Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 26
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Nat Metab ; 5(10): 1642-1645, 2023 Oct.
Article de Anglais | MEDLINE | ID: mdl-37794202
2.
Nat Rev Endocrinol ; 19(12): 691-707, 2023 Dec.
Article de Anglais | MEDLINE | ID: mdl-37749386

RÉSUMÉ

Adipose tissue is an endocrine organ and a crucial regulator of energy storage and systemic metabolic homeostasis. Additionally, adipose tissue is a pivotal regulator of cardiovascular health and disease, mediated in part by the endocrine and paracrine secretion of several bioactive products, such as adipokines. Adipose vasculature has an instrumental role in the modulation of adipose tissue expansion, homeostasis and metabolism. The role of the adipose vasculature has been extensively explored in the context of obesity, which is recognized as a global health problem. Obesity-induced accumulation of fat, in combination with vascular rarefaction, promotes adipocyte dysfunction and induces oxidative stress, hypoxia and inflammation. It is now recognized that obesity-associated endothelial dysfunction often precedes the development of cardiovascular diseases. Investigations have revealed heterogeneity within the vascular niche and dynamic reciprocity between vascular and adipose cells, which can become dysregulated in obesity. Here we provide a comprehensive overview of the evolving functions of the vasculature in regulating adipose tissue biology in health and obesity.


Sujet(s)
Tissu adipeux , Obésité , Humains , Tissu adipeux/métabolisme , Obésité/métabolisme , Adipocytes/métabolisme , Adipokines/métabolisme , Biologie
3.
Front Cell Neurosci ; 17: 1165887, 2023.
Article de Anglais | MEDLINE | ID: mdl-37201162

RÉSUMÉ

Pericytes (PCs) are essential components of the blood brain barrier. Brain PCs are critical for dynamically regulating blood flow, for maintaining vascular integrity and their dysregulation is associated with a myriad of disorders such as Alzheimer's disease. To understand their physiological and molecular functions, studies have increasingly focused on primary brain PC isolation and culture. Multiple methods for PC culture have been developed over the years, however, it is still unclear how primary PCs compare to their in vivo counterparts. To address this question, we compared cultured brain PCs at passage 5 and 20 to adult and embryonic brain PCs directly isolated from mouse brains via single cell RNA-seq. Cultured PCs were highly homogeneous, and were most similar to embryonic PCs, while displaying a significantly different transcriptional profile to adult brain PCs. Cultured PCs downregulated canonical PC markers and extracellular matrix (ECM) genes. Importantly, expression of PC markers and ECM genes could be improved by co-culture with brain endothelial cells, showing the importance of the endothelium in maintaining PC identity and function. Taken together, these results highlight key transcriptional differences between cultured and in vivo PCs which should be considered when performing in vitro experiments with brain PCs.

4.
Nat Metab ; 4(11): 1591-1610, 2022 11.
Article de Anglais | MEDLINE | ID: mdl-36400935

RÉSUMÉ

Obesity promotes diverse pathologies, including atherosclerosis and dementia, which frequently involve vascular defects and endothelial cell (EC) dysfunction. Each organ has distinct EC subtypes, but whether ECs are differentially affected by obesity is unknown. Here we use single-cell RNA sequencing to analyze transcriptomes of ~375,000 ECs from seven organs in male mice at progressive stages of obesity to identify organ-specific vulnerabilities. We find that obesity deregulates gene expression networks, including lipid handling, metabolic pathways and AP1 transcription factor and inflammatory signaling, in an organ- and EC-subtype-specific manner. The transcriptomic aberrations worsen with sustained obesity and are only partially mitigated by dietary intervention and weight loss. For example, dietary intervention substantially attenuates dysregulation of liver, but not kidney, EC transcriptomes. Through integration with human genome-wide association study data, we further identify a subset of vascular disease risk genes that are induced by obesity. Our work catalogs the impact of obesity on the endothelium, constitutes a useful resource and reveals leads for investigation as potential therapeutic targets.


Sujet(s)
Athérosclérose , Cellules endothéliales , Mâle , Animaux , Souris , Humains , Cellules endothéliales/métabolisme , Étude d'association pangénomique , Obésité/métabolisme , Perte de poids , Athérosclérose/génétique , Athérosclérose/métabolisme
5.
Proc Natl Acad Sci U S A ; 119(40): e2110374119, 2022 10 04.
Article de Anglais | MEDLINE | ID: mdl-36161905

RÉSUMÉ

Lipodystrophy syndromes (LDs) are characterized by loss of adipose tissue, metabolic complications such as dyslipidemia, insulin resistance, and fatty liver disease, as well as accelerated atherosclerosis. As a result of adipose tissue deficiency, the systemic concentration of the adipokine leptin is reduced. A current promising therapeutic option for patients with LD is treatment with recombinant leptin (metreleptin), resulting in reduced risk of mortality. Here, we investigate the effects of leptin on endothelial to mesenchymal transition (EndMT), which impair the functional properties of endothelial cells and promotes atherogenesis in LD. Leptin treatment reduced inflammation and TGF-ß2-induced expression of mesenchymal genes and prevented impairment of endothelial barrier function. Treatment of lipodystrophic- and atherosclerosis-prone animals (Ldlr-/-; aP2-nSrebp1c-Tg) with leptin reduced macrophage accumulation in atherosclerotic lesions, vascular plaque protrusion, and the number of endothelial cells with mesenchymal gene expression, confirming a reduction in EndMT in LD after leptin treatment. Treatment with leptin inhibited LD-mediated induction of the proatherosclerotic cytokine growth/differentiation factor 15 (GDF15). Inhibition of GDF15 reduced EndMT induction triggered by plasma from patients with LD. Our study reveals that in addition to the effects on adipose tissue function, leptin treatment exerts beneficial effects protecting endothelial function and identity in LD by reducing GDF15.


Sujet(s)
Cellules endothéliales , Transition épithélio-mésenchymateuse , Facteur-15 de croissance et de différenciation , Leptine , Lipodystrophie , Animaux , Athérosclérose/génétique , Cellules endothéliales/effets des médicaments et des substances chimiques , Transition épithélio-mésenchymateuse/effets des médicaments et des substances chimiques , Facteur-15 de croissance et de différenciation/métabolisme , Leptine/pharmacologie , Leptine/usage thérapeutique , Lipodystrophie/traitement médicamenteux , Lipodystrophie/génétique , Souris , Facteur de croissance transformant bêta-2/métabolisme
6.
Basic Res Cardiol ; 117(1): 32, 2022 06 23.
Article de Anglais | MEDLINE | ID: mdl-35737129

RÉSUMÉ

Alterations of RNA editing that affect the secondary structure of RNAs can cause human diseases. We therefore studied RNA editing in failing human hearts. Transcriptome sequencing showed that adenosine-to-inosine (A-to-I) RNA editing was responsible for 80% of the editing events in the myocardium. Failing human hearts were characterized by reduced RNA editing. This was primarily attributable to Alu elements in introns of protein-coding genes. In the failing left ventricle, 166 circRNAs were upregulated and 7 circRNAs were downregulated compared to non-failing controls. Most of the upregulated circRNAs were associated with reduced RNA editing in the host gene. ADAR2, which binds to RNA regions that are edited from A-to-I, was decreased in failing human hearts. In vitro, reduction of ADAR2 increased circRNA levels suggesting a causal effect of reduced ADAR2 levels on increased circRNAs in the failing human heart. To gain mechanistic insight, one of the identified upregulated circRNAs with a high reduction of editing in heart failure, AKAP13, was further characterized. ADAR2 reduced the formation of double-stranded structures in AKAP13 pre-mRNA, thereby reducing the stability of Alu elements and the circularization of the resulting circRNA. Overexpression of circAKAP13 impaired the sarcomere regularity of human induced pluripotent stem cell-derived cardiomyocytes. These data show that ADAR2 mediates A-to-I RNA editing in the human heart. A-to-I RNA editing represses the formation of dsRNA structures of Alu elements favoring canonical linear mRNA splicing and inhibiting the formation of circRNAs. The findings are relevant to diseases with reduced RNA editing and increased circRNA levels and provide insights into the human-specific regulation of circRNA formation.


Sujet(s)
Cellules souches pluripotentes induites , Édition des ARN , Humains , Cellules souches pluripotentes induites/métabolisme , ARN/composition chimique , ARN/génétique , ARN/métabolisme , ARN circulaire/génétique , Protéines de liaison à l'ARN/génétique , Protéines de liaison à l'ARN/métabolisme
7.
Int J Mol Sci ; 21(13)2020 Jun 30.
Article de Anglais | MEDLINE | ID: mdl-32630148

RÉSUMÉ

The vascular system is critical infrastructure that transports oxygen and nutrients around the body, and dynamically adapts its function to an array of environmental changes. To fulfil the demands of diverse organs, each with unique functions and requirements, the vascular system displays vast regional heterogeneity as well as specialized cell types. Our understanding of the heterogeneity of vascular cells and the molecular mechanisms that regulate their function is beginning to benefit greatly from the rapid development of single cell technologies. Recent studies have started to analyze and map vascular beds in a range of organs in healthy and diseased states at single cell resolution. The current review focuses on recent biological insights on the vascular system garnered from single cell analyses. We cover the themes of vascular heterogeneity, phenotypic plasticity of vascular cells in pathologies such as atherosclerosis and cardiovascular disease, as well as the contribution of defective microvasculature to the development of neurodegenerative disorders such as Alzheimer's disease. Further adaptation of single cell technologies to study the vascular system will be pivotal in uncovering the mechanisms that drive the array of diseases underpinned by vascular dysfunction.


Sujet(s)
Vaisseaux sanguins/cytologie , Inflammation/complications , Analyse sur cellule unique , Maladies vasculaires/étiologie , Animaux , Vaisseaux sanguins/physiologie , Humains
8.
Nat Cell Biol ; 22(7): 828-841, 2020 07.
Article de Anglais | MEDLINE | ID: mdl-32541879

RÉSUMÉ

Mutations in chromatin-modifying complexes and metabolic enzymes commonly underlie complex human developmental syndromes affecting multiple organs. A major challenge is to determine how disease-causing genetic lesions cause deregulation of homeostasis in unique cell types. Here we show that neural-specific depletion of three members of the non-specific lethal (NSL) chromatin complex-Mof, Kansl2 or Kansl3-unexpectedly leads to severe vascular defects and brain haemorrhaging. Deregulation of the epigenetic landscape induced by the loss of the NSL complex in neural cells causes widespread metabolic defects, including an accumulation of free long-chain fatty acids (LCFAs). Free LCFAs induce a Toll-like receptor 4 (TLR4)-NFκB-dependent pro-inflammatory signalling cascade in neighbouring vascular pericytes that is rescued by TLR4 inhibition. Pericytes display functional changes in response to LCFA-induced activation that result in vascular breakdown. Our work establishes that neurovascular function is determined by the neural metabolic environment.


Sujet(s)
Noyau de la cellule/anatomopathologie , Chromatine/métabolisme , Histone acetyltransferases/physiologie , Inflammation/anatomopathologie , Néovascularisation pathologique/anatomopathologie , Neurones/anatomopathologie , Péricytes/anatomopathologie , Animaux , Encéphale/cytologie , Encéphale/métabolisme , Noyau de la cellule/métabolisme , Chromatine/génétique , Acides gras/métabolisme , Femelle , Foetus/cytologie , Foetus/métabolisme , Humains , Inflammation/métabolisme , Mâle , Métabolome , Souris de lignée C57BL , Souris knockout , Néovascularisation pathologique/métabolisme , Neurones/métabolisme , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Péricytes/métabolisme
9.
Nat Commun ; 11(1): 2243, 2020 05 07.
Article de Anglais | MEDLINE | ID: mdl-32382029

RÉSUMÉ

Cells rely on a diverse repertoire of genes for maintaining homeostasis, but the transcriptional networks underlying their expression remain poorly understood. The MOF acetyltransferase-containing Non-Specific Lethal (NSL) complex is a broad transcription regulator. It is essential in Drosophila, and haploinsufficiency of the human KANSL1 subunit results in the Koolen-de Vries syndrome. Here, we perform a genome-wide RNAi screen and identify the BET protein BRD4 as an evolutionary conserved co-factor of the NSL complex. Using Drosophila and mouse embryonic stem cells, we characterise a recruitment hierarchy, where NSL-deposited histone acetylation enables BRD4 recruitment for transcription of constitutively active genes. Transcriptome analyses in Koolen-de Vries patient-derived fibroblasts reveals perturbations with a cellular homeostasis signature that are evoked by the NSL complex/BRD4 axis. We propose that BRD4 represents a conserved bridge between the NSL complex and transcription activation, and provide a new perspective in the understanding of their functions in healthy and diseased states.


Sujet(s)
Histone/métabolisme , Activation de la transcription/physiologie , Acétylation , Animaux , Cellules cultivées , Chromatine/métabolisme , Drosophila , Protéines de Drosophila/génétique , Protéines de Drosophila/métabolisme , Épigénomique , Femelle , Analyse de profil d'expression de gènes , Mâle , Souris , Protéines nucléaires/génétique , Protéines nucléaires/métabolisme , Grossesse , Régions promotrices (génétique)/génétique , Interférence par ARN/physiologie , Activation de la transcription/génétique
10.
iScience ; 21: 273-287, 2019 Nov 22.
Article de Anglais | MEDLINE | ID: mdl-31677479

RÉSUMÉ

Since the generation of cell-type specific knockout models, the importance of inter-cellular communication between neural, vascular, and microglial cells during neural development has been increasingly appreciated. However, the extent of communication between these major cell populations remains to be systematically mapped. Here, we describe EMBRACE (embryonic brain cell extraction using FACS), a method to simultaneously isolate neural, mural, endothelial, and microglial cells to more than 94% purity in ∼4 h. Utilizing EMBRACE we isolate, transcriptionally analyze, and build a cell-cell communication map of the developing mouse brain. We identify 1,710 unique ligand-receptor interactions between neural, endothelial, mural, and microglial cells in silico and experimentally confirm the APOE-LDLR, APOE-LRP1, VTN-KDR, and LAMA4-ITGB1 interactions in the E14.5 brain. We provide our data via the searchable "Brain interactome explorer", available at https://mpi-ie.shinyapps.io/braininteractomeexplorer/. Together, this study provides a comprehensive map that reveals the richness of communication within the developing brain.

11.
EMBO Rep ; 20(7): e47630, 2019 07.
Article de Anglais | MEDLINE | ID: mdl-31267707

RÉSUMÉ

The functionality of chromatin is tightly regulated by post-translational modifications that modulate transcriptional output from target loci. Among the post-translational modifications of chromatin, reversible ε-lysine acetylation of histone proteins is prominent at transcriptionally active genes. Lysine acetylation is catalyzed by lysine acetyltransferases (KATs), which utilize the central cellular metabolite acetyl-CoA as their substrate. Among the KATs that mediate lysine acetylation, males absent on the first (MOF/KAT8) is particularly notable for its ability to acetylate histone 4 lysine 16 (H4K16ac), a modification that decompacts chromatin structure. MOF and its non-specific lethal (NSL) complex members have been shown to localize to gene promoters and enhancers in the nucleus, as well as to microtubules and mitochondria to regulate key cellular processes. Highlighting their importance, mutations or deregulation of NSL complex members has been reported in both human neurodevelopmental disorders and cancer. Based on insight gained from studies in human, mouse, and Drosophila model systems, this review discusses the role of NSL-mediated lysine acetylation in a myriad of cellular functions in both health and disease. Through these studies, the importance of the NSL complex in regulating core transcriptional and signaling networks required for normal development and cellular homeostasis is beginning to emerge.


Sujet(s)
Assemblage et désassemblage de la chromatine , Épigenèse génétique , Histone acetyltransferases/métabolisme , Activation de la transcription , Animaux , Homéostasie , Humains
12.
Blood ; 133(16): 1729-1741, 2019 04 18.
Article de Anglais | MEDLINE | ID: mdl-30755422

RÉSUMÉ

Somatically acquired mutations in PHF6 (plant homeodomain finger 6) frequently occur in hematopoietic malignancies and often coincide with ectopic expression of TLX3. However, there is no functional evidence to demonstrate whether these mutations contribute to tumorigenesis. Similarly, the role of PHF6 in hematopoiesis is unknown. We report here that Phf6 deletion in mice resulted in a reduced number of hematopoietic stem cells (HSCs), an increased number of hematopoietic progenitor cells, and an increased proportion of cycling stem and progenitor cells. Loss of PHF6 caused increased and sustained hematopoietic reconstitution in serial transplantation experiments. Interferon-stimulated gene expression was upregulated in the absence of PHF6 in hematopoietic stem and progenitor cells. The numbers of hematopoietic progenitor cells and cycling hematopoietic stem and progenitor cells were restored to normal by combined loss of PHF6 and the interferon α and ß receptor subunit 1. Ectopic expression of TLX3 alone caused partially penetrant leukemia. TLX3 expression and loss of PHF6 combined caused fully penetrant early-onset leukemia. Our data suggest that PHF6 is a hematopoietic tumor suppressor and is important for fine-tuning hematopoietic stem and progenitor cell homeostasis.


Sujet(s)
Cellules souches hématopoïétiques/cytologie , Protéines à homéodomaine/métabolisme , Leucémies/étiologie , Protéines de répression/physiologie , Animaux , Carcinogenèse , Régulation de l'expression des gènes , Humains , Souris , Souris knockout , Récepteur interféron , Protéines de répression/génétique , Protéines suppresseurs de tumeurs
13.
Nat Rev Genet ; 20(1): 7-23, 2019 01.
Article de Anglais | MEDLINE | ID: mdl-30390049

RÉSUMÉ

Research over the past three decades has firmly established lysine acetyltransferases (KATs) as central players in regulating transcription. Recent advances in genomic sequencing, metabolomics, animal models and mass spectrometry technologies have uncovered unexpected new roles for KATs at the nexus between the environment and transcriptional regulation. Thousands of reversible acetylation sites have been mapped in the proteome that respond dynamically to the cellular milieu and maintain major processes such as metabolism, autophagy and stress response. Concurrently, researchers are continuously uncovering how deregulation of KAT activity drives disease, including cancer and developmental syndromes characterized by severe intellectual disability. These novel findings are reshaping our view of KATs away from mere modulators of chromatin to detectors of the cellular environment and integrators of diverse signalling pathways with the ability to modify cellular phenotype.


Sujet(s)
Chromatine/métabolisme , Lysine acetyltransferases/métabolisme , Maturation post-traductionnelle des protéines/physiologie , Transduction du signal/physiologie , Acétylation , Animaux , Chromatine/génétique , Humains , Lysine acetyltransferases/génétique
14.
Cell Rep ; 24(12): 3285-3295.e4, 2018 09 18.
Article de Anglais | MEDLINE | ID: mdl-30232009

RÉSUMÉ

Apoptotic cell death removes unwanted cells and is regulated by interactions between pro-survival and pro-apoptotic members of the BCL-2 protein family. The regulation of apoptosis is thought to be crucial for normal embryonic development. Accordingly, complete loss of pro-survival MCL-1 or BCL-XL (BCL2L1) causes embryonic lethality. However, it is not known whether minor reductions in pro-survival proteins could cause developmental abnormalities. We explored the rate-limiting roles of MCL-1 and BCL-XL in development and show that combined loss of single alleles of Mcl-1 and Bcl-x causes neonatal lethality. Mcl-1+/-;Bcl-x+/- mice display craniofacial anomalies, but additional loss of a single allele of pro-apoptotic Bim (Bcl2l11) restores normal development. These findings demonstrate that the control of cell survival during embryogenesis is finely balanced and suggest that some human craniofacial defects, for which causes are currently unknown, may be due to subtle imbalances between pro-survival and pro-apoptotic BCL-2 family members.


Sujet(s)
Protéine-11 analogue à Bcl-2/génétique , Malformations crâniofaciales/génétique , Protéine Mcl-1/génétique , Protéine bcl-X/génétique , Animaux , Apoptose , Protéine-11 analogue à Bcl-2/métabolisme , Cellules cultivées , Femelle , Hétérozygote , Mâle , Souris , Souris de lignée C57BL , Protéine Mcl-1/métabolisme , Protéine bcl-X/métabolisme
15.
Nature ; 560(7717): 253-257, 2018 08.
Article de Anglais | MEDLINE | ID: mdl-30069049

RÉSUMÉ

Acetylation of histones by lysine acetyltransferases (KATs) is essential for chromatin organization and function1. Among the genes coding for the MYST family of KATs (KAT5-KAT8) are the oncogenes KAT6A (also known as MOZ) and KAT6B (also known as MORF and QKF)2,3. KAT6A has essential roles in normal haematopoietic stem cells4-6 and is the target of recurrent chromosomal translocations, causing acute myeloid leukaemia7,8. Similarly, chromosomal translocations in KAT6B have been identified in diverse cancers8. KAT6A suppresses cellular senescence through the regulation of suppressors of the CDKN2A locus9,10, a function that requires its KAT activity10. Loss of one allele of KAT6A extends the median survival of mice with MYC-induced lymphoma from 105 to 413 days11. These findings suggest that inhibition of KAT6A and KAT6B may provide a therapeutic benefit in cancer. Here we present highly potent, selective inhibitors of KAT6A and KAT6B, denoted WM-8014 and WM-1119. Biochemical and structural studies demonstrate that these compounds are reversible competitors of acetyl coenzyme A and inhibit MYST-catalysed histone acetylation. WM-8014 and WM-1119 induce cell cycle exit and cellular senescence without causing DNA damage. Senescence is INK4A/ARF-dependent and is accompanied by changes in gene expression that are typical of loss of KAT6A function. WM-8014 potentiates oncogene-induced senescence in vitro and in a zebrafish model of hepatocellular carcinoma. WM-1119, which has increased bioavailability, arrests the progression of lymphoma in mice. We anticipate that this class of inhibitors will help to accelerate the development of therapeutics that target gene transcription regulated by histone acetylation.


Sujet(s)
Benzènesulfonates/pharmacologie , Vieillissement de la cellule/effets des médicaments et des substances chimiques , Histone acetyltransferases/antagonistes et inhibiteurs , Hydrazines/pharmacologie , Lymphomes/traitement médicamenteux , Lymphomes/anatomopathologie , Sulfonamides/pharmacologie , Acétylation/effets des médicaments et des substances chimiques , Animaux , Benzènesulfonates/usage thérapeutique , Prolifération cellulaire/effets des médicaments et des substances chimiques , Cellules cultivées , Développement de médicament , Fibroblastes , Régulation de l'expression des gènes tumoraux/effets des médicaments et des substances chimiques , Histone acetyltransferases/déficit , Histone acetyltransferases/génétique , Histone/composition chimique , Histone/métabolisme , Hydrazines/usage thérapeutique , Lymphomes/enzymologie , Lymphomes/génétique , Lysine/composition chimique , Lysine/métabolisme , Mâle , Souris , Souris de lignée C57BL , Modèles moléculaires , Sulfonamides/usage thérapeutique
16.
Cereb Cortex ; 27(1): 576-588, 2017 01 01.
Article de Anglais | MEDLINE | ID: mdl-26503265

RÉSUMÉ

Mutations of the reelin gene cause severe defects in cerebral cortex development and profound intellectual impairment. While many aspects of the reelin signaling pathway have been identified, the molecular and ultimate cellular consequences of reelin signaling remain unknown. Specifically, it is unclear if termination of reelin signaling is as important for normal cortical neuron migration as activation of reelin signaling. Using mice that are single or double deficient, we discovered that combined loss of the suppressors of cytokine signaling, SOCS6 and SOCS7, recapitulated the cortical layer inversion seen in mice lacking reelin and led to a dramatic increase in the reelin signaling molecule disabled (DAB1) in the cortex. The SRC homology domains of SOCS6 and SOCS7 bound DAB1 ex vivo. Mutation of DAB1 greatly diminished binding and protected from degradation by SOCS6. Phosphorylated DAB1 was elevated in cortical neurons in the absence of SOCS6 and SOCS7. Thus, constitutive activation of reelin signaling was observed to be equally detrimental as lack of activation. We hypothesize that, by terminating reelin signaling, SOCS6 and SOCS7 may allow new cycles of reelin signaling to occur and that these may be essential for cortical neuron migration.


Sujet(s)
Molécules d'adhérence cellulaire neuronale/métabolisme , Cortex cérébral/embryologie , Cortex cérébral/métabolisme , Protéines de la matrice extracellulaire/métabolisme , Protéines de tissu nerveux/métabolisme , Serine endopeptidases/métabolisme , Protéines SOCS/déficit , Animaux , Molécules d'adhérence cellulaire neuronale/génétique , Mouvement cellulaire/physiologie , Cortex cérébral/anatomopathologie , Protéines de la matrice extracellulaire/génétique , Cellules HEK293 , Humains , Souris de lignée BALB C , Souris de lignée C57BL , Souris knockout , Protéines de tissu nerveux/génétique , Neurones/métabolisme , Phosphorylation , Protéine reeline , Serine endopeptidases/génétique , Protéines SOCS/génétique
17.
Exp Hematol ; 47: 83-97.e8, 2017 03.
Article de Anglais | MEDLINE | ID: mdl-27773671

RÉSUMÉ

Chromatin plays a central role in maintaining hematopoietic stem cells and during their stepwise differentiation. Although a large number of histone modifications and chromatin-modifying enzymes have been identified, how these act in concert to produce specific phenotypic outcomes remains to be established. MOZ (KAT6A) is a lysine acetyltransferase and enhances transcription at target gene loci. In contrast, the Polycomb group protein BMI1 (PCGF4) is part of the transcriptionally repressive PRC1 complex. Despite their opposing effects on transcription, MOZ and BMI1 regulate biological systems in a similar manner. MOZ and BMI1 are required for the development of transplantable HSCs, for restraining cellular senescence, for the proper patterning of the anterior-posterior axis during development and for the specification and maintenance of the B-cell lineage. Thus, we set out to explore the relationship between MOZ and BMI1. We recently established that MOZ and BMI1 have opposing effects on the initiation of Hox gene expression during embryonic development and that defects in body segment identity specification observed in single Moz and Bmi1 mutants were rescued in compound mutants. We report here the relationship between MOZ and BMI1 in hematopoiesis. Using Moz+/-;Bmi1+/- compound mutant mice, we found that MOZ and BMI1, but not the BMI1-related protein MEL18 (PCGF2), play synergistic roles in maintaining adult HSCs. Although BMI1 restrains premature senescence, we established that MOZ acts to maintain the quiescent state of HSCs. Our work revealed that MOZ and BMI1 regulate HSCs in a synergistic manner by acting on distinct processes required to maintain HSCs.


Sujet(s)
Auto-renouvellement cellulaire/génétique , Épistasie , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Histone acetyltransferases/génétique , Complexe répresseur Polycomb-1/génétique , Protéines proto-oncogènes/génétique , Animaux , Marqueurs biologiques , Transplantation de moelle osseuse , Différenciation cellulaire/génétique , Inhibiteur p16 de kinase cycline-dépendante/métabolisme , Dosage génique , Génotype , Immunophénotypage , Souris , Souris knockout , Phénotype
18.
Blood ; 128(19): 2307-2318, 2016 11 10.
Article de Anglais | MEDLINE | ID: mdl-27663673

RÉSUMÉ

Hematopoietic stem cells (HSCs) are conventionally thought to be at the apex of a hierarchy that produces all mature cells of the blood. The quintessential property of these cells is their ability to reconstitute the entire hematopoietic system of hemoablated recipients. This characteristic has enabled HSCs to be used to replenish the hematopoietic system of patients after chemotherapy or radiotherapy. Here, we use deletion of the monocytic leukemia zinc finger gene (Moz/Kat6a/Myst3) to examine the effects of removing HSCs. Loss of MOZ in adult mice leads to the rapid loss of HSCs as defined by transplantation. This is accompanied by a reduction of the LSK-CD48-CD150+ and LSK-CD34-Flt3- populations in the bone marrow and a reduction in quiescent cells in G0 Surprisingly, the loss of classically defined HSCs did not affect mouse viability, and there was no recovery of the LSK-CD48-CD150+ and LSK-CD34-Flt3- populations 15 to 18 months after Moz deletion. Clonal analysis of myeloid progenitors, which produce short-lived granulocytes, demonstrate that these are derived from cells that had undergone recombination at the Moz locus up to 2 years earlier, suggesting that early progenitors have acquired extended self-renewal. Our results establish that there are essential differences in HSC requirement for steady-state blood cell production compared with the artificial situation of reconstitution after transplantation into a hemoablated host. A better understanding of steady-state hematopoiesis may facilitate the development of novel therapies engaging hematopoietic cell populations with previously unrecognized traits, as well as characterizing potential vulnerability to oncogenic transformation.


Sujet(s)
Cellules souches adultes/cytologie , Cellules souches adultes/métabolisme , Cellules souches hématopoïétiques/cytologie , Cellules souches hématopoïétiques/métabolisme , Histone acetyltransferases/métabolisme , Animaux , Marqueurs biologiques/métabolisme , Cellules de la moelle osseuse/anatomopathologie , Numération cellulaire , Différenciation cellulaire , Vieillissement de la cellule , Test clonogénique , Délétion de gène , Integrases/métabolisme , Souris de lignée C57BL , Phénotype , Phase G0 , Transplantation de cellules souches
19.
Proc Natl Acad Sci U S A ; 112(17): 5437-42, 2015 Apr 28.
Article de Anglais | MEDLINE | ID: mdl-25922517

RÉSUMÉ

Hox genes underlie the specification of body segment identity in the anterior-posterior axis. They are activated during gastrulation and undergo a dynamic shift from a transcriptionally repressed to an active chromatin state in a sequence that reflects their chromosomal location. Nevertheless, the precise role of chromatin modifying complexes during the initial activation phase remains unclear. In the current study, we examined the role of chromatin regulators during Hox gene activation. Using embryonic stem cell lines lacking the transcriptional activator MOZ and the polycomb-family repressor BMI1, we showed that MOZ and BMI1, respectively, promoted and repressed Hox genes during the shift from the transcriptionally repressed to the active state. Strikingly however, MOZ but not BMI1 was required to regulate Hox mRNA levels after the initial activation phase. To determine the interaction of MOZ and BMI1 in vivo, we interrogated their role in regulating Hox genes and body segment identity using Moz;Bmi1 double deficient mice. We found that the homeotic transformations and shifts in Hox gene expression boundaries observed in single Moz and Bmi1 mutant mice were rescued to a wild type identity in Moz;Bmi1 double knockout animals. Together, our findings establish that MOZ and BMI1 play opposing roles during the onset of Hox gene expression in the ES cell model and during body segment identity specification in vivo. We propose that chromatin-modifying complexes have a previously unappreciated role during the initiation phase of Hox gene expression, which is critical for the correct specification of body segment identity.


Sujet(s)
Plan d'organisation du corps/physiologie , Embryon de mammifère/embryologie , Cellules souches embryonnaires/métabolisme , Histone acetyltransferases/métabolisme , Protéines à homéodomaine/biosynthèse , Complexe répresseur Polycomb-1/métabolisme , Protéines proto-oncogènes/métabolisme , Animaux , Embryon de mammifère/cytologie , Cellules souches embryonnaires/cytologie , Régulation de l'expression des gènes au cours du développement/physiologie , Histone acetyltransferases/génétique , Protéines à homéodomaine/génétique , Souris , Souris de lignée BALB C , Souris knockout , Complexe répresseur Polycomb-1/génétique , Protéines proto-oncogènes/génétique
20.
Blood ; 125(12): 1910-21, 2015 Mar 19.
Article de Anglais | MEDLINE | ID: mdl-25605372

RÉSUMÉ

The histone acetyltransferase MOZ (MYST3, KAT6A) is the target of recurrent chromosomal translocations fusing the MOZ gene to CBP, p300, NCOA3, or TIF2 in particularly aggressive cases of acute myeloid leukemia. In this study, we report the role of wild-type MOZ in regulating B-cell progenitor proliferation and hematopoietic malignancy. In the Eµ-Myc model of aggressive pre-B/B-cell lymphoma, the loss of just one allele of Moz increased the median survival of mice by 3.9-fold. MOZ was required to maintain the proliferative capacity of B-cell progenitors, even in the presence of c-MYC overexpression, by directly maintaining the transcriptional activity of genes required for normal B-cell development. Hence, B-cell progenitor numbers were significantly reduced in Moz haploinsufficient animals. Interestingly, we find a significant overlap in genes regulated by MOZ, mixed lineage leukemia 1, and mixed lineage leukemia 1 cofactor menin. This includes Meis1, a TALE class homeobox transcription factor required for B-cell development, characteristically upregulated as a result of MLL1 translocations in leukemia. We demonstrate that MOZ localizes to the Meis1 locus in pre-B-cells and maintains Meis1 expression. Our results suggest that even partial inhibition of MOZ may reduce the proliferative capacity of MEIS1, and HOX-driven lymphoma and leukemia cells.


Sujet(s)
Lymphocytes B/cytologie , Régulation de l'expression des gènes tumoraux , Histone acetyltransferases/génétique , Lymphomes/métabolisme , Protéines proto-oncogènes c-myc/métabolisme , Cellules souches/cytologie , Allèles , Animaux , Différenciation cellulaire , Survie cellulaire , Cellules cultivées , Vieillissement de la cellule , Femelle , Haploinsuffisance , Souris , Souris de lignée C57BL , Souris transgéniques , Analyse de séquence d'ARN , Transcription génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...