Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 31
Filtrar
4.
Immunity ; 55(8): 1431-1447.e11, 2022 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-35830859

RESUMEN

Conventional dendritic cells (cDCs) consist of two major functionally and phenotypically distinct subsets, cDC1 and cDC2, whose development is dependent on distinct sets of transcription factors. Interferon regulatory factor 8 (IRF8) is required at multiple stages of cDC1 development, but its role in committed cDC1 remains unclear. Here, we used Xcr1-cre to delete Irf8 in committed cDC1 and demonstrate that Irf8 is required for maintaining the identity of cDC1. In the absence of Irf8, committed cDC1 acquired the transcriptional, functional, and chromatin accessibility properties of cDC2. This conversion was independent of Irf4 and was associated with the decreased accessibility of putative IRF8, Batf3, and composite AP-1-IRF (AICE)-binding elements, together with increased accessibility of cDC2-associated transcription-factor-binding elements. Thus, IRF8 expression by committed cDC1 is required for preventing their conversion into cDC2-like cells.


Asunto(s)
Células Dendríticas , Factores Reguladores del Interferón , Células Dendríticas/metabolismo , Epigénesis Genética , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo
5.
Blood Adv ; 6(24): 6228-6241, 2022 12 27.
Artículo en Inglés | MEDLINE | ID: mdl-35584393

RESUMEN

The fetal-to-adult switch in hematopoietic stem cell (HSC) behavior is characterized by alterations in lineage output and entry into deep quiescence. Here we identify the emergence of megakaryocyte (Mk)-biased HSCs as an event coinciding with this developmental switch. Single-cell chromatin accessibility analysis reveals a ubiquitous acquisition of Mk lineage priming signatures in HSCs during the fetal-to-adult transition. These molecular changes functionally coincide with increased amplitude of early Mk differentiation events after acute inflammatory insult. Importantly, we identify LIN28B, known for its role in promoting fetal-like self-renewal, as an insulator against the establishment of an Mk-biased HSC pool. LIN28B protein is developmentally silenced in the third week of life, and its prolonged expression delays emergency platelet output in young adult mice. We propose that developmental regulation of Mk priming may represent a switch for HSCs to toggle between prioritizing self-renewal in the fetus and increased host protection in postnatal life.


Asunto(s)
Señales (Psicología) , Megacariocitos , Animales , Ratones , Megacariocitos/metabolismo , Células Madre Hematopoyéticas/metabolismo , Plaquetas/metabolismo , Hematopoyesis
6.
Cell Rep ; 39(6): 110798, 2022 05 10.
Artículo en Inglés | MEDLINE | ID: mdl-35545037

RESUMEN

The emerging notion of hematopoietic stem and progenitor cells (HSPCs) as a low-primed cloud without sharply demarcated gene expression programs raises the question on how cellular-fate options emerge and at which stem-like stage lineage priming is initiated. Here, we investigate single-cell chromatin accessibility of Lineage-, cKit+, and Sca1+ (LSK) HSPCs spanning the early differentiation landscape. Application of a signal-processing algorithm to detect transition points corresponding to massive alterations in accessibility of 571 transcription factor motifs reveals a population of LSK FMS-like tyrosine kinase 3 (Flt3)intCD9high cells that concurrently display stem-like and lineage-affiliated chromatin signatures, pointing to a simultaneous gain of both lympho-myeloid and megakaryocyte-erythroid programs. Molecularly and functionally, these cells position between stem cells and committed progenitors and display multi-lineage capacity in vitro and in vivo but lack self-renewal activity. This integrative molecular analysis resolves the heterogeneity of cells along hematopoietic differentiation and permits investigation of chromatin-mediated transition between multipotency and lineage restriction.


