Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Immunity ; 45(1): 185-97, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27438771

RESUMEN

Group 3 innate lymphoid cells (ILC3s) expressing the transcription factor (TF) RORγt are important for the defense and homeostasis of host intestinal tissues. The zinc finger TF Ikaros, encoded by Ikzf1, is essential for the development of RORγt(+) fetal lymphoid tissue inducer (LTi) cells and lymphoid organogenesis, but its role in postnatal ILC3s is unknown. Here, we show that small-intestinal ILC3s had lower Ikaros expression than ILC precursors and other ILC subsets. Ikaros inhibited ILC3s in a cell-intrinsic manner through zinc-finger-dependent inhibition of transcriptional activity of the aryl hydrocarbon receptor, a key regulator of ILC3 maintenance and function. Ablation of Ikzf1 in RORγt(+) ILC3s resulted in increased expansion and cytokine production of intestinal ILC3s and protection against infection and colitis. Therefore, in contrast to being required for LTi development, Ikaros inhibits postnatal ILC3 development and function to regulate gut immune responses at steady state and in disease.


Asunto(s)
Colitis/inmunología , Factor de Transcripción Ikaros/metabolismo , Mucosa Intestinal/inmunología , Linfocitos/fisiología , Receptores de Hidrocarburo de Aril/metabolismo , Animales , Diferenciación Celular , Células Cultivadas , Colitis/inducido químicamente , Sulfato de Dextran , Homeostasis , Factor de Transcripción Ikaros/genética , Inmunidad Innata , Mucosa Intestinal/microbiología , Activación de Linfocitos , Linfocitos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores de Hidrocarburo de Aril/genética , Transducción de Señal , Activación Transcripcional
2.
Biochem Biophys Res Commun ; 694: 149399, 2024 01 29.
Artículo en Inglés | MEDLINE | ID: mdl-38134477

RESUMEN

Ikaros family proteins (Ikaros, Helios, Aiolos, Eos) are zinc finger transcription factors essential for the development and function of the adaptive immune system. They also control developmental events in neurons and other cell types, suggesting that they possess crucial functions across disparate cell types. These functions are likely shared among the organisms in which these factors exist, and it is thus important to obtain a view of their distribution and conservation across organisms. How this family evolved remains poorly understood. Here we mined protein, mRNA and DNA databases to identify proteins with DNA-binding domains homologous to that of Ikaros. We show that Ikaros-related proteins exist in organisms from all four deuterostome phyla (chordates, echinoderms, hemichordates, xenacoelomorpha), but not in more distant groups. While most non-vertebrates have a single family member, this family grew to six members in the acoel worm Hofstenia miamia, three in jawless and four in jawed vertebrates. Most residues involved in DNA contact from zinc fingers 2 to 4 were identical across the Ikaros family, suggesting conserved mechanisms for target sequence recognition. Further, we identified a novel KRKxxxPxK/R motif that inhibits DNA binding in vitro which was conserved across the deuterostome phyla. We also identified a EψψxxxψM(D/E)QAIxxAIxYLGA(D/E)xL motif conserved among human Ikaros, Aiolos, Helios and subsets of chordate proteins, and motifs that are specific to subsets of vertebrate family members. Some of these motifs are targets of mutations in human patients. Finally we show that the atypical family member Pegasus emerged only in vertebrates, which is consistent with its function in bone. Our data provide a novel evolutionary perspective for Ikaros family proteins and suggest that they have conserved regulatory functions across deuterostomes.


Asunto(s)
Factor de Transcripción Ikaros , Dedos de Zinc , Animales , Humanos , ADN , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , ARN Mensajero , Dedos de Zinc/genética
3.
Nat Immunol ; 13(10): 972-80, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22961053

RESUMEN

The transcription factor Foxp3 participates dominantly in the specification and function of Foxp3(+)CD4(+) regulatory T cells (T(reg) cells) but is neither strictly necessary nor sufficient to determine the characteristic T(reg) cell signature. Here we used computational network inference and experimental testing to assess the contribution of other transcription factors to this. Enforced expression of Helios or Xbp1 elicited distinct signatures, but Eos, IRF4, Satb1, Lef1 and GATA-1 elicited exactly the same outcome, acting in synergy with Foxp3 to activate expression of most of the T(reg) cell signature, including key transcription factors, and enhancing occupancy by Foxp3 at its genomic targets. Conversely, the T(reg) cell signature was robust after inactivation of any single cofactor. A redundant genetic switch thus 'locked in' the T(reg) cell phenotype, a model that would account for several aspects of T(reg) cell physiology, differentiation and stability.


Asunto(s)
Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Animales , Antígenos CD4/biosíntesis , Diferenciación Celular , Biología Computacional , Proteínas de Unión al ADN/biosíntesis , Proteínas de Unión al ADN/genética , Factores de Transcripción Forkhead/genética , Factor de Transcripción GATA1/genética , Humanos , Factor de Transcripción Ikaros/biosíntesis , Factor de Transcripción Ikaros/genética , Factores Reguladores del Interferón/genética , Activación de Linfocitos , Factor de Unión 1 al Potenciador Linfoide/genética , Proteínas de Unión a la Región de Fijación a la Matriz/genética , Ratones , Ratones Endogámicos C57BL , Factores de Transcripción del Factor Regulador X , Serina Endopeptidasas/genética , Linfocitos T Reguladores/citología , Factores de Transcripción/biosíntesis , Factores de Transcripción/genética , Proteína 1 de Unión a la X-Box
4.
Cell ; 138(2): 300-13, 2009 Jul 23.
Artículo en Inglés | MEDLINE | ID: mdl-19632180

RESUMEN

While hematopoietic stem cell (HSC) self-renewal is well studied, it remains unknown whether distinct control mechanisms enable HSC divisions that generate progeny cells with specific lineage bias. Here, we report that the monocytic transcription factor MafB specifically restricts the ability of M-CSF to instruct myeloid commitment divisions in HSCs. MafB deficiency specifically enhanced sensitivity to M-CSF and caused activation of the myeloid master-regulator PU.1 in HSCs in vivo. Single-cell analysis revealed that reduced MafB levels enabled M-CSF to instruct divisions producing asymmetric daughter pairs with one PU.1(+) cell. As a consequence, MafB(-/-) HSCs showed a PU.1 and M-CSF receptor-dependent competitive repopulation advantage specifically in the myelomonocytic, but not T lymphoid or erythroid, compartment. Lineage-biased repopulation advantage was progressive, maintained long term, and serially transplantable. Together, this indicates that an integrated transcription factor/cytokine circuit can control the rate of specific HSC commitment divisions without compromising other lineages or self-renewal.


Asunto(s)
Linaje de la Célula , Células Madre Hematopoyéticas/citología , Factor Estimulante de Colonias de Macrófagos/metabolismo , Factor de Transcripción MafB/metabolismo , Células Mieloides/citología , Animales , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Madre Hematopoyéticas/metabolismo , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas/metabolismo , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Transactivadores/metabolismo
5.
Adv Exp Med Biol ; 1459: 33-52, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39017838

RESUMEN

The IKAROS family of transcription factors comprises four zinc-finger proteins (IKAROS, HELIOS, AIOLOS, and EOS), which over the last decades have been established to be critical regulators of the development and function of lymphoid cells. These factors act as homo- or heterodimers and are involved both in gene activation and repression. Their function often involves cross-talk with other regulatory circuits, such as the JAK/STAT, NF-κB, and NOTCH pathways. They control lymphocyte differentiation at multiple stages and are notably critical for lymphoid commitment in multipotent hematopoietic progenitors and for T and B cell differentiation downstream of pre-TCR and pre-BCR signaling. They also control many aspects of effector functions in mature B and T cells. They are dysregulated or mutated in multiple pathologies affecting the lymphoid system, which range from leukemia to immunodeficiencies. In this chapter, we review the molecular and physiological function of these factors in lymphocytes and their implications in human pathologies.


Asunto(s)
Diferenciación Celular , Factor de Transcripción Ikaros , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Animales , Transducción de Señal , Linfocitos/metabolismo , Linfocitos/inmunología
6.
Proc Natl Acad Sci U S A ; 118(17)2021 04 27.
Artículo en Inglés | MEDLINE | ID: mdl-33893236

RESUMEN

The production of proinflammatory cytokines, particularly granulocyte-macrophage colony-stimulating factor (GM-CSF), by pathogenic CD4+ T cells is central for mediating tissue injury in inflammatory and autoimmune diseases. However, the factors regulating the T cell pathogenic gene expression program remain unclear. Here, we investigated how the Ikaros transcription factor regulates the global gene expression and chromatin accessibility changes in murine T cells during Th17 polarization and after activation via the T cell receptor (TCR) and CD28. We found that, in both conditions, Ikaros represses the expression of genes from the pathogenic signature, particularly Csf2, which encodes GM-CSF. We show that, in TCR/CD28-activated T cells, Ikaros binds a critical enhancer downstream of Csf2 and is required to regulate chromatin accessibility at multiple regions across this locus. Genome-wide Ikaros binding is associated with more compact chromatin, notably at multiple sites containing NFκB or STAT5 target motifs, and STAT5 or NFκB inhibition prevents GM-CSF production in Ikaros-deficient cells. Importantly, Ikaros also limits GM-CSF production in TCR/CD28-activated human T cells. Our data therefore highlight a critical conserved transcriptional mechanism that antagonizes GM-CSF expression in T cells.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Factor de Transcripción Ikaros/metabolismo , Activación de Linfocitos , Diferenciación Celular , Células Cultivadas , Epigenoma , Regulación de la Expresión Génica , Humanos
7.
Biochem Biophys Res Commun ; 674: 83-89, 2023 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-37413709

RESUMEN

The transcriptional regulators that drive regulatory T (Treg) cell development and function remain partially understood. Helios (Ikzf2) and Eos (Ikzf4) are closely-related members of the Ikaros family of transcription factors. They are highly expressed in CD4+ Treg cells and functionally important for Treg cell biology, as mice deficient for either Helios or Eos are susceptible to autoimmune diseases. However, it remains unknown if these factors exhibit specific or partially redundant functions in Treg cells. Here we show that mice with germline deletions of both Ikzf2 and Ikzf4 are not very different from animals with single Ikzf2 or Ikzf4 deletions. Double knockout Treg cells differentiate normally, and efficiently suppress effector T cell proliferation in vitro. Both Helios and Eos are required for optimal Foxp3 protein expression. Surprisingly, Helios and Eos regulate different, largely non-overlapping, sets of genes. Only Helios is required for proper Treg cell aging, as Helios deficiency results in reduced Treg cell frequencies in the spleen of older animals. These results indicate that Helios and Eos are required for distinct aspects of Treg cell function.


Asunto(s)
Factor de Transcripción Ikaros , Linfocitos T Reguladores , Animales , Ratones , Enfermedades Autoinmunes/genética , Susceptibilidad a Enfermedades/metabolismo , Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Factores de Transcripción/metabolismo
8.
PLoS Genet ; 14(7): e1007485, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-30001316

RESUMEN

Plasmacytoid and conventional dendritic cells (pDCs and cDCs) arise from monocyte and dendritic progenitors (MDPs) and common dendritic progenitors (CDPs) through gene expression changes that remain partially understood. Here we show that the Ikaros transcription factor is required for DC development at multiple stages. Ikaros cooperates with Notch pathway activation to maintain the homeostasis of MDPs and CDPs. Ikaros then antagonizes TGFß function to promote pDC differentiation from CDPs. Strikingly, Ikaros-deficient CDPs and pDCs express a cDC-like transcriptional signature that is correlated with TGFß activation, suggesting that Ikaros is an upstream negative regulator of the TGFß pathway and a repressor of cDC-lineage genes in pDCs. Almost all of these phenotypes can be rescued by short-term in vitro treatment with γ-secretase inhibitors, which affects both TGFß-dependent and -independent pathways, but is Notch-independent. We conclude that Ikaros is a crucial differentiation factor in early dendritic progenitors that is required for pDC identity.


Asunto(s)
Diferenciación Celular/genética , Células Dendríticas/fisiología , Factor de Transcripción Ikaros/metabolismo , Receptores Notch/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Secretasas de la Proteína Precursora del Amiloide/metabolismo , Animales , Trasplante de Médula Ósea , Línea Celular , Regulación hacia Abajo , Células Madre Hematopoyéticas/fisiología , Factor de Transcripción Ikaros/genética , Ratones , Ratones Transgénicos , Monocitos/fisiología , Mutación , Transducción de Señal/genética , Regulación hacia Arriba
9.
Development ; 144(8): 1566-1577, 2017 04 15.
Artículo en Inglés | MEDLINE | ID: mdl-28289129

RESUMEN

Here, we unravel the mechanism of action of the Ikaros family zinc finger protein Helios (He) during the development of striatal medium spiny neurons (MSNs). He regulates the second wave of striatal neurogenesis involved in the generation of striatopallidal neurons, which express dopamine 2 receptor and enkephalin. To exert this effect, He is expressed in neural progenitor cells (NPCs) keeping them in the G1/G0 phase of the cell cycle. Thus, a lack of He results in an increase of S-phase entry and S-phase length of NPCs, which in turn impairs striatal neurogenesis and produces an accumulation of the number of cycling NPCs in the germinal zone (GZ), which end up dying at postnatal stages. Therefore, He-/- mice show a reduction in the number of dorso-medial striatal MSNs in the adult that produces deficits in motor skills acquisition. In addition, overexpression of He in NPCs induces misexpression of DARPP-32 when transplanted in mouse striatum. These findings demonstrate that He is involved in the correct development of a subset of striatopallidal MSNs and reveal new cellular mechanisms for neuronal development.


Asunto(s)
Cuerpo Estriado/citología , Proteínas de Unión al ADN/metabolismo , Globo Pálido/citología , Neuronas/citología , Neuronas/metabolismo , Factores de Transcripción/metabolismo , Animales , Animales Recién Nacidos , Recuento de Células , Puntos de Control del Ciclo Celular , Muerte Celular , Proliferación Celular , Ciclina E/metabolismo , Fase G1 , Ratones Noqueados , Actividad Motora , Células-Madre Neurales/citología , Células-Madre Neurales/metabolismo , Neurogénesis , Fenotipo , Fase S
10.
J Biol Chem ; 291(17): 9073-86, 2016 Apr 22.
Artículo en Inglés | MEDLINE | ID: mdl-26841869

RESUMEN

B1 B cells secrete most of the circulating natural antibodies and are considered key effector cells of the innate immune response. However, B1 cell-associated antibodies often cross-react with self-antigens, which leads to autoimmunity, and B1 cells have been implicated in cancer. How B1 cell activity is regulated remains unclear. We show that the Ikaros transcription factor is a major negative regulator of B1 cell development and function. Using conditional knock-out mouse models to delete Ikaros at different locations, we show that Ikaros-deficient mice exhibit specific and significant increases in splenic and bone marrow B1 cell numbers, and that the B1 progenitor cell pool is increased ∼10-fold in the bone marrow. Ikaros-null B1 cells resemble WT B1 cells at the molecular and cellular levels, but show a down-regulation of signaling components important for inhibiting proliferation and immunoglobulin production. Ikaros-null B1 cells hyper-react to TLR4 stimulation and secrete high amounts of IgM autoantibodies. These results indicate that Ikaros is required to limit B1 cell homeostasis in the adult.


Asunto(s)
Autoanticuerpos/inmunología , Subgrupos de Linfocitos B/inmunología , Células de la Médula Ósea/inmunología , Factor de Transcripción Ikaros/inmunología , Inmunoglobulina M/inmunología , Células Precursoras de Linfocitos B/inmunología , Animales , Factor de Transcripción Ikaros/genética , Ratones , Ratones Noqueados , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/inmunología
12.
BMC Bioinformatics ; 17(1): 462, 2016 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-27846811

RESUMEN

We published a new method (BMC Bioinformatics 2014, 15:14) for searching for differentially expressed genes from two biological conditions datasets. The presentation of theorem 1 in this paper was incomplete. We received an anonymous comment about our publication that motivates the present work. Here, we present a complementary result which is necessary from the theoretical point of view to demonstrate our theorem. We also show that this result has no negative impact on our conclusions obtained with synthetic and experimental microarrays datasets.


