Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 19 de 19
Filter
Add more filters










Publication year range
1.
Proc Natl Acad Sci U S A ; 120(20): e2210058120, 2023 05 16.
Article in English | MEDLINE | ID: mdl-37155858

ABSTRACT

Notch signaling is essential for the emergence of definitive hematopoietic stem cells (HSCs) in the embryo and their development in the fetal liver niche. However, how Notch signaling is activated and which fetal liver cell type provides the ligand for receptor activation in HSCs is unknown. Here we provide evidence that endothelial Jagged1 (Jag1) has a critical early role in fetal liver vascular development but is not required for hematopoietic function during fetal HSC expansion. We demonstrate that Jag1 is expressed in many hematopoietic cells in the fetal liver, including HSCs, and that its expression is lost in adult bone marrow HSCs. Deletion of hematopoietic Jag1 does not affect fetal liver development; however, Jag1-deficient fetal liver HSCs exhibit a significant transplantation defect. Bulk and single-cell transcriptomic analysis of HSCs during peak expansion in the fetal liver indicates that loss of hematopoietic Jag1 leads to the downregulation of critical hematopoietic factors such as GATA2, Mllt3, and HoxA7, but does not perturb Notch receptor expression. Ex vivo activation of Notch signaling in Jag1-deficient fetal HSCs partially rescues the functional defect in a transplant setting. These findings indicate a new fetal-specific niche that is based on juxtracrine hematopoietic Notch signaling and reveal Jag1 as a fetal-specific niche factor essential for HSC function.


Subject(s)
Fetus , Hematopoietic Stem Cells , Adult , Humans , Endothelium/metabolism , Fetus/metabolism , Hematopoietic Stem Cells/metabolism , Liver/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism
2.
Angiogenesis ; 26(2): 249-263, 2023 05.
Article in English | MEDLINE | ID: mdl-36376768

ABSTRACT

The Notch signaling pathway is an important therapeutic target for the treatment of inflammatory diseases and cancer. We previously created ligand-specific inhibitors of Notch signaling comprised of Fc fusions to specific EGF-like repeats of the Notch1 extracellular domain, called Notch decoys, which bound ligands, blocked Notch signaling, and showed anti-tumor activity with low toxicity. However, the study of their function depended on virally mediated expression, which precluded dosage control and limited clinical applicability. We have refined the decoy design to create peptibody-based Notch inhibitors comprising the core binding domains, EGF-like repeats 10-14, of either Notch1 or Notch4. These Notch peptibodies showed high secretion properties and production yields that were improved by nearly 100-fold compared to previous Notch decoys. Using surface plasmon resonance spectroscopy coupled with co-immunoprecipitation assays, we observed that Notch1 and Notch4 peptibodies demonstrate strong but distinct binding properties to Notch ligands DLL4 and JAG1. Both Notch1 and Notch4 peptibodies interfere with Notch signaling in endothelial cells and reduce expression of canonical Notch targets after treatment. While prior DLL4 inhibitors cause hyper-sprouting, the Notch1 peptibody reduced angiogenesis in a 3-dimensional in vitro sprouting assay. Administration of Notch1 peptibodies to neonate mice resulted in reduced radial outgrowth of retinal vasculature, confirming anti-angiogenic properties. We conclude that purified Notch peptibodies comprising EGF-like repeats 10-14 bind to both DLL4 and JAG1 ligands and exhibit anti-angiogenic properties. Based on their secretion profile, unique Notch inhibitory activities, and anti-angiogenic properties, Notch peptibodies present new opportunities for therapeutic Notch inhibition.


Subject(s)
Angiogenesis Inhibitors , Endothelial Cells , Receptor, Notch1 , Receptor, Notch4 , Animals , Mice , Angiogenesis Inhibitors/genetics , Angiogenesis Inhibitors/metabolism , Angiogenesis Inhibitors/pharmacology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , Epidermal Growth Factor/metabolism , Immunoprecipitation , Inflammation/drug therapy , Inflammation/genetics , Inflammation/metabolism , Ligands , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/metabolism , Receptor, Notch1/antagonists & inhibitors , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Receptor, Notch4/genetics , Receptor, Notch4/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Retinal Vessels/drug effects , Surface Plasmon Resonance
3.
Proc Natl Acad Sci U S A ; 119(35): e2121251119, 2022 08 30.
Article in English | MEDLINE | ID: mdl-35994670

ABSTRACT

GCN2 (general control nonderepressible 2) is a serine/threonine-protein kinase that controls messenger RNA translation in response to amino acid availability and ribosome stalling. Here, we show that GCN2 controls erythrocyte clearance and iron recycling during stress. Our data highlight the importance of liver macrophages as the primary cell type mediating these effects. During different stress conditions, such as hemolysis, amino acid deficiency or hypoxia, GCN2 knockout (GCN2-/-) mice displayed resistance to anemia compared with wild-type (GCN2+/+) mice. GCN2-/- liver macrophages exhibited defective erythrophagocytosis and lysosome maturation. Molecular analysis of GCN2-/- cells demonstrated that the ATF4-NRF2 pathway is a critical downstream mediator of GCN2 in regulating red blood cell clearance and iron recycling.


Subject(s)
Amino Acids , Erythrocytes , Iron , Liver , Macrophages , Protein Serine-Threonine Kinases , Activating Transcription Factor 4/metabolism , Amino Acids/deficiency , Amino Acids/metabolism , Anemia/metabolism , Animals , Cytophagocytosis , Erythrocytes/metabolism , Gene Deletion , Hemolysis , Hypoxia/metabolism , Iron/metabolism , Liver/cytology , Lysosomes/metabolism , Macrophages/metabolism , Mice , Mice, Knockout , NF-E2-Related Factor 2/metabolism , Protein Serine-Threonine Kinases/deficiency , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Stress, Physiological
4.
JCI Insight ; 7(9)2022 05 09.
Article in English | MEDLINE | ID: mdl-35349492

ABSTRACT

Long-term impairment in T cell-mediated adaptive immunity is a major clinical obstacle following treatment of blood disorders with hematopoietic stem cell transplantation. Although T cell development in the thymus has been extensively characterized, there are significant gaps in our understanding of prethymic processes that influence early T cell potential. We have uncovered a Notch/IL-21 signaling axis in bone marrow common lymphoid progenitor (CLP) cells. IL-21 receptor expression was driven by Notch activation in CLPs, and in vivo treatment with IL-21 induced Notch-dependent CLP proliferation. Taking advantage of this potentially novel signaling axis, we generated T cell progenitors ex vivo, which improved repopulation of the thymus and peripheral lymphoid organs of mice in an allogeneic transplant model. Importantly, Notch and IL-21 activation were equally effective in the priming and expansion of human cord blood cells toward the T cell fate, confirming the translational potential of the combined treatment.


Subject(s)
Hematopoietic Stem Cells , T-Lymphocytes , Animals , Bone Marrow/metabolism , Hematopoietic Stem Cells/metabolism , Interleukins , Mice , Signal Transduction
5.
J Physiol ; 600(3): 509-530, 2022 02.
Article in English | MEDLINE | ID: mdl-34921404

ABSTRACT

Here we genetically and functionally addressed potential pathways of Notch signalling in mediating vascular regeneration in mouse models. We first used transgenic adult mice with either gain- or loss-of-function Notch signalling in vascular endothelial cells and monitored perfusion in the hindlimb following ischaemia induced by femoral artery ligation. Mice deficient in Notch signalling showed defective perfusion recovery and expansion of collateral arteries. Transcriptomics analysis of arterial endothelial cells in the Notch mutants identified the guidance factor Sema3g as a candidate gene mediating reperfusion downstream of Notch. Studies in the retinal circulation showed the central role of SEMA3G downstream of Notch signalling in the orderly regulation of vascular patterning. These studies in multiple vascular beds show the primacy of Notch signalling and downstream generation of guidance peptides such as SEMA3G in promoting well-ordered vascular regeneration. KEY POINTS: Notch signalling is a critical mediator of revascularization. Yet the cellular processes activated during recovery following vascular injury are incompletely understood. Here we used genetic and cellular approaches in two different vascular beds and cultured endothelial cells to address the generalizability of mechanisms. By utilizing a highly reproducible murine model of hindlimb ischaemia in transgenic mice in which Notch signalling was inhibited at the transcriptional level, we demonstrated the centrality of Notch signalling in perfusion recovery and revascularization. RNA-sequencing of Notch mutants identified class 3 Semaphorins regulated by Notch signalling as downstream targets. Studies in retinal vessels and endothelial cells showed an essential role of guidance peptide Sema3g as a modulator of angiogenesis and orderly vascular patterning. The Notch to Sema3g signalling axis functions as a feedback mechanism to sculpt the growing vasculature in multiple beds.


Subject(s)
Semaphorins , Animals , Endothelial Cells/metabolism , Hindlimb/blood supply , Mice , Neovascularization, Physiologic/physiology , Receptor, Notch1 , Receptors, Notch/metabolism , Semaphorins/genetics , Semaphorins/metabolism , Signal Transduction
6.
Front Immunol ; 12: 723055, 2021.
Article in English | MEDLINE | ID: mdl-34394130

ABSTRACT

Lifelong mammalian hematopoiesis requires continuous generation of mature blood cells that originate from Hematopoietic Stem and Progenitor Cells (HSPCs) situated in the post-natal Bone Marrow (BM). The BM microenvironment is inherently complex and extensive studies have been devoted to identifying the niche that maintains HSPC homeostasis and supports hematopoietic potential. The Notch signaling pathway is required for the emergence of the definitive Hematopoietic Stem Cell (HSC) during embryonic development, but its role in BM HSC homeostasis is convoluted. Recent work has begun to explore novel roles for the Notch signaling pathway in downstream progenitor populations. In this review, we will focus an important role for Notch signaling in the establishment of a T cell primed sub-population of Common Lymphoid Progenitors (CLPs). Given that its activation mechanism relies primarily on cell-to-cell contact, Notch signaling is an ideal means to investigate and define a novel BM lymphopoietic niche. We will discuss how new genetic model systems indicate a pre-thymic, BM-specific role for Notch activation in early T cell development and what this means to the paradigm of lymphoid lineage commitment. Lastly, we will examine how leukemic T-cell acute lymphoblastic leukemia (T-ALL) blasts take advantage of Notch and downstream lymphoid signals in the pathological BM niche.


Subject(s)
Bone Marrow/metabolism , Hematopoietic Stem Cells/metabolism , Lymphopoiesis/physiology , Osteoblasts/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Animals , Bone Marrow/pathology , Cell Differentiation/physiology , Hematopoiesis/physiology , Hematopoietic Stem Cells/pathology , Homeostasis , Humans , Osteoblasts/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Signal Transduction/physiology , Tumor Microenvironment
7.
Blood ; 137(6): 775-787, 2021 02 11.
Article in English | MEDLINE | ID: mdl-32881992

ABSTRACT

Hematopoietic and nervous systems are linked via innervation of bone marrow (BM) niche cells. Hematopoietic stem/progenitor cells (HSPCs) express neurotransmitter receptors, such as the γ-aminobutyric acid (GABA) type B receptor subunit 1 (GABBR1), suggesting that HSPCs could be directly regulated by neurotransmitters like GABA that directly bind to GABBR1. We performed imaging mass spectrometry and found that the endogenous GABA molecule is regionally localized and concentrated near the endosteum of the BM niche. To better understand the role of GABBR1 in regulating HSPCs, we generated a constitutive Gabbr1-knockout mouse model. Analysis revealed that HSPC numbers were significantly reduced in the BM compared with wild-type littermates. Moreover, Gabbr1-null hematopoietic stem cells had diminished capacity to reconstitute irradiated recipients in a competitive transplantation model. Gabbr1-null HSPCs were less proliferative under steady-state conditions and upon stress. Colony-forming unit assays demonstrated that almost all Gabbr1-null HSPCs were in a slow or noncycling state. In vitro differentiation of Gabbr1-null HSPCs in cocultures produced fewer overall cell numbers with significant defects in differentiation and expansion of the B-cell lineage. To determine whether a GABBR1 agonist could stimulate human umbilical cord blood (UCB) HSPCs, we performed brief ex vivo treatment prior to transplant into immunodeficient mice, with significant increases in long-term engraftment of HSPCs compared with GABBR1 antagonist or vehicle treatments. Our results indicate a direct role for GABBR1 in HSPC proliferation, and identify a potential target to improve HSPC engraftment in clinical transplantation.


Subject(s)
Hematopoietic Stem Cells/cytology , Receptors, GABA-B/physiology , Animals , B-Lymphocytes/pathology , Baclofen/analogs & derivatives , Baclofen/pharmacology , Bone Marrow/innervation , Bone Marrow/metabolism , Bone Marrow Transplantation , Cell Division , Cell Lineage , Female , Gene Expression Regulation , Hematopoietic Stem Cells/metabolism , Human Umbilical Vein Endothelial Cells/transplantation , Humans , Lymphopenia/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Radiation Chimera , Receptors, GABA-B/deficiency , Receptors, GABA-B/genetics , Stem Cell Niche
8.
J Biol Chem ; 295(15): 4796-4808, 2020 04 10.
Article in English | MEDLINE | ID: mdl-32071080

ABSTRACT

Endothelial cells (ECs) lining the vasculature of vertebrates respond to low oxygen (hypoxia) by maintaining vascular homeostasis and initiating adaptive growth of new vasculature through angiogenesis. Previous studies have uncovered the molecular underpinnings of the hypoxic response in ECs; however, there is a need for comprehensive temporal analysis of the transcriptome during hypoxia. Here, we sought to investigate the early transcriptional programs of hypoxic ECs by using RNA-Seq of primary cultured human umbilical vein ECs exposed to progressively increasing severity and duration of hypoxia. We observed that hypoxia modulates the expression levels of approximately one-third of the EC transcriptome. Intriguingly, expression of the gene encoding the developmental transcription factor SOX7 (SRY-box transcription factor 7) rapidly and transiently increased during hypoxia. Transcriptomic and functional analyses of ECs following SOX7 depletion established its critical role in regulating hypoxia-induced angiogenesis. We also observed that depletion of the hypoxia-inducible factor (HIF) genes, HIF1A (encoding HIF-1α) and endothelial PAS domain protein 1 (EPAS1 encoding HIF-2α), inhibited both distinct and overlapping transcriptional programs. Our results indicated a role for HIF-1α in down-regulating mitochondrial metabolism while concomitantly up-regulating glycolytic genes, whereas HIF-2α primarily up-regulated the angiogenesis transcriptional program. These results identify the concentration and time dependence of the endothelial transcriptomic response to hypoxia and an early key role for SOX7 in mediating angiogenesis.


Subject(s)
Gene Expression Profiling/methods , Gene Expression Regulation , Human Umbilical Vein Endothelial Cells/pathology , Hypoxia/physiopathology , Neovascularization, Pathologic/pathology , SOXF Transcription Factors/metabolism , Transcriptome , Human Umbilical Vein Endothelial Cells/metabolism , Humans , Neovascularization, Pathologic/genetics , SOXF Transcription Factors/genetics
9.
Cell Rep ; 26(11): 2942-2954.e5, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30865885

ABSTRACT

Lung alveolar type I cells (AT1) and alveolar type II cells (AT2) regulate the structural integrity and function of alveoli. AT1, covering ∼95% of the surface area, are responsible for gas exchange, whereas AT2 serve multiple functions, including alveolar repair through proliferation and differentiation into AT1. However, the signaling mechanisms for alveolar repair remain unclear. Here, we demonstrate, in Pseudomonas aeruginosa-induced acute lung injury in mice, that non-canonical Notch ligand Dlk1 (delta-like 1 homolog) is essential for AT2-to-AT1 differentiation. Notch signaling was activated in AT2 at the onset of repair but later suppressed by Dlk1. Deletion of Dlk1 in AT2 induced persistent Notch activation, resulting in stalled transition to AT1 and accumulation of an intermediate cell population that expressed low levels of both AT1 and AT2 markers. Thus, Dlk1 expression leads to precisely timed inhibition of Notch signaling and activates AT2-to-AT1 differentiation, leading to alveolar repair.


Subject(s)
Alveolar Epithelial Cells/metabolism , Calcium-Binding Proteins/metabolism , Pneumonia, Bacterial/metabolism , Pseudomonas Infections/metabolism , Receptors, Notch/metabolism , Alveolar Epithelial Cells/cytology , Alveolar Epithelial Cells/microbiology , Animals , Calcium-Binding Proteins/genetics , Cell Differentiation , Cells, Cultured , Female , Male , Mice , Mice, Inbred C57BL , Regeneration , Signal Transduction
10.
Haematologica ; 104(11): 2164-2177, 2019 11.
Article in English | MEDLINE | ID: mdl-30923091

ABSTRACT

Loss-of-function studies have determined that Notch signaling is essential for hematopoietic and endothelial development. By deleting a single allele of the Notch1 transcriptional activation domain we generated viable, post-natal mice exhibiting hypomorphic Notch signaling. These heterozygous mice, which lack only one copy of the transcriptional activation domain, appear normal and have no endothelial or hematopoietic phenotype, apart from an inherent, cell-autonomous defect in T-cell lineage development. Following chemotherapy, these hypomorphs exhibited severe pancytopenia, weight loss and morbidity. This phenotype was confirmed in an endothelial-specific, loss-of-function Notch1 model system. Ang1, secreted by hematopoietic progenitors after damage, activated endothelial Tie2 signaling, which in turn enhanced expression of Notch ligands and potentiated Notch1 receptor activation. In our heterozygous, hypomorphic model system, the mutant protein that lacks the Notch1 transcriptional activation domain accumulated in endothelial cells and interfered with optimal activity of the wildtype Notch1 transcriptional complex. Failure of the hypomorphic mutant to efficiently drive transcription of key gene targets such as Hes1 and Myc prolonged apoptosis and limited regeneration of the bone marrow niche. Thus, basal Notch1 signaling is sufficient for niche development, but robust Notch activity is required for regeneration of the bone marrow endothelial niche and hematopoietic recovery.


Subject(s)
Cellular Microenvironment , Endothelial Cells/physiology , Receptor, Notch1/metabolism , Receptor, TIE-2/metabolism , Regeneration , Signal Transduction , Animals , Bone Marrow/drug effects , Bone Marrow/metabolism , Cellular Microenvironment/drug effects , Endothelial Cells/drug effects , Fluorouracil/pharmacology , Gamma Rays/adverse effects , Gene Expression Profiling , Gene Expression Regulation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Mice , Mice, Knockout , Pancytopenia/etiology , Pancytopenia/metabolism , Pancytopenia/pathology , Signal Transduction/drug effects
11.
Sci Signal ; 10(505)2017 11 14.
Article in English | MEDLINE | ID: mdl-29138297

ABSTRACT

Activating mutations in the gene encoding the cell-cell contact signaling protein Notch1 are common in human T cell acute lymphoblastic leukemias (T-ALLs). However, expressing Notch1 mutant alleles in mice fails to efficiently induce the development of leukemia. We performed a gain-of-function screen to identify proteins that enhanced signaling by leukemia-associated Notch1 mutants. The transcription factors MAFB and ETS2 emerged as candidates that individually enhanced Notch1 signaling, and when coexpressed, they synergistically increased signaling to an extent similar to that induced by core components of the Notch transcriptional complex. In mouse models of T-ALL, MAFB enhanced leukemogenesis by the naturally occurring Notch1 mutants, decreased disease latency, and increased disease penetrance. Decreasing MAFB abundance in mouse and human T-ALL cells reduced the expression of Notch1 target genes, including MYC and HES1, and sustained MAFB knockdown impaired T-ALL growth in a competitive setting. MAFB bound to ETS2 and interacted with the acetyltransferases PCAF and P300, highlighting its importance in recruiting coactivators that enhance Notch1 signaling. Together, these data identify a mechanism for enhancing the oncogenic potential of weak Notch1 mutants in leukemia models, and they reveal the MAFB-ETS2 transcriptional axis as a potential therapeutic target in T-ALL.


Subject(s)
Carcinogenesis , Gene Expression Regulation, Leukemic , MafB Transcription Factor/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Receptor, Notch1/genetics , Signal Transduction , Animals , Disease Models, Animal , Female , Humans , MafB Transcription Factor/genetics , Mice , Mice, Inbred C57BL , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Proto-Oncogene Protein c-ets-2/genetics , Proto-Oncogene Protein c-ets-2/metabolism , Receptor, Notch1/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Cells, Cultured
12.
Stem Cell Reports ; 9(3): 796-806, 2017 09 12.
Article in English | MEDLINE | ID: mdl-28781077

ABSTRACT

The generation of functional arterial endothelial cells (aECs) from embryonic stem cells (ESCs) holds great promise for vascular tissue engineering. However, the mechanisms underlying their generation and the potential of aECs in revascularizing ischemic tissue are not fully understood. Here, we observed that hypoxia exposure of mouse ESCs induced an initial phase of HIF1α-mediated upregulation of the transcription factor Etv2, which in turn induced the commitment to the EC fate. However, sustained activation of HIF1α in these EC progenitors thereafter induced NOTCH1 signaling that promoted the transition to aEC fate. We observed that transplantation of aECs mediated arteriogenesis in the mouse hindlimb ischemia model. Furthermore, transplantation of aECs in mice showed engraftment in ischemic myocardium and restored cardiac function in contrast to ECs derived under normoxia. Thus, HIF1α activation of Etv2 in ESCs followed by NOTCH1 signaling is required for the generation aECs that are capable of arteriogenesis and revascularization of ischemic tissue.


Subject(s)
Arteries/cytology , Cell Differentiation , Endothelial Cells/cytology , Endothelial Cells/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Mouse Embryonic Stem Cells/cytology , Receptors, Notch/metabolism , Transcription Factors/metabolism , Animals , Cell Hypoxia , Cell Lineage , Endothelial Cells/transplantation , Hindlimb/pathology , Ischemia/pathology , Mice , Mice, Inbred C57BL , Mouse Embryonic Stem Cells/metabolism , Myocardial Infarction/pathology , Myocardial Infarction/physiopathology , Organogenesis , Signal Transduction , Transcription Factors/genetics , Up-Regulation/genetics
13.
PLoS One ; 11(9): e0161157, 2016.
Article in English | MEDLINE | ID: mdl-27622512

ABSTRACT

Delta/Notch-like EGF-related receptor (DNER) has been reported to act as a Notch ligand, despite lacking a Delta/Serrate/Lag (DSL) binding domain common to all other known ligands. The established Notch ligand Delta-like 1 (DLL1), but not DNER, activated Notch1 in a luciferase assay, prevented the differentiation of myoblasts through Notch signaling, and bound Notch-fc in a cell-based assay. DNER is not a Notch ligand and its true function remains unknown.


Subject(s)
Nerve Tissue Proteins/metabolism , Receptors, Cell Surface/metabolism , Receptors, Notch/metabolism , Cell Differentiation/physiology , Cell Line , HEK293 Cells , Humans , Ligands , Myoblasts/metabolism , Receptor, Notch1/metabolism , Signal Transduction/physiology
14.
PLoS One ; 11(5): e0155408, 2016.
Article in English | MEDLINE | ID: mdl-27191957

ABSTRACT

Trib2 is highly expressed in human T cell acute lymphoblastic leukemia (T-ALL) and is a direct transcriptional target of the oncogenic drivers Notch and TAL1. In human TAL1-driven T-ALL cell lines, Trib2 is proposed to function as an important survival factor, but there is limited information about the role of Trib2 in primary T-ALL. In this study, we investigated the role of Trib2 in the initiation and maintenance of Notch-dependent T-ALL. Trib2 had no effect on the growth and survival of murine T-ALL cell lines in vitro when expression was blocked by shRNAs. To test the function of Trib2 on leukemogenesis in vivo, we generated Trib2 knockout mice. Mice were born at the expected Mendelian frequencies without gross developmental anomalies. Adult mice did not develop pathology or shortened survival, and hematopoiesis, including T cell development, was unperturbed. Using a retroviral model of Notch-induced T-ALL, deletion of Trib2 unexpectedly decreased the latency and increased the penetrance of T-ALL development in vivo. Immunoblotting of primary murine T-ALL cells showed that the absence of Trib2 increased C/EBPα expression, a known regulator of cell proliferation, and did not alter AKT or ERK phosphorylation. Although Trib2 was suggested to be highly expressed in T-ALL, transcriptomic analysis of two independent T-ALL cohorts showed that low Trib2 expression correlated with the TLX1-expressing cortical mature T-ALL subtype, whereas high Trib2 expression correlated with the LYL1-expressing early immature T-ALL subtype. These data indicate that Trib2 has a complex role in the pathogenesis of Notch-driven T-ALL, which may vary between different T-ALL subtypes.


Subject(s)
Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Protein Serine-Threonine Kinases/antagonists & inhibitors , Receptors, Notch/metabolism , Animals , CCAAT-Enhancer-Binding Proteins/genetics , CCAAT-Enhancer-Binding Proteins/metabolism , Cell Line, Tumor , Disease Models, Animal , Gene Expression , Gene Targeting , Genetic Loci , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Knockout , Penetrance , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Signal Transduction
15.
Genes Dev ; 28(6): 576-93, 2014 Mar 15.
Article in English | MEDLINE | ID: mdl-24637115

ABSTRACT

Notch1 is required to generate the earliest embryonic hematopoietic stem cells (HSCs); however since Notch-deficient embryos die early in gestation, additional functions for Notch in embryonic HSC biology have not been described. We used two complementary genetic models to address this important biological question. Unlike Notch1-deficient mice, mice lacking the conserved Notch1 transcriptional activation domain (TAD) show attenuated Notch1 function in vivo and survive until late gestation, succumbing to multiple cardiac abnormalities. Notch1 TAD-deficient HSCs emerge and successfully migrate to the fetal liver but are decreased in frequency by embryonic day 14.5. In addition, TAD-deficient fetal liver HSCs fail to compete with wild-type HSCs in bone marrow transplant experiments. This phenotype is independently recapitulated by conditional knockout of Rbpj, a core Notch pathway component. In vitro analysis of Notch1 TAD-deficient cells shows that the Notch1 TAD is important to properly assemble the Notch1/Rbpj/Maml trimolecular transcription complex. Together, these studies reveal an essential role for the Notch1 TAD in fetal development and identify important cell-autonomous functions for Notch1 signaling in fetal HSC homeostasis.


Subject(s)
Gene Expression Regulation, Developmental , Hematopoietic Stem Cells/physiology , Receptor, Notch1/metabolism , Signal Transduction , Animals , Cell Line , Fetal Stem Cells , Gene Knock-In Techniques , Gene Knockout Techniques , Hematopoietic Stem Cells/metabolism , Immunoglobulin J Recombination Signal Sequence-Binding Protein/genetics , Immunoglobulin J Recombination Signal Sequence-Binding Protein/metabolism , Mice , Mutation , Protein Structure, Tertiary/genetics , Receptor, Notch1/genetics , Survival Analysis
16.
Cancer Cell ; 23(3): 390-405, 2013 Mar 18.
Article in English | MEDLINE | ID: mdl-23434461

ABSTRACT

Notch1 is a rational therapeutic target in several human cancers, but as a transcriptional regulator, it poses a drug discovery challenge. To identify Notch1 modulators, we performed two cell-based, high-throughput screens for small-molecule inhibitors and cDNA enhancers of a NOTCH1 allele bearing a leukemia-associated mutation. Sarco/endoplasmic reticulum calcium ATPase (SERCA) channels emerged at the intersection of these complementary screens. SERCA inhibition preferentially impairs the maturation and activity of mutated Notch1 receptors and induces a G0/G1 arrest in NOTCH1-mutated human leukemia cells. A small-molecule SERCA inhibitor has on-target activity in two mouse models of human leukemia and interferes with Notch signaling in Drosophila. These studies "credential" SERCA as a therapeutic target in cancers associated with NOTCH1 mutations.


Subject(s)
Leukemia/genetics , Leukemia/metabolism , Receptor, Notch1/genetics , Sarcoplasmic Reticulum Calcium-Transporting ATPases/genetics , Alleles , Animals , Calcium Channels/genetics , Cell Line, Tumor , Drosophila/genetics , Drosophila/metabolism , Drug Screening Assays, Antitumor , Enzyme Inhibitors/pharmacology , Female , G1 Phase Cell Cycle Checkpoints/genetics , Gene Library , High-Throughput Screening Assays , Humans , Mice , Mice, SCID , Mutation , Neoplasm Transplantation , Receptor, Notch1/antagonists & inhibitors , Sarcoplasmic Reticulum Calcium-Transporting ATPases/antagonists & inhibitors , Signal Transduction/genetics , Small Molecule Libraries , Thapsigargin/pharmacology , Transplantation, Heterologous
17.
Cell Stem Cell ; 7(2): 198-213, 2010 Aug 06.
Article in English | MEDLINE | ID: mdl-20682446

ABSTRACT

An outstanding biological question is why tissue regeneration in mammals is limited, whereas urodele amphibians and teleost fish regenerate major structures, largely by cell cycle reentry. Upon inactivation of Rb, proliferation of postmitotic urodele skeletal muscle is induced, whereas in mammalian muscle this mechanism does not exist. We postulated that a tumor suppressor present in mammals but absent in regenerative vertebrates, the Ink4a product ARF (alternative reading frame), is a regeneration suppressor. Concomitant inactivation of Arf and Rb led to mammalian muscle cell cycle reentry, loss of differentiation properties, and upregulation of cytokinetic machinery. Single postmitotic myocytes were isolated by laser micro-dissection-catapulting, and transient suppression of Arf and Rb yielded myoblast colonies that retained the ability to differentiate and fuse into myofibers upon transplantation in vivo. These results show that differentiation of mammalian cells is reversed by inactivation of Arf and Rb and support the hypothesis that Arf evolved at the expense of regeneration.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Mammals/metabolism , Mitosis , Muscles/cytology , Muscles/metabolism , Regeneration/physiology , Retinoblastoma Protein/metabolism , Animals , Cell Dedifferentiation , Cell Nucleus/metabolism , Cell Proliferation , Cells, Cultured , Clone Cells , Cytokinesis , Lasers , Mice , Microdissection , Muscle Cells/cytology , Muscle Cells/metabolism , Muscle Fibers, Skeletal/cytology , Muscle Fibers, Skeletal/metabolism , Pressure , RNA, Small Interfering/metabolism , S Phase , Serum , Up-Regulation
18.
J Cell Biol ; 180(5): 1005-19, 2008 Mar 10.
Article in English | MEDLINE | ID: mdl-18332221

ABSTRACT

Cell-cell fusion is critical to the normal development of certain tissues, yet the nature and degree of conservation of the underlying molecular components remains largely unknown. Here we show that the two guanine-nucleotide exchange factors Brag2 and Dock180 have evolutionarily conserved functions in the fusion of mammalian myoblasts. Their effects on muscle cell formation are distinct and are a result of the activation of the GTPases ARF6 and Rac, respectively. Inhibition of ARF6 activity results in a lack of physical association between paxillin and beta(1)-integrin, and disruption of paxillin transport to sites of focal adhesion. We show that fusion machinery is conserved among distinct cell types because Dock180 deficiency prevented fusion of macrophages and the formation of multinucleated giant cells. Our results are the first to demonstrate a role for a single protein in the fusion of two different cell types, and provide novel mechanistic insight into the function of GEFs in the morphological maturation of multinucleated cells.


Subject(s)
Giant Cells/metabolism , Guanine Nucleotide Exchange Factors/metabolism , Macrophages/metabolism , Myoblasts/metabolism , ADP-Ribosylation Factor 6 , ADP-Ribosylation Factors/genetics , ADP-Ribosylation Factors/metabolism , Animals , Cell Adhesion/physiology , Cell Communication/physiology , Cell Fusion , Cell Line , Focal Adhesions/metabolism , Focal Adhesions/ultrastructure , Giant Cells/ultrastructure , Guanine Nucleotide Exchange Factors/genetics , Integrin beta1/metabolism , Macrophages/ultrastructure , Mice , Myoblasts/ultrastructure , Neuropeptides/genetics , Neuropeptides/metabolism , Paxillin/metabolism , RNA, Small Interfering , rac GTP-Binding Proteins/genetics , rac GTP-Binding Proteins/metabolism , rac1 GTP-Binding Protein
19.
PLoS Genet ; 2(6): e92, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16789831

ABSTRACT

Bacteriophages are the most abundant forms of life in the biosphere and carry genomes characterized by high genetic diversity and mosaic architectures. The complete sequences of 30 mycobacteriophage genomes show them collectively to encode 101 tRNAs, three tmRNAs, and 3,357 proteins belonging to 1,536 "phamilies" of related sequences, and a statistical analysis predicts that these represent approximately 50% of the total number of phamilies in the mycobacteriophage population. These phamilies contain 2.19 proteins on average; more than half (774) of them contain just a single protein sequence. Only six phamilies have representatives in more than half of the 30 genomes, and only three-encoding tape-measure proteins, lysins, and minor tail proteins-are present in all 30 phages, although these phamilies are themselves highly modular, such that no single amino acid sequence element is present in all 30 mycobacteriophage genomes. Of the 1,536 phamilies, only 230 (15%) have amino acid sequence similarity to previously reported proteins, reflecting the enormous genetic diversity of the entire phage population. The abundance and diversity of phages, the simplicity of phage isolation, and the relatively small size of phage genomes support bacteriophage isolation and comparative genomic analysis as a highly suitable platform for discovery-based education.


Subject(s)
Mycobacteriophages/genetics , Proteome , Virology/education , DNA, Viral/genetics , Genes, Viral , Genome, Viral , Genomics , Molecular Sequence Data , RNA, Messenger/genetics , RNA, Transfer/genetics , RNA, Viral/genetics , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...