Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Proc Natl Acad Sci U S A ; 114(2): 376-381, 2017 01 10.
Artículo en Inglés | MEDLINE | ID: mdl-28028233

RESUMEN

The unrestrained growth of tumor cells is generally attributed to mutations in essential growth control genes, but tumor cells are also affected by, or even addicted to, signals from the microenvironment. As therapeutic targets, these extrinsic signals may be equally significant as mutated oncogenes. In multiple myeloma (MM), a plasma cell malignancy, most tumors display hallmarks of active Wnt signaling but lack activating Wnt-pathway mutations, suggesting activation by autocrine Wnt ligands and/or paracrine Wnts emanating from the bone marrow (BM) niche. Here, we report a pivotal role for the R-spondin/leucine-rich repeat-containing G protein-coupled receptor 4 (LGR4) axis in driving aberrant Wnt/ß-catenin signaling in MM. We show that LGR4 is expressed by MM plasma cells, but not by normal plasma cells or B cells. This aberrant LGR4 expression is driven by IL-6/STAT3 signaling and allows MM cells to hijack R-spondins produced by (pre)osteoblasts in the BM niche, resulting in Wnt (co)receptor stabilization and a dramatically increased sensitivity to auto- and paracrine Wnts. Our study identifies aberrant R-spondin/LGR4 signaling with consequent deregulation of Wnt (co)receptor turnover as a driver of oncogenic Wnt/ß-catenin signaling in MM cells. These results advocate targeting of the LGR4/R-spondin interaction as a therapeutic strategy in MM.


Asunto(s)
Glicoproteínas de Membrana/metabolismo , Mieloma Múltiple/metabolismo , Osteoblastos/metabolismo , Receptores Acoplados a Proteínas G/metabolismo , Proteínas Wnt/metabolismo , Vía de Señalización Wnt/fisiología , Animales , Línea Celular Tumoral , Células HEK293 , Humanos , Interleucina-6/metabolismo , Ligandos , Ratones , Unión Proteica/fisiología , Factor de Transcripción STAT3/metabolismo , beta Catenina/metabolismo
2.
J Immunol ; 197(7): 2918-29, 2016 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-27559048

RESUMEN

The recombination activating gene (RAG) 1 and RAG2 protein complex introduces DNA breaks at Tcr and Ig gene segments that are required for V(D)J recombination in developing lymphocytes. Proper regulation of RAG1/2 expression safeguards the ordered assembly of Ag receptors and the development of lymphocytes, while minimizing the risk for collateral damage. The ataxia telangiectasia mutated (ATM) kinase is involved in the repair of RAG1/2-mediated DNA breaks and prevents their propagation. The simultaneous occurrence of RAG1/2-dependent and -independent DNA breaks in developing lymphocytes exposed to genotoxic stress increases the risk for aberrant recombinations. In this study, we assessed the effect of genotoxic stress on RAG1/2 expression in pre-B cells and show that activation of the DNA damage response resulted in the rapid ATM-dependent downregulation of RAG1/2 mRNA and protein expression. We show that DNA damage led to the loss of FOXO1 binding to the enhancer region of the RAG1/2 locus (Erag) and provoked FOXO1 cleavage. We also show that DNA damage caused by RAG1/2 activity in pre-B cells was able to downmodulate RAG1/2 expression and activity, confirming the existence of a negative feedback regulatory mechanism. Our data suggest that pre-B cells are endowed with a protective mechanism that reduces the risk for aberrant recombinations and chromosomal translocations when exposed to DNA damage, involving the ATM-dependent regulation of FOXO1 binding to the Erag enhancer region.


Asunto(s)
Proteínas de la Ataxia Telangiectasia Mutada/metabolismo , Daño del ADN , Proteínas de Unión al ADN/genética , Proteína Forkhead Box O1/metabolismo , Proteínas de Homeodominio/genética , Proteínas Nucleares/genética , Células Precursoras de Linfocitos B/metabolismo , Transducción de Señal , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Células HEK293 , Proteínas de Homeodominio/metabolismo , Humanos , Proteínas Nucleares/metabolismo
3.
Blood ; 126(11): 1324-35, 2015 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-26153519

RESUMEN

In developing lymphocytes, expression and activity of the recombination activation gene protein 1 (RAG1) and RAG2 endonuclease complex is tightly regulated to ensure ordered recombination of the immunoglobulin genes and to avoid genomic instability. Aberrant RAG activity has been implicated in the generation of secondary genetic events in human B-cell acute lymphoblastic leukemias (B-ALLs), illustrating the oncogenic potential of the RAG complex. Several layers of regulation prevent collateral genomic DNA damage by restricting RAG activity to the G1 phase of the cell cycle. In this study, we show a novel pathway that suppresses RAG expression in cycling-transformed mouse pre-B cells and human pre-B B-ALL cells that involves the negative regulation of FOXO1 by nuclear factor κB (NF-κB). Inhibition of NF-κB in cycling pre-B cells resulted in upregulation of RAG expression and recombination activity, which provoked RAG-dependent DNA damage. In agreement, we observe a negative correlation between NF-κB activity and the expression of RAG1, RAG2, and TdT in B-ALL patients. Our data suggest that targeting NF-κB in B-ALL increases the risk of RAG-dependent genomic instability.


Asunto(s)
Daño del ADN , FN-kappa B/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Animales , Secuencia de Bases , Línea Celular , ADN/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/metabolismo , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/antagonistas & inhibidores , Factores de Transcripción Forkhead/metabolismo , Expresión Génica , Genes RAG-1 , Genes abl , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Humanos , Cadenas kappa de Inmunoglobulina/genética , Ratones , Ratones Noqueados , Modelos Biológicos , Datos de Secuencia Molecular , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/inmunología , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/inmunología , Transducción de Señal , Transformación Genética
4.
Blood ; 124(16): 2479-86, 2014 Oct 16.
Artículo en Inglés | MEDLINE | ID: mdl-25185711

RESUMEN

The proinflammatory cytokine interferon-γ (IFN-γ) is well known for its important role in innate and adaptive immunity against intracellular infections and for tumor control. Yet, it has become clear that IFN-γ also has a strong impact on bone marrow (BM) output during inflammation, as it affects the differentiation of most hematopoietic progenitor cells. Here, we review the impact of IFN-γ on hematopoiesis, including the function of hematopoietic stem cells (HSCs) and more downstream progenitors. We discuss which hematopoietic lineages are functionally modulated by IFN-γ and through which underlying molecular mechanism(s). We propose the novel concept that IFN-γ acts through upregulation of suppressor of cytokine signaling molecules, which impairs signaling of several cytokine receptors. IFN-γ has also gained clinical interest from different angles, and we discuss how chronic IFN-γ production can lead to the development of anemia and BM failure and how it is involved in malignant hematopoiesis. Overall, this review illustrates the wide-ranging effect of IFN-γ on the (patho-)physiological processes in the BM.


Asunto(s)
Hematopoyesis , Células Madre Hematopoyéticas/citología , Interferón gamma/metabolismo , Animales , Médula Ósea/inmunología , Médula Ósea/metabolismo , Médula Ósea/patología , Citocinas/inmunología , Citocinas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Células Madre Hematopoyéticas/inmunología , Células Madre Hematopoyéticas/metabolismo , Células Madre Hematopoyéticas/patología , Humanos , Interferón gamma/inmunología , Transducción de Señal , Factores de Transcripción/genética
5.
Blood ; 121(18): 3578-85, 2013 May 02.
Artículo en Inglés | MEDLINE | ID: mdl-23487025

RESUMEN

Balancing the processes of hematopoietic stem cell (HSC) differentiation and self-renewal is critical for maintaining a lifelong supply of blood cells. The bone marrow (BM) produces a stable output of newly generated cells, but immunologic stress conditions inducing leukopenia increase the demand for peripheral blood cell supply. Here we demonstrate that the proinflammatory cytokine interferon-γ (IFN-γ) impairs maintenance of HSCs by directly reducing their proliferative capacity and that IFN-γ impairs restoration of HSC numbers upon viral infection. We show that IFN-γ reduces thrombopoietin (TPO)-mediated phosphorylation of signal transducer and activator of transcription (STAT) 5, an important positive regulator of HSC self-renewal. IFN-γ also induced expression of suppressor of cytokine signaling (SOCS) 1 in HSCs, and we demonstrate that SOCS1 expression is sufficient to inhibit TPO-induced STAT5 phosphorylation. Furthermore, IFN-γ deregulates expression of STAT5-mediated cell-cycle genes cyclin D1 and p57. These findings suggest that IFN-γ is a negative modulator of HSC self-renewal by modifying cytokine responses and expression of genes involved in HSC proliferation. We postulate that the occurrence of BM failure in chronic inflammatory conditions, such as aplastic anemia, HIV, and graft-versus-host disease, is related to a sustained impairment of HSC self-renewal caused by chronic IFN-γ signaling in these disorders.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Células Madre Hematopoyéticas/efectos de los fármacos , Interferón gamma/farmacología , Animales , Células Cultivadas , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/fisiología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Biológicos , Receptores de Interferón/genética , Receptores de Interferón/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Trombopoyetina/metabolismo , Virosis/genética , Virosis/inmunología , Virosis/metabolismo , Virosis/patología , Receptor de Interferón gamma
6.
Blood ; 119(6): 1543-54, 2012 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-22117048

RESUMEN

Steady-state hematopoiesis is altered on infection, but the cellular and molecular mechanisms driving these changes are largely unknown. Modulation of hematopoiesis is essential to increase the output of the appropriate type of effector cell required to combat the invading pathogen. In the present study, we demonstrate that the pro-inflammatory cytokine IFNγ is involved in orchestrating inflammation-induced myelopoiesis. Using both mouse models and in vitro assays, we show that IFNγ induces the differentiation of monocytes over neutrophils at the level of myeloid progenitors. Infection with lymphocytic choriomeningitis virus induces monopoiesis in wild-type mice, but causes increased neutrophil production in IFNγ(-/-) mice. We demonstrate that IFNγ enhances the expression of the monopoiesis-inducing transcription factors IRF8 and PU.1 in myeloid progenitor cells, whereas it reduces G-CSF-driven neutrophil differentiation via a SOCS3-dependent inhibition of STAT3 phosphorylation. These results establish a critical role for IFNγ in directing monocyte versus neutrophil development during immune activation.


Asunto(s)
Mediadores de Inflamación/fisiología , Inflamación/metabolismo , Interferón gamma/fisiología , Neutrófilos/metabolismo , Animales , Células de la Médula Ósea/inmunología , Células de la Médula Ósea/metabolismo , Ligando CD27/genética , Ligando CD27/metabolismo , Diferenciación Celular/efectos de los fármacos , Células Cultivadas , Factor Estimulante de Colonias de Granulocitos/farmacología , Inflamación/genética , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/farmacología , Factores Reguladores del Interferón/metabolismo , Interferón gamma/genética , Interferón gamma/farmacología , Coriomeningitis Linfocítica/genética , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Monocitos/citología , Monocitos/metabolismo , Células Progenitoras Mieloides/metabolismo , Mielopoyesis/efectos de los fármacos , Neutrófilos/citología , Fosforilación , Proteínas Proto-Oncogénicas/metabolismo , Factor de Transcripción STAT3/metabolismo , Proteína 3 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transactivadores/metabolismo
7.
Blood ; 118(9): 2578-88, 2011 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-21725055

RESUMEN

Anemia of chronic disease is a complication accompanying many inflammatory diseases. The proinflammatory cytokine IFN-γ has been implicated in this form of anemia, but the underlying mechanism remains unclear. Here we describe a novel mouse model for anemia of chronic disease, in which enhanced CD27-mediated costimulation strongly increases the formation of IFN-γ-producing effector T cells, leading to a progressive anemia. We demonstrate that the anemia in these mice is fully dependent on IFN-γ and that this cytokine reduces both the life span and the formation of red blood cells. Molecular analysis revealed that IFN-γ induces expression of the transcription factors of interferon regulatory factor-1 (IRF-1) and PU.1 in both murine and human erythroid precursors. We found that, on IFN-γ stimulation, IRF-1 binds to the promoter of SPI.1 (PU.1) and induces PU.1 expression, leading to inhibition of erythropoiesis. Notably, down-regulation of either IRF-1 or PU.1 expression is sufficient to overcome IFN-γ-induced inhibition of erythropoiesis. These findings reveal a molecular mechanism by which chronic exposure to IFN-γ induces anemia.


Asunto(s)
Anemia/etiología , Modelos Animales de Enfermedad , Envejecimiento Eritrocítico/efectos de los fármacos , Células Precursoras Eritroides/efectos de los fármacos , Eritropoyesis/efectos de los fármacos , Factor 1 Regulador del Interferón/fisiología , Interferón gamma/farmacología , Proteínas Proto-Oncogénicas/fisiología , Transactivadores/fisiología , Animales , Ligando CD27/genética , Ligando CD27/fisiología , Enfermedad Crónica , Ensayo de Unidades Formadoras de Colonias , Células Precursoras Eritroides/citología , Factor 1 Regulador del Interferón/antagonistas & inhibidores , Factor 1 Regulador del Interferón/biosíntesis , Factor 1 Regulador del Interferón/sangre , Factor 1 Regulador del Interferón/genética , Macrófagos/fisiología , Ratones , Ratones Endogámicos C57BL , Fagocitosis , Proteínas Proto-Oncogénicas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas/biosíntesis , Proteínas Proto-Oncogénicas/sangre , Proteínas Proto-Oncogénicas/genética , ARN Interferente Pequeño/farmacología , Proteínas Recombinantes de Fusión/fisiología , Organismos Libres de Patógenos Específicos , Transactivadores/antagonistas & inhibidores , Transactivadores/biosíntesis , Transactivadores/sangre , Transactivadores/genética
8.
Blood ; 116(14): 2559-69, 2010 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-20587787

RESUMEN

To explore whether and how T cells can affect myelopoiesis, we investigated myeloid differentiation in a model for T cell-mediated immune activation. We found that CD70-transgenic (CD70TG) mice, which have elevated numbers of interferon-γ (IFN-γ)-producing effector T cells in the periphery and bone marrow, are almost devoid of eosinophilic granulocytes. Induction of allergic airway inflammation in these mice failed to induce eosinophilia as well as airway hyperresponsiveness. CD70TG mice also have strongly reduced numbers of eosinophil lineage-committed progenitors, whereas granulocyte/macrophage progenitors from these mice are unable to generate eosinophils in vitro. We found that granulocyte/macrophage progenitors express IFN-γR1 and that IFN-γ is sufficient to inhibit eosinophil differentiation of both murine and human progenitor cells in vitro. We demonstrate that inhibition of eosinophil development in CD70TG mice is IFN-γ-dependent and that T cell-derived IFN-γ is sufficient to inhibit eosinophil formation in vivo. Finally, we found that IFN-γ produced on anti-CD40 treatment and during viral infection can also suppress eosinophil formation in wild-type mice. These data demonstrate that IFN-γ inhibits the differentiation of myeloid progenitors to eosinophils, indicating that the adaptive immune system plays an important role in orchestrating the formation of the appropriate type of myeloid cells during immune activation.


Asunto(s)
Células de la Médula Ósea/citología , Eosinófilos/citología , Interferón gamma/inmunología , Leucopoyesis , Linfocitos T/inmunología , Animales , Asma/inmunología , Células de la Médula Ósea/inmunología , Ligando CD27/inmunología , Línea Celular , Células Cultivadas , Eosinófilos/inmunología , Humanos , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/citología , Células Progenitoras Mieloides/citología , Neutrófilos/citología , Linfocitos T/citología
9.
Blood ; 115(7): 1453-60, 2010 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-20008786

RESUMEN

In this study, the effect of human erythropoietin Delta (Epo) on smooth muscle cell (SMC)-rich lesions was evaluated. Mice, of which the left carotid artery was ligated, were treated with suberythropoietic as well as erythropoietic doses of Epo and both doses of Epo enhanced SMC-rich lesion formation. No association was observed between hemoglobin levels and lesion size. Moreover, endothelial progenitor cell (EPC) numbers in the peripheral blood increased only in the erythropoietic dosing group, indicating that EPC numbers did not correlate with lesion size. Immunohistochemical analysis revealed that Epo-mediated enhancement of lesion formation correlates with increased signal transducer and activator of transcription 5 (Stat5) phosphorylation in the vessel wall. Experiments performed in cultured vascular cells demonstrated that Epo robustly induced phosphorylation of Stat5 in human umbilical vein endothelial cells (HUVECs), but only very weakly in SMCs. In tumor necrosis factor-alpha (TNFalpha)-activated HUVECS, Epo induced expression of platelet-derived growth factor B (PDGF-B), which was at least partially responsible for the induction of Stat5 phosphorylation in SMCs by HUVEC-conditioned medium. In conclusion, in mice Epo accelerates SMC-rich neointima formation, which correlates with increased Stat5 phosphorylation in the vessel wall but is independent of erythrocyte and EPC numbers.


Asunto(s)
Arterias Carótidas/efectos de los fármacos , Enfermedades de las Arterias Carótidas/tratamiento farmacológico , Eritropoyetina/farmacología , Músculo Liso Vascular/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Inductores de la Angiogénesis/farmacología , Animales , Becaplermina , Arterias Carótidas/patología , Arterias Carótidas/fisiología , Enfermedades de las Arterias Carótidas/patología , Células Cultivadas , Medios de Cultivo Condicionados/farmacología , Relación Dosis-Respuesta a Droga , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Endoteliales/patología , Hemoglobinas/metabolismo , Humanos , Ligadura , Masculino , Ratones , Ratones Endogámicos , Músculo Liso Vascular/patología , Músculo Liso Vascular/fisiología , Fosforilación/efectos de los fármacos , Factor de Crecimiento Derivado de Plaquetas/farmacología , Proteínas Proto-Oncogénicas c-sis , Factor de Transcripción STAT5/metabolismo , Factor de Necrosis Tumoral alfa/farmacología , Túnica Íntima/efectos de los fármacos , Túnica Íntima/patología , Túnica Íntima/fisiología , Arterias Umbilicales/citología , Venas Umbilicales/citología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA