Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Biochem J ; 476(19): 2869-2882, 2019 10 15.
Artículo en Inglés | MEDLINE | ID: mdl-31488575

RESUMEN

Interleukin 6 (IL6) is a cytokine that regulates a number of important immune and inflammatory pathways. We used the ability of IL6 to inhibit the clonal proliferation of the mouse M1 myeloid leukemia cell line in agar to positively screen a cDNA expression library for proteins that inhibited IL6 activity. We found three clones completely resistant to IL6 that contained the cDNA for the Membrane-Associated RING-CH E3 ubiquitin ligase MARCH2. MARCH2 is a member of a family of membrane-bound E3 ubiquitin ligases that target cell surface receptors for degradation. MARCH2 overexpressing M1 clones retained responsiveness to the related cytokines leukemia inhibitory factor and oncostatin M and we showed that its inhibitory effect was a result of selective down-regulation of the IL6 receptor alpha chain and not the shared receptor subunit, gp130 or other signalling molecules. This activity of MARCH2 was also shared with related proteins MARCH4, MARCH9 and an isoform of MARCH3. The transmembrane domains and C-terminal domains, as well as a functional RING domain, of MARCH proteins were all required for substrate recognition and down-regulation. Genetic deletion of individual MARCH proteins in mice had no or little effect on IL6Rα levels but combined deletions of MARCH2,3 and 4 displayed elevated steady-state levels of IL6Rα in selected haemopoietic cell subsets including CD8+ and CD4+ T cells. These studies extend the potential immunosuppressive roles of MARCH proteins to include down-regulation of IL6 inflammatory responses.


Asunto(s)
Membrana Celular/metabolismo , Proteínas de la Membrana/fisiología , Receptores de Interleucina-6/metabolismo , Ubiquitina-Proteína Ligasas/fisiología , Animales , Línea Celular Tumoral , Regulación hacia Abajo , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Dominios Proteicos , Transporte de Proteínas
2.
J Immunol ; 193(4): 1766-77, 2014 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-25024380

RESUMEN

The IFN regulatory factor family member 8 (IRF8) regulates differentiation of lymphoid and myeloid lineage cells by promoting or suppressing lineage-specific genes. How IRF8 promotes hematopoietic progenitors to commit to one lineage while preventing the development of alternative lineages is not known. In this study, we report an IRF8-EGFP fusion protein reporter mouse that revealed previously unrecognized patterns of IRF8 expression. Differentiation of hematopoietic stem cells into oligopotent progenitors is associated with progressive increases in IRF8-EGFP expression. However, significant induction of IRF8-EGFP is found in granulocyte-myeloid progenitors and the common lymphoid progenitors but not the megakaryocytic-erythroid progenitors. Surprisingly, IRF8-EGFP identifies three subsets of the seemingly homogeneous granulocyte-myeloid progenitors with an intermediate level of expression of EGFP defining bipotent progenitors that differentiation into either EGFP(hi) monocytic progenitors or EGFP(lo) granulocytic progenitors. Also surprisingly, IRF8-EGFP revealed a highly heterogeneous pre-pro-B population with a fluorescence intensity ranging from background to 4 orders above background. Interestingly, IRF8-EGFP readily distinguishes true B cell committed (EGFP(int)) from those that are noncommitted. Moreover, dendritic cell progenitors expressed extremely high levels of IRF8-EGFP. Taken together, the IRF8-EGFP reporter revealed previously unrecognized subsets with distinct developmental potentials in phenotypically well-defined oligopotent progenitors, providing new insights into the dynamic heterogeneity of developing hematopoietic progenitors.


Asunto(s)
Factores Reguladores del Interferón/genética , Linfopoyesis/inmunología , Mielopoyesis/inmunología , Animales , Linfocitos B/citología , Diferenciación Celular/inmunología , Linaje de la Célula/inmunología , Genes Reporteros , Genotipo , Factor Estimulante de Colonias de Granulocitos/farmacología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/farmacología , Granulocitos/citología , Proteínas Fluorescentes Verdes/genética , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/inmunología , Factores Reguladores del Interferón/biosíntesis , Interleucina-3/farmacología , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos/citología , Ratones , Ratones Endogámicos C57BL , Células Mieloides/citología , Proteínas Recombinantes de Fusión/genética , Linfocitos T/citología
3.
Sci Adv ; 10(10): eadj8803, 2024 Mar 08.
Artículo en Inglés | MEDLINE | ID: mdl-38457494

RESUMEN

Philadelphia chromosome-positive B cell acute lymphoblastic leukemia (B-ALL), characterized by the BCR::ABL1 fusion gene, remains a poor prognosis cancer needing new therapeutic approaches. Transcriptomic profiling identified up-regulation of oncogenic transcription factors ERG and c-MYC in BCR::ABL1 B-ALL with ERG and c-MYC required for BCR::ABL1 B-ALL in murine and human models. Profiling of ERG- and c-MYC-dependent gene expression and analysis of ChIP-seq data established ERG and c-MYC coordinate a regulatory network in BCR::ABL1 B-ALL that controls expression of genes involved in several biological processes. Prominent was control of ribosome biogenesis, including expression of RNA polymerase I (POL I) subunits, the importance of which was validated by inhibition of BCR::ABL1 cells by POL I inhibitors, including CX-5461, that prevents promoter recruitment and transcription initiation by POL I. Our results reveal an essential ERG- and c-MYC-dependent transcriptional network involved in regulation of metabolic and ribosome biogenesis pathways in BCR::ABL1 B-ALL, from which previously unidentified vulnerabilities and therapeutic targets may emerge.


Asunto(s)
Proteínas de Fusión bcr-abl , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Regulador Transcripcional ERG , Animales , Humanos , Ratones , Proteínas de Fusión bcr-abl/genética , Proteínas de Fusión bcr-abl/metabolismo , Proteínas de Fusión bcr-abl/uso terapéutico , Redes Reguladoras de Genes , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras , Factores de Transcripción/genética , Regulador Transcripcional ERG/genética
4.
Nat Commun ; 11(1): 3013, 2020 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-32541654

RESUMEN

B lymphoid development is initiated by the differentiation of hematopoietic stem cells into lineage committed progenitors, ultimately generating mature B cells. This highly regulated process generates clonal immunological diversity via recombination of immunoglobulin V, D and J gene segments. While several transcription factors that control B cell development and V(D)J recombination have been defined, how these processes are initiated and coordinated into a precise regulatory network remains poorly understood. Here, we show that the transcription factor ETS Related Gene (Erg) is essential for early B lymphoid differentiation. Erg initiates a transcriptional network involving the B cell lineage defining genes, Ebf1 and Pax5, which directly promotes expression of key genes involved in V(D)J recombination and formation of the B cell receptor. Complementation of Erg deficiency with a productively rearranged immunoglobulin gene rescued B lineage development, demonstrating that Erg is an essential and stage-specific regulator of the gene regulatory network controlling B lymphopoiesis.


Asunto(s)
Linfocitos B/metabolismo , Diferenciación Celular/genética , Células Madre Hematopoyéticas/metabolismo , Linfopoyesis/genética , Proteínas Oncogénicas/genética , Transcripción Genética , Regulador Transcripcional ERG/genética , Animales , Linfocitos B/citología , Linaje de la Célula/genética , Células Cultivadas , Redes Reguladoras de Genes/genética , Células Madre Hematopoyéticas/citología , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Oncogénicas/metabolismo , Factor de Transcripción PAX5/genética , Factor de Transcripción PAX5/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Regulador Transcripcional ERG/metabolismo , Recombinación V(D)J/genética
5.
Cell Stem Cell ; 25(2): 258-272.e9, 2019 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-31374198

RESUMEN

Tumors are composed of phenotypically heterogeneous cancer cells that often resemble various differentiation states of their lineage of origin. Within this hierarchy, it is thought that an immature subpopulation of tumor-propagating cancer stem cells (CSCs) differentiates into non-tumorigenic progeny, providing a rationale for therapeutic strategies that specifically eradicate CSCs or induce their differentiation. The clinical success of these approaches depends on CSC differentiation being unidirectional rather than reversible, yet this question remains unresolved even in prototypically hierarchical malignancies, such as acute myeloid leukemia (AML). Here, we show in murine and human models of AML that, upon perturbation of endogenous expression of the lineage-determining transcription factor PU.1 or withdrawal of established differentiation therapies, some mature leukemia cells can de-differentiate and reacquire clonogenic and leukemogenic properties. Our results reveal plasticity of CSC maturation in AML, highlighting the need to therapeutically eradicate cancer cells across a range of differentiation states.


Asunto(s)
Diferenciación Celular/fisiología , Transdiferenciación Celular/fisiología , Leucemia Mieloide Aguda/patología , Células Madre Neoplásicas/fisiología , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Animales , Carcinogénesis , Plasticidad de la Célula , Células Cultivadas , Humanos , Leucemia Mieloide Aguda/metabolismo , Ratones , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Tretinoina/metabolismo
6.
Cell Metab ; 23(1): 155-64, 2016 Jan 12.
Artículo en Inglés | MEDLINE | ID: mdl-26603191

RESUMEN

Interleukin-18 (IL-18) is activated by Caspase-1 in inflammasome complexes and has anti-obesity effects; however, it is not known which inflammasome regulates this process. We found that mice lacking the NLRP1 inflammasome phenocopy mice lacking IL-18, with spontaneous obesity due to intrinsic lipid accumulation. This is exacerbated when the mice are fed a high-fat diet (HFD) or a high-protein diet, but not when mice are fed a HFD with low energy density (high fiber). Furthermore, mice with an activating mutation in NLRP1, and hence increased IL-18, have decreased adiposity and are resistant to diet-induced metabolic dysfunction. Feeding these mice a HFD further increased plasma IL-18 concentrations and strikingly resulted in loss of adipose tissue mass and fatal cachexia, which could be prevented by genetic deletion of IL-18. Thus, NLRP1 is an innate immune sensor that functions in the context of metabolic stress to produce IL-18, preventing obesity and metabolic syndrome.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Proteínas Reguladoras de la Apoptosis/metabolismo , Inflamasomas/metabolismo , Interleucina-18/biosíntesis , Síndrome Metabólico/metabolismo , Obesidad/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Proteínas Reguladoras de la Apoptosis/genética , Peso Corporal , Dieta Alta en Grasa/efectos adversos , Interleucina-18/genética , Hígado/metabolismo , Hígado/patología , Masculino , Síndrome Metabólico/prevención & control , Ratones Noqueados , Obesidad/etiología , Obesidad/prevención & control
7.
J Exp Med ; 212(6): 927-38, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-26008898

RESUMEN

Gain-of-function mutations that activate the innate immune system can cause systemic autoinflammatory diseases associated with increased IL-1ß production. This cytokine is activated identically to IL-18 by an intracellular protein complex known as the inflammasome; however, IL-18 has not yet been specifically implicated in the pathogenesis of hereditary autoinflammatory disorders. We have now identified an autoinflammatory disease in mice driven by IL-18, but not IL-1ß, resulting from an inactivating mutation of the actin-depolymerizing cofactor Wdr1. This perturbation of actin polymerization leads to systemic autoinflammation that is reduced when IL-18 is deleted but not when IL-1 signaling is removed. Remarkably, inflammasome activation in mature macrophages is unaltered, but IL-18 production from monocytes is greatly exaggerated, and depletion of monocytes in vivo prevents the disease. Small-molecule inhibition of actin polymerization can remove potential danger signals from the system and prevents monocyte IL-18 production. Finally, we show that the inflammasome sensor of actin dynamics in this system requires caspase-1, apoptosis-associated speck-like protein containing a caspase recruitment domain, and the innate immune receptor pyrin. Previously, perturbation of actin polymerization by pathogens was shown to activate the pyrin inflammasome, so our data now extend this guard hypothesis to host-regulated actin-dependent processes and autoinflammatory disease.


Asunto(s)
Actinas/fisiología , Proteínas del Citoesqueleto/metabolismo , Enfermedades Autoinflamatorias Hereditarias/metabolismo , Inflamación/metabolismo , Interleucina-1beta/metabolismo , Proteínas de Microfilamentos/metabolismo , Actinas/química , Animales , Células de la Médula Ósea/citología , Caspasa 1/metabolismo , Caspasas/metabolismo , Ácido Clodrónico/química , Cruzamientos Genéticos , Medios de Cultivo Condicionados/química , Ensayo de Inmunoadsorción Enzimática , Interleucina-18/metabolismo , Lipopolisacáridos/metabolismo , Liposomas/química , Hígado/embriología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Fluorescente , Monocitos/citología , Pirina , Transducción de Señal
8.
Proc Natl Acad Sci U S A ; 103(44): 16460-5, 2006 Oct 31.
Artículo en Inglés | MEDLINE | ID: mdl-17060613

RESUMEN

Mixl1, the sole murine homologue of the Xenopus Mix/Bix family of homeobox transcription factors, is essential for the patterning of axial mesendodermal structures during early embryogenesis. Gene targeting and overexpression studies have implicated Mixl1 as a regulator of hematopoiesis arising in differentiating embryonic stem cells. To assess the role of Mixl1 in the regulation of adult hematopoiesis, we overexpressed Mixl1 in murine bone marrow using a retroviral transduction/transplantation model. Enforced expression of Mixl1 profoundly perturbed hematopoietic lineage commitment and differentiation, giving rise to abnormal myeloid progenitors and impairing erythroid and lymphoid differentiation. Moreover, all mice reconstituted with Mixl1-transduced bone marrow developed fatal, transplantable acute myeloid leukemia with a mean latency period of 200 days. These observations establish a link between enforced Mixl1 expression and leukemogenesis in the mouse.


Asunto(s)
Diferenciación Celular , Hematopoyesis , Proteínas de Homeodominio/metabolismo , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patología , Animales , Médula Ósea/metabolismo , Transformación Celular Neoplásica/metabolismo , Transformación Celular Neoplásica/patología , Regulación de la Expresión Génica , Proteínas de Homeodominio/genética , Leucemia Mieloide Aguda/genética , Ratones , Ratones Transgénicos , Células Mieloides/citología , Células Mieloides/metabolismo , Fenotipo , Tasa de Supervivencia
9.
Blood ; 105(9): 3480-7, 2005 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-15665109

RESUMEN

Mpl(-/-) mice bearing the Plt3 or Plt4 mutations in the c-Myb gene exhibit thrombopoietin (TPO)-independent supraphysiological platelet production accompanied by excessive megakaryocytopoiesis and defective erythroid and lymphoid cell production. To better define the cellular basis for the thrombocytosis in these mice, we analyzed the production and characteristics of megakaryocytes and their progenitors. Consistent with thrombocytosis arising from hyperactive production, the high platelet counts in mice carrying the c-Myb(Plt4) allele were not accompanied by any significant alteration in platelet half-life. Megakaryocytes in c-Myb mutant mice displayed reduced modal DNA ploidy and, among the excessive numbers of megakaryocyte progenitor cells, more mature precursors were particularly evident. Megakaryocyte progenitor cells carrying the Plt3 or Plt4 c-Myb mutations, but not granulocyte-macrophage progenitors, exhibited 200-fold enhanced responsiveness to granulocyte-macrophage colony-stimulating factor (GM-CSF), suggesting that altered responses to cytokines may contribute to expanded megakaryocytopoiesis. Mutant preprogenitor (blast colony-forming) cells appeared to have little capacity to form megakaryocyte progenitor cells. In contrast, the spleens of irradiated mice 12 days after transplantation with mutant bone marrow contained abundant megakaryocyte progenitor cells, suggesting that altered c-Myb activity skews differentiation commitment in spleen colony-forming units (CFU-S) in favor of excess megakaryocytopoiesis.


Asunto(s)
Genes myb/fisiología , Trombopoyesis/genética , Alelos , Animales , Células de la Médula Ósea/citología , Trasplante de Médula Ósea , Células Cultivadas , Genes myb/genética , Células Madre Hematopoyéticas/citología , Ratones , Ratones Noqueados , Células Madre Multipotentes/citología , Mutación Missense , Bazo/citología , Trombocitosis/etiología , Trombocitosis/genética
10.
Immunity ; 20(2): 153-65, 2004 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-14975238

RESUMEN

To determine the importance of suppressor of cytokine signaling-3 (SOCS3) in the regulation of hematopoietic growth factor signaling generally, and of G-CSF-induced cellular responses specifically, we created mice in which the Socs3 gene was deleted in all hematopoietic cells. Although normal until young adulthood, these mice then developed neutrophilia and a spectrum of inflammatory pathologies. When stimulated with G-CSF in vitro, SOCS3-deficient cells of the neutrophilic granulocyte lineage exhibited prolonged STAT3 activation and enhanced cellular responses to G-CSF, including an increase in cloning frequency, survival, and proliferative capacity. Consistent with the in vitro findings, mutant mice injected with G-CSF displayed enhanced neutrophilia, progenitor cell mobilization, and splenomegaly, but unexpectedly also developed inflammatory neutrophil infiltration into multiple tissues and consequent hind-leg paresis. We conclude that SOCS3 is a key negative regulator of G-CSF signaling in myeloid cells and that this is of particular significance during G-CSF-driven emergency granulopoiesis.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos/inmunología , Granulocitos/inmunología , Inflamación/patología , Proteínas Represoras/inmunología , Factores de Transcripción/inmunología , Animales , Diferenciación Celular , Células Cultivadas , Proteínas de Unión al ADN/efectos de los fármacos , Proteínas de Unión al ADN/inmunología , Proteínas de Unión al ADN/metabolismo , Factor Estimulante de Colonias de Granulocitos/farmacología , Granulocitos/efectos de los fármacos , Granulocitos/metabolismo , Hematopoyesis/fisiología , Células Madre Hematopoyéticas/efectos de los fármacos , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Inflamación/inmunología , Ratones , Ratones Transgénicos , Neutrófilos/efectos de los fármacos , Neutrófilos/inmunología , Neutrófilos/metabolismo , Factor de Transcripción STAT3 , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas , Transactivadores/efectos de los fármacos , Transactivadores/inmunología , Transactivadores/metabolismo , Factores de Transcripción/deficiencia
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA