Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 32
Filtrar
1.
Blood ; 139(24): 3505-3518, 2022 06 16.
Artículo en Inglés | MEDLINE | ID: mdl-35316324

RESUMEN

Oncogenic alterations underlying B-cell acute lymphoblastic leukemia (B-ALL) in adults remain incompletely elucidated. To uncover novel oncogenic drivers, we performed RNA sequencing and whole-genome analyses in a large cohort of unresolved B-ALL. We identified a novel subtype characterized by a distinct gene expression signature and the unique association of 2 genomic microdeletions. The 17q21.31 microdeletion resulted in a UBTF::ATXN7L3 fusion transcript encoding a chimeric protein. The 13q12.2 deletion resulted in monoallelic ectopic expression of the homeobox transcription factor CDX2, located 138 kb in cis from the deletion. Using 4C-sequencing and CRISPR interference experiments, we elucidated the mechanism of CDX2 cis-deregulation, involving PAN3 enhancer hijacking. CDX2/UBTF ALL (n = 26) harbored a distinct pattern of additional alterations including 1q gain and CXCR4 activating mutations. Within adult patients with Ph- B-ALL enrolled in GRAALL trials, patients with CDX2/UBTF ALL (n = 17/723, 2.4%) were young (median age, 31 years) and dramatically enriched in females (male/female ratio, 0.2, P = .002). They commonly presented with a pro-B phenotype ALL and moderate blast cell infiltration. They had poor response to treatment including a higher risk of failure to first induction course (19% vs 3%, P = .017) and higher post-induction minimal residual disease (MRD) levels (MRD ≥ 10-4, 93% vs 46%, P < .001). This early resistance to treatment translated into a significantly higher cumulative incidence of relapse (75.0% vs 32.4%, P = .004) in univariate and multivariate analyses. In conclusion, we discovered a novel B-ALL entity defined by the unique combination of CDX2 cis-deregulation and UBTF::ATXN7L3 fusion, representing a high-risk disease in young adults.


Asunto(s)
Factor de Transcripción CDX2 , Proteínas del Complejo de Iniciación de Transcripción Pol1 , Leucemia-Linfoma Linfoblástico de Células Precursoras B , Leucemia-Linfoma Linfoblástico de Células Precursoras , Factores de Transcripción , Adulto , Factor de Transcripción CDX2/genética , Femenino , Genes Homeobox , Humanos , Masculino , Neoplasia Residual/genética , Proteínas de Fusión Oncogénica , Proteínas del Complejo de Iniciación de Transcripción Pol1/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras B/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/metabolismo , Factores de Transcripción/genética
2.
Genome Res ; 29(7): 1087-1099, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31175153

RESUMEN

To initiate X-Chromosome inactivation (XCI), the long noncoding RNA Xist mediates chromosome-wide gene silencing of one X Chromosome in female mammals to equalize gene dosage between the sexes. The efficiency of gene silencing is highly variable across genes, with some genes even escaping XCI in somatic cells. A gene's susceptibility to Xist-mediated silencing appears to be determined by a complex interplay of epigenetic and genomic features; however, the underlying rules remain poorly understood. We have quantified chromosome-wide gene silencing kinetics at the level of the nascent transcriptome using allele-specific Precision nuclear Run-On sequencing (PRO-seq). We have developed a Random Forest machine-learning model that can predict the measured silencing dynamics based on a large set of epigenetic and genomic features and tested its predictive power experimentally. The genomic distance to the Xist locus, followed by gene density and distance to LINE elements, are the prime determinants of the speed of gene silencing. Moreover, we find two distinct gene classes associated with different silencing pathways: a class that requires Xist-repeat A for silencing, which is known to activate the SPEN pathway, and a second class in which genes are premarked by Polycomb complexes and tend to rely on the B repeat in Xist for silencing, known to recruit Polycomb complexes during XCI. Moreover, a series of features associated with active transcriptional elongation and chromatin 3D structure are enriched at rapidly silenced genes. Our machine-learning approach can thus uncover the complex combinatorial rules underlying gene silencing during X inactivation.


Asunto(s)
Epigénesis Genética , Silenciador del Gen , Aprendizaje Automático , ARN Largo no Codificante/fisiología , Inactivación del Cromosoma X/genética , Animales , Línea Celular , Células Madre Embrionarias , Femenino , Genes Ligados a X , Genoma , Cinética , Ratones , Modelos Genéticos
3.
Nat Immunol ; 10(6): 655-64, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19448632

RESUMEN

Coordinated recombination of homologous antigen receptor loci is thought to be important for allelic exclusion. Here we show that homologous immunoglobulin alleles pair in a stage-specific way that mirrors the recombination patterns of these loci. The frequency of homologous immunoglobulin pairing was much lower in the absence of the RAG-1-RAG-2 recombinase and was restored in Rag1-/- developing B cells with a transgene expressing a RAG-1 active-site mutant that supported DNA binding but not cleavage. The introduction of DNA breaks on one immunoglobulin allele induced ATM-dependent repositioning of the other allele to pericentromeric heterochromatin. ATM activated by the cleaved allele acts in trans on the uncleaved allele to prevent biallelic recombination and chromosome breaks or translocations.


Asunto(s)
Proteínas de Ciclo Celular/genética , Proteínas de Unión al ADN/genética , Proteínas de Homeodominio/genética , Inmunoglobulinas/genética , Proteínas Serina-Treonina Quinasas/genética , Recombinación Genética , Proteínas Supresoras de Tumor/genética , Alelos , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Linfocitos B/metabolismo , Células Cultivadas , Roturas del ADN , Reordenamiento Génico , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , VDJ Recombinasas/metabolismo
4.
Immunity ; 34(3): 303-14, 2011 Mar 25.
Artículo en Inglés | MEDLINE | ID: mdl-21435585

RESUMEN

T cell fate is associated with mutually exclusive expression of CD4 or CD8 in helper and cytotoxic T cells, respectively. How expression of one locus is temporally coordinated with repression of the other has been a long-standing enigma, though we know RUNX transcription factors activate the Cd8 locus, silence the Cd4 locus, and repress the Zbtb7b locus (encoding the transcription factor ThPOK), which is required for CD4 expression. Here we found that nuclear organization was altered by interplay among members of this transcription factor circuitry: RUNX binding mediated association of Cd4 and Cd8 whereas ThPOK binding kept the loci apart. Moreover, targeted deletions within Cd4 modulated CD8 expression and pericentromeric repositioning of Cd8. Communication between Cd4 and Cd8 thus appears to enable long-range epigenetic regulation to ensure that expression of one excludes the other in mature CD4 or CD8 single-positive (SP) cells.


Asunto(s)
Linfocitos B/inmunología , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Subunidades alfa del Factor de Unión al Sitio Principal/inmunología , Regulación de la Expresión Génica/inmunología , Animales , Epigenómica , Citometría de Flujo , Hibridación Fluorescente in Situ , Ratones , Ratones Endogámicos C57BL
5.
Nature ; 471(7336): 119-23, 2011 Mar 03.
Artículo en Inglés | MEDLINE | ID: mdl-21368836

RESUMEN

Misrepair of DNA double-strand breaks produced by the V(D)J recombinase (the RAG1/RAG2 proteins) at immunoglobulin (Ig) and T cell receptor (Tcr) loci has been implicated in pathogenesis of lymphoid malignancies in humans and in mice. Defects in DNA damage response factors such as ataxia telangiectasia mutated (ATM) protein and combined deficiencies in classical non-homologous end joining and p53 predispose to RAG-initiated genomic rearrangements and lymphomagenesis. Although we showed previously that RAG1/RAG2 shepherd the broken DNA ends to classical non-homologous end joining for proper repair, roles for the RAG proteins in preserving genomic stability remain poorly defined. Here we show that the RAG2 carboxy (C) terminus, although dispensable for recombination, is critical for maintaining genomic stability. Thymocytes from 'core' Rag2 homozygotes (Rag2(c/c) mice) show dramatic disruption of Tcrα/δ locus integrity. Furthermore, all Rag2(c/c) p53(-/-) mice, unlike Rag1(c/c) p53(-/-) and p53(-/-) animals, rapidly develop thymic lymphomas bearing complex chromosomal translocations, amplifications and deletions involving the Tcrα/δ and Igh loci. We also find these features in lymphomas from Atm(-/-) mice. We show that, like ATM-deficiency, core RAG2 severely destabilizes the RAG post-cleavage complex. These results reveal a novel genome guardian role for RAG2 and suggest that similar 'end release/end persistence' mechanisms underlie genomic instability and lymphomagenesis in Rag2(c/c) p53(-/-) and Atm(-/-) mice.


Asunto(s)
Proteínas de Unión al ADN/química , Proteínas de Unión al ADN/metabolismo , Progresión de la Enfermedad , Inestabilidad Genómica , Linfoma/genética , Linfoma/patología , Animales , Proteínas de la Ataxia Telangiectasia Mutada , Proteínas de Ciclo Celular/genética , Deleción Cromosómica , Cromosomas de los Mamíferos/genética , Proteínas de Unión al ADN/deficiencia , Proteínas de Unión al ADN/genética , Reordenamiento Génico de Linfocito T/genética , Genes de las Cadenas Pesadas de las Inmunoglobulinas/genética , Genes p53/genética , Hibridación Fluorescente in Situ , Estimación de Kaplan-Meier , Ratones , Proteínas Serina-Treonina Quinasas/deficiencia , Proteínas Serina-Treonina Quinasas/genética , Receptores de Antígenos de Linfocitos T/genética , Recombinación Genética/genética , Timo/citología , Translocación Genética/genética , Proteínas Supresoras de Tumor/química , Proteínas Supresoras de Tumor/deficiencia , Proteínas Supresoras de Tumor/genética , Proteínas Supresoras de Tumor/metabolismo
6.
Proc Natl Acad Sci U S A ; 111(1): 391-6, 2014 Jan 07.
Artículo en Inglés | MEDLINE | ID: mdl-24371308

RESUMEN

Deregulated activation of ß-catenin in cancer has been correlated with genomic instability. During thymocyte development, ß-catenin activates transcription in partnership with T-cell-specific transcription factor 1 (Tcf-1). We previously reported that targeted activation of ß-catenin in thymocytes (CAT mice) induces lymphomas that depend on recombination activating gene (RAG) and myelocytomatosis oncogene (Myc) activities. Here we show that these lymphomas have recurring Tcra/Myc translocations that resulted from illegitimate RAG recombination events and resembled oncogenic translocations previously described in human T-ALL. We therefore used the CAT animal model to obtain mechanistic insights into the transformation process. ChIP-seq analysis uncovered a link between Tcf-1 and RAG2 showing that the two proteins shared binding sites marked by trimethylated histone-3 lysine-4 (H3K4me3) throughout the genome, including near the translocation sites. Pretransformed CAT thymocytes had increased DNA damage at the translocating loci and showed altered repair of RAG-induced DNA double strand breaks. These cells were able to survive despite DNA damage because activated ß-catenin promoted an antiapoptosis gene expression profile. Thus, activated ß-catenin promotes genomic instability that leads to T-cell lymphomas as a consequence of altered double strand break repair and increased survival of thymocytes with damaged DNA.


Asunto(s)
Inestabilidad Genómica , Activación de Linfocitos , Linfoma/genética , Linfocitos T/citología , beta Catenina/metabolismo , Animales , Apoptosis , Secuencia de Bases , Supervivencia Celular , Roturas del ADN de Doble Cadena , Metilación de ADN , Reparación del ADN , Modelos Animales de Enfermedad , Genes RAG-1/genética , Factor Nuclear 1-alfa del Hepatocito , Histonas/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Datos de Secuencia Molecular , Recombinación Genética , Factor 1 de Transcripción de Linfocitos T/metabolismo , Timocitos/citología , Translocación Genética , beta Catenina/genética
7.
J Immunol ; 188(12): 6084-92, 2012 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-22581861

RESUMEN

Ag receptor diversity involves the introduction of DNA double-stranded breaks during lymphocyte development. To ensure fidelity, cleavage is confined to the G(0)-G(1) phase of the cell cycle. One established mechanism of regulation is through periodic degradation of the RAG2 recombinase protein. However, there are additional levels of protection. In this paper, we show that cyclical changes in the IL-7R signaling pathway functionally segregate pro-B cells according to cell cycle status. In consequence, the level of a downstream effector of IL-7 signaling, phospho-STAT5, is inversely correlated with cell cycle expression of Rag, a key gene involved in recombination. Higher levels of phopho-STAT5 in S-G(2) correlate with decreased Rag expression and Rag relocalization to pericentromeric heterochromatin. These cyclical changes in transcription and locus repositioning are ablated upon transformation with v-Abl, which renders STAT5 constitutively active across the cell cycle. We propose that this activity of the IL-7R/STAT5 pathway plays a critical protective role in development, complementing regulation of RAG2 at the protein level, to ensure that recombination does not occur during replication. Our data, suggesting that pro-B cells are not a single homogeneous population, explain inconsistencies in the role of IL-7 signaling in regulating Igh recombination.


Asunto(s)
Subgrupos de Linfocitos B/inmunología , Ciclo Celular/inmunología , Interleucina-7/inmunología , Células Precursoras de Linfocitos B/inmunología , Animales , Subgrupos de Linfocitos B/citología , Subgrupos de Linfocitos B/metabolismo , Ciclo Celular/genética , Diferenciación Celular/genética , Diferenciación Celular/inmunología , Citometría de Flujo , Reordenamiento Génico de Cadena Pesada de Linfocito B/genética , Reordenamiento Génico de Cadena Pesada de Linfocito B/inmunología , Genes RAG-1 , Proteínas de Homeodominio/biosíntesis , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Hibridación Fluorescente in Situ , Interleucina-7/metabolismo , Ratones , Microscopía Confocal , Análisis de Secuencia por Matrices de Oligonucleótidos , Células Precursoras de Linfocitos B/citología , Células Precursoras de Linfocitos B/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT5/inmunología , Factor de Transcripción STAT5/metabolismo , Transducción de Señal/inmunología , Transcripción Genética
8.
Immunol Rev ; 237(1): 43-54, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20727028

RESUMEN

Perhaps no process has provided more insight into the fine manipulation of locus accessibility than antigen receptor rearrangement. V(D)J recombination is carried out by the lymphoid-specific recombination-activating (RAG 1 and 2) proteins and the non-homologous end joining machinery; yet, it occurs only at specific loci (or portions of loci) during specific developmental stages. This spatiotemporal restriction of recombination is achieved through precise alterations in locus accessibility. In this article, we discuss the work of our laboratory in elucidating how nuclear sublocalization, chromosome conformation, and locus interactions contribute to regulating this complex process. We also discuss what is known about how key factors in B-cell development (such as the ubiquitously expressed helix loop helix protein E2A, the B-cell specific transcription factors EBF1 and Pax5, and the interleukin-7 cytokine signaling pathway) exert their effects through changes in nuclear dynamics.


Asunto(s)
Linfocitos B/inmunología , Cromosomas/genética , Reordenamiento Génico , Región de Unión de la Inmunoglobulina/genética , Región Variable de Inmunoglobulina/genética , Linfocitos T/inmunología , Animales , Cromosomas/metabolismo , Humanos , VDJ Recombinasas/fisiología
9.
Hemasphere ; 8(6): e90, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38903535

RESUMEN

Transcriptional cofactors of the ETO family are recurrent fusion partners in acute leukemia. We characterized the ETO2 regulome by integrating transcriptomic and chromatin binding analyses in human erythroleukemia xenografts and controlled ETO2 depletion models. We demonstrate that beyond its well-established repressive activity, ETO2 directly activates transcription of MYB, among other genes. The ETO2-activated signature is associated with a poorer prognosis in erythroleukemia but also in other acute myeloid and lymphoid leukemia subtypes. Mechanistically, ETO2 colocalizes with EP300 and MYB at enhancers supporting the existence of an ETO2/MYB feedforward transcription activation loop (e.g., on MYB itself). Both small-molecule and PROTAC-mediated inhibition of EP300 acetyltransferases strongly reduced ETO2 protein, chromatin binding, and ETO2-activated transcripts. Taken together, our data show that ETO2 positively enforces a leukemia maintenance program that is mediated in part by the MYB transcription factor and that relies on acetyltransferase cofactors to stabilize ETO2 scaffolding activity.

10.
Front Immunol ; 14: 1156077, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37215121

RESUMEN

Over the past decades, it has become increasingly clear that higher order chromatin folding and organization within the nucleus is involved in the regulation of genome activity and serves as an additional epigenetic mechanism that modulates cellular functions and gene expression programs in diverse biological processes. In particular, dynamic allelic interactions and nuclear locations can be of functional importance during the process of lymphoid differentiation and the regulation of immune responses. Analyses of the proximity between chromatin and/or nuclear regions can be performed on populations of cells with high-throughput sequencing approaches such as chromatin conformation capture ("3C"-based) or DNA adenine methyltransferase identification (DamID) methods, or, in individual cells, by the simultaneous visualization of genomic loci, their primary transcripts and nuclear compartments within the 3-dimensional nuclear space using Fluorescence In Situ Hybridization (FISH) and immunostaining. Here, we present a detailed protocol to simultaneously detect nascent RNA transcripts (3D RNA FISH), their genomic loci (3D DNA FISH) and/or their chromosome territories (CT paint DNA FISH) combined with the antibody-based detection of various nuclear factors (immunofluorescence). We delineate the application and effectiveness of this robust and reproducible protocol in several murine T lymphocyte subtypes (from differentiating thymic T cells, to activated splenic and peripheral T cells) as well as other murine cells, including embryonic stem cells, B cells, megakaryocytes and macrophages.


Asunto(s)
Cromatina , Linfocitos T , Animales , Ratones , Hibridación Fluorescente in Situ/métodos , Linfocitos T/metabolismo , Cromatina/genética , ADN/metabolismo , Genómica
11.
Biol Sex Differ ; 14(1): 60, 2023 09 18.
Artículo en Inglés | MEDLINE | ID: mdl-37723501

RESUMEN

BACKGROUND: Human endosomal Toll-like receptors TLR7 and TLR8 recognize self and non-self RNA ligands, and are important mediators of innate immunity and autoimmune pathogenesis. TLR7 and TLR8 are, respectively, encoded by adjacent X-linked genes. We previously established that TLR7 evades X chromosome inactivation (XCI) in female immune cells. Whether TLR8 also evades XCI, however, has not yet been explored. METHOD: In the current study, we used RNA fluorescence in situ hybridization (RNA FISH) to directly visualize, on a single-cell basis, primary transcripts of TLR7 and TLR8 relative to X chromosome territories in CD14+ monocytes and CD4+ T lymphocytes from women, Klinefelter syndrome (KS) men, and euploid men. To assign X chromosome territories in cells lacking robust expression of a XIST compartment, we designed probes specific for X-linked genes that do not escape XCI and therefore robustly label the active X chromosome. We also assessed whether XCI escape of TLR8 was associated with sexual dimorphism in TLR8 protein expression by western blot and flow cytometry. RESULTS: Using RNA FISH, we show that TLR8, like TLR7, evades XCI in immune cells, and that cells harboring simultaneously TLR7 and TLR8 transcript foci are more frequent in women and KS men than in euploid men, resulting in a sevenfold difference in frequency. This transcriptional bias was again observable when comparing the single X of XY males with the active X of cells from females or KS males. Interestingly, TLR8 protein expression was significantly higher in female mononuclear blood cells, including all monocyte subsets, than in male cells. CONCLUSIONS: TLR8, mirroring TLR7, escapes XCI in human monocytes and CD4+ T cells. Co-dependent transcription from the active X chromosome and escape from XCI could both contribute to higher TLR8 protein abundance in female cells, which may have implications for the response to viruses and bacteria, and the risk of developing inflammatory and autoimmune diseases.


Human endosomal Toll-like receptors TLR7 and TLR8, encoded by two adjacent X-linked genes, recognize self and non-self RNA ligands, and are important mediators of innate immunity and autoimmune pathogenesis. We previously reported that TLR7 evades X chromosome inactivation (XCI) in female immune cells, correlating with enhanced functional properties in B cells harboring biallelic expression of this gene. Here, we conducted a comprehensive single-cell resolution analysis of the transcriptional regulation of both TLR7 and TLR8, in CD14+ monocytes and CD4+ T lymphocytes. We unequivocally demonstrated that TLR8, like TLR7, escapes XCI in immune cells from female and Klinefelter syndrome males. When we analyzed TLR7 and TLR8 transcripts together, cells from women and KS men exhibited higher frequencies of cells co-transcribing the two genes. Surprisingly, these differences were attributable not only to the ability of TLR7 and TLR8 to be expressed on the Xi, but also to the joint transcriptional behavior of the TLR7­TLR8 gene pair on the active X chromosome specifically. This contrasted with a striking pattern of mutually exclusive transcription on the single X of euploid men. Corroborating our RNA FISH results, we found higher TLR8 protein expression in female than in male leukocytes, including all monocyte subpopulations. In summary, our data suggest that sex-biased co-regulation of the Toll-like receptor locus and XCI escape of TLR8 contribute to the sexual dimorphism in TLR8 expression, which may have important consequences for the functional make-up of monocyte and T cell populations.


Asunto(s)
Monocitos , Inactivación del Cromosoma X , Humanos , Femenino , Masculino , Receptor Toll-Like 8/genética , Linfocitos T , Hibridación Fluorescente in Situ , Receptor Toll-Like 7/genética , Linfocitos T CD4-Positivos
12.
Sci Adv ; 8(6): eabg9455, 2022 Feb 11.
Artículo en Inglés | MEDLINE | ID: mdl-35138899

RESUMEN

Super Enhancers (SEs) are clusters of regulatory elements associated with cell identity and disease. However, whether these elements are induced by oncogenes and can regulate gene modules cooperating for cancer cell transformation or maintenance remains elusive. To address this question, we conducted a genome-wide CRISPRi-based screening of SEs in ETO2-GLIS2+ acute megakaryoblastic leukemia. This approach revealed SEs essential for leukemic cell growth and survival that are induced by ETO2-GLIS2 expression. In particular, we identified a de novo SE specific of this leukemia subtype and regulating expression of tyrosine kinase-associated receptors KIT and PDGFRA. Combined expression of these two receptors was required for leukemic cell growth, and CRISPRi-mediated inhibition of this SE or treatment with tyrosine kinase inhibitors impaired progression of leukemia in vivo in patient-derived xenografts experiments. Our results show that fusion oncogenes, such as ETO2-GLIS2, can induce activation of SEs regulating essential gene modules synergizing for leukemia progression.

14.
Chromosome Res ; 17(1): 115-26, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19214764

RESUMEN

The inactivation of one of the two X chromosomes in female placental mammals represents a remarkable example of epigenetic silencing. X inactivation occurs also in marsupial mammals, but is phenotypically different, being incomplete, tissue-specific and paternal. Paternal X inactivation occurs also in the extraembryonic cells of rodents, suggesting that imprinted X inactivation represents a simpler ancestral mechanism. This evolved into a complex and random process in placental mammals under the control of the XIST gene, involving notably variant and modified histones. Molecular mechanisms of X inactivation in marsupials are poorly known, but occur in the absence of an XIST homologue. We analysed the specific pattern of histone modifications using immunofluorescence on metaphasic chromosomes of a model kangaroo, the tammar wallaby. We found that all active marks are excluded from the inactive X in marsupials, as in placental mammals, so this represents a common feature of X inactivation throughout mammals. However, we were unable to demonstrate the accumulation of inactive histone marks, suggesting some fundamental differences in the molecular mechanism of X inactivation between marsupial and placental mammals. A better understanding of the epigenetic mechanisms underlying X inactivation in marsupials will provide important insights into the evolution of this complex process.


Asunto(s)
Histonas/metabolismo , Marsupiales/genética , Inactivación del Cromosoma X/genética , Cromosoma X/metabolismo , Animales , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Histonas/genética , Hibridación Fluorescente in Situ , Marsupiales/metabolismo , Cromosoma X/genética
15.
Chromosome Res ; 17(5): 671-85, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19802707

RESUMEN

Determining the evolutionary origin of X inactivation mechanisms in mammals requires knowledge of features of X inactivation across all three major mammal lineages; monotremes, marsupials and eutherians. In the past, research into X inactivation in marsupials and monotremes lagged far behind the major advances made in understanding the mechanisms of X inactivation in human and mouse. Fragmentary knowledge of the genic content and sequence of marsupial and monotreme X chromosomes has been alleviated by the recent release of genome sequences for two marsupials and one monotreme. This has lead to a number of important findings, among which is the absence of XIST in marsupials and monotremes, and the surprising finding that X-borne genes in platypus are subject to stochastic transcriptional inhibition rather than whole chromosome inactivation. Availability of sequence data, and new techniques for studying expression and chromatin modification, now make rapid advance possible.


Asunto(s)
Evolución Biológica , Marsupiales/genética , Monotremata/genética , Inactivación del Cromosoma X , Animales , Epigénesis Genética , Cromosomas Sexuales
16.
Semin Immunopathol ; 41(2): 153-164, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30276444

RESUMEN

Women develop stronger immune responses than men, with positive effects on the resistance to viral or bacterial infections but magnifying also the susceptibility to autoimmune diseases like systemic lupus erythematosus (SLE). In SLE, the dosage of the endosomal Toll-like receptor 7 (TLR7) is crucial. Murine models have shown that TLR7 overexpression suffices to induce spontaneous lupus-like disease. Conversely, suppressing TLR7 in lupus-prone mice abolishes SLE development. TLR7 is encoded by a gene on the X chromosome gene, denoted TLR7 in humans and Tlr7 in the mouse, and expressed in plasmacytoid dendritic cells (pDC), monocytes/macrophages, and B cells. The receptor recognizes single-stranded RNA, and its engagement promotes B cell maturation and the production of pro-inflammatory cytokines and antibodies. In female mammals, each cell randomly inactivates one of its two X chromosomes to equalize gene dosage with XY males. However, 15 to 23% of X-linked human genes escape X chromosome inactivation so that both alleles can be expressed simultaneously. It has been hypothesized that biallelic expression of X-linked genes could occur in female immune cells, hence fostering harmful autoreactive and inflammatory responses. We review here the current knowledge of the role of TLR7 in SLE, and recent evidence demonstrating that TLR7 escapes from X chromosome inactivation in pDCs, monocytes, and B lymphocytes from women and Klinefelter syndrome men. Female B cells where TLR7 is thus biallelically expressed display higher TLR7-driven functional responses, connecting the presence of two X chromosomes with the enhanced immunity of women and their increased susceptibility to TLR7-dependent autoimmune syndromes.


Asunto(s)
Cromosomas Humanos X , Dosificación de Gen/inmunología , Predisposición Genética a la Enfermedad , Lupus Eritematoso Sistémico , Caracteres Sexuales , Receptor Toll-Like 7 , Inactivación del Cromosoma X/inmunología , Animales , Cromosomas Humanos X/genética , Cromosomas Humanos X/inmunología , Femenino , Humanos , Lupus Eritematoso Sistémico/genética , Lupus Eritematoso Sistémico/inmunología , Masculino , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/inmunología , Ratones , Receptor Toll-Like 7/genética , Receptor Toll-Like 7/inmunología
17.
Front Immunol ; 10: 443, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30923523

RESUMEN

Xlf/Cernunnos is unique among the core factors of the non-homologous end joining (NHEJ) DNA double strand breaks (DSBs) repair pathway, in the sense that it is not essential for V(D)J recombination in vivo and in vitro. Unlike other NHEJ deficient mice showing a SCID phenotype, Xlf-/- mice present a unique immune phenotype with a moderate B- and T-cell lymphopenia, a decreased cellularity in the thymus, and a characteristic TCRα repertoire bias associated with the P53-dependent apoptosis of CD4+CD8+ DP thymocytes. Here, we thoroughly analyzed Xlf-/- mice immune phenotype and showed that it is specifically related to the DP stage but independent of the MHC-driven antigen presentation and T-cell activation during positive selection. Instead, we show that V(D)J recombination is subefficient in Xlf-/- mice in vivo, exemplified by the presence of unrepaired DSBs in the thymus. This results in a moderate developmental delay of both B- and T-lymphocytes at key V(D)J recombination dependent stages. Furthermore, subefficient V(D)J recombination waves are accumulating during TCRα rearrangement, causing the typical TCRα repertoire bias with loss of distal Vα and Jα rearrangements.


Asunto(s)
Linfocitos B/inmunología , Proteínas de Unión al ADN/deficiencia , Reordenamiento Génico de la Cadena alfa de los Receptores de Antígenos de los Linfocitos T/inmunología , Transducción de Señal , Linfocitos T/inmunología , Recombinación V(D)J/inmunología , Animales , Linfocitos B/citología , Proteínas de Unión al ADN/inmunología , Ratones , Ratones Noqueados , Transducción de Señal/genética , Transducción de Señal/inmunología , Linfocitos T/citología , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/inmunología
18.
Cell Stem Cell ; 24(4): 551-565.e8, 2019 04 04.
Artículo en Inglés | MEDLINE | ID: mdl-30905619

RESUMEN

Precise gene editing in hematopoietic stem and progenitor cells (HSPCs) holds promise for treating genetic diseases. However, responses triggered by programmable nucleases in HSPCs are poorly characterized and may negatively impact HSPC engraftment and long-term repopulation capacity. Here, we induced either one or several DNA double-stranded breaks (DSBs) with optimized zinc-finger and CRISPR/Cas9 nucleases and monitored DNA damage response (DDR) foci induction, cell-cycle progression, and transcriptional responses in HSPC subpopulations, with up to single-cell resolution. p53-mediated DDR pathway activation was the predominant response to even single-nuclease-induced DSBs across all HSPC subtypes analyzed. Excess DSB load and/or adeno-associated virus (AAV)-mediated delivery of DNA repair templates induced cumulative p53 pathway activation, constraining proliferation, yield, and engraftment of edited HSPCs. However, functional impairment was reversible when DDR burden was low and could be overcome by transient p53 inhibition. These findings provide molecular and functional evidence for feasible and seamless gene editing in HSPCs.


Asunto(s)
Daño del ADN , Edición Génica , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Línea Celular , Humanos , Células K562 , Ratones , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID
19.
Sci Immunol ; 3(19)2018 01 26.
Artículo en Inglés | MEDLINE | ID: mdl-29374079

RESUMEN

Toll-like receptor 7 (TLR7) is critical to the induction of antiviral immunity, but TLR7 dosage is also a key pathogenic factor in systemic lupus erythematosus (SLE), an autoimmune disease with strong female bias. SLE prevalence is also elevated in individuals with Klinefelter syndrome, who carry one or more supernumerary X chromosomes, suggesting that the X chromosome complement contributes to SLE susceptibility. TLR7 is encoded by an X chromosome locus, and we examined here whether the TLR7 gene evades silencing by X chromosome inactivation in immune cells from women and Klinefelter syndrome males. Single-cell analyses of TLR7 allelic expression demonstrated that substantial fractions of primary B lymphocytes, monocytes, and plasmacytoid dendritic cells not only in women but also in Klinefelter syndrome males express TLR7 on both X chromosomes. Biallelic B lymphocytes from women displayed greater TLR7 transcriptional expression than the monoallelic cells, correlated with higher TLR7 protein expression in female than in male leukocyte populations. Biallelic B cells were preferentially enriched during the TLR7-driven proliferation of CD27+ plasma cells. In addition, biallelic cells showed a greater than twofold increase over monoallelic cells in the propensity to immunoglobulin G class switch during the TLR7-driven, T cell-dependent differentiation of naive B lymphocytes into immunoglobulin-secreting cells. TLR7 escape from X inactivation endows the B cell compartment with added responsiveness to TLR7 ligands. This finding supports the hypothesis that enhanced TLR7 expression owing to biallelism contributes to the higher risk of developing SLE and other autoimmune disorders in women and in men with Klinefelter syndrome.


Asunto(s)
Activación de Linfocitos/inmunología , Receptor Toll-Like 7/inmunología , Inactivación del Cromosoma X/inmunología , Linfocitos B/inmunología , Diferenciación Celular/inmunología , Proliferación Celular/fisiología , Células Dendríticas/inmunología , Femenino , Humanos , Cambio de Clase de Inmunoglobulina/inmunología , Inmunoglobulina G/inmunología , Ligandos , Lupus Eritematoso Sistémico/inmunología , Masculino , Miembro 7 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/inmunología
20.
Mol Cell Biol ; 24(12): 5475-84, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15169908

RESUMEN

Histone H3 tail modifications are among the earliest chromatin changes in the X-chromosome inactivation process. In this study we investigated the relative profiles of two important repressive marks on the X chromosome: methylation of H3 lysine 9 (K9) and 27 (K27). We found that both H3K9 dimethylation and K27 trimethylation characterize the inactive X in somatic cells and that their relative kinetics of enrichment on the X chromosome as it undergoes inactivation are similar. However, dynamic changes of H3K9 and H3K27 methylation on the inactivating X chromosome compared to the rest of the genome are distinct, suggesting that these two modifications play complementary and perhaps nonredundant roles in the establishment and/or maintenance of X inactivation. Furthermore, we show that a hotspot of H3K9 dimethylation 5' to Xist also displays high levels of H3 tri-meK27. However, analysis of this region in G9a mutant embryonic stem cells shows that these two methyl marks are dependent on different histone methyltransferases.


Asunto(s)
Histonas/química , Cromosoma X/química , Animales , Células Cultivadas , Compensación de Dosificación (Genética) , Femenino , Histona Metiltransferasas , N-Metiltransferasa de Histona-Lisina/genética , N-Metiltransferasa de Histona-Lisina/metabolismo , Histonas/metabolismo , Cinética , Lisina/química , Masculino , Metilación , Ratones , Mutación , Proteína Metiltransferasas , ARN Largo no Codificante , ARN no Traducido/genética , ARN no Traducido/metabolismo , Cromosoma X/genética , Cromosoma X/metabolismo
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda