Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Immunol ; 25(1): 129-141, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37985858

RESUMEN

Lymphocyte development consists of sequential and mutually exclusive cell states of proliferative selection and antigen receptor gene recombination. Transitions between each state require large, coordinated changes in epigenetic landscapes and transcriptional programs. How this occurs remains unclear. Here we demonstrate that in small pre-B cells, the lineage and stage-specific epigenetic reader bromodomain and WD repeat-containing protein 1 (BRWD1) reorders three-dimensional chromatin topology to affect the transition between proliferative and gene recombination molecular programs. BRWD1 regulated the switch between poised and active enhancers interacting with promoters, and coordinated this switch with Igk locus contraction. BRWD1 did so by converting chromatin-bound static to dynamic cohesin competent to mediate long-range looping. ATP-depletion revealed cohesin conversion to be the main energetic mechanism dictating dynamic chromatin looping. Our findings provide a new mechanism of cohesin regulation and reveal how cohesin function can be dictated by lineage contextual mechanisms to facilitate specific cell fate transitions.


Asunto(s)
Cromatina , Cohesinas , Cromatina/genética , Células Precursoras de Linfocitos B , Regulación de la Expresión Génica , Diferenciación Celular , Proteínas de Ciclo Celular/genética , Proteínas de Ciclo Celular/metabolismo
2.
Sci Immunol ; 7(74): eabm1664, 2022 08 05.
Artículo en Inglés | MEDLINE | ID: mdl-35930652

RESUMEN

During B lymphopoiesis, B cell progenitors progress through alternating and mutually exclusive stages of clonal expansion and immunoglobulin (Ig) gene rearrangements. Great diversity is generated through the stochastic recombination of Ig gene segments encoding heavy and light chain variable domains. However, this commonly generates autoreactivity. Receptor editing is the predominant tolerance mechanism for self-reactive B cells in the bone marrow (BM). B cell receptor editing rescues autoreactive B cells from negative selection through renewed light chain recombination first at Igκ then Igλ loci. Receptor editing depends on BM microenvironment cues and key transcription factors such as NF-κB, FOXO, and E2A. The specific BM factor required for receptor editing is unknown. Furthermore, how transcription factors coordinate these developmental programs to promote usage of the λ chain remains poorly defined. Therefore, we used two mouse models that recapitulate pathways by which Igλ light chain-positive B cells develop. The first has deleted J kappa (Jκ) genes and hence models Igλ expression resulting from failed Igκ recombination (Igκdel). The second models autoreactivity by ubiquitous expression of a single-chain chimeric anti-Igκ antibody (κ-mac). Here, we demonstrated that autoreactive B cells transit asymmetric forward and reverse developmental trajectories. This imparted a unique epigenetic landscape on small pre-B cells, which opened chromatin to transcription factors essential for Igλ recombination. The consequences of this asymmetric developmental path were both amplified and complemented by CXCR4 signaling. These findings reveal how intrinsic molecular programs integrate with extrinsic signals to drive receptor editing.


Asunto(s)
Linfocitos B , Receptores de Antígenos de Linfocitos B , Animales , Cromatina/metabolismo , Ratones , Receptores de Antígenos de Linfocitos B/genética , Recombinación Genética , Factores de Transcripción/genética
3.
Nat Immunol ; 21(6): 660-670, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32341509

RESUMEN

Within germinal centers (GCs), complex and highly orchestrated molecular programs must balance proliferation, somatic hypermutation and selection to both provide effective humoral immunity and to protect against genomic instability and neoplastic transformation. In contrast to this complexity, GC B cells are canonically divided into two principal populations, dark zone (DZ) and light zone (LZ) cells. We now demonstrate that, following selection in the LZ, B cells migrated to specialized sites within the canonical DZ that contained tingible body macrophages and were sites of ongoing cell division. Proliferating DZ (DZp) cells then transited into the larger DZ to become differentiating DZ (DZd) cells before re-entering the LZ. Multidimensional analysis revealed distinct molecular programs in each population commensurate with observed compartmentalization of noncompatible functions. These data provide a new three-cell population model that both orders critical GC functions and reveals essential molecular programs of humoral adaptive immunity.


Asunto(s)
Microambiente Celular/genética , Microambiente Celular/inmunología , Centro Germinal/citología , Centro Germinal/fisiología , Animales , Biomarcadores , Biología Computacional/métodos , Técnica del Anticuerpo Fluorescente , Perfilación de la Expresión Génica , Genómica/métodos , Ratones , Fosforilación , Proteoma , Proteómica/métodos , Transcriptoma
5.
Nat Immunol ; 20(10): 1393-1403, 2019 10.
Artículo en Inglés | MEDLINE | ID: mdl-31477919

RESUMEN

In B lymphopoiesis, activation of the pre-B cell antigen receptor (pre-BCR) is associated with both cell cycle exit and Igk recombination. Yet how the pre-BCR mediates these functions remains unclear. Here, we demonstrate that the pre-BCR initiates a feed-forward amplification loop mediated by the transcription factor interferon regulatory factor 4 and the chemokine receptor C-X-C motif chemokine receptor 4 (CXCR4). CXCR4 ligation by C-X-C motif chemokine ligand 12 activates the mitogen-activated protein kinase extracellular-signal-regulated kinase, which then directs the development of small pre- and immature B cells, including orchestrating cell cycle exit, pre-BCR repression, Igk recombination and BCR expression. In contrast, pre-BCR expression and escape from interleukin-7 have only modest effects on B cell developmental transcriptional and epigenetic programs. These data show a direct and central role for CXCR4 in orchestrating late B cell lymphopoiesis. Furthermore, in the context of previous findings, our data provide a three-receptor system sufficient to recapitulate the essential features of B lymphopoiesis in vitro.


Asunto(s)
Linfocitos B/inmunología , Cadenas kappa de Inmunoglobulina/genética , Células Precursoras de Linfocitos B/fisiología , Receptores de Antígenos de Linfocitos B/metabolismo , Receptores CXCR4/metabolismo , Animales , Puntos de Control del Ciclo Celular , Células Cultivadas , Quimiocina CXCL12/metabolismo , Femenino , Factores Reguladores del Interferón/genética , Linfopoyesis , Masculino , Ratones , Receptores de Antígenos de Linfocitos B/genética , Receptores CXCR4/genética , Recombinación Genética
6.
J Immunol ; 202(10): 2837-2842, 2019 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-30962294

RESUMEN

Lymphoid specification is the process by which hematopoietic stem cells (HSCs) and their progeny become restricted to differentiation through the lymphoid lineages. The basic helix-loop-helix transcription factors E2A and Lyl1 form a complex that promotes lymphoid specification. In this study, we demonstrate that Tal1, a Lyl1-related basic helix-loop-helix transcription factor that promotes T acute lymphoblastic leukemia and is required for HSC specification, erythropoiesis, and megakaryopoiesis, is a negative regulator of murine lymphoid specification. We demonstrate that Tal1 limits the expression of multiple E2A target genes in HSCs and controls the balance of myeloid versus T lymphocyte differentiation potential in lymphomyeloid-primed progenitors. Our data provide insight into the mechanisms controlling lymphocyte specification and may reveal a basis for the unique functions of Tal1 and Lyl1 in T acute lymphoblastic leukemia.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Diferenciación Celular/inmunología , Células Progenitoras Linfoides/inmunología , Células Progenitoras Mieloides/inmunología , Proteínas de Neoplasias/inmunología , Proteína 1 de la Leucemia Linfocítica T Aguda/inmunología , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Diferenciación Celular/genética , Ratones , Ratones Noqueados , Proteínas de Neoplasias/genética , Proteína 1 de la Leucemia Linfocítica T Aguda/genética
7.
Nat Immunol ; 19(12): 1366-1378, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30420627

RESUMEN

Thymocyte development requires a complex orchestration of multiple transcription factors. Ablating either TCF-1 or HEB in CD4+CD8+ thymocytes elicits similar developmental outcomes including increased proliferation, decreased survival, and fewer late Tcra rearrangements. Here, we provide a mechanistic explanation for these similarities by showing that TCF-1 and HEB share ~7,000 DNA-binding sites genome wide and promote chromatin accessibility. The binding of both TCF-1 and HEB was required at these shared sites for epigenetic and transcriptional gene regulation. Binding of TCF-1 and HEB to their conserved motifs in the enhancer regions of genes associated with T cell differentiation promoted their expression. Binding to sites lacking conserved motifs in the promoter regions of cell-cycle-associated genes limited proliferation. TCF-1 displaced nucleosomes, allowing for chromatin accessibility. Importantly, TCF-1 inhibited Notch signaling and consequently protected HEB from Notch-mediated proteasomal degradation. Thus, TCF-1 shifts nucleosomes and safeguards HEB, thereby enabling their cooperation in establishing the epigenetic and transcription profiles of CD4+CD8+ thymocytes.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Regulación de la Expresión Génica/inmunología , Factor Nuclear 1-alfa del Hepatocito/inmunología , Linfopoyesis/inmunología , Timocitos/inmunología , Animales , Antígenos CD4/inmunología , Antígenos CD8/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
8.
Nat Commun ; 9(1): 3888, 2018 09 24.
Artículo en Inglés | MEDLINE | ID: mdl-30250168

RESUMEN

Transcription factor (TF) networks determine cell fate in hematopoiesis. However, how TFs cooperate with other regulatory mechanisms to instruct transcription remains poorly understood. Here we show that in small pre-B cells, the lineage restricted epigenetic reader BRWD1 closes early development enhancers and opens the enhancers of late B lymphopoiesis to TF binding. BRWD1 regulates over 7000 genes to repress proliferative and induce differentiation programs. However, BRWD1 does not regulate the expression of TFs required for B lymphopoiesis. Hypogammaglobulinemia patients with BRWD1 mutations have B-cell transcriptional profiles and enhancer landscapes similar to those observed in Brwd1-/- mice. These data indicate that, in both mice and humans, BRWD1 is a master orchestrator of enhancer accessibility that cooperates with TF networks to drive late B-cell development.


Asunto(s)
Agammaglobulinemia/genética , Proteínas Portadoras/metabolismo , Epigénesis Genética/fisiología , Linfopoyesis/genética , Proteínas Nucleares/metabolismo , Adolescente , Adulto , Agammaglobulinemia/sangre , Animales , Proteínas Portadoras/genética , Diferenciación Celular/genética , Niño , Elementos de Facilitación Genéticos/genética , Perfilación de la Expresión Génica , Redes Reguladoras de Genes/fisiología , Humanos , Leucocitos Mononucleares , Masculino , Ratones , Ratones Noqueados , Proteínas Nucleares/genética , Células Precursoras de Linfocitos B , Cultivo Primario de Células , Proteínas Proto-Oncogénicas c-myc/genética , Proteínas Proto-Oncogénicas c-myc/metabolismo , ARN Interferente Pequeño/metabolismo , Análisis de Secuencia de ARN , Secuenciación del Exoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...