Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 5 de 5
1.
Stem Cells Dev ; 27(21): 1494-1506, 2018 11 01.
Article En | MEDLINE | ID: mdl-30084753

The interactions of hematopoietic stem and progenitor cells (HSPCs) with extracellular matrix (ECM) components and cells from the bone marrow (BM) microenvironment control their homeostasis. Regenerative BM conditions can induce expression of the ECM protein transforming growth factor beta-induced gene H3 (TGFBI or BIGH3) in murine HSPCs. In this study, we examined how increased or reduced TGFBI expression in human HSPCs and BM mesenchymal stromal cells (MSCs) affects HSPC maintenance, differentiation, and migration. HSPCs that overexpressed TGFBI showed accelerated megakaryopoiesis, whereas granulocyte differentiation and proliferation of granulocyte, erythrocyte, and monocyte cultures were reduced. In addition, both upregulation and downregulation of TGFBI expression impaired HSPC colony-forming capacity of HSPCs. Interestingly, the colony-forming capacity of HSPCs with reduced TGFBI levels was increased after long-term co-culture with MSCs, as measured by long-term culture-colony forming cell (LTC-CFC) formation. Moreover, TGFBI downregulation in HSPCs resulted in increased cobblestone area-forming cell (CAFC) frequency, a measure for hematopoietic stem cell (HSC) capacity. Concordantly, TGFBI upregulation in HSPCs resulted in a decrease of CAFC and LTC-CFC frequency. These results indicate that reduced TGFBI levels in HSPCs enhanced HSC maintenance, but only in the presence of MSCs. In addition, reduced levels of TGFBI in MSCs affected MSC/HSPC interaction, as observed by an increased migration of HSPCs under the stromal layer. In conclusion, tight regulation of TGFBI expression in the BM niche is essential for balanced HSPC proliferation and differentiation.


Cell Proliferation/genetics , Extracellular Matrix Proteins/genetics , Hematopoietic Stem Cells/cytology , Mesenchymal Stem Cells/cytology , Stem Cells/cytology , Transforming Growth Factor beta/genetics , Bone Marrow Cells/cytology , Cell Differentiation/genetics , Cell Line , Cell Movement/genetics , Cobblestone Lissencephaly/genetics , Coculture Techniques , Flow Cytometry , Gene Expression Regulation, Developmental , Genetic Vectors , Hematopoiesis/genetics , Hematopoietic Stem Cells/metabolism , Humans , Lentivirus/genetics , Mesenchymal Stem Cells/metabolism , Stem Cells/metabolism
2.
Radiat Res ; 181(5): 445-51, 2014 May.
Article En | MEDLINE | ID: mdl-24720754

Exposure to a nuclear accident or radiological attack can cause death from acute radiation syndrome (ARS), which results from radiation injury to vital organs such as the hematopoietic system. However, the U.S. Food and Drug Administration (FDA) has not approved any medical countermeasures for this specific purpose. With growing concern over nuclear terrorism, there is an urgent need to develop small molecule deliverables that mitigate mortality from ARS. One emerging modulator of hematopoietic stem/progenitor cell (HSPC) activity is glycogen synthase kinase-3 (GSK-3). The inhibition of GSK-3 has been shown to augment hematopoietic repopulation in mouse models of bone marrow transplantation. In this study, we performed an in vitro screen using irradiated bone marrow mononuclear cells (BM-MNCs) to test the effects of four GSK-3 inhibitors: CHIR99021; 6-Bromoindirubin-3'-oxime (BIO); SB415286; and SB216763. This screen showed that SB216763 significantly increased the frequency of c-Kit(+) Lin(-) Sca1(+) (KLS) cells and hematopoietic colony-forming cells in irradiated BM-MNCs. Importantly, administration of a single dose of SB216763 to C57BL/6J mice by subcutaneous injection 24 h after total-body irradiation significantly improved hematopoietic recovery and mitigated hematopoietic ARS. Collectively, our results demonstrate that the GSK-3 inhibitor SB216763 is an effective medical countermeasure against acute radiation injury of the hematopoietic system.


Acute Radiation Syndrome/drug therapy , Glycogen Synthase Kinase 3/antagonists & inhibitors , Hematopoietic Stem Cells/drug effects , Protein Kinase Inhibitors/therapeutic use , Radiation Injuries, Experimental/drug therapy , Acute Radiation Syndrome/enzymology , Acute Radiation Syndrome/pathology , Aminophenols/pharmacology , Aminophenols/therapeutic use , Animals , Apoptosis/drug effects , Apoptosis/radiation effects , Bone Marrow/pathology , Cells, Cultured , Colony-Forming Units Assay , Drug Evaluation, Preclinical , Glycogen Synthase Kinase 3/physiology , Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/radiation effects , Indoles/pharmacology , Indoles/therapeutic use , Injections, Subcutaneous , Maleimides/pharmacology , Maleimides/therapeutic use , Mice , Mice, Inbred C57BL , Oximes/pharmacology , Oximes/therapeutic use , Protein Kinase Inhibitors/pharmacology , Pyridines/pharmacology , Pyridines/therapeutic use , Pyrimidines/pharmacology , Pyrimidines/therapeutic use , Radiation Injuries, Experimental/enzymology , Radiation Injuries, Experimental/pathology , Whole-Body Irradiation/adverse effects
3.
J Leukoc Biol ; 94(6): 1243-51, 2013 Dec.
Article En | MEDLINE | ID: mdl-23935208

Chemokine receptor interactions coordinate leukocyte migration in inflammation. Chemokine receptors are GPCRs that when activated, are phosphorylated by GRKs to turn off G protein-mediated signaling yet recruit additional signaling machinery. Recently, GRK3 was identified as a negative regulator of CXCL12/CXCR4 signaling that is defective in human WHIM syndrome. Here, we report that GRK3-/- mice exhibit numerous features of human WHIM, such as impaired CXCL12-mediated desensitization, enhanced CXCR4 signaling to ERK activation, altered granulocyte migration, and a mild myelokathexis. Moreover, GRK3-/- protects mice from two acute models of inflammatory arthritis (K/BxN serum transfer and CAIA). In these granulocyte-dependent disease models, protection of GRK3-/- mice is mediated by retention of cells in the marrow, fewer circulating granulocytes in the peripheral blood, and reduced granulocytes in the joints during active inflammation. In contrast to WHIM, GRK3-/- mice have minimal hypogammaglobulinemia and a peripheral leukocytosis with increased lymphocytes and absent neutropenia. Thus, we conclude that the loss of GRK3-mediated regulation of CXCL12/CXCR4 signaling contributes to some, but not all, of the complete WHIM phenotype and that GRK3 inhibition may be beneficial in the treatment of inflammatory arthritis.


G-Protein-Coupled Receptor Kinase 3/immunology , Immunologic Deficiency Syndromes/immunology , MAP Kinase Signaling System/immunology , Warts/immunology , Animals , Cell Line, Transformed , Chemokine CXCL12/genetics , Chemokine CXCL12/immunology , Chemokine CXCL12/metabolism , Disease Models, Animal , G-Protein-Coupled Receptor Kinase 3/genetics , G-Protein-Coupled Receptor Kinase 3/metabolism , Granulocytes/enzymology , Granulocytes/immunology , Granulocytes/pathology , Humans , Immunologic Deficiency Syndromes/enzymology , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/pathology , Inflammation/enzymology , Inflammation/genetics , Inflammation/immunology , Inflammation/pathology , MAP Kinase Signaling System/genetics , Mice , Mice, Knockout , Primary Immunodeficiency Diseases , Receptors, CXCR4/genetics , Receptors, CXCR4/immunology , Receptors, CXCR4/metabolism , Warts/enzymology , Warts/genetics , Warts/pathology
4.
Nature ; 466(7307): 765-8, 2010 Aug 05.
Article En | MEDLINE | ID: mdl-20639863

Chronic myelogenous leukaemia (CML) can progress from a slow growing chronic phase to an aggressive blast crisis phase, but the molecular basis of this transition remains poorly understood. Here we have used mouse models of CML to show that disease progression is regulated by the Musashi-Numb signalling axis. Specifically, we find that the chronic phase is marked by high levels of Numb expression whereas the blast crisis phase has low levels of Numb expression, and that ectopic expression of Numb promotes differentiation and impairs advanced-phase disease in vivo. As a possible explanation for the decreased levels of Numb in the blast crisis phase, we show that NUP98-HOXA9, an oncogene associated with blast crisis CML, can trigger expression of the RNA-binding protein Musashi2 (Msi2), which in turn represses Numb. Notably, loss of Msi2 restores Numb expression and significantly impairs the development and propagation of blast crisis CML in vitro and in vivo. Finally we show that Msi2 expression is not only highly upregulated during human CML progression but is also an early indicator of poorer prognosis. These data show that the Musashi-Numb pathway can control the differentiation of CML cells, and raise the possibility that targeting this pathway may provide a new strategy for the therapy of aggressive leukaemias.


Cell Differentiation , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , RNA-Binding Proteins/metabolism , Animals , Blast Crisis/genetics , Blast Crisis/metabolism , Blast Crisis/pathology , Cell Differentiation/genetics , Disease Progression , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Gene Expression Regulation, Neoplastic , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Membrane Proteins/biosynthesis , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Nerve Tissue Proteins/biosynthesis , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Nuclear Pore Complex Proteins/genetics , Nuclear Pore Complex Proteins/metabolism , Oncogene Proteins, Fusion/genetics , Oncogene Proteins, Fusion/metabolism , Prognosis , RNA-Binding Proteins/biosynthesis , RNA-Binding Proteins/genetics , Receptor, Notch1/metabolism , Signal Transduction , Tumor Suppressor Protein p53/metabolism , Up-Regulation
5.
Nat Med ; 16(4): 475-82, 2010 Apr.
Article En | MEDLINE | ID: mdl-20305662

Hematopoietic stem cell (HSC) self-renewal is regulated by both intrinsic and extrinsic signals. Although some of the pathways that regulate HSC self-renewal have been uncovered, it remains largely unknown whether these pathways can be triggered by deliverable growth factors to induce HSC growth or regeneration. Here we show that pleiotrophin, a neurite outgrowth factor with no known function in hematopoiesis, efficiently promotes HSC expansion in vitro and HSC regeneration in vivo. Treatment of mouse bone marrow HSCs with pleiotrophin caused a marked increase in long-term repopulating HSC numbers in culture, as measured in competitive repopulating assays. Treatment of human cord blood CD34(+)CDCD38(-)Lin(-) cells with pleiotrophin also substantially increased severe combined immunodeficient (SCID)-repopulating cell counts in culture, compared to input and cytokine-treated cultures. Systemic administration of pleiotrophin to irradiated mice caused a pronounced expansion of bone marrow stem and progenitor cells in vivo, indicating that pleiotrophin is a regenerative growth factor for HSCs. Mechanistically, pleiotrophin activated phosphoinositide 3-kinase (PI3K) signaling in HSCs; antagonism of PI3K or Notch signaling inhibited pleiotrophin-mediated expansion of HSCs in culture. We identify the secreted growth factor pleiotrophin as a new regulator of both HSC expansion and regeneration.


Carrier Proteins/physiology , Cytokines/physiology , Hematopoietic Stem Cells/physiology , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Carrier Proteins/pharmacology , Cell Proliferation/drug effects , Cells, Cultured , Cytokines/pharmacology , Dose-Response Relationship, Drug , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred C57BL , Mice, SCID , Phosphatidylinositol 3-Kinases/metabolism , Phosphatidylinositol 3-Kinases/physiology , Receptors, Notch/physiology , Signal Transduction/drug effects , Signal Transduction/physiology , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology
...