Asunto(s)
Cromatina , Células Madre Hematopoyéticas , Diferenciación Celular , Linaje de la Célula , Cromatina/metabolismo , Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Megacariocitos
7.
J Immunol ; 206(11): 2700-2713, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34021049

RESUMEN

B lymphocyte development is dependent on the interplay between the chromatin landscape and lineage-specific transcription factors. It has been suggested that B lineage commitment is associated with major changes in the nuclear chromatin environment, proposing a critical role for lineage-specific transcription factors in the formation of the epigenetic landscape. In this report, we have used chromosome conformation capture in combination with assay for transposase-accessible chromatin sequencing analysis to enable highly efficient annotation of both proximal and distal transcriptional control elements to genes activated in B lineage specification in mice. A large majority of these genes were annotated to at least one regulatory element with an accessible chromatin configuration in multipotent progenitors. Furthermore, the majority of binding sites for the key regulators of B lineage specification, EBF1 and PAX5, occurred in already accessible regions. EBF1 did, however, cause a dynamic change in assay for transposase-accessible chromatin accessibility and was critical for an increase in distal promoter-enhancer interactions. Our data unravel an extensive epigenetic priming at regulatory elements annotated to lineage-restricted genes and provide insight into the interplay between the epigenetic landscape and transcription factors in cell specification.


Asunto(s)
Linfocitos B/inmunología , Epigénesis Genética/inmunología , Factor de Transcripción PAX5/inmunología , Transactivadores/inmunología , Animales , Epigénesis Genética/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor de Transcripción PAX5/deficiencia , Factor de Transcripción PAX5/genética , Transactivadores/deficiencia , Transactivadores/genética
8.
Blood ; 137(22): 3037-3049, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33619557

RESUMEN

Genes encoding B lineage-restricted transcription factors are frequently mutated in B-lymphoid leukemias, suggesting a close link between normal and malignant B-cell development. One of these transcription factors is early B-cell factor 1 (EBF1), a protein of critical importance for lineage specification and survival of B-lymphoid progenitors. Here, we report that impaired EBF1 function in mouse B-cell progenitors results in reduced expression of Myc. Ectopic expression of MYC partially rescued B-cell expansion in the absence of EBF1 both in vivo and in vitro. Using chromosome conformation analysis in combination with ATAC-sequencing, chromatin immunoprecipitation-sequencing, and reporter gene assays, six EBF1-responsive enhancer elements were identified within the Myc locus. CRISPR-Cas9-mediated targeting of EBF1-binding sites identified one element of key importance for Myc expression and pro-B cell expansion. These data provide evidence that Myc is a direct target of EBF1. Furthermore, chromatin immunoprecipitation-sequencing analysis revealed that several regulatory elements in the Myc locus are targets of PAX5. However, ectopic expression of PAX5 in EBF1-deficient cells inhibits the cell cycle and reduces Myc expression, suggesting that EBF1 and PAX5 act in an opposing manner to regulate Myc levels. This hypothesis is further substantiated by the finding that Pax5 inactivation reduces requirements for EBF1 in pro-B-cell expansion. The binding of EBF1 and PAX5 to regulatory elements in the human MYC gene in a B-cell acute lymphoblastic leukemia cell line indicates that the EBF1:PAX5:MYC regulatory loop is conserved and may control both normal and malignant B-cell development.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Factor de Transcripción PAX5/metabolismo , Leucemia-Linfoma Linfoblástico de Células Precursoras B/metabolismo , Células Precursoras de Linfocitos B/metabolismo , Proteínas Proto-Oncogénicas c-myc/biosíntesis , Transactivadores/metabolismo , Animales , Proliferación Celular , Ratones , Ratones Noqueados , Factor de Transcripción PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/patología , Células Precursoras de Linfocitos B/patología , Proteínas Proto-Oncogénicas c-myc/genética , Elementos de Respuesta , Transactivadores/genética
9.
J Immunol ; 205(5): 1419-1432, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32747500

RESUMEN

Maturation of lymphoid cells is controlled by the action of stage and lineage-restricted transcription factors working in concert with the general transcription and chromatin remodeling machinery to regulate gene expression. To better understand this functional interplay, we used Biotin Identification in human embryonic kidney cells to identify proximity interaction partners for GATA3, TCF7 (TCF1), SPI1, HLF, IKZF1, PAX5, ID1, and ID2. The proximity interaction partners shared among the lineage-restricted transcription factors included ARID1a, a BRG1-associated factor complex component. CUT&RUN analysis revealed that ARID1a shared binding with TCF7 and GATA3 at a substantial number of putative regulatory elements in mouse T cell progenitors. In support of an important function for ARID1a in lymphocyte development, deletion of Arid1a in early lymphoid progenitors in mice resulted in a pronounced developmental arrest in early T cell development with a reduction of CD4+CD8+ cells and a 20-fold reduction in thymic cellularity. Exploring gene expression patterns in DN3 cells from Wt and Arid1a-deficient mice suggested that the developmental block resided in the DN3a to DN3b transition, indicating a deficiency in ß-selection. Our work highlights the critical importance of functional interactions between stage and lineage-restricted factors and the basic transcription machinery during lymphocyte differentiation.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Linfocitos/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Animales , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Línea Celular , Linaje de la Célula/genética , Linaje de la Célula/inmunología , Cromatina/genética , Cromatina/inmunología , Factor de Transcripción GATA3/genética , Factor de Transcripción GATA3/inmunología , Expresión Génica/genética , Expresión Génica/inmunología , Células HEK293 , Humanos , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Ratones , Ratones Endogámicos C57BL , Transcripción Genética/genética , Transcripción Genética/inmunología
10.
Blood ; 136(8): 946-956, 2020 08 20.
Artículo en Inglés | MEDLINE | ID: mdl-32384149

RESUMEN

Mutations in the FMS-like tyrosine kinase 3 (FLT3) gene in 13q12.2 are among the most common driver events in acute leukemia, leading to increased cell proliferation and survival through activation of the phosphatidylinositol 3-kinase/AKT-, RAS/MAPK-, and STAT5-signaling pathways. In this study, we examine the pathogenetic impact of somatic hemizygous 13q12.2 microdeletions in B-cell precursor (BCP) acute lymphoblastic leukemia (ALL) using 5 different patient cohorts (in total including 1418 cases). The 13q12.2 deletions occur immediately 5' of FLT3 and involve the PAN3 locus. By detailed analysis of the 13q12.2 segment, we show that the deletions lead to loss of a topologically associating domain border and an enhancer of FLT3. This results in increased cis interactions between the FLT3 promoter and another enhancer located distally to the deletion breakpoints, with subsequent allele-specific upregulation of FLT3 expression, expected to lead to ligand-independent activation of the receptor and downstream signaling. The 13q12.2 deletions are highly enriched in the high-hyperdiploid BCP ALL subtype (frequency 3.9% vs 0.5% in other BCP ALL) and in cases that subsequently relapsed. Taken together, our study describes a novel mechanism of FLT3 involvement in leukemogenesis by upregulation via chromatin remodeling and enhancer hijacking. These data further emphasize the role of FLT3 as a driver gene in BCP ALL.


Asunto(s)
Trastornos de los Cromosomas/genética , Elementos de Facilitación Genéticos/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Tirosina Quinasa 3 Similar a fms/genética , Línea Celular , Ensamble y Desensamble de Cromatina/genética , Ensamble y Desensamble de Cromatina/fisiología , Deleción Cromosómica , Trastornos de los Cromosomas/complicaciones , Cromosomas Humanos Par 13/genética , Estudios de Cohortes , Variaciones en el Número de Copia de ADN/genética , Regulación Leucémica de la Expresión Génica , Humanos , Análisis por Micromatrices , Polimorfismo de Nucleótido Simple , RNA-Seq , Regulación hacia Arriba/genética , Secuenciación Completa del Genoma
11.
Sci Immunol ; 4(39)2019 09 27.
Artículo en Inglés | MEDLINE | ID: mdl-31562190

RESUMEN

The ability of B-1 cells to become positively selected into the mature B cell pool, despite being weakly self-reactive, has puzzled the field since its initial discovery. Here, we explore changes in B cell positive selection as a function of developmental time by exploiting a link between CD5 surface levels and the natural occurrence of self-reactive B cell receptors (BCRs) in BCR wild-type mice. We show that the heterochronic RNA binding protein Lin28b potentiates a neonatal mode of B cell selection characterized by enhanced overall positive selection in general and the developmental progression of CD5+ immature B cells in particular. Lin28b achieves this by amplifying the CD19/PI3K/c-Myc positive feedback loop, and ectopic Lin28b expression restores both positive selection and mature B cell numbers in CD19-/- adult mice. Thus, the temporally restricted expression of Lin28b relaxes the rules for B cell selection during ontogeny by modulating tonic signaling. We propose that this neonatal mode of B cell selection represents a cell-intrinsic cue to accelerate the de novo establishment of the adaptive immune system and incorporate a layer of natural antibody-mediated immunity throughout life.


Asunto(s)
Linfocitos B/inmunología , Proteínas de Unión al ARN/inmunología , Animales , Ratones , Ratones Noqueados
12.
PLoS Genet ; 15(8): e1008280, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31381561

RESUMEN

One of the most frequently mutated proteins in human B-lineage leukemia is the transcription factor PAX5. These mutations often result in partial rather than complete loss of function of the transcription factor. While the functional dose of PAX5 has a clear connection to human malignancy, there is limited evidence for that heterozygote loss of PAX5 have a dramatic effect on the development and function of B-cell progenitors. One possible explanation comes from the finding that PAX5 mutated B-ALL often display complex karyotypes and additional mutations. Thus, PAX5 might be one component of a larger transcription factor network targeted in B-ALL. To investigate the functional network associated with PAX5 we used BioID technology to isolate proteins associated with this transcription factor in the living cell. This identified 239 proteins out of which several could be found mutated in human B-ALL. Most prominently we identified the commonly mutated IKZF1 and RUNX1, involved in the formation of ETV6-AML1 fusion protein, among the interaction partners. ChIP- as well as PLAC-seq analysis supported the idea that these factors share a multitude of target genes in human B-ALL cells. Gene expression analysis of mouse models and primary human leukemia suggested that reduced function of PAX5 increased the ability of an oncogenic form of IKZF1 or ETV6-AML to modulate gene expression. Our data reveals that PAX5 belong to a regulatory network frequently targeted by multiple mutations in B-ALL shedding light on the molecular interplay in leukemia cells.


Asunto(s)
Regulación Leucémica de la Expresión Génica , Redes Reguladoras de Genes/genética , Factor de Transcripción PAX5/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Humanos , Factor de Transcripción Ikaros/genética , Ratones , Ratones Noqueados , Mutación , Proteínas de Fusión Oncogénica/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Células Precursoras de Linfocitos B , Cultivo Primario de Células , Células Tumorales Cultivadas
13.
Front Immunol ; 10: 228, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30842770

RESUMEN

PU.1 is an ETS-family transcription factor that plays a broad range of roles in hematopoiesis. A direct regulator of myeloid, dendritic-cell, and B cell functional programs, and a well-known antagonist of terminal erythroid cell differentiation, it is also expressed in the earliest stages of T-cell development of each cohort of intrathymic pro-T cells. Its expression in this context appears to give T-cell precursors initial, transient access to myeloid and dendritic cell developmental competence and therefore to represent a source of antagonism or delay of T-cell lineage commitment. However, it has remained uncertain until recently why T-cell development is also intensely dependent upon PU.1. Here, we review recent work that sheds light on the molecular biology of PU.1 action across the genome in pro-T cells and identifies the genes that depend on PU.1 for their correct regulation. This work indicates modes of chromatin engagement, pioneering, and cofactor recruitment ("coregulator theft") by PU.1 as well as gene network interactions that not only affect specific target genes but also have system-wide regulatory consequences, amplifying the impact of PU.1 beyond its own direct binding targets. The genes directly regulated by PU.1 also suggest a far-reaching transformation of cell biology and signaling potential between the early stages of T-cell development when PU.1 is expressed and when it is silenced. These cell-biological functions can be important to distinguish fetal from adult T-cell development and have the potential to illuminate aspects of thymic function that have so far remained the most mysterious.


Asunto(s)
Proteínas Proto-Oncogénicas/metabolismo , Linfocitos T/fisiología , Transactivadores/metabolismo , Animales , Diferenciación Celular , Linaje de la Célula , Redes Reguladoras de Genes , Hematopoyesis/genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Proteínas Proto-Oncogénicas/genética , Transactivadores/genética , Transcripción Genética
15.
Nat Immunol ; 19(12): 1427-1440, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30374131

RESUMEN

Multipotent progenitor cells confirm their T cell-lineage identity in the CD4-CD8- double-negative (DN) pro-T cell DN2 stages, when expression of the essential transcription factor Bcl11b begins. In vivo and in vitro stage-specific deletions globally identified Bcl11b-controlled target genes in pro-T cells. Proteomics analysis revealed that Bcl11b associated with multiple cofactors and that its direct action was needed to recruit those cofactors to selective target sites. Regions near functionally regulated target genes showed enrichment for those sites of Bcl11b-dependent recruitment of cofactors, and deletion of individual cofactors relieved the repression of many genes normally repressed by Bcl11b. Runx1 collaborated with Bcl11b most frequently for both activation and repression. In parallel, Bcl11b indirectly regulated a subset of target genes by a gene network circuit via the transcription inhibitor Id2 (encoded by Id2) and transcription factor PLZF (encoded by Zbtb16); Id2 and Zbtb16 were directly repressed by Bcl11b, and Id2 and PLZF controlled distinct alternative programs. Thus, our study defines the molecular basis of direct and indirect Bcl11b actions that promote T cell identity and block alternative potentials.


Asunto(s)
Linfopoyesis/inmunología , Células Precursoras de Linfocitos T/inmunología , Proteína de la Leucemia Promielocítica con Dedos de Zinc/biosíntesis , Proteínas Represoras/inmunología , Proteínas Supresoras de Tumor/inmunología , Animales , Diferenciación Celular/inmunología , Regulación de la Expresión Génica/inmunología , Proteína 2 Inhibidora de la Diferenciación/biosíntesis , Proteína 2 Inhibidora de la Diferenciación/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Células Precursoras de Linfocitos T/citología , Proteína de la Leucemia Promielocítica con Dedos de Zinc/inmunología
16.
Genome Res ; 28(10): 1508-1519, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30171019

RESUMEN

SPI1 (also known as PU.1) is a dominant but transient regulator in early T-cell precursors and a potent transcriptional controller of developmentally important pro-T-cell genes. Before T-lineage commitment, open chromatin is frequently occupied by PU.1, and many PU.1 sites lose accessibility when PU.1 is later down-regulated. Pioneering activity of PU.1 was tested in this developmentally dynamic context by quantitating the relationships between PU.1 occupancy and site quality and accessibility as PU.1 levels naturally declined in pro-T-cell development and by using stage-specific gain- and loss-of-function perturbations to relate binding to effects on target genes. PU.1 could bind closed genomic sites, but rapidly opened many of them, despite the absence of its frequent collaborator, CEBPA. RUNX motifs and RUNX1 binding were often linked to PU.1 at open sites, but highly expressed PU.1 could bind its sites without RUNX1. The dynamic properties of PU.1 engagements implied that PU.1 binding affinity and concentration determine its occupancy choices, but with quantitative trade-offs for occupancy between site sequence quality and stage-dependent site accessibility in chromatin. At nonpromoter sites, PU.1 binding criteria were more stringent than at promoters, and PU.1 was also much more effective as a transcriptional regulator at nonpromoter sites where local chromatin accessibility depended on the presence of PU.1. Notably, closed chromatin presented a qualitative barrier to occupancy by the PU.1 DNA-binding domain alone. Thus, effective pioneering at closed chromatin sites also depends on requirements beyond site recognition, served by non-DNA-binding domains of PU.1.


Asunto(s)
Cromatina/química , Cromatina/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Animales , Sitios de Unión , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Ensamble y Desensamble de Cromatina , Inmunoprecipitación de Cromatina , Epigénesis Genética , Regulación del Desarrollo de la Expresión Génica , Ratones , Regiones Promotoras Genéticas , Análisis de Secuencia de ARN
17.
Immunity ; 48(6): 1119-1134.e7, 2018 06 19.
Artículo en Inglés | MEDLINE | ID: mdl-29924977

RESUMEN

Transcription factors normally regulate gene expression through their action at sites where they bind to DNA. However, the balance of activating and repressive functions that a transcription factor can mediate is not completely understood. Here, we showed that the transcription factor PU.1 regulated gene expression in early T cell development both by recruiting partner transcription factors to its own binding sites and by depleting them from the binding sites that they preferred when PU.1 was absent. The removal of partner factors Satb1 and Runx1 occurred primarily from sites where PU.1 itself did not bind. Genes linked to sites of partner factor "theft" were enriched for genes that PU.1 represses despite lack of binding, both in a model cell line system and in normal T cell development. Thus, system-level competitive recruitment dynamics permit PU.1 to affect gene expression both through its own target sites and through action at a distance.


Asunto(s)
Diferenciación Celular/inmunología , Regulación de la Expresión Génica/inmunología , Linfopoyesis/fisiología , Proteínas Proto-Oncogénicas/inmunología , Linfocitos T/inmunología , Transactivadores/inmunología , Animales , Subunidad alfa 2 del Factor de Unión al Sitio Principal/inmunología , Subunidad alfa 2 del Factor de Unión al Sitio Principal/metabolismo , Linfopoyesis/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/inmunología , Proteínas de Unión a la Región de Fijación a la Matriz/metabolismo , Ratones , Proteínas Proto-Oncogénicas/metabolismo , Transactivadores/metabolismo , Factores de Transcripción/inmunología , Factores de Transcripción/metabolismo
18.
J Exp Med ; 215(7): 1947-1963, 2018 07 02.
Artículo en Inglés | MEDLINE | ID: mdl-29899037

RESUMEN

To understand the developmental trajectories in early lymphocyte differentiation, we identified differentially expressed surface markers on lineage-negative lymphoid progenitors (LPs). Single-cell polymerase chain reaction experiments allowed us to link surface marker expression to that of lineage-associated transcription factors (TFs) and identify GFRA2 and BST1 as markers of early B cells. Functional analyses in vitro and in vivo as well as single-cell gene expression analyses supported that surface expression of these proteins defined distinct subpopulations that include cells from both the classical common LPs (CLPs) and Fraction A compartments. The formation of the GFRA2-expressing stages of development depended on the TF EBF1, critical both for the activation of stage-specific target genes and modulation of the epigenetic landscape. Our data show that consecutive expression of Ly6D, GFRA2, and BST1 defines a developmental trajectory linking the CLP to the CD19+ progenitor compartment.


Asunto(s)
Linfocitos B/citología , Linfocitos B/inmunología , Compartimento Celular , Linfopoyesis , Células Madre/citología , ADP-Ribosil Ciclasa/metabolismo , Animales , Antígenos CD/metabolismo , Antígenos Ly/metabolismo , Médula Ósea/metabolismo , Linaje de la Célula , Membrana Celular/metabolismo , Proteínas Ligadas a GPI/metabolismo , Receptores del Factor Neurotrófico Derivado de la Línea Celular Glial/metabolismo , Ratones , Modelos Biológicos
19.
Genes Dev ; 30(22): 2486-2499, 2016 11 15.
Artículo en Inglés | MEDLINE | ID: mdl-27913602

RESUMEN

Even though leukemia is considered to be confined to one specific hematopoietic cell type, cases of acute leukemia of ambiguous lineage and patients relapsing in phenotypically altered disease suggest that a malignant state may be transferred between lineages. Because B-cell leukemia is associated with mutations in transcription factors of importance for stable preservation of lineage identity, we here investigated the potential lineage plasticity of leukemic cells. We report that primary pro-B leukemia cells from mice carrying heterozygous mutations in either or both the Pax5 and Ebf1 genes, commonly mutated in human leukemia, can be converted into T lineage leukemia cells. Even though the conversion process involved global changes in gene expression and lineage-restricted epigenetic reconfiguration, the malignant phenotype of the cells was preserved, enabling them to expand as T lineage leukemia cells in vivo. Furthermore, while the transformed pro-B cells displayed plasticity toward myeloid lineages, the converted cells failed to cause myeloid leukemia after transplantation. These data provide evidence that a malignant phenotype can be transferred between hematopoietic lineages. This has important implications for modern cancer medicine because lineage targeted treatment of leukemia patients can be predicted to provoke the emergence of phenotypically altered subclones, causing clinical relapse.


Asunto(s)
Linfocitos B/patología , Transformación Celular Neoplásica/genética , Leucemia Linfoide/fisiopatología , Animales , Línea Celular , Línea Celular Tumoral , Linaje de la Célula , Proliferación Celular/genética , Regulación Neoplásica de la Expresión Génica , Humanos , Leucemia de Células T/fisiopatología , Ratones , Ratones Endogámicos C57BL , Mutación , Células Mieloides/patología , Células Precursoras de Linfocitos B/metabolismo , Unión Proteica , Receptores Notch/genética , Receptores Notch/metabolismo , Transducción de Señal
20.
Adv Immunol ; 129: 109-74, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26791859

RESUMEN

T-lymphocyte development branches off from other lymphoid developmental programs through its requirement for sustained environmental signals through the Notch pathway. In the thymus, Notch signaling induces a succession of T-lineage regulatory factors that collectively create the T-cell identity through distinct steps. This process involves both the staged activation of T-cell identity genes and the staged repression of progenitor-cell-inherited regulatory genes once their roles in self-renewal and population expansion are no longer needed. With the recent characterization of innate lymphoid cells (ILCs) that share transcriptional regulation programs extensively with T-cell subsets, T-cell identity can increasingly be seen as defined in modular terms, as the processes selecting and actuating effector function are potentially detachable from the processes generating and selecting clonally unique T-cell receptor structures. The developmental pathways of different classes of T cells and ILCs are distinguished by the numbers of prerequisites of gene rearrangement, selection, and antigen contact before the cells gain access to nearly common regulatory mechanisms for choosing effector function. Here, the major classes of transcription factors that interact with Notch signals during T-lineage specification are discussed in terms of their roles in these programs, the evidence for their spectra of target genes at different stages, and their cross-regulatory and cooperative actions with each other. Specific topics include Notch modulation of PU.1 and GATA-3, PU.1-Notch competition, the relationship between PU.1 and GATA-3, and the roles of E proteins, Bcl11b, and GATA-3 in guiding acquisition of T-cell identity while avoiding redirection to an ILC fate.


Asunto(s)
Regulación de la Expresión Génica/inmunología , Células Precursoras de Linfocitos T/fisiología , Receptores Notch/metabolismo , Subgrupos de Linfocitos T/fisiología , Factores de Transcripción/metabolismo , Transcripción Genética/inmunología , Animales , Diferenciación Celular , Microambiente Celular , Humanos , Receptores Notch/inmunología , Transducción de Señal , Factores de Transcripción/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...