Asunto(s)
Algoritmos , Biología Computacional/métodos , Interpretación Estadística de Datos , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Simulación por Computador , Bases de Datos Genéticas , Humanos
13.
Biochem Biophys Res Commun ; 470(3): 714-720, 2016 Feb 12.
Artículo en Inglés | MEDLINE | ID: mdl-26775846

RESUMEN

The Ikaros transcription factor is essential for early B cell development, but its effect on mature B cells is debated. We show that Ikaros is required to limit the response of naive splenic B cells to B cell receptor signals. Ikaros deficient follicular B cells grow larger and enter cell cycle faster after anti-IgM stimulation. Unstimulated mutant B cells show deregulation of positive and negative regulators of signal transduction at the mRNA level, and constitutive phosphorylation of ERK, p38, SYK, BTK, AKT and LYN. Stimulation results in enhanced and prolonged ERK and p38 phosphorylation, followed by hyper-proliferation. Pharmacological inhibition of ERK and p38 abrogates the increased proliferative response of Ikaros deficient cells. These results suggest that Ikaros functions as a negative regulator of follicular B cell activation.


Asunto(s)
Factor de Transcripción Ikaros/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal/fisiología , Bazo/citología , Animales , Linfocitos B , Proliferación Celular/fisiología , Células Cultivadas , Regulación de la Expresión Génica/fisiología , Ratones , Ratones Endogámicos C57BL
14.
Blood ; 122(5): 694-704, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23801632

RESUMEN

Wnt signaling is important for T-cell differentiation at the early CD4(-)CD8(-) stage and is subsequently downregulated with maturation. To assess the importance of this downregulation, we generated a mouse line (R26-ßcat) in which high levels of active ß-catenin are maintained throughout T-cell development. Young R26-ßcat mice show a differentiation block at the CD4(+)CD8(+) double-positive (DP) stage. These DP cells exhibit impaired apoptosis upon irradiation or dexamethasone treatment. All R26-ßcat mice develop T-cell leukemias at 5 to 6 months of age. R26-ßcat leukemias remain dependent on ß-catenin function but lack Notch pathway activation. They exhibit recurrent secondary genomic rearrangements that lead to Myc overexpression and loss of Pten activity. Because ß-catenin activation and Myc translocations were previously found in murine T-cell acute lymphoblastic leukemias (T-ALLs) deficient for Pten, our results suggest that activation of the canonical Wnt pathway is associated with a subtype of Notch-independent T-ALLs that bear Myc gene rearrangements and Pten mutations.


Asunto(s)
Genes myc/genética , Fosfohidrolasa PTEN/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Receptores Notch/fisiología , beta Catenina/fisiología , Animales , Diferenciación Celular/genética , Eliminación de Gen , Regulación Leucémica de la Expresión Génica , Ratones , Ratones Transgénicos , Mutación/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/mortalidad , Receptores Notch/genética , Linfocitos T/metabolismo , Linfocitos T/fisiología , Regulación hacia Arriba/genética , Vía de Señalización Wnt/genética , Vía de Señalización Wnt/fisiología , beta Catenina/agonistas , beta Catenina/genética , beta Catenina/metabolismo
15.
BMC Bioinformatics ; 15: 14, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24423217

RESUMEN

BACKGROUND: Different methods have been proposed for analyzing differentially expressed (DE) genes in microarray data. Methods based on statistical tests that incorporate expression level variability are used more commonly than those based on fold change (FC). However, FC based results are more reproducible and biologically relevant. RESULTS: We propose a new method based on fold change rank ordering statistics (FCROS). We exploit the variation in calculated FC levels using combinatorial pairs of biological conditions in the datasets. A statistic is associated with the ranks of the FC values for each gene, and the resulting probability is used to identify the DE genes within an error level. The FCROS method is deterministic, requires a low computational runtime and also solves the problem of multiple tests which usually arises with microarray datasets. CONCLUSION: We compared the performance of FCROS with those of other methods using synthetic and real microarray datasets. We found that FCROS is well suited for DE gene identification from noisy datasets when compared with existing FC based methods.


Asunto(s)
Biología Computacional/métodos , Perfilación de la Expresión Génica/métodos , Análisis de Secuencia por Matrices de Oligonucleótidos/métodos , Algoritmos , Simulación por Computador , Bases de Datos Genéticas
17.
Blood ; 120(1): 90-9, 2012 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-22611152

RESUMEN

The physiologic role played by plasmacytoid dendritic cells (pDCs) in the induction of innate responses and inflammation in response to pathogen signaling is not well understood. Here, we describe a new mouse model lacking pDCs and establish that pDCs are essential for the in vivo induction of NK-cell activity in response to Toll-like receptor 9 (TLR9) triggering. Furthermore, we provide the first evidence that pDCs are critical for the systemic production of a wide variety of chemokines in response to TLR9 activation. Consequently, we observed a profound alteration in monocyte, macrophage, neutrophil, and NK-cell recruitment at the site of inflammation in the absence of pDCs in response to CpG-Dotap and stimulation by microbial pathogens, such as Leishmania major, Escherichia coli, and Mycobacterium bovis. This study, which is based on the development of a constitutively pDC-deficient mouse model, highlights the pivotal role played by pDCs in the induction of innate immune responses and inflammation after TLR9 triggering.


Asunto(s)
Células Dendríticas/inmunología , Infecciones/inmunología , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/microbiología , Receptor Toll-Like 9/inmunología , Animales , Linfocitos B/citología , Linfocitos B/inmunología , Movimiento Celular/inmunología , Quimiocinas/inmunología , Citocinas/inmunología , Proteínas de Unión al ADN/genética , Células Dendríticas/citología , Infecciones por Escherichia coli/inmunología , Inmunidad Innata/inmunología , Leishmania major/inmunología , Macrófagos/citología , Macrófagos/inmunología , Ratones , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Monocitos/citología , Monocitos/inmunología , Neutrófilos/citología , Neutrófilos/inmunología , Transducción de Señal/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Receptor Toll-Like 9/metabolismo , Tuberculosis/inmunología
18.
Sci Immunol ; 8(88): eabq3109, 2023 10 27.
Artículo en Inglés | MEDLINE | ID: mdl-37889983

RESUMEN

Mutations in the gene encoding the zinc-finger transcription factor Ikaros (IKZF1) are found in patients with immunodeficiency, leukemia, and autoimmunity. Although Ikaros has a well-established function in modulating gene expression programs important for hematopoietic development, its role in other cell types is less well defined. Here, we uncover functions for Ikaros in thymic epithelial lineage development in mice and show that Ikzf1 expression in medullary thymic epithelial cells (mTECs) is required for both autoimmune regulator-positive (Aire+) mTEC development and tissue-specific antigen (TSA) gene expression. Accordingly, TEC-specific deletion of Ikzf1 in mice results in a profound decrease in Aire+ mTECs, a global loss of TSA gene expression, and the development of autoimmunity. Moreover, Ikaros shapes thymic mimetic cell diversity, and its deletion results in a marked expansion of thymic tuft cells and muscle-like mTECs and a loss of other Aire-dependent mimetic populations. Single-cell analysis reveals that Ikaros modulates core transcriptional programs in TECs that correlate with the observed cellular changes. Our findings highlight a previously undescribed role for Ikaros in regulating epithelial lineage development and function and suggest that failed thymic central tolerance could contribute to the autoimmunity seen in humans with IKZF1 mutations.


Asunto(s)
Tolerancia Central , Timo , Humanos , Ratones , Animales , Diferenciación Celular , Factores de Transcripción , Regulación de la Expresión Génica
19.
Blood ; 116(25): 5443-54, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20829372

RESUMEN

The Notch pathway is frequently activated in T-cell acute lymphoblastic leukemias (T-ALLs). Of the Notch receptors, Notch1 is a recurrent target of gain-of-function mutations and Notch3 is expressed in all T-ALLs, but it is currently unclear how these receptors contribute to T-cell transformation in vivo. We investigated the role of Notch1 and Notch3 in T-ALL progression by a genetic approach, in mice bearing a knockdown mutation in the Ikaros gene that spontaneously develop Notch-dependent T-ALL. While deletion of Notch3 has little effect, T cell-specific deletion of floxed Notch1 promoter/exon 1 sequences significantly accelerates leukemogenesis. Notch1-deleted tumors lack surface Notch1 but express γ-secretase-cleaved intracellular Notch1 proteins. In addition, these tumors accumulate high levels of truncated Notch1 transcripts that are caused by aberrant transcription from cryptic initiation sites in the 3' part of the gene. Deletion of the floxed sequences directly reprograms the Notch1 locus to begin transcription from these 3' promoters and is accompanied by an epigenetic reorganization of the Notch1 locus that is consistent with transcriptional activation. Further, spontaneous deletion of 5' Notch1 sequences occurs in approximately 75% of Ikaros-deficient T-ALLs. These results reveal a novel mechanism for the oncogenic activation of the Notch1 gene after deletion of its main promoter.


Asunto(s)
Factor de Transcripción Ikaros/fisiología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Regiones Promotoras Genéticas/genética , Receptor Notch1/genética , Activación Transcripcional/fisiología , Animales , Northern Blotting , Western Blotting , Transformación Celular Neoplásica , Cartilla de ADN/química , Cartilla de ADN/genética , Citometría de Flujo , Regulación Neoplásica de la Expresión Génica , Proteína de Unión a la Señal Recombinante J de las Inmunoglobulinas/fisiología , Ratones , Ratones Noqueados , Mutación/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , ARN Mensajero/genética , Receptor Notch3 , Receptores Notch/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia , Tasa de Supervivencia
20.
Blood ; 116(25): 5455-64, 2010 Dec 16.
Artículo en Inglés | MEDLINE | ID: mdl-20852131

RESUMEN

Point mutations that trigger ligand-independent proteolysis of the Notch1 ectodomain occur frequently in human T-cell acute lymphoblastic leukemia (T-ALL) but are rare in murine T-ALL, suggesting that other mechanisms account for Notch1 activation in murine tumors. Here we show that most murine T-ALLs harbor Notch1 deletions that fall into 2 types, both leading to ligand-independent Notch1 activation. Type 1 deletions remove exon 1 and the proximal promoter, appear to be RAG-mediated, and are associated with mRNA transcripts that initiate from 3' regions of Notch1. In line with the RAG dependency of these rearrangements, RAG2 binds to the 5' end of Notch1 in normal thymocytes near the deletion breakpoints. Type 2 deletions remove sequences between exon 1 and exons 26 to 28 of Notch1, appear to be RAG-independent, and are associated with transcripts in which exon 1 is spliced out of frame to 3' Notch1 exons. Translation of both types of transcripts initiates at a conserved methionine residue, M1727, which lies within the Notch1 transmembrane domain. Polypeptides initiating at M1727 insert into membranes and are subject to constitutive cleavage by γ-secretase. Thus, like human T-ALL, murine T-ALL is often associated with acquired mutations that cause ligand-independent Notch1 activation.


Asunto(s)
Proteínas de Homeodominio/fisiología , Iniciación de la Cadena Peptídica Traduccional/genética , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Regiones Promotoras Genéticas/genética , Receptor Notch1/genética , Activación Transcripcional/fisiología , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Northern Blotting , Southern Blotting , Western Blotting , Neoplasias Óseas/genética , Neoplasias Óseas/metabolismo , Neoplasias Óseas/patología , Regulación Neoplásica de la Expresión Génica , Humanos , Factor de Transcripción Ikaros/fisiología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Mutación/genética , Osteosarcoma/genética , Osteosarcoma/metabolismo , Osteosarcoma/patología , Leucemia-Linfoma Linfoblástico de Células T Precursoras/metabolismo , Leucemia-Linfoma Linfoblástico de Células T Precursoras/patología , ARN Mensajero/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Eliminación de Secuencia , Homología de Secuencia de Ácido Nucleico , Células Tumorales Cultivadas
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA