli1 expressing cells (fli1-) with these two (fli1+) cell populations, we identified several up-regulated genes, not previously recognized as important, during endothelial development. Compared with fli1- and low-fli1+ cells, high-fli1+ cells showed up-regulated expression of the zinc finger transcription factor PRDI-BF1 and RIZ homology domain containing 16 (prdm16). Prdm16 knockdown (KD) by morpholino in the zebrafish larva was associated with impaired angiogenesis and increased number of low-fli1+ cells at the expense of high-fli1+ cells. In addition, PRDM16 KD in endothelial cells derived from human-induced pluripotent stem cells impaired their differentiation and migration in vitro. Moreover, zebrafish mutants (mut) with loss of function for the oncogene LIM domain only 2 (lmo2) also showed reduced prdm16 gene expression combined with impaired angiogenesis. Prdm16 expression was reduced further in endothelial (CD31+) cells compared with CD31- cells isolated from lmo2-mutants (lmo2-mut) embryos. Chromatin immunoprecipitation-PCR demonstrated that Lmo2 binds to the promoter and directly regulates the transcription of prdm16 This work unveils a mechanism by which prdm16 expression is activated in endothelial cells by Lmo2 and highlights a possible therapeutic pathway by which to modulate endothelial cell growth and repair.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Células Endoteliales/fisiología , Células Madre Pluripotentes Inducidas/fisiología , Neovascularización Fisiológica/fisiología , Proteína Proto-Oncogénica c-fli-1/fisiología , Proteínas de Pez Cebra/metabolismo , Animales , Animales Modificados Genéticamente , Diferenciación Celular , Proteínas de Unión al ADN/genética , Embrión no Mamífero , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Humanos , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/genética , Molécula-1 de Adhesión Celular Endotelial de Plaqueta/metabolismo , RNA-Seq , Transcriptoma , Regulación hacia Arriba , Pez Cebra , Proteínas de Pez Cebra/genética
2.
Nucleic Acids Res ; 48(20): 11434-11451, 2020 11 18.
Artículo en Inglés | MEDLINE | ID: mdl-33080033

RESUMEN

Core regulatory circuitry (CRC)-dependent transcriptional network is critical for developmental tumors in children and adolescents carrying few gene mutations. However, whether and how CRC contributes to transcription regulation in Ewing sarcoma is unknown. Here, we identify and functionally validate a CRC 'trio' constituted by three transcription factors (TFs): KLF15, TCF4 and NKX2-2, in Ewing sarcoma cells. Epigenomic analyses demonstrate that EWS-FLI1, the primary fusion driver for this cancer, directly establishes super-enhancers of each of these three TFs to activate their transcription. In turn, KLF15, TCF4 and NKX2-2 co-bind to their own and each other's super-enhancers and promoters, forming an inter-connected auto-regulatory loop. Functionally, CRC factors contribute significantly to cell proliferation of Ewing sarcoma both in vitro and in vivo. Mechanistically, CRC factors exhibit prominent capacity of co-regulating the epigenome in cooperation with EWS-FLI1, occupying 77.2% of promoters and 55.6% of enhancers genome-wide. Downstream, CRC TFs coordinately regulate gene expression networks in Ewing sarcoma, controlling important signaling pathways for cancer, such as lipid metabolism pathway, PI3K/AKT and MAPK signaling pathways. Together, molecular characterization of the oncogenic CRC model advances our understanding of the biology of Ewing sarcoma. Moreover, CRC-downstream genes and signaling pathways may contain potential therapeutic targets for this malignancy.


Asunto(s)
Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN/metabolismo , Sarcoma de Ewing/genética , Animales , Línea Celular Tumoral , Proliferación Celular , Proteína Homeobox Nkx-2.2 , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Factores de Transcripción de Tipo Kruppel/genética , Factores de Transcripción de Tipo Kruppel/metabolismo , Metabolismo de los Lípidos/genética , Ratones Desnudos , Proteínas Nucleares , Proteínas de Fusión Oncogénica/fisiología , Proteína Proto-Oncogénica c-fli-1/fisiología , Proteína EWS de Unión a ARN/fisiología , Sarcoma de Ewing/metabolismo , Sarcoma de Ewing/patología , Transducción de Señal , Factor de Transcripción 4/genética , Factor de Transcripción 4/metabolismo , Factores de Transcripción , Proteínas de Pez Cebra/genética , Proteínas de Pez Cebra/metabolismo
3.
Arterioscler Thromb Vasc Biol ; 40(4): e105-e113, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-32075417

RESUMEN

OBJECTIVE: Vascular progenitor cells (VPCs), which are able to differentiate into both endothelial cells and smooth muscle cells, have the potential for treatment of ischemic diseases. Generated by pluripotent stem cells, VPCs carry the risk of tumorigenicity in clinical application. This issue could be resolved by direct lineage conversion, the induction of functional cells from another lineage by using only lineage-restricted transcription factors. Here, we show that induced VPCs (iVPCs) can be generated from fibroblasts by ETS (E-twenty six) transcription factors, Etv2 and Fli1. Approach and Results: Mouse fibroblasts were infected with lentivirus encoding Etv2 and Fli1. Cell colonies appeared in Fli1- and Etv2/Fli1-infected groups and were mechanically picked. The identity of cell colonies was confirmed by proliferation assay and reverse-transcription polymerase chain reaction with vascular markers. Etv2/Fli1- infected cell colonies were sorted by CD144 (also known as CDH5, VE-cadherin). We defined that CD144-positive iVPCs maintained its own population and expanded stably at multiple passages. iVPCs could differentiate into functional endothelial cells and smooth muscle cells by a defined medium. The functionalities of iVPC-derived endothelial cells and smooth muscle cells were confirmed by analyzing LDL (low-density lipoprotein) uptake, carbachol-induced contraction, and tube formation in vitro. Transplantation of iVPCs into the ischemic hindlimb model enhanced blood flow without tumor formation in vivo. Human iVPCs were generated by human ETS transcription factors ETV2 and FLI1. CONCLUSIONS: We demonstrate that ischemic disease curable iVPCs, which have self-renewal and bipotency, can be generated from mouse fibroblasts by enforced ETS family transcription factors, Etv2 and Fli1 expression. Our simple strategy opens insights into stem cell-based ischemic disease therapy.


Asunto(s)
Fibroblastos/citología , Isquemia/fisiopatología , Proteína Proto-Oncogénica c-fli-1/fisiología , Células Madre/fisiología , Factores de Transcripción/fisiología , Animales , Antígenos CD , Cadherinas , Diferenciación Celular , Línea Celular , Proliferación Celular , Modelos Animales de Enfermedad , Células Endoteliales/citología , Miembro Posterior/irrigación sanguínea , Isquemia/terapia , Miocitos del Músculo Liso/citología , Trasplante de Células Madre , Células Madre/inmunología
4.
Cytokine ; 134: 155180, 2020 10.
Artículo en Inglés | MEDLINE | ID: mdl-32673994

RESUMEN

The chemokine fractalkine is synthesized as a membrane-bound protein, but studies have shown that serum levels of soluble fractalkine are elevated in inflammatory and autoimmune diseases. Patients with autoimmune diseases also have increased serum levels of neuropeptide substance P (SP). The shedding activity of the ADAM family is induced by peroxynitrite, but that of SP is unclear. Treatment of human macrophages with SP upregulated levels of membrane-bound fractalkine. Interestingly, small interfering RNA (siRNA) for DUOX2 further increased membrane-bound fractalkine but decreased soluble fractalkine compared with cells treated with SP alone. SP induced nitric oxide 2/inducible nitric oxide synthase (NOS2/iNOS) mRNA and increased levels of nitrotyrosine, a biomarker of peroxynitrite, whereas transfection with DUOX2 siRNA blunted upregulation of nitrotyrosine. Most importantly, N(ω)-nitro-L-arginine methyl ester (L-NAME, a nitric oxide synthase inhibitor) decreased protein levels of nitrotyrosine and concomitantly increased expression of membrane-bound fractalkine after exposure to SP. As for the signaling pathway of TGFß1 (an inhibitor of iNOS mRNA expression), silencing of RNA for TAK-1 upregulated membrane-bound fractalkine, but silencing of RNA for the Smad family did not. Interfering RNA of transcription factor specificity protein 1 (Sp1) upregulated protein levels of TGFß1/LAP. Most importantly, double transfection with siRNA for Sp1 and TRIM28/TIF1ßor Fli-1 led to a significant increase in TGFß1/LAP levels and a corresponding reduction of NOS2/iNOS, which inhibited the shedding of membrane-bound fractalkine. In conclusion, TRIM28/TIF1ß and Fli-1 negatively regulate TGFß1 expression to upregulate the generation of peroxynitrite, leading to increased shedding of membrane-bound fractalkine induced by SP.


Asunto(s)
Quimiocina CX3CL1/metabolismo , Oxidasas Duales/metabolismo , Macrófagos/metabolismo , Ácido Peroxinitroso/metabolismo , Proteína Proto-Oncogénica c-fli-1/fisiología , Sustancia P/fisiología , Proteína 28 que Contiene Motivos Tripartito/fisiología , Humanos , Macrófagos/enzimología , Proteínas de la Membrana/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo
5.
Circ Res ; 114(11): 1690-9, 2014 May 23.
Artículo en Inglés | MEDLINE | ID: mdl-24727028

RESUMEN

RATIONALE: Cardiovascular health depends on proper development and integrity of blood vessels. Ets variant 2 (Etv2), a member of the E26 transforming-specific family of transcription factors, is essential to initiate a transcriptional program leading to vascular morphogenesis in early mouse embryos. However, endothelial expression of the Etv2 gene ceases at midgestation; therefore, vascular development past this stage must continue independent of Etv2. OBJECTIVE: To identify molecular mechanisms underlying transcriptional regulation of vascular morphogenesis and homeostasis in the absence of Etv2. METHODS AND RESULTS: Using loss- and gain-of-function strategies and a series of molecular techniques, we identify Friend leukemia integration 1 (Fli1), another E26 transforming-specific family transcription factor, as a downstream target of Etv2. We demonstrate that Etv2 binds to conserved Ets-binding sites within the promoter region of the Fli1 gene and governs Fli1 expression. Importantly, in the absence of Etv2 at midgestation, binding of Etv2 at Ets-binding sites in the Fli1 promoter is replaced by Fli1 protein itself, sustaining expression of Fli1 as well as selective Etv2-regulated endothelial genes to promote endothelial cell survival and vascular integrity. Consistent with this, we report that Fli1 binds to the conserved Ets-binding sites within promoter and enhancer regions of other Etv2-regulated endothelial genes, including Tie2, to control their expression at and beyond midgestation. CONCLUSIONS: We have identified a novel positive feed-forward regulatory loop in which Etv2 activates expression of genes involved in vasculogenesis, including Fli1. Once the program is activated in early embryos, Fli1 then takes over to sustain the process in the absence of Etv2.


Asunto(s)
Endotelio Vascular/citología , Homeostasis/fisiología , Neovascularización Fisiológica/fisiología , Proteína Proto-Oncogénica c-fli-1/fisiología , Factores de Transcripción/fisiología , Animales , Supervivencia Celular/fisiología , Desarrollo Embrionario/fisiología , Endotelio Vascular/fisiología , Femenino , Hemorragia/etiología , Hemorragia/fisiopatología , Masculino , Ratones , Ratones Noqueados , Ratones Transgénicos , Modelos Animales , Morfogénesis/fisiología , Proteína Proto-Oncogénica c-fli-1/deficiencia , Proteína Proto-Oncogénica c-fli-1/genética
6.
Blood ; 121(12): 2289-300, 2013 Mar 21.
Artículo en Inglés | MEDLINE | ID: mdl-23327922

RESUMEN

Aberrant transcriptional programs in combination with abnormal proliferative signaling drive leukemic transformation. These programs operate in normal hematopoiesis where they are involved in hematopoietic stem cell (HSC) proliferation and maintenance. Ets Related Gene (ERG) is a component of normal and leukemic stem cell signatures and high ERG expression is a risk factor for poor prognosis in acute myeloid leukemia (AML). However, mechanisms that underlie ERG expression in AML and how its expression relates to leukemic stemness are unknown. We report that ERG expression in AML is associated with activity of the ERG promoters and +85 stem cell enhancer and a heptad of transcription factors that combinatorially regulate genes in HSCs. Gene expression signatures derived from ERG promoter-stem cell enhancer and heptad activity are associated with clinical outcome when ERG expression alone fails. We also show that the heptad signature is associated with AMLs that lack somatic mutations in NPM1 and confers an adverse prognosis when associated with FLT3 mutations. Taken together, these results suggest that transcriptional regulators cooperate to establish or maintain primitive stem cell-like signatures in leukemic cells and that the underlying pattern of somatic mutations contributes to the development of these signatures and modulate their influence on clinical outcome.


Asunto(s)
Células Madre Hematopoyéticas/metabolismo , Leucemia Mieloide Aguda/diagnóstico , Leucemia Mieloide Aguda/genética , Células Madre Neoplásicas/metabolismo , Factores de Transcripción/fisiología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Adaptadoras Transductoras de Señales/fisiología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/fisiología , Células Cultivadas , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Subunidad alfa 2 del Factor de Unión al Sitio Principal/fisiología , Elementos de Facilitación Genéticos/genética , Factor de Transcripción GATA2/genética , Factor de Transcripción GATA2/metabolismo , Factor de Transcripción GATA2/fisiología , Regulación Leucémica de la Expresión Génica , Células Madre Hematopoyéticas/fisiología , Humanos , Células K562 , Proteínas con Dominio LIM/genética , Proteínas con Dominio LIM/metabolismo , Proteínas con Dominio LIM/fisiología , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Ratones , Ratones Transgénicos , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo , Proteínas de Neoplasias/fisiología , Células Madre Neoplásicas/fisiología , Nucleofosmina , Pronóstico , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína Proto-Oncogénica c-fli-1/fisiología , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/metabolismo , Proteínas Proto-Oncogénicas/fisiología , Proteína 1 de la Leucemia Linfocítica T Aguda , Transactivadores/genética , Transactivadores/metabolismo , Transactivadores/fisiología , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Activación Transcripcional/genética , Regulador Transcripcional ERG
7.
J Proteome Res ; 13(8): 3783-91, 2014 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-24999758

RESUMEN

Ewing sarcoma is a cancer of bone and soft tissue in children that is characterized by a chromosomal translocation involving EWS and an Ets family transcription factor, most commonly Fli-1. EWS-Fli-1 fusion accounts for 85% of cases. The growth and survival of Ewing sarcoma cells are critically dependent on EWS-Fli-1. A large body of evidence has established that EWS-Fli-1 functions as a DNA-binding transcription factor that regulates the expression of a number of genes important for cell proliferation and transformation. However, little is known about the biochemical properties of the EWS-Fli-1 protein. We undertook a series of proteomic analyses to dissect the EWS-Fli-1 interactome. Employing a proximity-dependent biotinylation technique, BioID, we identified cation-independent mannose 6-phosphate receptor (CIMPR) as a protein located in the vicinity of EWS-Fli-1 within a cell. CIMPR is a cargo that mediates the delivery of lysosomal hydrolases from the trans-Golgi network to the endosome, which are subsequently transferred to the lysosomes. Further molecular cell biological analyses uncovered a role for lysosomes in the turnover of the EWS-Fli-1 protein. We demonstrate that an mTORC1 active-site inhibitor, torin 1, which stimulates the TFEB-lysosome pathway, can induce the degradation of EWS-Fli-1, suggesting a potential therapeutic approach to target EWS-Fli-1 for degradation.


Asunto(s)
Lisosomas/metabolismo , Proteínas de Fusión Oncogénica/fisiología , Proteómica , Proteína Proto-Oncogénica c-fli-1/fisiología , Proteína EWS de Unión a ARN/fisiología , Sarcoma de Ewing/tratamiento farmacológico , Biotinilación , Dominio Catalítico , Línea Celular Tumoral , Proliferación Celular , Transformación Celular Neoplásica/genética , Regulación Neoplásica de la Expresión Génica , Células HEK293 , Células HeLa , Humanos , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/metabolismo , Proteoma/metabolismo , Sarcoma de Ewing/patología , Serina-Treonina Quinasas TOR/metabolismo , Espectrometría de Masas en Tándem , Factores de Transcripción/metabolismo , Red trans-Golgi/metabolismo
8.
Blood ; 118(17): 4666-73, 2011 Oct 27.
Artículo en Inglés | MEDLINE | ID: mdl-21873545

RESUMEN

Large chromosomal deletions are among the most common molecular abnormalities in cancer, yet the identification of relevant genes has proven difficult. The 5q- syndrome, a subtype of myelodysplastic syndrome (MDS), is a chromosomal deletion syndrome characterized by anemia and thrombocytosis. Although we have previously shown that hemizygous loss of RPS14 recapitulates the failed erythroid differentiation seen in 5q- syndrome, it does not affect thrombocytosis. Here we show that a microRNA located in the common deletion region of 5q- syndrome, miR-145, affects megakaryocyte and erythroid differentiation. We find that miR-145 functions through repression of Fli-1, a megakaryocyte and erythroid regulatory transcription factor. Patients with del(5q) MDS have decreased expression of miR-145 and increased expression of Fli-1. Overexpression of miR-145 or inhibition of Fli-1 decreases the production of megakaryocytic cells relative to erythroid cells, whereas inhibition of miR-145 or overexpression of Fli-1 has a reciprocal effect. Moreover, combined loss of miR-145 and RPS14 cooperates to alter erythroid-megakaryocytic differentiation in a manner similar to the 5q- syndrome. Taken together, these findings demonstrate that coordinate deletion of a miRNA and a protein-coding gene contributes to the phenotype of a human malignancy, the 5q- syndrome.


Asunto(s)
Anemia Macrocítica/genética , MicroARNs/genética , Sistemas de Lectura Abierta/genética , Anemia Macrocítica/etiología , Animales , Estudios de Casos y Controles , Diferenciación Celular/genética , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Células Eritroides/metabolismo , Eritropoyesis/genética , Eritropoyesis/fisiología , Humanos , Pérdida de Heterocigocidad , Megacariocitos/metabolismo , Megacariocitos/fisiología , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , MicroARNs/metabolismo , MicroARNs/fisiología , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/patología , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína Proto-Oncogénica c-fli-1/fisiología , Proteínas Ribosómicas/genética , Proteínas Ribosómicas/metabolismo , Proteínas Ribosómicas/fisiología , Células Tumorales Cultivadas
9.
Ann Hematol ; 92(1): 11-8, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22965552

RESUMEN

Friend leukemia virus integration 1 (Fli1) and erythroid Krüppel-like factor (EKLF) participate under experimental conditions in the differentiation of megakaryocytic and erythroid progenitor in cooperation with other transcription factors, cytokines, cytokine receptors, and microRNAs. Defective erythropoiesis with refractory anemia and effective megakaryopoiesis with normal or increased platelet count is typical for 5q- syndrome. We decided to evaluate the roles of EKLF and Fli1 in the pathogenesis of this syndrome and of another ribosomopathy, Diamond-Blackfan anemia (DBA). Fli1 and EKLF mRNA levels were examined in mononuclear blood and bone marrow cells from patients with 5q- syndrome, low-risk MDS patients with normal chromosome 5, DBA patients, and healthy controls. In 5q- syndrome, high Fli1 mRNA levels in the blood and bone marrow mononuclear cells were found. In DBA, Fli1 expression did not differ from the controls. EKLF mRNA level was significantly decreased in the blood and bone marrow of 5q- syndrome and in all DBA patients. We propose that the elevated Fli1 in 5q- syndrome protects megakaryocytic cells from ribosomal stress contrary to erythroid cells and contributes to effective though dysplastic megakaryopoiesis.


Asunto(s)
Anemia de Diamond-Blackfan/genética , Anemia Macrocítica/genética , Eritropoyesis/genética , Factores de Transcripción de Tipo Kruppel/fisiología , Proteína Proto-Oncogénica c-fli-1/fisiología , Trombopoyesis/genética , Adolescente , Adulto , Anemia de Diamond-Blackfan/metabolismo , Anemia Macrocítica/metabolismo , Células de la Médula Ósea/metabolismo , Niño , Deleción Cromosómica , Cromosomas Humanos Par 5/genética , Cromosomas Humanos Par 5/metabolismo , Islas de CpG , Femenino , Humanos , Factores de Transcripción de Tipo Kruppel/biosíntesis , Factores de Transcripción de Tipo Kruppel/genética , Leucocitos Mononucleares/metabolismo , Masculino , Síndromes Mielodisplásicos/genética , Síndromes Mielodisplásicos/metabolismo , Proteína Proto-Oncogénica c-fli-1/biosíntesis , Proteína Proto-Oncogénica c-fli-1/genética , ARN Mensajero/biosíntesis , ARN Mensajero/sangre , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Ribosómicas/fisiología , Transcripción Genética , Adulto Joven
10.
Am J Pathol ; 176(4): 1983-98, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-20228226

RESUMEN

Systemic sclerosis or scleroderma (SSc) is a complex autoimmune connective tissue disease characterized by obliterative vasculopathy and tissue fibrosis. The molecular mechanisms underlying SSc vasculopathy are largely unknown. Friend leukemia integration factor 1 (Fli1), an important regulator of immune function and collagen fibrillogenesis, is expressed at reduced levels in endothelial cells in affected skin of patients with SSc. To develop a disease model and to investigate the function of Fli1 in the vasculature, we generated mice with a conditional deletion of Fli1 in endothelial cells (Fli1 CKO). Fli1 CKO mice showed a disorganized dermal vascular network with greatly compromised vessel integrity and markedly increased vessel permeability. We show that Fli1 regulates expression of genes involved in maintaining vascular homeostasis including VE-cadherin, platelet endothelial cell adhesion molecule 1, type IV collagen, matrix metalloproteinase 9, platelet-derived growth factor B, and S1P(1) receptor. Accordingly, Fli1 CKO mice are characterized by down-regulation of VE-cadherin and platelet endothelial cell adhesion molecule 1, impaired development of basement membrane, and a decreased presence of alpha-smooth muscle actin-positive cells in dermal microvessels. This phenotype is consistent with a role of Fli1 as a regulator of vessel maturation and stabilization. Importantly, vascular characteristics of Fli1 CKO mice are recapitulated by SSc microvasculature. Thus, persistently reduced levels of Fli1 in endothelial cells may play a critical role in the development of SSc vasculopathy.


Asunto(s)
Proteína Proto-Oncogénica c-fli-1/genética , Esclerodermia Sistémica/metabolismo , Enfermedades Vasculares/patología , Actinas/metabolismo , Alelos , Animales , Humanos , Inmunohistoquímica/métodos , Ratones , Ratones Noqueados , Microcirculación , Permeabilidad , Fenotipo , Proteína Proto-Oncogénica c-fli-1/fisiología , Piel/patología
11.
Infect Immun ; 78(6): 2734-44, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20368343

RESUMEN

Genetic linkage studies of the host response to Leishmania major, the causative agent of cutaneous leishmaniasis, have identified significant genetic complexity in humans and mice. In the mouse model, multiple loci have been implicated in susceptibility to infection, but to date, the genes underlying these loci have not been identified. We now describe the contribution of a novel candidate gene, Fli1, to both L. major resistance and enhanced wound healing. We have previously mapped the L. major response locus, lmr2, to proximal chromosome 9 in a genetic cross between the resistant C57BL/6 strain and the susceptible BALB/c strain. We now show that the presence of the resistant C57BL/6 lmr2 allele in susceptible BALB/c mice confers an enhanced L. major resistance and wound healing phenotype. Fine mapping of the lmr2 locus permitted the localization of the lmr2 quantitative trait locus to a 5-Mb interval comprising 21 genes, of which microarray analysis was able to identify differential expression in 1 gene-Fli1. Analysis of Fli1 expression in wounded and L. major-infected skin and naïve and infected lymph nodes validated the importance of Fli1 in lesion resolution and wound healing and identified 3 polymorphisms in the Fli1 promoter, among which a GA repeat element may be the important contributor.


Asunto(s)
Predisposición Genética a la Enfermedad , Leishmania major/inmunología , Leishmaniasis Cutánea/inmunología , Proteína Proto-Oncogénica c-fli-1/fisiología , Cicatrización de Heridas , Animales , Mapeo Cromosómico , Cruzamientos Genéticos , Femenino , Perfilación de la Expresión Génica , Sitios Genéticos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Polimorfismo Genético , Regiones Promotoras Genéticas
12.
Clin Exp Immunol ; 160(2): 275-82, 2010 May.
Artículo en Inglés | MEDLINE | ID: mdl-20015093

RESUMEN

The transcription factor Fli-1 is implicated in the pathogenesis of both murine and human lupus. Decreased expression of Fli-1 in heterozygous (Fli-1(+/-)) Murphy Roths Large (MRL)/lpr mice resulted in significantly lower kidney pathological scores and markedly increased survival. In this study, bone marrow (BM) transplantation was used to investigate the role of decreased expression of Fli-1 in haematopoietic versus non-haematopoietic cell lineages in autoimmune disease development. Wild-type (WT) MRL/lpr that received BM from Fli-1(+/-) MRL/lpr mice had statistically significantly lower autoantibodies, less proteinuria, reduced renal disease and prolonged survival compared to WT MRL/lpr mice that received BM from WT MRL/lpr mice. Although not statistically significant, Fli-1(+/-) MRL/lpr mice that received BM from WT MRL/lpr mice also had lower autoantibodies and improved survival compared to WT MRL/lpr mice that received BM from WT MRL/lpr mice. Our data indicate that expression of Fli-1 in haematopoietic cell lineages has a significant effect on disease development in MRL/lpr mice.


Asunto(s)
Trasplante de Médula Ósea , Lupus Eritematoso Sistémico/terapia , Proteína Proto-Oncogénica c-fli-1/fisiología , Animales , Autoanticuerpos/biosíntesis , Autoanticuerpos/sangre , Linaje de la Célula , Progresión de la Enfermedad , Femenino , Células Madre Hematopoyéticas/metabolismo , Heterocigoto , Riñón/patología , Lupus Eritematoso Sistémico/sangre , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Nefritis Lúpica/genética , Nefritis Lúpica/inmunología , Nefritis Lúpica/patología , Nefritis Lúpica/terapia , Masculino , Ratones , Ratones Endogámicos MRL lpr , Proteína Proto-Oncogénica c-fli-1/biosíntesis , Proteína Proto-Oncogénica c-fli-1/deficiencia , Proteína Proto-Oncogénica c-fli-1/genética , Quimera por Radiación , Organismos Libres de Patógenos Específicos
13.
Biochimie ; 168: 1-9, 2020 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-31626853

RESUMEN

Friend leukemia integration 1 (Fli-1) is a member of the E26 transformation-specific (ETS) transcription factor family. Fli-1 regulates normal hematopoiesis and vasculogenesis, and its aberrant expression underlies virus-induced leukemias and various types of human cancers. NANOGP8, a retro-pseudogene of stem cell mediator NANOG, is expressed predominantly in cancer cells and plays a role in tumorigenesis. In this study, we demonstrate that Fli-1 expression enhances human acute T-cell leukemia Jurkat cell proliferation and that Fli-1 acts as a transcriptional activator of NANOGP8 expression in these cells. NANOGP8 and Fli-1 are highly expressed in Jurkat cells, whereas NANOG was undetectable at both the RNA and protein levels. Moreover, the expression of endogenous NANOGP8 was significantly influenced by gain of function and loss of function of Fli-1. Promoter-reporter assays showed that NANOGP8 transcription was significantly upregulated by dose-dependent Fli-1 overexpression. A series of deletion mutagenesis of NANOGP8 promoter sequence revealed that NANOGP8 promoter activity was tightly regulated and found the minimal promoter region sufficient to activate NANOGP8 transcription mediated by Fli-1. Moreover, site-directed mutagenesis of the putative binding site abolished both NANOGP8 full-length and minimal promoter activities. Binding assays revealed that Fli-1 directly interacts with the potent binding site in NANOG promoter region. Taken together, our data demonstrate that Fli-1 is a novel upstream transcriptional activator of NANOGP8 and provide the molecular details of Fli-1-mediated NANOGP8 gene expression. Ultimately, these findings may contribute to understanding the expanded regulatory mechanisms of oncogenic NANOGP8 and ETS family transcription factors in leukemogenesis.


Asunto(s)
Leucemia de Células T/metabolismo , Proteína Homeótica Nanog/metabolismo , Proteína Proto-Oncogénica c-fli-1/fisiología , Proliferación Celular , Regulación Leucémica de la Expresión Génica , Células HEK293 , Humanos , Células Jurkat , Leucemia de Células T/genética , Oncogenes
14.
Oncogene ; 26(12): 1802-10, 2007 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-16964281

RESUMEN

It was reported that short interfering RNA (siRNA) of EWS/Fli-1 downregulated phospholipase D (PLD)2 in Ewing's sarcoma (EWS) cell line, suggesting that PLD2 is the target of aberrant transcription factor, EWS/Fli-1. Here, we further investigated the regulation of PLD2 gene expression by EWS/Fli-1 and Fli-1 in another EWS cell line, and also in EWS/Fli-1- or Fli-1-transfected cell line. EWS/Fli-1- or Fli-1-overexpressed cells showed higher PLD2 but not PLD1 protein expression and enhanced cell proliferation as compared to mock transfectant. The treatment of these cells with 1-butanol or siRNA of PLD2 inhibited cell growth, suggesting the pivotal role of PLD in cell growth promotion. PLD2 but not PLD1 mRNA level was also increased in EWS/Fli-1 or Fli-1-transfectants. After determining the transcription initiation points, we cloned the 5' promoter of both PLD1 and PLD2 and analysed promoter activities. Results showed that EWS/Fli-1 and Fli-1 increase PLD2 gene expression by binding to an erythroblast transformation-specific domain (-126 to -120 bp from the transcription initiation site) of PLD2 promoter, which is the minimal and most powerful region. Electrophoresis mobility shift assay using truncated proteins showed that both DNA-binding domain and trans-activating domain were necessary for the enhanced gene expression of PLD2.


Asunto(s)
Proteínas de Microfilamentos/fisiología , Proteínas de Fusión Oncogénica/fisiología , Fosfolipasa D/genética , Regiones Promotoras Genéticas , Proteína Proto-Oncogénica c-fli-1/fisiología , Receptores Citoplasmáticos y Nucleares/fisiología , Secuencia de Bases , Línea Celular , Cartilla de ADN , Ensayo de Cambio de Movilidad Electroforética , Humanos , Inmunoprecipitación , Proteínas de Microfilamentos/metabolismo , Proteínas de Fusión Oncogénica/metabolismo , Unión Proteica , Proteína Proto-Oncogénica c-fli-1/metabolismo , Proteína EWS de Unión a ARN , Receptores Citoplasmáticos y Nucleares/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transactivadores , Transfección
15.
Clin Immunol ; 129(1): 19-30, 2008 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-18692443

RESUMEN

Transgenic overexpression of Fli-1 in normal mice leads to SLE-like disease and increased expression was reported in SLE-affected human and murine lymphocytes. Reducing Fli-1 expression in MRL/lpr mice decreased antibody production, proteinuria, renal pathology, and mortality. Compared to those with wild-type expression of Fli-1, we report here that proliferative responses of Fli-1-deficient naïve B cells to several mitogens were reduced in lupus-prone and control mice. Expression of mitogen receptors, including BCR, TLR4, and TLR9, was not significantly impacted in Fli-1-deficient naïve B cells. IL12a transcripts were upregulated and NFAT transcripts were downregulated in Fli-1-deficient MRL/lpr B cells. These results demonstrate that Fli-1 deficiency affects B cell proliferative responses to mitogens, independent of BCR and TLR expression. IL12a and NFAT, known to influence proliferation, were identified as potential mediators of this effect. This may be a mechanism by which overexpression of Fli-1 contributes to B cell hyperactivity and subsequent SLE pathogenesis.


Asunto(s)
Linfocitos B/inmunología , Lupus Eritematoso Sistémico/inmunología , Proteína Proto-Oncogénica c-fli-1/fisiología , Animales , Linfocitos B/metabolismo , Proliferación Celular , Regulación hacia Abajo , Expresión Génica , Subunidad p35 de la Interleucina-12/inmunología , Subunidad p35 de la Interleucina-12/metabolismo , Lupus Eritematoso Sistémico/patología , Activación de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Mutantes , Factores de Transcripción NFATC/inmunología , Factores de Transcripción NFATC/metabolismo , Proteína Proto-Oncogénica c-fli-1/deficiencia , Receptores de Antígenos de Linfocitos B/inmunología , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 9/inmunología , Receptor Toll-Like 9/metabolismo , Regulación hacia Arriba
16.
Cancer Res ; 66(2): 803-11, 2006 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-16424012

RESUMEN

The chromosomal translocation t(11;22) yields the EWS-Fli1 fusion gene and is associated with oncogenesis of Ewing family tumors (EFT). In this study, using the RNA interference method, we show that EWS-Fli1-targeting small interfering RNAs (siRNA) depleted EWS-Fli1 protein and caused growth inhibition in EFT cells with the accumulation of p27 protein and the down-regulation of Skp2 protein in dose-dependent, time-dependent, and sequence-specific manners. Depletion of EWS-Fli1 subacutely elicited a senescence-like phenotype, but not apoptosis, in EFT cells. Furthermore, not only the knockdown of p27, but also the forced expression of Skp2, reduced the expression levels of p27 protein and partially rescued senescence-like phenotype caused by EWS-Fli1-targeting siRNAs. The accumulation of p27 protein in EWS-Fli1-depleted cells inhibited cdk2 kinase activity and was related to the stability of p27 protein, which resulted from a decrease in Skp2 protein. Immunohistochemical analysis of p27 and Skp2 proteins in EFT samples revealed that there was an inverse relationship between the expression profiles of p27 and Skp2 proteins. These findings indicate an important role of EWS-Fli1 in the prevention of senescence, leading to the unlimited growth and oncogenesis of EFT cells through a decrease in the stability of p27 protein due to increased action of Skp2-mediated 26S proteasome degradation.


Asunto(s)
Senescencia Celular , Proteínas de Fusión Oncogénica/fisiología , Proteína Proto-Oncogénica c-fli-1/fisiología , Sarcoma de Ewing/genética , Apoptosis , Transformación Celular Neoplásica , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/biosíntesis , Regulación hacia Abajo , Humanos , Fenotipo , Interferencia de ARN , ARN Interferente Pequeño , Proteína EWS de Unión a ARN , Proteínas Quinasas Asociadas a Fase-S/biosíntesis , Sarcoma de Ewing/fisiopatología
17.
Cancer Res ; 66(20): 9937-47, 2006 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-17047056

RESUMEN

Tumors of the Ewing's sarcoma family (ESFT), such as Ewing's sarcoma (EWS) and primitive neuroectodermal tumors (PNET), are highly aggressive malignancies predominantly affecting children and young adults. ESFT express chimeric transcription factors encoded by hybrid genes fusing the EWS gene with several ETS genes, most commonly FLI-1. EWS/FLI-1 proteins are responsible for the malignant phenotype of ESFT, but only few of their transcriptional targets are known. Using antisense and short hairpin RNA-mediated gene expression knockdown, array analyses, chromatin immunoprecipitation methods, and reexpression studies, we show that caveolin-1 (CAV1) is a new direct target of EWS/FLI-1 that is overexpressed in ESFT cell lines and tumor specimens and is necessary for ESFT tumorigenesis. CAV1 knockdown led to up-regulation of Snail and the concomitant loss of E-cadherin expression. Consistently, loss of CAV1 expression inhibited the anchorage-independent growth of EWS cells and markedly reduced the growth of EWS cell-derived tumors in nude mice xenografts, indicating that CAV1 promotes the malignant phenotype in EWS carcinogenesis. Reexpression of CAV1 or E-cadherin in CAV1 knockdown EWS cells rescued the oncogenic phenotype of the original EWS cells, showing that the CAV1/Snail/E-cadherin pathway plays a central role in the expression of the oncogenic transformation functions of EWS/FLI-1. Overall, these data identify CAV1 as a key determinant of the tumorigenicity of ESFT and imply that targeting CAV1 may allow the development of new molecular therapeutic strategies for ESFT patients.


Asunto(s)
Caveolina 1/fisiología , Proteínas de Fusión Oncogénica/fisiología , Proteína Proto-Oncogénica c-fli-1/fisiología , Sarcoma de Ewing/patología , Animales , Cadherinas/biosíntesis , Cadherinas/genética , Caveolina 1/antagonistas & inhibidores , Caveolina 1/genética , Caveolina 1/metabolismo , Adhesión Celular/genética , Procesos de Crecimiento Celular/genética , Línea Celular Tumoral , Inmunoprecipitación de Cromatina , Regulación hacia Abajo , Regulación Neoplásica de la Expresión Génica , Células HT29 , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteínas de Fusión Oncogénica/genética , Proteínas de Fusión Oncogénica/metabolismo , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/metabolismo , ARN sin Sentido/genética , Proteína EWS de Unión a ARN , Sarcoma de Ewing/genética , Sarcoma de Ewing/metabolismo , Factores de Transcripción de la Familia Snail , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Regulación hacia Arriba
18.
J Exp Med ; 214(4): 1129-1151, 2017 04 03.
Artículo en Inglés | MEDLINE | ID: mdl-28232470

RESUMEN

Systemic sclerosis (SSc), or scleroderma, is a multisystem autoimmune disorder characterized by vasculopathy and fibrosis in the skin and internal organs, most frequently in the esophagus and lungs. Hitherto, studies on SSc pathogenesis centered on immune cells, vascular cells, and fibroblasts. Although dysregulated keratinocytes in SSc have been recently reported, the contribution of epithelial cells to pathogenesis remains unexplored. In this study, we demonstrated the induction of SSc-like molecular phenotype in keratinocytes by gene silencing of transcription factor Friend leukemia virus integration 1 (Fli1), the deficiency of which is implicated in SSc pathogenesis. Keratin 14-expressing epithelial cell-specific Fli1 knockout mice spontaneously developed dermal and esophageal fibrosis with epithelial activation. Furthermore, they developed remarkable autoimmunity with interstitial lung disease derived from thymic defects with down-regulation of autoimmune regulator (Aire). Importantly, Fli1 directly regulated Aire expression in epithelial cells. Collectively, epithelial Fli1 deficiency might be involved in the systemic autoimmunity and selective organ fibrosis in SSc. This study uncovers unidentified roles of dysregulated epithelial cells in SSc pathogenesis.


Asunto(s)
Autoinmunidad , Proteína Proto-Oncogénica c-fli-1/fisiología , Esclerodermia Sistémica/etiología , Animales , Modelos Animales de Enfermedad , Células Epiteliales/fisiología , Esófago/patología , Fibrosis , Proteínas de Homeodominio/fisiología , Humanos , Queratina-14/análisis , Queratinocitos/metabolismo , Ratones , Piel/patología , Células Th17/fisiología , Células Th2/fisiología , Factores de Transcripción/genética , Factores de Transcripción/fisiología , Transcriptoma , Proteína AIRE
19.
Adv Exp Med Biol ; 587: 41-52, 2006.
Artículo en Inglés | MEDLINE | ID: mdl-17163154

RESUMEN

Ewing's sarcoma family of tumors (ESFT) are a clinically and scientifically very demanding group of tumors in children and young adults with still unknown histogenesis. The rate-limiting oncogenic mutation in this disease has been identified as a chromosomal translocation, t(11;22)(q24;q12), that leads to the expression of a chimeric transcription factor, EWS-FLI1. We have studied the downstream pathway of EWS-FLI1 by a dual strategy including the isolation of direct target genes from ESFT chromatin and the monitoring of transcriptomic changes after silencing of EWS-FLI1 by RNA interference. This study has lead to the identification of several directly EWS-FLI1-regulated genes and the characterization of their genomic distribution. By comparing several ESFT cell lines, not only variation in overall gene expression patterns downstream of EWS-FLIl was observed, but also differential regulation of directly EWS-FLI1-bound genes. Interestingly, there was variation between members of the same functional gene families. Studies on CD99, another diagnostic hallmark of ESFT, in relation to EWS-FLI1 provided additional evidence for context dependence of fusion protein function. Together, our study represents a first approach to the separation of essential molecular consequences from noise generated by the EWS-FLI1 gene rearrangement in ESFT.


Asunto(s)
Neoplasias Óseas/fisiopatología , Proteínas de Fusión Oncogénica/fisiología , Proteína Proto-Oncogénica c-fli-1/genética , Proteína Proto-Oncogénica c-fli-1/fisiología , Proteína EWS de Unión a ARN/genética , Sarcoma de Ewing/fisiopatología , Neoplasias Óseas/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Proteínas de Fusión Oncogénica/genética , Sarcoma de Ewing/genética , Translocación Genética
20.
Oncogene ; 35(30): 3944-54, 2016 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-26616853

RESUMEN

Sarcomas are mesenchymal tumors characterized by blocked differentiation process. In Ewing sarcoma (EWS) both CD99 and EWS-FLI1 concur to oncogenesis and inhibition of differentiation. Here, we demonstrate that uncoupling CD99 from EWS-FLI1 by silencing the former, nuclear factor-κB (NF-κB) signaling is inhibited and the neural differentiation program is re-established. NF-κB inhibition passes through miR-34a-mediated repression of Notch pathway. CD99 counteracts EWS-FLI1 in controlling NF-κB signaling through the miR-34a, which is increased and secreted into exosomes released by CD99-silenced EWS cells. Delivery of exosomes from CD99-silenced cells was sufficient to induce neural differentiation in recipient EWS cells through miR-34a inhibition of Notch-NF-κB signaling. Notably, even the partial delivery of CD99 small interfering RNA may have a broad effect on the entire tumor cell population owing to the spread operated by their miR-34a-enriched exosomes, a feature opening to a new therapeutic option.


Asunto(s)
Antígeno 12E7/fisiología , MicroARNs/fisiología , FN-kappa B/fisiología , Receptores Notch/fisiología , Sarcoma de Ewing/patología , Transducción de Señal/fisiología , Diferenciación Celular , Humanos , Proteínas de Fusión Oncogénica/fisiología , Proteína Proto-Oncogénica c-fli-1/fisiología , ARN Interferente Pequeño/genética , Proteína EWS de Unión a ARN/fisiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA