Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Cell ; 170(6): 1079-1095.e20, 2017 Sep 07.
Article in English | MEDLINE | ID: mdl-28823558

ABSTRACT

Loss-of-function mutations in TET2 occur frequently in patients with clonal hematopoiesis, myelodysplastic syndrome (MDS), and acute myeloid leukemia (AML) and are associated with a DNA hypermethylation phenotype. To determine the role of TET2 deficiency in leukemia stem cell maintenance, we generated a reversible transgenic RNAi mouse to model restoration of endogenous Tet2 expression. Tet2 restoration reverses aberrant hematopoietic stem and progenitor cell (HSPC) self-renewal in vitro and in vivo. Treatment with vitamin C, a co-factor of Fe2+ and α-KG-dependent dioxygenases, mimics TET2 restoration by enhancing 5-hydroxymethylcytosine formation in Tet2-deficient mouse HSPCs and suppresses human leukemic colony formation and leukemia progression of primary human leukemia PDXs. Vitamin C also drives DNA hypomethylation and expression of a TET2-dependent gene signature in human leukemia cell lines. Furthermore, TET-mediated DNA oxidation induced by vitamin C treatment in leukemia cells enhances their sensitivity to PARP inhibition and could provide a safe and effective combination strategy to selectively target TET deficiency in cancer. PAPERCLIP.


Subject(s)
Ascorbic Acid/pharmacology , DNA-Binding Proteins/metabolism , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Proto-Oncogene Proteins/metabolism , Vitamins/pharmacology , Animals , Ascorbic Acid/administration & dosage , Cell Death , Cell Line, Tumor , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , Gene Knockdown Techniques , Humans , Leukemia, Myeloid, Acute/genetics , Mice , Myelodysplastic Syndromes/genetics , Neoplasm Transplantation , Poly (ADP-Ribose) Polymerase-1/genetics , Proto-Oncogene Proteins/genetics , Transcription, Genetic , Transplantation, Heterologous , Vitamins/administration & dosage
2.
Nat Immunol ; 16(6): 653-62, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25867473

ABSTRACT

The methylcytosine dioxygenase TET1 ('ten-eleven translocation 1') is an important regulator of 5-hydroxymethylcytosine (5hmC) in embryonic stem cells. The diminished expression of TET proteins and loss of 5hmC in many tumors suggests a critical role for the maintenance of this epigenetic modification. Here we found that deletion of Tet1 promoted the development of B cell lymphoma in mice. TET1 was required for maintenance of the normal abundance and distribution of 5hmC, which prevented hypermethylation of DNA, and for regulation of the B cell lineage and of genes encoding molecules involved in chromosome maintenance and DNA repair. Whole-exome sequencing of TET1-deficient tumors revealed mutations frequently found in non-Hodgkin B cell lymphoma (B-NHL), in which TET1 was hypermethylated and transcriptionally silenced. Our findings provide in vivo evidence of a function for TET1 as a tumor suppressor of hematopoietic malignancy.


Subject(s)
B-Lymphocytes/physiology , Cytosine/analogs & derivatives , DNA-Binding Proteins/metabolism , Embryonic Stem Cells/physiology , Lymphoma, B-Cell/genetics , Proto-Oncogene Proteins/metabolism , Tumor Suppressor Proteins/metabolism , 5-Methylcytosine/analogs & derivatives , Animals , Cell Differentiation/genetics , Cell Lineage/genetics , Chromosomal Instability , Cytosine/metabolism , DNA Methylation , DNA Repair , DNA-Binding Proteins/genetics , Epigenesis, Genetic , Exome/genetics , Gene Expression Profiling , Humans , Mice , Mutation/genetics , Proto-Oncogene Proteins/genetics , Tumor Suppressor Proteins/genetics
3.
Immunity ; 42(4): 613-26, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25862091

ABSTRACT

Epigenetic regulation of lineage-specific genes is important for the differentiation and function of T cells. Ten-eleven translocation (Tet) proteins catalyze 5-methylcytosine (5 mC) conversion to 5-hydroxymethylcytosine (5 hmC) to mediate DNA demethylation. However, the roles of Tet proteins in the immune response are unknown. Here, we characterized the genome-wide distribution of 5 hmC in CD4(+) T cells and found that 5 hmC marks putative regulatory elements in signature genes associated with effector cell differentiation. Moreover, Tet2 protein was recruited to 5 hmC-containing regions, dependent on lineage-specific transcription factors. Deletion of Tet2 in T cells decreased their cytokine expression, associated with reduced p300 recruitment. In vivo, Tet2 plays a critical role in the control of cytokine gene expression in autoimmune disease. Collectively, our findings suggest that Tet2 promotes DNA demethylation and activation of cytokine gene expression in T cells.


Subject(s)
Cytokines/biosynthesis , DNA-Binding Proteins/immunology , Epigenesis, Genetic/immunology , Proto-Oncogene Proteins/immunology , Th1 Cells/immunology , Th17 Cells/immunology , 5-Methylcytosine/analogs & derivatives , Animals , Cell Differentiation , Cytokines/immunology , Cytosine/analogs & derivatives , Cytosine/immunology , Cytosine/metabolism , DNA/immunology , DNA/metabolism , DNA Methylation , DNA-Binding Proteins/genetics , Dioxygenases , E1A-Associated p300 Protein/genetics , E1A-Associated p300 Protein/immunology , Gene Expression Regulation , Genome , Humans , Mice , Mice, Transgenic , Proto-Oncogene Proteins/genetics , STAT4 Transcription Factor/genetics , STAT4 Transcription Factor/immunology , T-Box Domain Proteins/genetics , T-Box Domain Proteins/immunology , Th1 Cells/cytology , Th1 Cells/enzymology , Th17 Cells/cytology , Th17 Cells/enzymology
5.
FASEB J ; 35(1): e21161, 2021 01.
Article in English | MEDLINE | ID: mdl-33156577

ABSTRACT

Association of G protein-coupled receptors into heterodimeric complexes has been reported for over 50 receptor pairs in vitro but functional in vivo validation remains a challenge. Our recent in vitro studies defined the functional fingerprint of heteromers composed of Gi -coupled melatonin MT2 receptors and Gq -coupled serotonin 5-HT2C receptors, in which melatonin transactivates phospholipase C (PLC) through 5-HT2C . Here, we identified this functional fingerprint in the mouse brain. Gq protein activation was probed by [35 S]GTPγS incorporation followed by Gq immunoprecipitation, and PLC activation by determining the inositol phosphate levels in brain lysates of animals previously treated with melatonin. Melatonin concentration-dependently activated Gq proteins and PLC in the hypothalamus and cerebellum but not in cortex. These effects were inhibited by the 5-HT2C receptor-specific inverse agonist SB-243213, and were absent in MT2 and 5-HT2C knockout mice, fully recapitulating previous in vitro data and indicating the involvement of MT2 /5-HT2C heteromers. The antidepressant agomelatine had a similar effect than melatonin when applied alone but blocked the melatonin-promoted Gq activation due to its 5-HT2C antagonistic component. Collectively, we provide strong functional evidence for the existence of MT2 /5-HT2C heteromeric complexes in mouse brain. These heteromers might participate in the in vivo effects of agomelatine.


Subject(s)
Brain/metabolism , Gene Expression Regulation, Enzymologic , Protein Multimerization , Receptor, Melatonin, MT2/metabolism , Receptor, Serotonin, 5-HT2C/metabolism , Transcriptional Activation , Type C Phospholipases/biosynthesis , Acetamides/pharmacology , Animals , Indoles/pharmacology , Male , Mice , Mice, Knockout , Pyridines/pharmacology , Receptor, Melatonin, MT2/genetics , Receptor, Serotonin, 5-HT2C/genetics , Type C Phospholipases/genetics
6.
Blood ; 120(8): 1601-12, 2012 Aug 23.
Article in English | MEDLINE | ID: mdl-22776820

ABSTRACT

We recently defined a critical role for p53 in regulating the quiescence of adult hematopoietic stem cells (HSCs) and identified necdin as a candidate p53 target gene. Necdin is a growth-suppressing protein and the gene encoding it is one of several that are deleted in patients with Prader-Willi syndrome. To define the intrinsic role of necdin in adult hematopoiesis, in the present study, we transplanted necdin-null fetal liver cells into lethally irradiated recipients. We show that necdin-null adult HSCs are less quiescent and more proliferative than normal HSCs, demonstrating the similar role of necdin and p53 in promoting HSC quiescence during steady-state conditions. However, wild-type recipients repopulated with necdin-null hematopoietic stem/progenitor cells show enhanced sensitivity to irradiation and chemotherapy, with increased p53-dependent apoptosis, myelosuppression, and mortality. Necdin controls the HSC response to genotoxic stress via both cell-cycle-dependent and cell-cycle-independent mechanisms, with the latter occurring in a Gas2L3-dependent manner. We conclude that necdin functions as a molecular switch in adult hematopoiesis, acting in a p53-like manner to promote HSC quiescence in the steady state, but suppressing p53-dependent apoptosis in response to genotoxic stress.


Subject(s)
DNA Damage , Hematopoiesis , Hematopoietic Stem Cells/cytology , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Apoptosis , Cell Proliferation , Cells, Cultured , Drug Therapy , Gene Deletion , Genes, p53 , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/radiation effects , Liver/cytology , Liver/embryology , Mice , Mice, Inbred C57BL
7.
Elife ; 122023 03 14.
Article in English | MEDLINE | ID: mdl-36917493

ABSTRACT

Aggregates of the tau protein are a well-known hallmark of several neurodegenerative diseases, collectively referred to as tauopathies, including frontal temporal dementia and Alzheimer's disease (AD). Monitoring the transformation process of tau from physiological monomers into pathological oligomers or aggregates in a high-throughput, quantitative manner and in a cellular context is still a major challenge in the field. Identifying molecules able to interfere with those processes is of high therapeutic interest. Here, we developed a series of inter- and intramolecular tau biosensors based on the highly sensitive Nanoluciferase (Nluc) binary technology (NanoBiT) able to monitor the pathological conformational change and self-interaction of tau in living cells. Our repertoire of tau biosensors reliably reports i. molecular proximity of physiological full-length tau at microtubules; ii. changes in tau conformation and self-interaction associated with tau phosphorylation, as well as iii. tau interaction induced by seeds of recombinant tau or from mouse brain lysates of a mouse model of tau pathology. By comparing biosensors comprising different tau forms (i.e. full-length or short fragments, wild-type, or the disease-associated tau(P301L) variant) further insights into the tau transformation process are obtained. Proof-of-concept data for the high-throughput suitability and identification of molecules interfering with the pathological tau transformation processes are presented. This novel repertoire of tau biosensors is aimed to boost the disclosure of molecular mechanisms underlying pathological tau transformation in living cells and to discover new drug candidates for tau-related neurodegenerative diseases.


Subject(s)
Alzheimer Disease , Tauopathies , Mice , Animals , tau Proteins/genetics , tau Proteins/metabolism , Alzheimer Disease/metabolism , Tauopathies/pathology , Microtubules/metabolism , Neurons/physiology , Brain/metabolism
8.
Sci Adv ; 8(6): eabg9455, 2022 Feb 11.
Article in English | MEDLINE | ID: mdl-35138899

ABSTRACT

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.

9.
Proc Natl Acad Sci U S A ; 105(32): 11212-7, 2008 Aug 12.
Article in English | MEDLINE | ID: mdl-18676613

ABSTRACT

Genetic studies have shown that ubiquitination and endocytosis of the Drosophila ligand Delta in signal-sending cells are required for activation of Notch signaling, but how these events promote Notch activation remains poorly understood. Here, we show that an ubiquitination-defective mutant of the murine Delta-homologue Dll1 is endocytosed but, in contrast to the wild-type Dll1, is unable to subsequently recycle back to the cell surface or to bind Notch1 efficiently. These results demonstrate that ubiquitination, although not required for endocytosis, is essential for Dll1 recycling and that recycling is required to acquire affinity for the receptor. On the other hand, a chimeric molecule encompassing the extracellular domain of Dll1 and the transmembrane/intracellular domain of Dll3, which contains no lysine, is endocytosed, recycled, and interacts with Notch1 but is unable to induce transendocytosis of the extracellular region of Notch1 or to signal. These observations suggest that the chimera uses an ubiquitination-independent signal to recycle, allowing it to acquire affinity for Notch1. Our results support the idea that ligand recycling determines its competence to bind efficiently to the receptor but that this is insufficient to allow it to perform transendocytosis, an event required for activation of Notch signaling. Finally, the present study indicates that Dll1 partially localizes to lipid microdomains, whereas both ubiquitination-defective Dll1 and the Dll1-3 chimera are excluded from these compartments, suggesting that these microdomains provide the environment necessary for Dll1 to activate Notch signaling.


Subject(s)
Intercellular Signaling Peptides and Proteins/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Membrane Proteins/metabolism , Signal Transduction/physiology , Ubiquitination/physiology , Animals , Calcium-Binding Proteins , Drosophila , Endocytosis/physiology , HeLa Cells , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Ligands , Membrane Microdomains/genetics , Membrane Microdomains/metabolism , Membrane Proteins/genetics , Mice , Protein Transport/physiology , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
10.
Cell Stem Cell ; 24(4): 551-565.e8, 2019 04 04.
Article in English | MEDLINE | ID: mdl-30905619

ABSTRACT

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.


Subject(s)
DNA Damage , Gene Editing , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Line , Humans , K562 Cells , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID
11.
Nat Commun ; 9(1): 1216, 2018 03 23.
Article in English | MEDLINE | ID: mdl-29572483

ABSTRACT

Transforming growth factor-ß (TGFß) signaling is initiated by the type I, II TGFß receptor (TßRI/TßRII) complex. Here we report the formation of an alternative complex between TßRI and the orphan GPR50, belonging to the G protein-coupled receptor super-family. The interaction of GPR50 with TßRI induces spontaneous TßRI-dependent Smad and non-Smad signaling by stabilizing the active TßRI conformation and competing for the binding of the negative regulator FKBP12 to TßRI. GPR50 overexpression in MDA-MB-231 cells mimics the anti-proliferative effect of TßRI and decreases tumor growth in a xenograft mouse model. Inversely, targeted deletion of GPR50 in the MMTV/Neu spontaneous mammary cancer model shows decreased survival after tumor onset and increased tumor growth. Low GPR50 expression is associated with poor survival prognosis in human breast cancer irrespective of the breast cancer subtype. This describes a previously unappreciated spontaneous TGFß-independent activation mode of TßRI and identifies GPR50 as a TßRI co-receptor with potential impact on cancer development.


Subject(s)
Mammary Neoplasms, Animal/prevention & control , Nerve Tissue Proteins/physiology , Receptor, Transforming Growth Factor-beta Type I/physiology , Receptors, G-Protein-Coupled/physiology , Animals , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation , Endosomes/metabolism , Female , Gene Expression Profiling , HEK293 Cells , HeLa Cells , Humans , Mammary Neoplasms, Animal/metabolism , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Nerve Tissue Proteins/genetics , Oligonucleotide Array Sequence Analysis , Receptors, G-Protein-Coupled/genetics , Signal Transduction , Smad Proteins/metabolism , Tacrolimus Binding Protein 1A/metabolism
12.
Sci Rep ; 6: 22760, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26961797

ABSTRACT

Multiple myeloma is a plasma cell neoplasm with an extremely variable clinical course. Animal models are needed to better understand its pathophysiology and for preclinical testing of potential therapeutic agents. Hematopoietic cells expressing the hypermorphic Rad50(s) allele show hematopoietic failure, which can be mitigated by the lack of a transcription factor, Mef/Elf4. However, we find that 70% of Mef(-/-)Rad50(s/s) mice die from multiple myeloma or other plasma cell neoplasms. These mice initially show an abnormal plasma cell proliferation and monoclonal protein production, and then develop anemia and a decreased bone mineral density. Tumor cells can be serially transplanted and according to array CGH and whole exome sequencing, the pathogenesis of plasma cell neoplasms in these mice is not linked to activation of a specific oncogene, or inactivation of a specific tumor suppressor. This model recapitulates the systemic manifestations of human plasma cell neoplasms, and implicates cooperativity between the Rad50(s) and Mef/Elf4 pathways in initiating myelomagenic mutations that promote plasma cell transformation.


Subject(s)
ATP-Binding Cassette Transporters/genetics , DNA-Binding Proteins/genetics , Multiple Myeloma/genetics , Phenotype , Transcription Factors/genetics , Acid Anhydride Hydrolases , Animals , Blood Cells/metabolism , Blood Cells/pathology , Blood Cells/physiology , Blood Cells/transplantation , Bone Density , Cell Proliferation , Cells, Cultured , Disease Models, Animal , Exome , Mice , Mice, Inbred C57BL , Multiple Myeloma/pathology
13.
PLoS One ; 8(9): e74392, 2013.
Article in English | MEDLINE | ID: mdl-24069306

ABSTRACT

The activity of Notch ligands is tightly regulated by trafficking events occurring both before and after ligand-receptor interaction. In particular endocytosis and recycling have been shown to be required for full signaling activity of the ligands before they encounter the Notch receptor. However little is known about the precise endocytic processes that contribute to ligand internalization. Here we demonstrate that endocytosis contributes to Dll1 signaling activity by preserving the ligand from shedding and degradation. We further show that the glycosphingolipid-binding motif originally identified in Drosophila Notch ligands is conserved in mammals and is necessary for Dll1 internalization. Mutation of its conserved tryptophan residue results in a Dll1 molecule which is rapidly inactivated by shedding and degradation, does not recycle to the cell surface and does not activate Notch signaling. Finally, silencing in the signal-sending cells of glucosylceramide synthase, the enzyme implicated in the initial phase of glycosphingolipid synthesis, down-regulates Notch activation. Our data indicate that glycosphingolipids, by interacting with Dll1, may act as functional co-factors to promote its biological activity.


Subject(s)
Glycosphingolipids/metabolism , Intercellular Signaling Peptides and Proteins/chemistry , Intercellular Signaling Peptides and Proteins/metabolism , Protein Interaction Domains and Motifs , Receptors, Notch/metabolism , Amino Acid Sequence , Animals , Cell Membrane/chemistry , Cell Membrane/metabolism , Gene Silencing , Glucosyltransferases/genetics , Glycosphingolipids/chemistry , Humans , Ligands , Membrane Lipids/metabolism , Mice , Molecular Sequence Data , Protein Binding , Protein Stability , Protein Transport , RNA Interference , Signal Transduction
14.
Cancer Cell ; 23(3): 362-75, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23518350

ABSTRACT

The molecular mechanisms regulating leukemia-initiating cell (LIC) function are of important clinical significance. We use chronic myelogenous leukemia (CML) as a model of LIC-dependent malignancy and identify the interaction between the ubiquitin ligase Fbw7 and its substrate c-Myc as a regulator of LIC homeostasis. Deletion of Fbw7 leads to c-Myc overexpression, p53-dependent LIC-specific apoptosis, and the eventual inhibition of tumor progression. A decrease of either c-Myc protein levels or attenuation of the p53 response rescues LIC activity and disease progression. Further experiments showed that Fbw7 expression is required for survival and maintenance of human CML LIC. These studies identify a ubiquitin ligase:substrate pair regulating LIC activity, suggesting that targeting of the Fbw7:c-Myc axis is an attractive therapy target in refractory CML.


Subject(s)
F-Box Proteins/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Tumor Suppressor Protein p53/metabolism , Ubiquitin-Protein Ligases/metabolism , Animals , Apoptosis , Disease Progression , F-Box Proteins/genetics , F-Box-WD Repeat-Containing Protein 7 , Gene Expression Regulation, Neoplastic , Hematopoietic Stem Cells , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Mice , Mice, Inbred C57BL , RNA Interference , RNA, Small Interfering , Tumor Cells, Cultured , Ubiquitin-Protein Ligases/genetics , Ubiquitination
15.
J Exp Med ; 210(2): 301-19, 2013 Feb 11.
Article in English | MEDLINE | ID: mdl-23359070

ABSTRACT

Notch signaling pathway activation is known to contribute to the pathogenesis of a spectrum of human malignancies, including T cell leukemia. However, recent studies have implicated the Notch pathway as a tumor suppressor in myeloproliferative neoplasms and several solid tumors. Here we report a novel tumor suppressor role for Notch signaling in acute myeloid leukemia (AML) and demonstrate that Notch pathway activation could represent a therapeutic strategy in this disease. We show that Notch signaling is silenced in human AML samples, as well as in AML-initiating cells in an animal model of the disease. In vivo activation of Notch signaling using genetic Notch gain of function models or in vitro using synthetic Notch ligand induces rapid cell cycle arrest, differentiation, and apoptosis of AML-initiating cells. Moreover, we demonstrate that Notch inactivation cooperates in vivo with loss of the myeloid tumor suppressor Tet2 to induce AML-like disease. These data demonstrate a novel tumor suppressor role for Notch signaling in AML and elucidate the potential therapeutic use of Notch receptor agonists in the treatment of this devastating leukemia.


Subject(s)
Leukemia, Myeloid, Acute/metabolism , Leukemia, Myeloid, Acute/pathology , Receptors, Notch/metabolism , Animals , Cell Differentiation , Cell Survival , DNA-Binding Proteins/metabolism , Dioxygenases , Disease Models, Animal , Gene Silencing , Homeostasis , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/therapy , Ligands , Mice , Mice, Transgenic , Mutation , Proto-Oncogene Proteins/metabolism , Receptors, Notch/agonists , Signal Transduction , Tumor Suppressor Proteins/metabolism
16.
J Exp Med ; 210(12): 2641-59, 2013 Nov 18.
Article in English | MEDLINE | ID: mdl-24218140

ABSTRACT

Somatic Addition of Sex Combs Like 1 (ASXL1) mutations occur in 10-30% of patients with myeloid malignancies, most commonly in myelodysplastic syndromes (MDSs), and are associated with adverse outcome. Germline ASXL1 mutations occur in patients with Bohring-Opitz syndrome. Here, we show that constitutive loss of Asxl1 results in developmental abnormalities, including anophthalmia, microcephaly, cleft palates, and mandibular malformations. In contrast, hematopoietic-specific deletion of Asxl1 results in progressive, multilineage cytopenias and dysplasia in the context of increased numbers of hematopoietic stem/progenitor cells, characteristic features of human MDS. Serial transplantation of Asxl1-null hematopoietic cells results in a lethal myeloid disorder at a shorter latency than primary Asxl1 knockout (KO) mice. Asxl1 deletion reduces hematopoietic stem cell self-renewal, which is restored by concomitant deletion of Tet2, a gene commonly co-mutated with ASXL1 in MDS patients. Moreover, compound Asxl1/Tet2 deletion results in an MDS phenotype with hastened death compared with single-gene KO mice. Asxl1 loss results in a global reduction of H3K27 trimethylation and dysregulated expression of known regulators of hematopoiesis. RNA-Seq/ChIP-Seq analyses of Asxl1 in hematopoietic cells identify a subset of differentially expressed genes as direct targets of Asxl1. These findings underscore the importance of Asxl1 in Polycomb group function, development, and hematopoiesis.


Subject(s)
Abnormalities, Multiple/etiology , Myelodysplastic Syndromes/etiology , Repressor Proteins/deficiency , Repressor Proteins/genetics , Abnormalities, Multiple/genetics , Animals , DNA-Binding Proteins/deficiency , DNA-Binding Proteins/genetics , Dioxygenases , Disease Models, Animal , Epigenesis, Genetic , Female , Gene Deletion , Germ-Line Mutation , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Humans , Mice , Mice, 129 Strain , Mice, Inbred C57BL , Mice, Knockout , Mutation , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/pathology , Phenotype , Pregnancy , Protein Binding , Proto-Oncogene Proteins/deficiency , Proto-Oncogene Proteins/genetics , Repressor Proteins/metabolism
17.
Cancer Cell ; 22(4): 452-65, 2012 Oct 16.
Article in English | MEDLINE | ID: mdl-23079656

ABSTRACT

D-type cyclins form complexes with cyclin-dependent kinases (CDK4/6) and promote cell cycle progression. Although cyclin D functions appear largely tissue specific, we demonstrate that cyclin D3 has unique functions in lymphocyte development and cannot be replaced by cyclin D2, which is also expressed during blood differentiation. We show that only combined deletion of p27(Kip1) and retinoblastoma tumor suppressor (Rb) is sufficient to rescue the development of Ccnd3(-/-) thymocytes. Furthermore, we show that a small molecule targeting the kinase function of cyclin D3:CDK4/6 inhibits both cell cycle entry in human T cell acute lymphoblastic leukemia (T-ALL) and disease progression in animal models of T-ALL. These studies identify unique functions for cyclin D3:CDK4/6 complexes and suggest potential therapeutic protocols for this devastating blood tumor.


Subject(s)
Cyclin D3/antagonists & inhibitors , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Animals , Cyclin D2/physiology , Cyclin D3/physiology , Cyclin-Dependent Kinase 4/physiology , Cyclin-Dependent Kinase 6/physiology , Cyclin-Dependent Kinase Inhibitor p27/physiology , Humans , Lymphocytes/physiology , Mice , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/etiology , Receptor, Notch1/physiology , Retinoblastoma Protein/physiology
18.
Cancer Cell ; 20(1): 11-24, 2011 Jul 12.
Article in English | MEDLINE | ID: mdl-21723200

ABSTRACT

Somatic loss-of-function mutations in the ten-eleven translocation 2 (TET2) gene occur in a significant proportion of patients with myeloid malignancies. Although there are extensive genetic data implicating TET2 mutations in myeloid transformation, the consequences of Tet2 loss in hematopoietic development have not been delineated. We report here an animal model of conditional Tet2 loss in the hematopoietic compartment that leads to increased stem cell self-renewal in vivo as assessed by competitive transplant assays. Tet2 loss leads to a progressive enlargement of the hematopoietic stem cell compartment and eventual myeloproliferation in vivo, including splenomegaly, monocytosis, and extramedullary hematopoiesis. In addition, Tet2(+/-) mice also displayed increased stem cell self-renewal and extramedullary hematopoiesis, suggesting that Tet2 haploinsufficiency contributes to hematopoietic transformation in vivo.


Subject(s)
Cell Transformation, Neoplastic/pathology , DNA-Binding Proteins/deficiency , Hematopoietic Stem Cells/pathology , Myeloid Cells/pathology , Proto-Oncogene Proteins/deficiency , Alleles , Animals , Cell Proliferation , Cell Transformation, Neoplastic/metabolism , DNA-Binding Proteins/metabolism , Dioxygenases , Gene Deletion , Gene Knockout Techniques , Gene Silencing , Haploinsufficiency/genetics , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, Myelomonocytic, Chronic/pathology , Mice , Myeloid Cells/metabolism , Proto-Oncogene Proteins/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL