Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Nat Immunol ; 16(5): 505-16, 2015 May.
Article in English | MEDLINE | ID: mdl-25751747

ABSTRACT

A cytosolic role for the histone methyltransferase Ezh2 in regulating lymphocyte activation has been suggested, but the molecular mechanisms underpinning this extranuclear function have remained unclear. Here we found that Ezh2 regulated the integrin signaling and adhesion dynamics of neutrophils and dendritic cells (DCs). Ezh2 deficiency impaired the integrin-dependent transendothelial migration of innate leukocytes and restricted disease progression in an animal model of multiple sclerosis. Direct methylation of talin, a key regulatory molecule in cell migration, by Ezh2 disrupted the binding of talin to F-actin and thereby promoted the turnover of adhesion structures. This regulatory effect was abolished by targeted disruption of the interactions of Ezh2 with the cytoskeletal-reorganization effector Vav1. Our studies reveal an unforeseen extranuclear function for Ezh2 in regulating adhesion dynamics, with implications for leukocyte migration, immune responses and potentially pathogenic processes.


Subject(s)
Cell Nucleus/metabolism , Dendritic Cells/immunology , Encephalomyelitis, Autoimmune, Experimental/immunology , Multiple Sclerosis/immunology , Neutrophils/immunology , Polycomb Repressive Complex 2/metabolism , Talin/metabolism , Actins/metabolism , Animals , Cell Adhesion/genetics , Cell Movement , Cells, Cultured , Disease Models, Animal , Enhancer of Zeste Homolog 2 Protein , Humans , Lymphocyte Activation/genetics , Methylation , Mice , Mice, Knockout , Polycomb Repressive Complex 2/genetics , Protein Binding/genetics , Proto-Oncogene Proteins c-vav/metabolism , Talin/genetics , Transendothelial and Transepithelial Migration/genetics
2.
Blood ; 139(3): 343-356, 2022 01 20.
Article in English | MEDLINE | ID: mdl-34517413

ABSTRACT

In vitro generation and expansion of hematopoietic stem cells (HSCs) holds great promise for the treatment of any ailment that relies on bone marrow or blood transplantation. To achieve this, it is essential to resolve the molecular and cellular pathways that govern HSC formation in the embryo. HSCs first emerge in the aorta-gonad-mesonephros (AGM) region, where a rare subset of endothelial cells, hemogenic endothelium (HE), undergoes an endothelial-to-hematopoietic transition (EHT). Here, we present full-length single-cell RNA sequencing (scRNA-seq) of the EHT process with a focus on HE and dorsal aorta niche cells. By using Runx1b and Gfi1/1b transgenic reporter mouse models to isolate HE, we uncovered that the pre-HE to HE continuum is specifically marked by angiotensin-I converting enzyme (ACE) expression. We established that HE cells begin to enter the cell cycle near the time of EHT initiation when their morphology still resembles endothelial cells. We further demonstrated that RUNX1 AGM niche cells consist of vascular smooth muscle cells and PDGFRa+ mesenchymal cells and can functionally support hematopoiesis. Overall, our study provides new insights into HE differentiation toward HSC and the role of AGM RUNX1+ niche cells in this process. Our expansive scRNA-seq datasets represents a powerful resource to investigate these processes further.


Subject(s)
Embryo, Mammalian/embryology , Hemangioblasts/cytology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Animals , Cell Differentiation , Embryo, Mammalian/cytology , Embryo, Mammalian/metabolism , Hemangioblasts/metabolism , Hematopoietic Stem Cells/metabolism , Mesonephros/cytology , Mesonephros/embryology , Mesonephros/metabolism , Mice , Single-Cell Analysis , Transcriptome , Zebrafish
3.
Blood ; 131(20): 2223-2234, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29555646

ABSTRACT

Despite the well-established cell-intrinsic role of epigenetic factors in normal and malignant hematopoiesis, their cell-extrinsic role remains largely unexplored. Herein we investigated the hematopoietic impact of inactivating Ezh2, a key component of polycomb repressive complex 2 (PRC2), in the fetal liver (FL) vascular niche. Hematopoietic specific (Vav-iCre) Ezh2 inactivation enhanced FL hematopoietic stem cell (HSC) expansion with normal FL erythropoiesis. In contrast, endothelium (Tie2-Cre) targeted Ezh2 inactivation resulted in embryonic lethality with severe anemia at embryonic day 13.5 despite normal emergence of functional HSCs. Ezh2-deficient FL endothelium overexpressed Mmp9, which cell-extrinsically depleted the membrane-bound form of Kit ligand (mKitL), an essential hematopoietic cytokine, in FL. Furthermore, Mmp9 inhibition in vitro restored mKitL expression along with the erythropoiesis supporting capacity of FL endothelial cells. These data establish that Ezh2 is intrinsically dispensable for FL HSCs and provides proof of principle that modulation of epigenetic regulators in niche components can exert a marked cell-extrinsic impact.


Subject(s)
Endothelial Cells/cytology , Endothelial Cells/metabolism , Enhancer of Zeste Homolog 2 Protein/genetics , Fetus , Hematopoiesis, Extramedullary , Liver/physiology , Anemia/genetics , Anemia/metabolism , Animals , Biomarkers , Cells, Cultured , Enhancer of Zeste Homolog 2 Protein/metabolism , Fluorescent Antibody Technique , Gene Expression , Gene Silencing , Hematopoiesis, Extramedullary/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Immunohistochemistry , Matrix Metalloproteinase 9/genetics , Matrix Metalloproteinase 9/metabolism , Mice , Phenotype , Receptor, TIE-2/genetics , Receptor, TIE-2/metabolism , Stem Cell Factor/metabolism
4.
Haematologica ; 104(11): 2215-2224, 2019 11.
Article in English | MEDLINE | ID: mdl-30975913

ABSTRACT

Somatic mutations in acute myeloid leukemia are acquired sequentially and hierarchically. First, pre-leukemic mutations, such as t(8;21) that encodes AML1-ETO, are acquired within the hematopoietic stem cell (HSC) compartment, while signaling pathway mutations, including KRAS activating mutations, are late events acquired during transformation of leukemic progenitor cells and are rarely detectable in HSC. This raises the possibility that signaling pathway mutations are detrimental to clonal expansion of pre-leukemic HSC. To address this hypothesis, we used conditional genetics to introduce Aml1-ETO and K-RasG12D into murine HSC, either individually or in combination. In the absence of activated Ras, Aml1-ETO-expressing HSC conferred a competitive advantage. However, activated K-Ras had a marked detrimental effect on Aml1-ETO-expressing HSC, leading to loss of both phenotypic and functional HSC. Cell cycle analysis revealed a loss of quiescence in HSC co-expressing Aml1-ETO and K-RasG12D, accompanied by an enrichment in E2F and Myc target gene expression and depletion of HSC self-renewal-associated gene expression. These findings provide a mechanistic basis for the observed absence of KRAS signaling mutations in the pre-malignant HSC compartment.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Core Binding Factor Alpha 2 Subunit/genetics , Core Binding Factor Alpha 2 Subunit/metabolism , Hematopoietic Stem Cells/metabolism , Mutation , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , RUNX1 Translocation Partner 1 Protein/genetics , RUNX1 Translocation Partner 1 Protein/metabolism , Animals , Cell Proliferation/genetics , Gene Expression , Gene Expression Profiling , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, Transgenic , Models, Animal , Models, Biological , Precancerous Conditions/genetics , Precancerous Conditions/metabolism
5.
J Biol Chem ; 289(46): 31693-31707, 2014 Nov 14.
Article in English | MEDLINE | ID: mdl-25266721

ABSTRACT

The polycomb group protein Ezh2 is a histone methyltransferase that modifies chromatin structure to alter gene expression during embryonic development, lymphocyte activation, and tumorigenesis. The mechanism by which Ezh2 expression is regulated is not well defined. In the current study, we report that c-Rel is a critical activator of Ezh2 transcription in lymphoid cells. In activated primary murine B and T cells, plus human leukemia and multiple myeloma cell lines, recruitment of c-Rel to the first intron of the Ezh2 locus promoted Ezh2 mRNA expression. This up-regulation was abolished in activated c-Rel-deficient lymphocytes and by c-Rel knockdown in Jurkat T cells. Treatment of malignant cells with the c-Rel inhibitor pentoxifylline not only reduced c-Rel nuclear translocation and Ezh2 expression, but also enhanced their sensitivity to the Ezh2-specific drug, GSK126 through increased growth inhibition and cell death. In summary, our demonstration that c-Rel regulates Ezh2 expression in lymphocytes and malignant lymphoid cells reveals a novel transcriptional network in transformed lymphoid cells expressing high levels of Ezh2 that provides a molecular justification for combinatorial drug therapy.


Subject(s)
Gene Expression Regulation, Neoplastic , Polycomb Repressive Complex 2/metabolism , Proto-Oncogene Proteins c-rel/metabolism , Animals , Base Sequence , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Survival , Enhancer of Zeste Homolog 2 Protein , HEK293 Cells , Humans , Indoles/chemistry , Jurkat Cells , Lymphocyte Activation , Lymphocytes/cytology , Mice , Mice, Inbred C57BL , Molecular Sequence Data , Neoplasms/metabolism , Pentoxifylline/chemistry , Pyridones/chemistry , Transcription, Genetic , Up-Regulation
6.
J Biol Chem ; 289(30): 20788-801, 2014 Jul 25.
Article in English | MEDLINE | ID: mdl-24878960

ABSTRACT

Polycomb group protein Ezh2 is a histone H3 Lys-27 histone methyltransferase orchestrating an extensive epigenetic regulatory program. Several nervous system-specific genes are known to be repressed by Ezh2 in stem cells and derepressed during neuronal differentiation. However, the molecular mechanisms underlying this regulation remain poorly understood. Here we show that Ezh2 levels are dampened during neuronal differentiation by brain-enriched microRNA miR-124. Expression of miR-124 in a neuroblastoma cells line was sufficient to up-regulate a significant fraction of nervous system-specific Ezh2 target genes. On the other hand, naturally elevated expression of miR-124 in embryonic carcinoma cells undergoing neuronal differentiation correlated with down-regulation of Ezh2 levels. Importantly, overexpression of Ezh2 mRNA with a 3'-untranslated region (3'-UTR) lacking a functional miR-124 binding site, but not with the wild-type Ezh2 3'-UTR, hampered neuronal and promoted astrocyte-specific differentiation in P19 and embryonic mouse neural stem cells. Overall, our results uncover a molecular mechanism that allows miR-124 to balance the choice between alternative differentiation possibilities through fine-tuning the expression of a critical epigenetic regulator.


Subject(s)
Astrocytes/metabolism , Cell Differentiation/physiology , Gene Expression Regulation/physiology , MicroRNAs/metabolism , Neurons/metabolism , Polycomb Repressive Complex 2/biosynthesis , 3' Untranslated Regions/physiology , Animals , Astrocytes/cytology , Cell Line, Tumor , Enhancer of Zeste Homolog 2 Protein , HEK293 Cells , Humans , Mice , MicroRNAs/genetics , Neurons/cytology , Polycomb Repressive Complex 2/genetics
7.
Nat Commun ; 15(1): 1604, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-38383534

ABSTRACT

Hematopoietic stem cells (HSCs) develop from the hemogenic endothelium (HE) in the aorta- gonads-and mesonephros (AGM) region and reside within Intra-aortic hematopoietic clusters (IAHC) along with hematopoietic progenitors (HPC). The signalling mechanisms that distinguish HSCs from HPCs are unknown. Notch signaling is essential for arterial specification, IAHC formation and HSC activity, but current studies on how Notch segregates these different fates are inconsistent. We now demonstrate that Notch activity is highest in a subset of, GFI1 + , HSC-primed HE cells, and is gradually lost with HSC maturation. We uncover that the HSC phenotype is maintained due to increasing levels of NOTCH1 and JAG1 interactions on the surface of the same cell (cis) that renders the NOTCH1 receptor from being activated. Forced activation of the NOTCH1 receptor in IAHC activates a hematopoietic differentiation program. Our results indicate that NOTCH1-JAG1 cis-inhibition preserves the HSC phenotype in the hematopoietic clusters of the embryonic aorta.


Subject(s)
Hematopoietic Stem Cells , Receptor, Notch1 , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Hematopoietic Stem Cells/metabolism , Cell Differentiation/genetics , Aorta/metabolism , Arteries/metabolism , Mesonephros , Gonads/metabolism
8.
Nat Commun ; 15(1): 4673, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38824124

ABSTRACT

Recent findings suggest that Hematopoietic Stem Cells (HSC) and progenitors arise simultaneously and independently of each other already in the embryonic aorta-gonad mesonephros region, but it is still unknown how their different features are established. Here, we uncover IκBα (Nfkbia, the inhibitor of NF-κB) as a critical regulator of HSC proliferation throughout development. IκBα balances retinoic acid signaling levels together with the epigenetic silencer, PRC2, specifically in HSCs. Loss of IκBα decreases proliferation of HSC and induces a dormancy related gene expression signature instead. Also, IκBα deficient HSCs respond with superior activation to in vitro culture and in serial transplantation. At the molecular level, chromatin regions harboring binding motifs for retinoic acid signaling are hypo-methylated for the PRC2 dependent H3K27me3 mark in IκBα deficient HSCs. Overall, we show that the proliferation index in the developing HSCs is regulated by a IκBα-PRC2 axis, which controls retinoic acid signaling.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells , NF-KappaB Inhibitor alpha , Signal Transduction , Tretinoin , Animals , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Tretinoin/metabolism , NF-KappaB Inhibitor alpha/metabolism , NF-KappaB Inhibitor alpha/genetics , Mice , Embryonic Development/genetics , Mice, Knockout , Polycomb Repressive Complex 2/metabolism , Polycomb Repressive Complex 2/genetics , Mice, Inbred C57BL , Gene Expression Regulation, Developmental , Female
9.
Front Cell Dev Biol ; 9: 631699, 2021.
Article in English | MEDLINE | ID: mdl-33681211

ABSTRACT

During ontogeny, the establishment of the hematopoietic system takes place in several phases, separated both in time and location. The process is initiated extra-embryonically in the yolk sac (YS) and concludes in the main arteries of the embryo with the formation of hematopoietic stem cells (HSC). Initially, it was thought that HSC-independent hematopoietic YS cells were transient, and only required to bridge the gap to HSC activity. However, in recent years it has become clear that these cells also contribute to embryonic organogenesis, including the emergence of HSCs. Furthermore, some of these early HSC-independent YS cells persist into adulthood as distinct hematopoietic populations. These previously unrecognized abilities of embryonic HSC-independent hematopoietic cells constitute a new field of interest. Here, we aim to provide a succinct overview of the current knowledge regarding the contribution of YS-derived hematopoietic cells to the development of the embryo and the adult hematopoietic system.

10.
Nat Commun ; 12(1): 7019, 2021 12 02.
Article in English | MEDLINE | ID: mdl-34857757

ABSTRACT

Yolk sac (YS) hematopoiesis is critical for the survival of the embryo and a major source of tissue-resident macrophages that persist into adulthood. Yet, the transcriptional and epigenetic regulation of YS hematopoiesis remains poorly characterized. Here we report that the epigenetic regulator Ezh2 is essential for YS hematopoiesis but dispensable for subsequent aorta-gonad-mesonephros (AGM) blood development. Loss of EZH2 activity in hemogenic endothelium (HE) leads to the generation of phenotypically intact but functionally deficient erythro-myeloid progenitors (EMPs), while the generation of primitive erythroid cells is not affected. EZH2 activity is critical for the generation of functional EMPs at the onset of the endothelial-to-hematopoietic transition but subsequently dispensable. We identify a lack of Wnt signaling downregulation as the primary reason for the production of non-functional EMPs. Together, our findings demonstrate a critical and stage-specific role of Ezh2 in modulating Wnt signaling during the generation of EMPs from YS HE.


Subject(s)
Enhancer of Zeste Homolog 2 Protein/genetics , Erythroid Cells/metabolism , Gene Expression Regulation, Developmental , Mouse Embryonic Stem Cells/metabolism , Myeloid Progenitor Cells/metabolism , Vesicular Transport Proteins/genetics , Yolk Sac/metabolism , Animals , Cell Differentiation , Embryo, Mammalian , Enhancer of Zeste Homolog 2 Protein/deficiency , Epigenesis, Genetic , Erythroid Cells/cytology , Female , Fetus , Genes, Reporter , Hematopoiesis/genetics , Liver/cytology , Liver/growth & development , Liver/metabolism , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Mouse Embryonic Stem Cells/cytology , Myeloid Progenitor Cells/pathology , Primary Cell Culture , Vesicular Transport Proteins/metabolism , Wnt Signaling Pathway , Yolk Sac/cytology , Yolk Sac/growth & development , Red Fluorescent Protein
11.
Cancer Cell ; 33(2): 274-291.e8, 2018 02 12.
Article in English | MEDLINE | ID: mdl-29438697

ABSTRACT

Lympho-myeloid restricted early thymic progenitors (ETPs) are postulated to be the cell of origin for ETP leukemias, a therapy-resistant leukemia associated with frequent co-occurrence of EZH2 and RUNX1 inactivating mutations, and constitutively activating signaling pathway mutations. In a mouse model, we demonstrate that Ezh2 and Runx1 inactivation targeted to early lymphoid progenitors causes a marked expansion of pre-leukemic ETPs, showing transcriptional signatures characteristic of ETP leukemia. Addition of a RAS-signaling pathway mutation (Flt3-ITD) results in an aggressive leukemia co-expressing myeloid and lymphoid genes, which can be established and propagated in vivo by the expanded ETPs. Both mouse and human ETP leukemias show sensitivity to BET inhibition in vitro and in vivo, which reverses aberrant gene expression induced by Ezh2 inactivation.


Subject(s)
Core Binding Factor Alpha 2 Subunit/genetics , Enhancer of Zeste Homolog 2 Protein/genetics , Leukemia, Myeloid, Acute/genetics , Mutation/genetics , Animals , Gene Expression Regulation, Leukemic , Mice, Knockout , Myeloid Cells/metabolism , Signal Transduction/genetics , Stem Cells
12.
J Exp Med ; 214(7): 2005-2021, 2017 Jul 03.
Article in English | MEDLINE | ID: mdl-28637883

ABSTRACT

Although previous studies suggested that the expression of FMS-like tyrosine kinase 3 (Flt3) initiates downstream of mouse hematopoietic stem cells (HSCs), FLT3 internal tandem duplications (FLT3 ITDs) have recently been suggested to intrinsically suppress HSCs. Herein, single-cell interrogation found Flt3 mRNA expression to be absent in the large majority of phenotypic HSCs, with a strong negative correlation between Flt3 and HSC-associated gene expression. Flt3-ITD knock-in mice showed reduced numbers of phenotypic HSCs, with an even more severe loss of long-term repopulating HSCs, likely reflecting the presence of non-HSCs within the phenotypic HSC compartment. Competitive transplantation experiments established that Flt3-ITD compromises HSCs through an extrinsically mediated mechanism of disrupting HSC-supporting bone marrow stromal cells, with reduced numbers of endothelial and mesenchymal stromal cells showing increased inflammation-associated gene expression. Tumor necrosis factor (TNF), a cell-extrinsic potent negative regulator of HSCs, was overexpressed in bone marrow niche cells from FLT3-ITD mice, and anti-TNF treatment partially rescued the HSC phenotype. These findings, which establish that Flt3-ITD-driven myeloproliferation results in cell-extrinsic suppression of the normal HSC reservoir, are of relevance for several aspects of acute myeloid leukemia biology.


Subject(s)
Cell Proliferation/genetics , Hematopoietic Stem Cells/metabolism , Mutation , Stem Cell Niche/genetics , fms-Like Tyrosine Kinase 3/genetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cells, Cultured , Etanercept/pharmacology , Gene Expression Profiling/methods , Hematopoietic Stem Cells/drug effects , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Reverse Transcriptase Polymerase Chain Reaction , Single-Cell Analysis/methods , Tandem Repeat Sequences/genetics , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL