Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 34
Filtrer
1.
Nat Commun ; 15(1): 1090, 2024 Feb 05.
Article de Anglais | MEDLINE | ID: mdl-38316788

RÉSUMÉ

Macrophages are fundamental cells of the innate immune system that support normal haematopoiesis and play roles in both anti-cancer immunity and tumour progression. Here we use a chimeric mouse model of chronic myeloid leukaemia (CML) and human bone marrow (BM) derived macrophages to study the impact of the dysregulated BM microenvironment on bystander macrophages. Utilising single-cell RNA sequencing (scRNA-seq) of Philadelphia chromosome (Ph) negative macrophages we reveal unique subpopulations of immature macrophages residing in the CML BM microenvironment. CML exposed macrophages separate from their normal counterparts by reduced expression of the surface marker CD36, which significantly reduces clearance of apoptotic cells. We uncover aberrant production of CML-secreted factors, including the immune modulatory protein lactotransferrin (LTF), that suppresses efferocytosis, phagocytosis, and CD36 surface expression in BM macrophages, indicating that the elevated secretion of LTF is, at least partially responsible for the supressed clearance function of Ph- macrophages.


Sujet(s)
Leucémie myéloïde chronique BCR-ABL positive , Leucémie myéloïde , Animaux , Souris , Humains , Moelle osseuse/anatomopathologie , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Leucémie myéloïde/anatomopathologie , Chromosome Philadelphie , Macrophages/métabolisme , Protéines de fusion bcr-abl/génétique , Protéines de fusion bcr-abl/métabolisme , Microenvironnement tumoral/génétique
2.
Nat Commun ; 15(1): 651, 2024 Jan 22.
Article de Anglais | MEDLINE | ID: mdl-38246924

RÉSUMÉ

Whilst it is recognised that targeting self-renewal is an effective way to functionally impair the quiescent leukaemic stem cells (LSC) that persist as residual disease in chronic myeloid leukaemia (CML), developing therapeutic strategies to achieve this have proved challenging. We demonstrate that the regulatory programmes of quiescent LSC in chronic phase CML are similar to that of embryonic stem cells, pointing to a role for wild type p53 in LSC self-renewal. In support of this, increasing p53 activity in primitive CML cells using an MDM2 inhibitor in combination with a tyrosine kinase inhibitor resulted in reduced CFC outputs and engraftment potential, followed by loss of multilineage priming potential and LSC exhaustion when combination treatment was discontinued. Our work provides evidence that targeting LSC self-renewal is exploitable in the clinic to irreversibly impair quiescent LSC function in CML residual disease - with the potential to enable more CML patients to discontinue therapy and remain in therapy-free remission.


Sujet(s)
Leucémie myéloïde chronique BCR-ABL positive , Leucémie myéloïde , Humains , Division cellulaire , Cellules souches embryonnaires , Maladie résiduelle , Protéine p53 suppresseur de tumeur/génétique
3.
Cancers (Basel) ; 14(21)2022 Oct 26.
Article de Anglais | MEDLINE | ID: mdl-36358672

RÉSUMÉ

Tyrosine kinase inhibitors (TKI) have revolutionised the treatment of CML. However, TKI do not eliminate the leukaemia stem cells (LSC), which can re-initiate the disease. Thus, finding new therapeutic targets in CML LSC is key to finding a curative treatment. Using microarray datasets, we defined a list of 227 genes that were differentially expressed in CML LSC compared to the healthy controls but were not affected by TKI in vitro. Two of them, CD33 and PPIF, are targeted by gemtuzumab-ozogamicin and cyclosporin A, respectively. We treated CML and the control CD34+ cells with either drug with or without imatinib to investigate the therapeutic potential of the TKI-independent gene expression programme. Cyclosporine A, in combination with imatinib, reduced the number of CML CFC compared with non-CML controls, but only at supra-therapeutic concentrations. Gemtuzumab-ozogamicin showed an EC50 of 146 ng/mL, below the plasma peak concentration of 630 ng/mL observed in the AML patients and below the EC50 of 3247 ng/mL observed in the non-CML cells. Interestingly, gemtuzumab-ozogamicin seems to promote cell cycle progression in CML CD34+ cells and demonstrated activation of the RUNX1 pathway in an RNAseq experiment. This suggests that targeting the TKI-independent genes in CML LSC could be exploited for the development of new therapies in CML.

4.
Cancers (Basel) ; 14(19)2022 Sep 27.
Article de Anglais | MEDLINE | ID: mdl-36230624

RÉSUMÉ

RNA splicing factors are frequently altered in cancer and can act as both oncoproteins and tumour suppressors. They have been found mutated or deregulated, justifying the growing interest in the targeting of splicing catalysis, splicing regulatory proteins, and/or specific, key altered splicing events. We recently showed that the DNA methylation alterations of CD34+CD15- chronic myeloid leukaemia (CML) cells affect, among others, alternative splicing genes, suggesting that spliceosome actors might be altered in chronic-phase (CP)-CML. We investigated the expression of 12 spliceosome genes known to be oncogenes or tumour suppressor genes in primary CP-CML CD34+ cells at diagnosis (n = 15). We found that CP-CML CD34+ cells had a distinct splicing signature profile as compared with healthy donor CD34+ cells or whole CP-CML cells, suggesting: (i) a spliceosome deregulation from the diagnosis time and (ii) an intraclonal heterogeneity. We could identify three profile types, but there was no relationship with a patient's characteristics. By incubating cells with TKI and/or a spliceosome-targeted drug (TG003), we showed that CP-CML CD34+ cells are both BCR::ABL and spliceosome dependent, with the combination of the two drugs showing an additive effect while sparing healthy donors cells. Our results suggest that the spliceosome may be a new potential target for the treatment of CML.

5.
Sci Transl Med ; 13(613): eabd5016, 2021 Sep 29.
Article de Anglais | MEDLINE | ID: mdl-34586834

RÉSUMÉ

Inhibition of autophagy has been proposed as a potential therapy for individuals with cancer. However, current lysosomotropic autophagy inhibitors have demonstrated limited efficacy in clinical trials. Therefore, validation of novel specific autophagy inhibitors using robust preclinical models is critical. In chronic myeloid leukemia (CML), minimal residual disease is maintained by persistent leukemic stem cells (LSCs), which drive tyrosine kinase inhibitor (TKI) resistance and patient relapse. Here, we show that deletion of autophagy-inducing kinase ULK1 (unc-51­like autophagy activating kinase 1) reduces growth of cell line and patient-derived xenografted CML cells in mouse models. Using primitive cells, isolated from individuals with CML, we demonstrate that pharmacological inhibition of ULK1 selectively targets CML LSCs ex vivo and in vivo, when combined with TKI treatment. The enhanced TKI sensitivity after ULK1-mediated autophagy inhibition is driven by increased mitochondrial respiration and loss of quiescence and points to oxidative stress­induced differentiation of CML LSCs, proposing an alternative strategy for treating patients with CML.


Sujet(s)
Autophagie , Stress oxydatif , Homologue de la protéine-1 associée à l'autophagie/métabolisme , Différenciation cellulaire , Cellules souches/métabolisme
6.
Cell Death Dis ; 12(6): 573, 2021 06 03.
Article de Anglais | MEDLINE | ID: mdl-34083515

RÉSUMÉ

Epigenomic dysregulation is a common pathological feature in human hematological malignancies. H3K9me3 emerges as an important epigenomic marker in acute myeloid leukemia (AML). Its associated methyltransferases, such as SETDB1, suppress AML leukemogenesis, whilst H3K9me3 demethylases KDM4C is required for mixed-lineage leukemia rearranged AML. However, the specific role and molecular mechanism of action of another member of the KDM4 family, KDM4A has not previously been clearly defined. In this study, we delineated and functionally validated the epigenomic network regulated by KDM4A. We show that selective loss of KDM4A is sufficient to induce apoptosis in a broad spectrum of human AML cells. This detrimental phenotype results from a global accumulation of H3K9me3 and H3K27me3 at KDM4A targeted genomic loci thereby causing downregulation of a KDM4A-PAF1 controlled transcriptional program essential for leukemogenesis, distinct from that of KDM4C. From this regulatory network, we further extracted a KDM4A-9 gene signature enriched with leukemia stem cell activity; the KDM4A-9 score alone or in combination with the known LSC17 score, effectively stratifies high-risk AML patients. Together, these results establish the essential and unique role of KDM4A for AML self-renewal and survival, supporting further investigation of KDM4A and its targets as a potential therapeutic vulnerability in AML.


Sujet(s)
Auto-renouvellement cellulaire/génétique , Survie cellulaire/génétique , Épigénomique/méthodes , Histone Demethylases/métabolisme , Leucémie aigüe myéloïde/génétique , Animaux , Apoptose , Humains , Souris
7.
Nat Rev Cancer ; 20(3): 158-173, 2020 03.
Article de Anglais | MEDLINE | ID: mdl-31907378

RÉSUMÉ

For two decades, leukaemia stem cells (LSCs) in chronic myeloid leukaemia (CML) and acute myeloid leukaemia (AML) have been advanced paradigms for the cancer stem cell field. In CML, the acquisition of the fusion tyrosine kinase BCR-ABL1 in a haematopoietic stem cell drives its transformation to become a LSC. In AML, LSCs can arise from multiple cell types through the activity of a number of oncogenic drivers and pre-leukaemic events, adding further layers of context and genetic and cellular heterogeneity to AML LSCs not observed in most cases of CML. Furthermore, LSCs from both AML and CML can be refractory to standard-of-care therapies and persist in patients, diversify clonally and serve as reservoirs to drive relapse, recurrence or progression to more aggressive forms. Despite these complexities, LSCs in both diseases share biological features, making them distinct from other CML or AML progenitor cells and from normal haematopoietic stem cells. These features may represent Achilles' heels against which novel therapies can be developed. Here, we review many of the similarities and differences that exist between LSCs in CML and AML and examine the therapeutic strategies that could be used to eradicate them.


Sujet(s)
Leucémie myéloïde chronique BCR-ABL positive/étiologie , Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Leucémie aigüe myéloïde/étiologie , Leucémie aigüe myéloïde/métabolisme , Cellules souches tumorales/métabolisme , Animaux , Marqueurs biologiques tumoraux , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/immunologie , Transformation cellulaire néoplasique/métabolisme , Prise en charge de la maladie , Prédisposition aux maladies , Développement de médicament , Histoire du 20ème siècle , Histoire du 21ème siècle , Humains , Leucémie myéloïde chronique BCR-ABL positive/diagnostic , Leucémie myéloïde chronique BCR-ABL positive/thérapie , Leucémie aigüe myéloïde/diagnostic , Leucémie aigüe myéloïde/thérapie , Thérapie moléculaire ciblée , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Recherche/histoire , Recherche/tendances
8.
Front Cell Dev Biol ; 7: 136, 2019.
Article de Anglais | MEDLINE | ID: mdl-31380371

RÉSUMÉ

Chronic myeloid leukemia (CML) is a hematopoietic stem cell disorder characterized by BCR-ABL1, an oncogenic fusion gene arising from the Philadelphia chromosome. The development of tyrosine kinase inhibitors (TKIs) to overcome the constitutive tyrosine kinase activity of the BCR-ABL protein has dramatically improved disease management and patient outcomes over the past 20 years. However, the majority of patients are not cured and developing novel therapeutic strategies that target epigenetic processes are a promising avenue to improve cure rates. A number of epigenetic mechanisms are altered or reprogrammed during the development and progression of CML, resulting in alterations in histone modifications, DNA methylation and dysregulation of the transcriptional machinery. In this review these epigenetic alterations are examined and the potential of epigenetic therapies are discussed as a means of eradicating residual disease and offering a potential cure for CML in combination with current therapies.

9.
Semin Cancer Biol ; 51: 180-197, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-28778403

RÉSUMÉ

The onset of global epigenetic changes in chromatin that drive tumor proliferation and heterogeneity is a hallmark of many forms of cancer. Identifying the epigenetic mechanisms that govern these changes and developing therapeutic approaches to modulate them, is a well-established avenue pursued in translational cancer medicine. Chronic myeloid leukemia (CML) arises clonally when a hematopoietic stem cell (HSC) acquires the capacity to produce the constitutively active tyrosine kinase BCR-ABL1 fusion protein which drives tumor development. Treatment with tyrosine kinase inhibitors (TKI) that target BCR-ABL1 has been transformative in CML management but it does not lead to cure in the vast majority of patients. Thus novel therapeutic approaches are required and these must target changes to biological pathways that are aberrant in CML - including those that occur when epigenetic mechanisms are altered. These changes may be due to alterations in DNA or histones, their biochemical modifications and requisite 'writer' proteins, or to dysregulation of various types of non-coding RNAs that collectively function as modulators of transcriptional control and DNA integrity. Here, we review the evidence for subverted epigenetic mechanisms in CML and how these impact on a diverse set of biological pathways, on disease progression, prognosis and drug resistance. We will also discuss recent progress towards developing epigenetic therapies that show promise to improve CML patient care and may lead to improved cure rates.


Sujet(s)
Méthylation de l'ADN , Épigenèse génétique , Régulation de l'expression des gènes tumoraux , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/thérapie , Animaux , Humains , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie
10.
Blood ; 129(12): 1595-1606, 2017 03 23.
Article de Anglais | MEDLINE | ID: mdl-28159740

RÉSUMÉ

Chronic myeloid leukemia (CML) is caused by the acquisition of the tyrosine kinase BCR-ABL1 in a hemopoietic stem cell, transforming it into a leukemic stem cell (LSC) that self-renews, proliferates, and differentiates to give rise to a myeloproliferative disease. Although tyrosine kinase inhibitors (TKIs) that target the kinase activity of BCR-ABL1 have transformed CML from a once-fatal disease to a manageable one for the vast majority of patients, only ∼10% of those who present in chronic phase (CP) can discontinue TKI treatment and maintain a therapy-free remission. Strong evidence now shows that CML LSCs are resistant to the effects of TKIs and persist in all patients on long-term therapy, where they may promote acquired TKI resistance, drive relapse or disease progression, and inevitably represent a bottleneck to cure. Since their discovery in patients almost 2 decades ago, CML LSCs have become a well-recognized exemplar of the cancer stem cell and have been characterized extensively, with the aim of developing new curative therapeutic approaches based on LSC eradication. This review summarizes our current understanding of many of the pathways and mechanisms that promote the survival of the CP CML LSCs and how they can be a source of new gene coding mutations that impact in the clinic. We also review recent preclinical approaches that show promise to eradicate the LSC, and future challenges on the path to cure.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Leucémie myéloïde en phase chronique/traitement médicamenteux , Cellules souches tumorales/effets des médicaments et des substances chimiques , Protéines de fusion bcr-abl/génétique , Humains , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde en phase chronique/anatomopathologie , Cellules souches tumorales/anatomopathologie , Inhibiteurs de protéines kinases/usage thérapeutique
11.
Blood ; 129(2): 199-208, 2017 01 12.
Article de Anglais | MEDLINE | ID: mdl-27793879

RÉSUMÉ

Targeting the fusion oncoprotein BCR-ABL with tyrosine kinase inhibitors has significantly affected chronic myeloid leukemia (CML) treatment, transforming the life expectancy of patients; however the risk for relapse remains, due to persistence of leukemic stem cells (LSCs). Therefore it is imperative to explore the mechanisms that result in LSC survival and develop new therapeutic approaches. We now show that major histocompatibility complex (MHC)-II and its master regulator class II transactivator (CIITA) are downregulated in CML compared with non-CML stem/progenitor cells in a BCR-ABL kinase-independent manner. Interferon γ (IFN-γ) stimulation resulted in an upregulation of CIITA and MHC-II in CML stem/progenitor cells; however, the extent of IFN-γ-induced MHC-II upregulation was significantly lower than when compared with non-CML CD34+ cells. Interestingly, the expression levels of CIITA and MHC-II significantly increased when CML stem/progenitor cells were treated with the JAK1/2 inhibitor ruxolitinib (RUX). Moreover, mixed lymphocyte reactions revealed that exposure of CD34+ CML cells to IFN-γ or RUX significantly enhanced proliferation of the responder CD4+CD69+ T cells. Taken together, these data suggest that cytokine-driven JAK-mediated signals, provided by CML cells and/or the microenvironment, antagonize MHC-II expression, highlighting the potential for developing novel immunomodulatory-based therapies to enable host-mediated immunity to assist in the detection and eradication of CML stem/progenitor cells.


Sujet(s)
Antigènes d'histocompatibilité de classe II/biosynthèse , Leucémie myéloïde chronique BCR-ABL positive/immunologie , Cellules souches tumorales/immunologie , Échappement de la tumeur à la surveillance immunitaire/immunologie , Cellules cultivées , Cytokines/immunologie , Cytokines/métabolisme , Régulation négative , Femelle , Cytométrie en flux , Régulation de l'expression des gènes/immunologie , Humains , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Test de culture lymphocytaire mixte , Mâle , Cellules souches tumorales/anatomopathologie , Séquençage par oligonucléotides en batterie , RT-PCR , Transduction du signal/immunologie
12.
Cancer Discov ; 6(11): 1248-1257, 2016 11.
Article de Anglais | MEDLINE | ID: mdl-27630125

RÉSUMÉ

A major obstacle to curing chronic myeloid leukemia (CML) is residual disease maintained by tyrosine kinase inhibitor (TKI)-persistent leukemic stem cells (LSC). These are BCR-ABL1 kinase independent, refractory to apoptosis, and serve as a reservoir to drive relapse or TKI resistance. We demonstrate that Polycomb Repressive Complex 2 is misregulated in chronic phase CML LSCs. This is associated with extensive reprogramming of H3K27me3 targets in LSCs, thus sensitizing them to apoptosis upon treatment with an EZH2-specific inhibitor (EZH2i). EZH2i does not impair normal hematopoietic stem cell survival. Strikingly, treatment of primary CML cells with either EZH2i or TKI alone caused significant upregulation of H3K27me3 targets, and combined treatment further potentiated these effects and resulted in significant loss of LSCs compared to TKI alone, in vitro, and in long-term bone marrow murine xenografts. Our findings point to a promising epigenetic-based therapeutic strategy to more effectively target LSCs in patients with CML receiving TKIs. SIGNIFICANCE: In CML, TKI-persistent LSCs remain an obstacle to cure, and approaches to eradicate them remain a significant unmet clinical need. We demonstrate that EZH2 and H3K27me3 reprogramming is important for LSC survival, but renders LSCs sensitive to the combined effects of EZH2i and TKI. This represents a novel approach to more effectively target LSCs in patients receiving TKI treatment. Cancer Discov; 6(11); 1248-57. ©2016 AACR.See related article by Xie et al., p. 1237This article is highlighted in the In This Issue feature, p. 1197.


Sujet(s)
Protéine-2 homologue de l'activateur de Zeste/génétique , Protéines de fusion bcr-abl/génétique , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Inhibiteurs de protéines kinases/administration et posologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Reprogrammation cellulaire/génétique , Résistance aux médicaments antinéoplasiques/génétique , Protéine-2 homologue de l'activateur de Zeste/antagonistes et inhibiteurs , Épigenèse génétique/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/anatomopathologie , Humains , Leucémie myéloïde chronique BCR-ABL positive/génétique , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Souris , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie
13.
Nature ; 534(7607): 341-6, 2016 06 16.
Article de Anglais | MEDLINE | ID: mdl-27281222

RÉSUMÉ

Chronic myeloid leukaemia (CML) arises after transformation of a haemopoietic stem cell (HSC) by the protein-tyrosine kinase BCR-ABL. Direct inhibition of BCR-ABL kinase has revolutionized disease management, but fails to eradicate leukaemic stem cells (LSCs), which maintain CML. LSCs are independent of BCR-ABL for survival, providing a rationale for identifying and targeting kinase-independent pathways. Here we show--using proteomics, transcriptomics and network analyses--that in human LSCs, aberrantly expressed proteins, in both imatinib-responder and non-responder patients, are modulated in concert with p53 (also known as TP53) and c-MYC regulation. Perturbation of both p53 and c-MYC, and not BCR-ABL itself, leads to synergistic cell kill, differentiation, and near elimination of transplantable human LSCs in mice, while sparing normal HSCs. This unbiased systems approach targeting connected nodes exemplifies a novel precision medicine strategy providing evidence that LSCs can be eradicated.


Sujet(s)
Leucémie myéloïde chronique BCR-ABL positive/métabolisme , Leucémie myéloïde chronique BCR-ABL positive/anatomopathologie , Cellules souches tumorales/métabolisme , Cellules souches tumorales/anatomopathologie , Protéines proto-oncogènes c-myc/antagonistes et inhibiteurs , Protéine p53 suppresseur de tumeur/antagonistes et inhibiteurs , Acétamides/pharmacologie , Acétamides/usage thérapeutique , Animaux , Antigènes CD34/métabolisme , Azépines/pharmacologie , Azépines/usage thérapeutique , Mort cellulaire/effets des médicaments et des substances chimiques , Différenciation cellulaire/effets des médicaments et des substances chimiques , Protéines de liaison à l'ADN/métabolisme , Femelle , Protéines de fusion bcr-abl/métabolisme , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/métabolisme , Humains , Mésilate d'imatinib/pharmacologie , Mésilate d'imatinib/usage thérapeutique , Imidazolines/pharmacologie , Imidazolines/usage thérapeutique , Leucémie myéloïde chronique BCR-ABL positive/traitement médicamenteux , Leucémie myéloïde chronique BCR-ABL positive/génétique , Mâle , Souris , Protéines tumorales/métabolisme , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/transplantation , Protéomique , Protéines proto-oncogènes c-myc/déficit , Protéines proto-oncogènes c-myc/génétique , Protéines proto-oncogènes c-myc/métabolisme , Reproductibilité des résultats , Transduction du signal/effets des médicaments et des substances chimiques , Transcriptome , Protéine p53 suppresseur de tumeur/déficit , Protéine p53 suppresseur de tumeur/génétique , Protéine p53 suppresseur de tumeur/métabolisme
15.
Blood ; 122(26): 4199-209, 2013 Dec 19.
Article de Anglais | MEDLINE | ID: mdl-24200685

RÉSUMÉ

TAL1 is an important regulator of hematopoiesis and its expression is tightly controlled despite complexities in its genomic organization. It is frequently misregulated in T-cell acute lymphoblastic leukemia (T-ALL), often due to deletions between TAL1 and the neighboring STIL gene. To better understand the events that lead to TAL1 expression in hematopoiesis and in T-ALL, we studied looping interactions at the TAL1 locus. In TAL1-expressing erythroid cells, the locus adopts a looping "hub" which brings into close physical proximity all known TAL1 cis-regulatory elements including CTCF-bound insulators. Loss of GATA1 results in disassembly of the hub and loss of CTCF/RAD21 from one of its insulators. Genes flanking TAL1 are partly dependent on hub integrity for their transcriptional regulation. We identified looping patterns unique to TAL1-expressing T-ALL cells, and, intriguingly, loops occurring between the TAL1 and STIL genes at the common TAL1/STIL breakpoints found in T-ALL. These findings redefine how TAL1 and neighboring genes communicate within the nucleus, and indicate that looping facilitates both normal and aberrant TAL1 expression and may predispose to structural rearrangements in T-ALL. We also propose that GATA1-dependent looping mechanisms may facilitate the conservation of TAL1 regulation despite cis-regulatory remodeling during vertebrate evolution.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Chromatine/génétique , Régulation de l'expression des gènes tumoraux , Leucémie-lymphome lymphoblastique à précurseurs T/génétique , Protéines proto-oncogènes/génétique , Animaux , Chromatine/composition chimique , Éléments activateurs (génétique)/génétique , Facteur de transcription GATA-1/génétique , Humains , Protéines et peptides de signalisation intracellulaire/génétique , Cellules Jurkat , Cellules K562 , Lymphocytes/cytologie , Protéines membranaires/génétique , Souris , Souris de lignée ICR , Nucleoside phosphate kinase/génétique , Régions promotrices (génétique)/génétique , Conformation des protéines , Protéine-1 de la lleucémie lymphoïde aiguë à cellules T
17.
PLoS One ; 5(8): e12339, 2010 Aug 23.
Article de Anglais | MEDLINE | ID: mdl-20808788

RÉSUMÉ

It has recently been shown that nucleosome distribution, histone modifications and RNA polymerase II (Pol II) occupancy show preferential association with exons ("exon-intron marking"), linking chromatin structure and function to co-transcriptional splicing in a variety of eukaryotes. Previous ChIP-sequencing studies suggested that these marking patterns reflect the nucleosomal landscape. By analyzing ChIP-chip datasets across the human genome in three cell types, we have found that this marking system is far more complex than previously observed. We show here that a range of histone modifications and Pol II are preferentially associated with exons. However, there is noticeable cell-type specificity in the degree of exon marking by histone modifications and, surprisingly, this is also reflected in some histone modifications patterns showing biases towards introns. Exon-intron marking is laid down in the absence of transcription on silent genes, with some marking biases changing or becoming reversed for genes expressed at different levels. Furthermore, the relationship of this marking system with splicing is not simple, with only some histone modifications reflecting exon usage/inclusion, while others mirror patterns of exon exclusion. By examining nucleosomal distributions in all three cell types, we demonstrate that these histone modification patterns cannot solely be accounted for by differences in nucleosome levels between exons and introns. In addition, because of inherent differences between ChIP-chip array and ChIP-sequencing approaches, these platforms report different nucleosome distribution patterns across the human genome. Our findings confound existing views and point to active cellular mechanisms which dynamically regulate histone modification levels and account for exon-intron marking. We believe that these histone modification patterns provide links between chromatin accessibility, Pol II movement and co-transcriptional splicing.


Sujet(s)
Exons/génétique , Histone/métabolisme , Introns/génétique , Nucléosomes/génétique , Nucléosomes/métabolisme , Régulation de l'expression des gènes , Humains , Cellules K562 , RNA polymerase II/métabolisme , Transcription génétique
18.
PLoS One ; 5(2): e9059, 2010 Feb 05.
Article de Anglais | MEDLINE | ID: mdl-20140202

RÉSUMÉ

The SCL (TAL1) transcription factor is a critical regulator of haematopoiesis and its expression is tightly controlled by multiple cis-acting regulatory elements. To elaborate further the DNA elements which control its regulation, we used genomic tiling microarrays covering 256 kb of the human SCL locus to perform a concerted analysis of chromatin structure and binding of regulatory proteins in human haematopoietic cell lines. This approach allowed us to characterise further or redefine known human SCL regulatory elements and led to the identification of six novel elements with putative regulatory function both up and downstream of the SCL gene. They bind a number of haematopoietic transcription factors (GATA1, E2A LMO2, SCL, LDB1), CTCF or components of the transcriptional machinery and are associated with relevant histone modifications, accessible chromatin and low nucleosomal density. Functional characterisation shows that these novel elements are able to enhance or repress SCL promoter activity, have endogenous promoter function or enhancer-blocking insulator function. Our analysis opens up several areas for further investigation and adds new layers of complexity to our understanding of the regulation of SCL expression.


Sujet(s)
Facteurs de transcription à motif basique hélice-boucle-hélice/génétique , Régulation de l'expression des gènes , Protéines proto-oncogènes/génétique , Séquences d'acides nucléiques régulatrices/génétique , Facteurs de transcription/métabolisme , Acétylation , Sites de fixation/génétique , Facteur de liaison à la séquence CCCTC , Lignée cellulaire tumorale , Immunoprécipitation de la chromatine , Analyse de regroupements , Cellules HL-60 , Histone/métabolisme , Humains , Cellules K562 , Lysine/métabolisme , Méthylation , Séquençage par oligonucléotides en batterie , Régions promotrices (génétique)/génétique , Liaison aux protéines , Protéines de répression/métabolisme , Protéine-1 de la lleucémie lymphoïde aiguë à cellules T , Cellules U937
19.
Genome Res ; 19(6): 994-1005, 2009 Jun.
Article de Anglais | MEDLINE | ID: mdl-19401398

RÉSUMÉ

The molecular events that contribute to, and result from, the in vivo binding of transcription factors to their cognate DNA sequence motifs in mammalian genomes are poorly understood. We demonstrate that variations within the DNA sequence motifs that bind the transcriptional repressor REST (NRSF) encode in vivo DNA binding affinity hierarchies that contribute to regulatory function during lineage-specific and developmental programs in fundamental ways. First, canonical sequence motifs for REST facilitate strong REST binding and control functional classes of REST targets that are common to all cell types, whilst atypical motifs participate in weak interactions and control those targets, which are cell- or tissue-specific. Second, variations in REST binding relate directly to variations in expression and chromatin configurations of REST's target genes. Third, REST clearance from its binding sites is also associated with variations in the RE1 motif. Finally, and most surprisingly, weak REST binding sites reside in DNA sequences that show the highest levels of constraint through evolution, thus facilitating their roles in maintaining tissue-specific functions. These relationships have never been reported in mammalian systems for any transcription factor.


Sujet(s)
Protéines de liaison à l'ADN/métabolisme , ADN/métabolisme , Protéines de répression/métabolisme , Séquence nucléotidique , Sites de fixation/génétique , Fixation compétitive , Lignée cellulaire , Chromatine/métabolisme , Immunoprécipitation de la chromatine , ADN/génétique , Protéines de liaison à l'ADN/génétique , Analyse de profil d'expression de gènes , Cellules HeLa , Humains , Immunotransfert , Cellules K562 , Petit ARN interférent/génétique , Protéines de répression/génétique , RT-PCR , Transfection
20.
Am J Hum Genet ; 82(1): 181-7, 2008 Jan.
Article de Anglais | MEDLINE | ID: mdl-18179897

RÉSUMÉ

Recently, large-scale benign copy-number variations (CNVs)--encompassing over 12% of the genome and containing genes considered to be dosage tolerant for human development--were uncovered in the human population. Here we present a family with a novel autosomal-dominantly inherited syndrome characterized by microtia, eye coloboma, and imperforation of the nasolacrimal duct. This phenotype is linked to a cytogenetically visible alteration at 4pter consisting of five copies of a copy-number-variable region, encompassing a low-copy repeat (LCR)-rich sequence. We demonstrate that the approximately 750 kb amplicon occurs in exact tandem copies. This is the first example of an amplified CNV associated with a Mendelian disorder, a discovery that implies that genome screens for genetic disorders should include the analysis of so-called benign CNVs and LCRs.


Sujet(s)
Malformations multiples/génétique , Chromosomes humains de la paire 4 , Oreille externe/malformations , Dosage génique , Gènes dominants , Malformations multiples/anatomopathologie , Colobome/génétique , Colobome/anatomopathologie , Femelle , Humains , Mâle , Conduit nasolacrymal/malformations , Pedigree , Syndrome
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...