Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
Mol Cell ; 68(4): 645-658.e5, 2017 Nov 16.
Article in English | MEDLINE | ID: mdl-29149593

ABSTRACT

Hajdu-Cheney syndrome (HCS), a rare autosomal disorder caused by heterozygous mutations in NOTCH2, is clinically characterized by acro-osteolysis, severe osteoporosis, short stature, neurological symptoms, cardiovascular defects, and polycystic kidneys. Recent studies identified that aberrant NOTCH2 signaling and consequent osteoclast hyperactivity are closely associated with the bone-related disorder pathogenesis, but the exact molecular mechanisms remain unclear. Here, we demonstrate that sustained osteoclast activity is largely due to accumulation of NOTCH2 carrying a truncated C terminus that escapes FBW7-mediated ubiquitination and degradation. Mice with osteoclast-specific Fbw7 ablation revealed osteoporotic phenotypes reminiscent of HCS, due to elevated Notch2 signaling. Importantly, administration of Notch inhibitors in Fbw7 conditional knockout mice alleviated progressive bone resorption. These findings highlight the molecular basis of HCS pathogenesis and provide clinical insights into potential targeted therapeutic strategies for skeletal disorders associated with the aberrant FBW7/NOTCH2 pathway as observed in patients with HCS.


Subject(s)
F-Box-WD Repeat-Containing Protein 7 , Hajdu-Cheney Syndrome , Mutation , Osteoporosis , Proteolysis , Receptor, Notch2 , Animals , Cell Line , F-Box-WD Repeat-Containing Protein 7/genetics , F-Box-WD Repeat-Containing Protein 7/metabolism , Hajdu-Cheney Syndrome/genetics , Hajdu-Cheney Syndrome/metabolism , Mice, Knockout , Osteoporosis/genetics , Osteoporosis/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Ubiquitination/genetics
2.
PLoS One ; 9(9): e108535, 2014.
Article in English | MEDLINE | ID: mdl-25255288

ABSTRACT

Notch family members were first identified as cell adhesion molecules by cell aggregation assays in Drosophila studies. However, they are generally recognized as signaling molecules, and it was unclear if their adhesion function was restricted to Drosophila. We previously demonstrated that a mouse Notch ligand, Delta-like 1 (Dll1) functioned as a cell adhesion molecule. We here investigated whether this adhesion function was conserved in the diversified mammalian Notch ligands consisted of two families, Delta-like (Dll1, Dll3 and Dll4) and Jagged (Jag1 and Jag2). The forced expression of mouse Dll1, Dll4, Jag1, and Jag2, but not Dll3, on stromal cells induced the rapid and enhanced adhesion of cultured mast cells (MCs). This was attributed to the binding of Notch1 and Notch2 on MCs to each Notch ligand on the stromal cells themselves, and not the activation of Notch signaling. Notch receptor-ligand binding strongly supported the tethering of MCs to stromal cells, the first step of cell adhesion. However, the Jag2-mediated adhesion of MCs was weaker and unlike other ligands appeared to require additional factor(s) in addition to the receptor-ligand binding. Taken together, these results demonstrated that the function of cell adhesion was conserved in mammalian as well as Drosophila Notch family members. Since Notch receptor-ligand interaction plays important roles in a broad spectrum of biological processes ranging from embryogenesis to disorders, our finding will provide a new perspective on these issues from the aspect of cell adhesion.


Subject(s)
Cell Adhesion Molecules/metabolism , Receptors, Notch/metabolism , Animals , Cell Adhesion/genetics , Cell Adhesion Molecules/genetics , Cell Line , Gene Expression , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Ligands , Mast Cells/metabolism , Mice , Protein Binding , Receptors, Notch/genetics , Signal Transduction , Stromal Cells/metabolism
3.
J Immunol ; 185(7): 3905-12, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20810995

ABSTRACT

Mast cells (MCs) accumulate in chronic inflammatory sites; however, it is not clear which adhesion molecules are involved in this process. Recently, the expression of Notch ligands was reported to be upregulated in inflammatory sites. Although Notch receptors are known as signaling molecules that can activate integrins, their contributions to the adhesion of MCs have not been studied. In this study, we demonstrated that mouse MCs efficiently adhered to stromal cells forced to express a Notch ligand, Delta-like 1 (Dll1). Surprisingly, the adhesion was a consequence of direct cell-cell interaction between MCs and Dll1-expressing stromal cells rather than activation of downstream effectors of Notch receptor(s)-Dll1. The adhesion of MCs to Dll1-expressing stromal cells remained even when the cell metabolism was arrested. The recognition was blocked only by inhibition of Notch receptor(s)-Dll1 interaction by addition of soluble DLL1, or mAbs against Dll1 or Notch2. Taken together, these results indicate that Notch receptor(s) and Dll1 directly promote the adhesion of MCs to stromal cells by acting as adhesion molecules. This appreciation that Notch receptor-ligand interactions have an adhesion function will provide an important clue to molecular basis of accumulation of MCs to inflammatory sites.


Subject(s)
Cell Communication/immunology , Intercellular Signaling Peptides and Proteins/metabolism , Mast Cells/metabolism , Stromal Cells/metabolism , Animals , Calcium-Binding Proteins , Cell Adhesion Molecules/immunology , Cell Adhesion Molecules/metabolism , Cell Separation , Flow Cytometry , Gene Expression , Intercellular Signaling Peptides and Proteins/immunology , Mast Cells/immunology , Mice , Mice, Inbred C57BL , Receptors, Notch/immunology , Receptors, Notch/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/immunology
4.
Stem Cells ; 27(7): 1676-85, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19544442

ABSTRACT

Notch and its ligands regulate multiple cell fate decisions. However, several questions on the timing, durability, and reversibility of Notch signaling effects on human hematopoietic precursors are still unresolved. Here, we used recombinant Delta ligands to deliver temporally and dose-controlled signals to human immature cord blood CD34(+)CD38(low) cells at clonal cell levels. Notch activation increased the frequency of multipotent progenitors, skewed the T and natural killer (NK) cell potential of CD34(+)CD38(low) clones in a dose- and ligand-dependent manner, and inhibited the differentiation of B cell clones. Low doses of ligands were sufficient for significantly increasing the frequency of NK cell precursors, whereas higher doses were required for increasing the frequency of T-cell clones. Interestingly, we demonstrate that temporary Notch activation prevents the subsequent differentiation of CD34(+)CD38(low) cells beyond a pro-B CD79a(+)CD19(-) stage characterized as a common lymphoid progenitor (CLP). Moreover, the lymphoid potential of this pro-B/CLP was skewed toward NK cell potential while the B cell precursor frequency was dramatically reduced. These results indicate critical timing and quantitative aspects of Notch/Delta interactions, imprinting the potential of CD34(+)CD38(low) hematopoietic progenitors. These results may have implications both in physiology and for cell manipulation because they demonstrate a tight regulation of the fate of human progenitors by Notch signaling.


Subject(s)
B-Lymphocytes/cytology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Killer Cells, Natural/cytology , Receptors, Notch/metabolism , T-Lymphocytes/cytology , ADP-ribosyl Cyclase 1/metabolism , Animals , Antigens, CD34/immunology , B-Lymphocytes/metabolism , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/drug effects , Cell Line , Cells, Cultured , Fetal Blood/cytology , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins , Killer Cells, Natural/metabolism , Membrane Proteins/pharmacology , Mice , Polymerase Chain Reaction , T-Lymphocytes/metabolism
5.
J Immunol ; 182(10): 6168-78, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19414770

ABSTRACT

The development of NK cells from hematopoietic stem cells is thought to be dependent on IL-15. In this study, we demonstrate that stimulation of human cord blood CD34(+) cells by a Notch ligand, Delta4, along with IL-7, stem cell factor, and Fms-like tyrosine kinase 3 ligand, but no IL-15, in a stroma-free culture induced the generation of cells with characteristics of functional NK cells, including CD56 and CD161 Ag expression, IFN-gamma secretion, and cytotoxic activity against K562 and Jurkat cells. Addition of gamma-secretase inhibitor and anti-human Notch1 Ab to the culture medium almost completely blocked NK cell emergence. Addition of anti-human IL-15-neutralizing Ab did not affect NK cell development in these culture conditions. The presence of IL-15, however, augmented cytotoxicity and was required for a more mature NK cell phenotype. CD56(+) cells generated by culture with IL-15, but without Notch stimulation, were negative for CD7 and cytoplasmic CD3, whereas CD56(+) cells generated by culture with both Delta4 and IL-15 were CD7(+) and cytoplasmic CD3(+) from the beginning and therefore more similar to in vivo human NK cell progenitors. Together, these results suggest that Notch signaling is important for the physiologic development of NK cells at differentiation stages beyond those previously postulated.


Subject(s)
Cell Differentiation/immunology , Hematopoietic Stem Cells/cytology , Interleukin-15/metabolism , Killer Cells, Natural/cytology , Lymphocyte Subsets/cytology , Receptors, Notch/metabolism , Antigens, CD34/immunology , Antigens, CD34/metabolism , Fetal Blood , Flow Cytometry , Fluorescent Antibody Technique , Hematopoietic Stem Cells/immunology , Humans , Interleukin-15/immunology , Killer Cells, Natural/immunology , Lymphocyte Subsets/immunology , Receptors, Notch/immunology , Signal Transduction/immunology
6.
Eur J Immunol ; 39(5): 1231-40, 2009 May.
Article in English | MEDLINE | ID: mdl-19350552

ABSTRACT

Notch and IL-7 are both well-characterized factors involved in T-cell development. In contrast to the mouse model, their precise requirements in the differentiation and/or proliferation of various stages of human thymic development have not been fully explored. Here, we demonstrate that IL-7 alone is sufficient to induce the differentiation of ex vivo purified CD34(+) triple negative (TN) surface (s) CD3(-) CD4(-)CD8(-) (CD3(-)CD4(-)CD8(-)), CD4 immature single positive (ISP) (sCD3(-)CD4(+)CD8(-)) and double positive (DP) (sCD3(-)CD4(+)CD8(+)) human thymic precursors to mature DP expressing sCD3 (sCD3(+)CD4(+)CD8(+)). We show that activation of Notch signaling by its ligands Delta-1 or Delta-4 potentiates IL-7-driven proliferation and survival of CD34(+) TN and to a lesser extent of CD4(+) ISP precursors. This effect of Notch is related to a sustained induction of IL-7 receptor alpha chain expression on thymocytes through a decreased methylation of its gene promoter. Thus, we show here that proliferation and differentiation of T-cell precursors are differentially modulated by IL-7 depending on the presence or absence of external signals. These results may have important implications for the clinical use of this cytokine as a strategy aimed at improving immune restoration.


Subject(s)
Cell Differentiation/immunology , Intercellular Signaling Peptides and Proteins/immunology , Interleukin-7/immunology , Membrane Proteins/immunology , Receptors, Notch/immunology , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing , Antigens, CD34/immunology , Calcium-Binding Proteins , Cell Differentiation/drug effects , Cell Growth Processes/drug effects , Cell Growth Processes/immunology , Flow Cytometry , Humans , Immunophenotyping , Infant , Infant, Newborn , Intercellular Signaling Peptides and Proteins/pharmacology , Interleukin-7/pharmacology , Interleukin-7 Receptor alpha Subunit/biosynthesis , Interleukin-7 Receptor alpha Subunit/immunology , Intracellular Signaling Peptides and Proteins , Membrane Proteins/pharmacology , Receptors, Interleukin-7/genetics , Receptors, Interleukin-7/immunology , Signal Transduction , T-Lymphocytes/drug effects
7.
Mol Cell Biol ; 28(20): 6402-12, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18710934

ABSTRACT

Notch signaling plays a key role in various cell differentiation processes including bone homeostasis. However, the specific involvement of Notch in regulating osteoclastogenesis is still controversial. In the present study, we show that RANKL induces expression of Jagged1 and Notch2 in bone marrow macrophages during osteoclast differentiation. Suppression of Notch signaling by a selective gamma-secretase inhibitor or Notch2 short hairpin RNA suppresses RANKL-induced osteoclastogenesis. In contrast, induction of Notch signaling by Jagged1 or by ectopic expression of intracellular Notch2 enhances NFATc1 promoter activity and expression and promotes osteoclastogenesis. Finally, we found that Notch2 and p65 interact in the nuclei of RANKL-stimulated cells and that both proteins are recruited to the NFATc1 promoter, driving its expression. Taken together, our results show a new molecular cross talk between Notch and NF-kappaB pathways that is relevant in osteoclastogenesis.


Subject(s)
Cell Differentiation/drug effects , NF-kappa B/metabolism , Osteoclasts/cytology , Osteoclasts/drug effects , RANK Ligand/pharmacology , Receptor, Notch2/metabolism , Animals , Cell Line , Gene Silencing/drug effects , Humans , Ligands , Male , Mice , NFATC Transcription Factors/metabolism , Oligopeptides/pharmacology , Osteoblasts/cytology , Osteoblasts/drug effects , Osteoblasts/metabolism , Osteoclasts/metabolism , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Notch2/genetics , Stem Cells/cytology , Stem Cells/drug effects
8.
Biosci Biotechnol Biochem ; 72(5): 1242-8, 2008 May.
Article in English | MEDLINE | ID: mdl-18460821

ABSTRACT

Embryonic stem cells (ESCs), which have characteristics such as self-renewal, indefinite proliferation, and pluripotency, are thought to hold great promise for regenerative medicine. ESCs are generally cultured on mouse embryonic fibroblast (MEF) or MEF conditioned medium (MEF-CM). However, for therapeutic applications, it is preferable for ESCs to be cultured under chemically defined conditions. Here, we report synthetic compounds that allow expansion of undifferentiated mouse ESCs in the absence of MEF, Leukemia Inhibitory Factor (LIF), and Fetal Bovine Serum (FBS). ESCs cultured for more than 30 d in a serum-free medium supplemented with indole derivertives retained their characteristic morphology and expressed markers such as SSEA-1, OCT3/4, Rex-1, Sox2, and Nanog. They consistently differentiated into many types of cells, including neurons, muscle cells, and hepatocytes. These results indicate that our compounds provide a more efficient and safer large-scale culture system for pluripotent ESCs, and hence might contribute to the use of ESCs in therapeutic applications.


Subject(s)
Embryonic Stem Cells/cytology , Embryonic Stem Cells/drug effects , Indoles/chemistry , Indoles/pharmacology , Alkaline Phosphatase/metabolism , Animals , Cell Culture Techniques , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Culture Media, Serum-Free , Embryonic Stem Cells/metabolism , Mice , Organic Cation Transport Proteins/genetics , Regeneration , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Time Factors
9.
Exp Hematol ; 34(12): 1720-9, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17157169

ABSTRACT

OBJECTIVE: The Notch pathway plays a key role in cell fate choices and in T-cell development. The goal of our study was to evaluate whether a short in vitro stimulation of the Notch pathway may alter human progenitor cell behavior. METHODS: CD34+ cord blood progenitors were exposed for 4 days to either immobilized Notch ligand Delta-4 or in control conditions. Phenotypic and molecular changes induced by the short stimulation were assessed at day 4. Next, long-term alteration of the fate of these progenitors was assessed in culture conditions suitable for B (coculture with MS5 stromal cells) and T (FTOC and OP9 stromal cells expressing Delta-4 systems) cell differentiation. RESULTS: Notch activation was sufficient to trigger immunophenotypic and molecular changes consistent with early T-cell lineage differentiation. Delta-4 induced, in 4 days, CD7+cytCD3epsilon+ cells. This paralleled at the gene-transcription level with de novo expression of several T cell-related transcription factors and TCRgamma rearrangement, while B cell transcripts were simultaneous silenced. As compared to non-Delta-4 primed cells, these early changes translated to long-term alteration of the potential of cells. Delta-4 priming led to an acceleration of T-cell development, including a completion of the TCR rearrangement, when cells were cultured in systems suitable for T-cell development while B-cell development was inhibited. CONCLUSION: A transient Notch activation is sufficient to promote T-cell differentiation from cord blood CD34+ cells. This system may be a useful tool for the amplification and the quantification of the T potential of CD34+ cells in various disease conditions.


Subject(s)
Antigens, CD34/biosynthesis , Cell Differentiation/drug effects , Hematopoietic Stem Cells/drug effects , Intercellular Signaling Peptides and Proteins/pharmacology , T-Lymphocytes/drug effects , Adaptor Proteins, Signal Transducing , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Calcium-Binding Proteins , Cell Differentiation/immunology , Cells, Cultured , Hematopoietic Stem Cells/immunology , Humans , Signal Transduction/drug effects , T-Lymphocytes/immunology
10.
Eur J Haematol ; 77(4): 273-81, 2006 Oct.
Article in English | MEDLINE | ID: mdl-16930143

ABSTRACT

Notch signaling plays an important role in cell fate decisions in developmental systems. To clarify its role in committed hematopoietic progenitor cells, we investigated the effects of Notch signaling in erythroid colony forming cells (ECFCs) generated from peripheral blood. ECFCs express Notch receptors, Notch1 and Notch2, and Notch ligands Delta1, Delta4, and Jagged1. When we assayed the effects of Notch ligands on erythroid maturation by flow cytometry, we found that immobilized Delta1 and immobilized Delta4 in particular inhibited maturation, whereas Jagged1 had no effect. In addition, Delta4 inhibited proliferation without reducing cell viability. Increases in expression levels of the Notch target gene hairy enhancer of split (HES) -1 were evident by real-time PCR after stimulation with immobilized Delta4. The effect of soluble Delta4 on expression of HES-1 was less pronounced than that seen with the immobilized form, indicating that all surface-bound ligands are important for effective signal transduction. When ECFCs were cultured in the presence of soluble Delta4 at a low cell concentration, erythroid maturation was slightly inhibited, but at a high concentration, maturation was promoted via competition of soluble Delta4 with endogenous ligands. These results indicate a pivotal role of Notch signaling in regulating erythroid maturation and proliferation, and further suggest that cell-cell interactions modulate growth of erythroid progenitor cells via Notch system.


Subject(s)
Cell Proliferation , Erythroid Precursor Cells/cytology , Receptors, Notch/metabolism , Signal Transduction , Base Sequence , Cells, Cultured , Cloning, Molecular , Culture Media, Serum-Free , DNA Primers , Flow Cytometry , Humans , Receptors, Notch/genetics , Reverse Transcriptase Polymerase Chain Reaction
11.
Stem Cells ; 24(11): 2456-65, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16857897

ABSTRACT

Ex vivo expansion of hematopoietic stem cells (HSCs) has been explored in the fields of stem cell biology, gene therapy, and clinical transplantation. Here, we demonstrate efficient ex vivo expansion of HSCs measured by long-term severe combined immunodeficient (SCID) repopulating cells (SRCs) from human cord blood CD133-sorted cells using a soluble form of Delta1. After a 3-week culture on immobilized Delta1 supplemented with stem cell factor, thrombopoietin, Flt-3 ligand, interleukin (IL)-3, and IL-6/soluble IL-6 receptor chimeric protein (FP6) in a serum- and stromal cell-free condition, we achieved approximately sixfold expansion of SRCs when evaluated by limiting dilution/transplantation assays. The maintenance of full multipotency and self-renewal capacity during culture was confirmed by transplantation to nonobese diabetic/SCID/gammac(null) mice, which showed myeloid, B, T, and natural killer cells as well as CD133(+)CD34(+) cells, and hematopoietic reconstitution in the secondary recipients. Interestingly, the CD133-sorted cells contained approximately 4.5 times more SRCs than the CD34-sorted cells. The present study provides a promising method to expand HSCs and encourages future trials on clinical transplantation.


Subject(s)
Cell Proliferation , Hematopoietic Stem Cells , Membrane Proteins/metabolism , Receptors, Fc/metabolism , AC133 Antigen , ADP-ribosyl Cyclase 1/analysis , Animals , Antigens, CD/analysis , Antigens, CD34/analysis , Cell Differentiation/drug effects , Cell Lineage , Cell Proliferation/drug effects , Cells, Cultured , Cytokine Receptor gp130/metabolism , Fetal Blood/cytology , Fetal Blood/immunology , Fetal Blood/metabolism , Glycoproteins/analysis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Immunomagnetic Separation , Interleukin-3/metabolism , Interleukin-6/metabolism , Intracellular Signaling Peptides and Proteins , Membrane Proteins/genetics , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Peptides/analysis , Receptors, Fc/genetics , Receptors, Interleukin/metabolism , Receptors, Interleukin-6 , Receptors, Notch/metabolism , Recombinant Fusion Proteins/metabolism
12.
Leuk Res ; 30(11): 1385-90, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16780947

ABSTRACT

A novel lymphoma cell line, designated TMD8 was established from cells of a patient with diffuse large B-cell lymphoma. TMD8 cells expressed HES1 mRNA, suggesting constitutive activation of Notch signaling. TMD8 cells expressed normal-sized Notch1 protein, and showed no mutations in the NOTCH1 gene. Cell growth was suppressed by gamma-secretase inhibitors (GSI). It was reported that GSI suppressed growth of T-cell acute lymphoblastic leukemia (T-ALL) cell lines, which frequently had NOTCH1 mutations. In addition to T-ALL, TMD8 is another unique cell line sensitive to GSI, and is useful to study effects of GSI in molecular targeting therapy.


Subject(s)
Amyloid Precursor Protein Secretases/antagonists & inhibitors , Cell Line, Tumor , Cell Proliferation/drug effects , Dipeptides/pharmacology , Enzyme Inhibitors/pharmacology , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/pathology , Oligopeptides/pharmacology , Adaptor Proteins, Signal Transducing , Basic Helix-Loop-Helix Transcription Factors/drug effects , Basic Helix-Loop-Helix Transcription Factors/genetics , Calcium-Binding Proteins , Cell Survival/drug effects , Fatal Outcome , Homeodomain Proteins/drug effects , Homeodomain Proteins/genetics , Humans , Intercellular Signaling Peptides and Proteins/genetics , Jagged-2 Protein , Karyotyping , Ligands , Lymphoma, B-Cell/genetics , Male , Membrane Proteins/genetics , Middle Aged , RNA, Messenger/drug effects , RNA, Messenger/genetics , Receptor, Notch1/genetics , Receptor, Notch2/genetics , Reverse Transcriptase Polymerase Chain Reaction , Structure-Activity Relationship , Transcription Factor HES-1
13.
Eur J Immunol ; 35(8): 2443-51, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16047342

ABSTRACT

The Notch pathway is involved in cell differentiation processes in various organs and at several developmental stages. The importance of Notch for early T lymphocyte development is well established. Recently, Notch has been implicated in directing naive T helper cell differentiation towards the Th1, Th2 or regulatory T cell lineages. However, the molecular events underlying these processes are poorly understood. We show that the Notch ligands Delta-like1, Delta-like4 and Jagged1 differentially affect early T cell activation and proliferation following T cell receptor cross-linking. Delta-like1 and Jagged1 induce a dose-dependent inhibition of early activation markers CD69 and CD25, as well as inhibition of proliferation after anti-CD3 stimulation of purified CD4+ T cells. Similarly, the rapid activation of transcription factors NF-AT, AP-1 and NF-kappaB is suppressed. In contrast, triggering of Notch by Delta-like4 enhances T cell activation and proliferation. The observed effects are dependent on simultaneous cross-linking of TCR and Notch but independent of gamma-secretase-mediated cleavage of Notch. These data suggest direct interference between Notch and early TCR signal transduction events, independent of the classical Notch pathway via release of the Notch intracellular domain. A Notch-mediated alteration of TCR signaling strength may contribute to the recently described modulation of naïve T cell differentiation by Notch ligands.


Subject(s)
Blood Proteins/physiology , Glycoproteins/physiology , Intercellular Signaling Peptides and Proteins/physiology , Lymphocyte Activation/immunology , Membrane Proteins/physiology , T-Lymphocytes, Helper-Inducer/immunology , Adaptor Proteins, Signal Transducing , Animals , Calcium-Binding Proteins , Cell Proliferation , Humans , Jagged-1 Protein , Jurkat Cells , Ligands , Mice , Mice, Inbred BALB C , Receptors, Antigen, T-Cell/immunology , Receptors, Notch , Serrate-Jagged Proteins , Signal Transduction/immunology
14.
Exp Hematol ; 33(5): 558-63, 2005 May.
Article in English | MEDLINE | ID: mdl-15850833

ABSTRACT

OBJECTIVE: Notch signaling plays a role in regulating the self-renewal and differentiation of hematopoietic progenitors. Since acute myeloblastic leukemia (AML) originates from dysregulated hematopoietic progenitors, the Notch system may be involved in the abnormal growth. We previously reported that AML cells express Notch proteins. In this study, we examined the effects of recombinant human Notch ligand proteins, Jagged1 and Delta1, on the growth and differentiation of primary AML cells. MATERIALS AND METHODS: AML cells separated from blood from 12 patients were cultured in wells coated with Jagged1, Delta1, or control IgG. The short-term growth was evaluated using a colorimetric assay. The self-renewal capacity was evaluated by the clonogenic cells recovered, which were obtained via a colony assay involving cells cultured with the ligands or control IgG. Differentiation was evaluated by the morphology of the cultured cells and flow cytometric analysis. RESULTS: The ligand stimulation caused three types of response in the short-term growth of the primary AML cells, namely, promotion, suppression, or no significant effect. The self-renewal capacity was suppressed or not significantly affected by the ligands, even in cells showing short-term growth promotion. The ligand stimulation altered blast cells into macrophage-like cells from their morphology and increased the expression of differentiation markers such as CD13 or CD14 in some samples. CONCLUSIONS: The Notch ligands had diverse effects on the short-term growth of primary AML cells. The ligands did not promote the self-renewal capacity of any of the cells examined and instead tended to induce differentiation under the conditions used.


Subject(s)
Cell Differentiation/drug effects , Cell Division/drug effects , Leukemia, Myeloid, Acute/pathology , Membrane Proteins/pharmacology , Adult , Aged , Calcium-Binding Proteins , Female , Humans , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Jagged-1 Protein , Ligands , Male , Membrane Proteins/metabolism , Middle Aged , Receptors, Notch , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Serrate-Jagged Proteins , Signal Transduction
15.
J Biol Chem ; 280(16): 15842-8, 2005 Apr 22.
Article in English | MEDLINE | ID: mdl-15695512

ABSTRACT

Functional involvement of the Notch pathway in osteoblastic differentiation has been previously investigated using the truncated intracellular domain, which mimics Notch signaling by interacting with the DNA-binding protein CBF-1. However, it is unclear whether Notch ligands Delta1 and Jagged1 also induce an identical cellular response in osteoblastic differentiation. We have shown that both Delta1 and Jagged1 were expressed concomitantly with Notch1 in maturating osteoblastic cells during bone regeneration and that overexpressed and immobilized recombinant Delta1 and Jagged1 alone did not alter the differentiated state of MC3T3-E1 and C2C12 cells. However, they augmented bone morphogenetic protein-2 (BMP2)-induced alkaline phosphatase activity and the expression of several differentiation markers, except for osteocalcin, and ultimately enhanced calcified nodule and in vivo ectopic bone formation of MC3T3-E1. In addition, both ligands transmitted signal through the CBF-1-dependent pathway and stimulated the expression of HES-1, a direct target of Notch pathway. To test the necessity of Notch signaling in BMP2-induced differentiation, Notch signaling was inhibited by the dominant negative extracellular domain of Notch1, specific inhibitor, or small interference RNA. These treatments decreased alkaline phosphatase activity as well as the expression of other differentiation markers and inhibited the promoter activity of Id-1, a target gene of the BMP pathway. These results indicate the functional redundancy between Delta1 and Jagged1 in osteoblastic differentiation whereby Delta1/Jagged1-activated Notch1 enhances BMP2-induced differentiation through the identical signaling pathway. Furthermore, our data also suggest that functional Notch signaling is essential not only for BMP2-induced osteoblast differentiation but also for BMP signaling itself.


Subject(s)
Bone Morphogenetic Proteins/metabolism , Cell Differentiation/physiology , Membrane Proteins/metabolism , Osteoblasts/physiology , Proteins/metabolism , Receptors, Cell Surface/metabolism , Transcription Factors/metabolism , Alkaline Phosphatase/metabolism , Animals , Bone Regeneration/physiology , Calcium-Binding Proteins , Intercellular Signaling Peptides and Proteins , Intracellular Signaling Peptides and Proteins , Jagged-1 Protein , Ligands , Mice , Mice, Inbred C57BL , Osteoblasts/cytology , Osteocalcin/metabolism , RNA, Small Interfering , Receptor, Notch1 , Receptors, Cell Surface/antagonists & inhibitors , Receptors, Cell Surface/genetics , Serrate-Jagged Proteins , Transcription Factors/antagonists & inhibitors , Transcription Factors/genetics
16.
Leuk Res ; 29(2): 197-203, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15607369

ABSTRACT

Effects of Notch activation on retinoic acid (RA)-induced differentiation and apoptosis were investigated. NB4, an acute promyelocytic leukemia (APL) cell line, undergoes neutrophilic differentiation and apoptosis by RA. Notch activation induced by a recombinant Notch ligand, Delta-1, did not affect the growth by itself. Treatment with RA plus Delta-1 made part of NB4 cells monocyte-like shaped and reduced the apoptosis. Similar phenomenon was also observed in primary APL cells. RA treatment induced cleavage of caspase-8 and PARP in NB4. Delta-1 suppressed the RA-induced cleavage of them, which may be a possible mechanism through which Delta-1 suppressed the RA-induced apoptosis.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Membrane Proteins/metabolism , Membrane Proteins/pharmacology , Neutrophils/cytology , Tretinoin/antagonists & inhibitors , Antigens, CD/metabolism , Caspase 8 , Caspases/drug effects , Caspases/metabolism , Cell Differentiation/physiology , Cell Line, Tumor , Cell Lineage/drug effects , Flow Cytometry , Humans , Intracellular Signaling Peptides and Proteins , Leukemia, Promyelocytic, Acute/drug therapy , Leukemia, Promyelocytic, Acute/metabolism , Ligands , Monocytes/physiology , Neutrophils/drug effects , Neutrophils/metabolism , Poly(ADP-ribose) Polymerases/drug effects , RNA, Messenger/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptor, Macrophage Colony-Stimulating Factor/genetics , Receptors, Notch , Tretinoin/metabolism , Tretinoin/pharmacology
17.
Int J Mol Med ; 13(3): 419-23, 2004 Mar.
Article in English | MEDLINE | ID: mdl-14767573

ABSTRACT

Notch signaling plays an important role in the regulation of self-renewal and differentiation of hematopoietic cells. Human monoblastic U937 cells undergo differentiation into macrophage-like cells, growth suppression, and apoptosis following stimulation with GM-CSF. We examined the effects of Notch activation induced by Notch ligands on GM-CSF-induced differentiation and apoptosis in U937 cells. Furthermore, the molecular mechanism of the effects was investigated. A recombinant Notch ligand, Delta-1 protein did not affect the growth of U937 cells by itself. GM-CSF-induced growth suppression and apoptosis of U937 cells were partially rescued by incubation with Delta-1. Delta-1 also reduced the GM-CSF-induced differentiation. Incubation with Delta-1 did not affect the expression of GM-CSF receptor. GM-CSF stimulation induced the phosphorylation of ERK1/2 and STAT5 and the cleavage of caspase-8, which were not affected by Delta-1 incubation, either. GM-CSF stimulation induced the cleavage of PARP, which is the key molecule for differentiation and apoptosis. We found that incubation with Delta-1 significantly suppressed the GM-CSF-induced cleavage of PARP. Taken together, we found that Notch activation induced by Delta-1 partially inhibited GM-CSF-induced differentiation, growth suppression, and apoptosis, along with reducing the GM-CSF-induced cleavage of PARP. These findings suggest one of the mechanisms by which Notch activation inhibits differentiation and apoptosis.


Subject(s)
Apoptosis/drug effects , Cell Differentiation/drug effects , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Membrane Proteins/pharmacology , Milk Proteins , Poly(ADP-ribose) Polymerases/metabolism , Caspases/metabolism , DNA-Binding Proteins/metabolism , Enzyme Activation/drug effects , Humans , Intracellular Signaling Peptides and Proteins , Ligands , MAP Kinase Signaling System/drug effects , Membrane Proteins/metabolism , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3 , Mitogen-Activated Protein Kinases/metabolism , Phosphorylation , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Receptors, Notch , Recombinant Proteins/pharmacology , STAT5 Transcription Factor , Trans-Activators/metabolism , U937 Cells
18.
Nat Biotechnol ; 21(7): 763-70, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12819790

ABSTRACT

We show that transplantation of adult bone marrow-derived cells expressing c-kit reduces hyperglycemia in mice with streptozotocin-induced pancreatic damage. Although quantitative analysis of the pancreas revealed a low frequency of donor insulin-positive cells, these cells were not present at the onset of blood glucose reduction. Instead, the majority of transplanted cells were localized to ductal and islet structures, and their presence was accompanied by a proliferation of recipient pancreatic cells that resulted in insulin production. The capacity of transplanted bone marrow-derived stem cells to initiate endogenous pancreatic tissue regeneration represents a previously unrecognized means by which these cells can contribute to the restoration of organ function.


Subject(s)
Bone Marrow Transplantation/methods , Hematopoietic Stem Cell Transplantation/methods , Hyperglycemia/pathology , Hyperglycemia/surgery , Pancreas/pathology , Pancreas/surgery , Regeneration , Animals , Blood Glucose/analysis , Cell Division , Hyperglycemia/chemically induced , Hyperglycemia/metabolism , Insulin/metabolism , Mice , Mice, Inbred NOD , Mice, SCID , Pancreas/drug effects , Pancreas/metabolism , Pancreatic Diseases/chemically induced , Pancreatic Diseases/metabolism , Pancreatic Diseases/pathology , Pancreatic Diseases/surgery , Streptozocin , Treatment Outcome
19.
Int J Oncol ; 22(5): 1073-9, 2003 May.
Article in English | MEDLINE | ID: mdl-12684674

ABSTRACT

The self-renewal and differentiation of hematopoietic progenitors are regulated by the interaction between Notch receptors and Notch ligands. Since AML originates from dysregulated hematopoietic progenitors, some abnormalities in the Notch system may be involved in the abnormal proliferation of AML cells. However, the significance of the Notch system in AML is not known. We examined the functional roles of Notch activation on the in vitro growth of seven human AML cell lines using three kinds of recombinant Notch ligand proteins, Jagged-1, Delta-1 and Delta-4. The ligands significantly affected the growth of two cell lines. In TMD7 cells, Delta proteins promoted the short-term growth, however, suppressing the self-renewal capacity and long-term growth. In OCI/AML-6 cells, Delta proteins suppressed the growth and self-renewal capacity while inducing differentiation into macrophage-like cells. We additionally found that Notch ligands needed to be immobilized on culture wells to affect the cells. These findings were in contrast to our hypothesis that Notch activation in AML cells leads to excessive self-renewal capacity and proliferation. If the Notch system in AML cells is precisely understood, the control of Notch activation will become a novel therapeutic approach for AML.


Subject(s)
Blood Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Leukemia, Myeloid, Acute/pathology , Membrane Proteins/metabolism , Proteins/metabolism , Adaptor Proteins, Signal Transducing , Binding Sites , Blood Proteins/genetics , Calcium-Binding Proteins , Cell Division/drug effects , Cell Division/physiology , Granulocyte Colony-Stimulating Factor/pharmacology , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Homeodomain Proteins/metabolism , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins , Jagged-1 Protein , Kinetics , Ligands , Membrane Proteins/genetics , Proteins/genetics , Recombinant Proteins/metabolism , Serrate-Jagged Proteins , Tumor Cells, Cultured
20.
Blood ; 101(6): 2227-34, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12411305

ABSTRACT

Osteoclasts are derived from hematopoietic precursor cells belonging to the monocyte/macrophage lineage. Osteoclast development has been reported to be regulated by several molecules such as macrophage colony-stimulating factor (M-CSF), receptor activator of nuclear factor (NF)-kappaB ligand (RANKL), and a decoy receptor of RANKL, osteoprotegerin (OPG). Recently, it was demonstrated that the Notch signaling pathway regulates myeloid differentiation and antagonizes cell fate determination, however, the effect of Notch signaling on the osteoclast lineage has not been reported. In this study, we examined the effect of signaling via Notch receptors on the differentiation into osteoclasts by using cells from the bone marrow, spleen, and peritoneal cavity, and a cloned macrophagelike cell line. Osteoclastogenesis was inhibited by an immobilized Notch ligand, Delta-1. The dish-adherent bone marrow cells precultured with M-CSF expressed both Mac-1 and M-CSF receptors, c-Fms; osteoclastogenesis of these cells was efficiently inhibited. The immobilized Delta-1 also down-regulated the surface c-Fms expression, while the c-Fms gene expression was not changed. Genes for Notch receptors and Notch ligands are expressed in not only hematopoietic cells but also stromal cells that support osteoclast development. Constitutively active Notch1-transfected stromal cells showed increased expression of RANKL and OPG genes, and strong inhibition of M-CSF gene expression, resulting in reduction of their ability to support osteoclast development. Taken together, these findings indicate that Notch signaling affects both osteoclast precursors and stromal cells and thereby negatively regulates osteoclastogenesis.


Subject(s)
Cell Differentiation , Membrane Proteins/metabolism , Osteoclasts/cytology , Receptors, Cell Surface , Signal Transduction , Stem Cells/cytology , Stromal Cells/cytology , Transcription Factors , Animals , Basic Helix-Loop-Helix Transcription Factors , Bone Marrow Cells/chemistry , Bone Marrow Cells/metabolism , Carrier Proteins/genetics , Dendritic Cells/cytology , Gene Expression , Glycoproteins/genetics , Homeodomain Proteins/genetics , Intracellular Signaling Peptides and Proteins , Macrophage Colony-Stimulating Factor/genetics , Macrophage Colony-Stimulating Factor/pharmacology , Macrophage-1 Antigen/analysis , Membrane Glycoproteins/genetics , Membrane Proteins/genetics , Membrane Proteins/pharmacology , Mice , Mice, Inbred C57BL , Osteoprotegerin , Polymerase Chain Reaction , RANK Ligand , Receptor Activator of Nuclear Factor-kappa B , Receptor, Macrophage Colony-Stimulating Factor/analysis , Receptor, Notch1 , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Notch , Receptors, Tumor Necrosis Factor , Reverse Transcriptase Polymerase Chain Reaction , Stromal Cells/metabolism , Transcription Factor HES-1 , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...