Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Leukemia ; 25(10): 1525-32, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21647159

ABSTRACT

Notch is a crucial cell signaling pathway in metazoan development. By means of cell-cell interactions, Notch signaling regulates cellular identity, proliferation, differentiation and apoptosis. Within the last decade, numerous studies have shown an important role for this pathway in the development and homeostasis of mammalian stem cell populations. Hematopoietic stem cells (HSCs) constitute a well-defined population that shows self-renewal and multi-lineage differentiation potential, with the clinically relevant capacity to repopulate the hematopoietic system of an adult organism. Here, we review the emergence, development and maintenance of HSCs during mammalian embryogenesis and adulthood, with respect to the role of Notch signaling in hematopoietic biology.


Subject(s)
Hematopoietic Stem Cells/metabolism , Receptors, Notch/metabolism , Signal Transduction , Cell Lineage , Hematopoietic Stem Cells/cytology , Humans , Neovascularization, Physiologic
2.
J Exp Med ; 194(11): 1583-96, 2001 Dec 03.
Article in English | MEDLINE | ID: mdl-11733573

ABSTRACT

Signal transduction through the B cell antigen receptor (BCR) is determined by a balance of positive and negative regulators. This balance is shifted by aggregation that results from binding to extracellular ligand. Aggregation of the BCR is necessary for eliciting negative selection or activation by BCR-expressing B cells. However, ligand-independent signaling through intermediate and mature forms of the BCR has been postulated to regulate B cell development and peripheral homeostasis. To address the importance of ligand-independent BCR signaling functions and their regulation during B cell development, we have designed a model that allows us to isolate the basal signaling functions of immunoglobulin (Ig)alpha/Igbeta-containing BCR complexes from those that are dependent upon ligand-mediated aggregation. In vivo, we find that basal signaling is sufficient to facilitate pro-B --> pre-B cell transition and to generate immature/mature peripheral B cells. The ability to generate basal signals and to drive developmental progression were both dependent on plasma membrane association of Igalpha/Igbeta complexes and intact immunoregulatory tyrosine activation motifs (ITAM), thereby establishing a correlation between these processes. We believe that these studies are the first to directly demonstrate biologically relevant basal signaling through the BCR where the ability to interact with both conventional as well as nonconventional extracellular ligands is eliminated.


Subject(s)
Antigens, CD/immunology , B-Lymphocytes/cytology , Receptors, Antigen, B-Cell/immunology , Signal Transduction/immunology , Animals , Antigens, CD/genetics , B-Lymphocytes/immunology , CD79 Antigens , Cell Differentiation , Cell Membrane/immunology , HeLa Cells , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Ligands , Mice , Receptors, Antigen, B-Cell/genetics , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Tumor Cells, Cultured
3.
Curr Opin Hematol ; 8(4): 237-44, 2001 Jul.
Article in English | MEDLINE | ID: mdl-11561162

ABSTRACT

Mammalian Notch homologs were first identified from the involvement of Notch1 in a recurrent chromosomal translocation in a subset of human T-cell leukemias. The effect of the translocation was twofold: Notch expression was placed under the control of a T-cell-specific element, and Notch was truncated, resulting in a constitutively active protein. Subsequent work has shown that Notch1 is required for T cell commitment and is exclusively oncotropic for T cells. During the past year, several murine models have been used to dissect the function of Notch signaling in lymphoid development and leukemia. These models show that Notch1 drives the earliest stages of T cell commitment and that Notch signaling must be downregulated by the double positive stage for proper T cell development to occur. Constitutive Notch signaling mediated by Notch1, Notch2, or Notch3 predisposes to T-cell leukemia. Future studies are expected to elucidate the mechanisms by which Notch leads to transformation. Identification of the transcriptional targets of Notch signaling is likely to yield important insights.


Subject(s)
Leukemia-Lymphoma, Adult T-Cell/etiology , Membrane Proteins/physiology , Receptors, Cell Surface , Signal Transduction , Transcription Factors , Animals , Cell Transformation, Neoplastic , Hematopoietic Stem Cells/physiology , Humans , Leukemia, Experimental/etiology , Membrane Proteins/chemistry , Mice , Models, Biological , Protein Structure, Tertiary , Receptor, Notch1 , T-Lymphocytes/immunology
4.
J Exp Med ; 194(3): 275-84, 2001 Aug 06.
Article in English | MEDLINE | ID: mdl-11489947

ABSTRACT

p62(dok) has been identified as a substrate of many oncogenic tyrosine kinases such as the chronic myelogenous leukemia (CML) chimeric p210(bcr-abl) oncoprotein. It is also phosphorylated upon activation of many receptors and cytoplamic tyrosine kinases. However, the biological functions of p62(dok) in normal cell signaling as well as in p210(bcr-abl) leukemogenesis are as yet not fully understood. Here we show, in hemopoietic and nonhemopoietic cells derived from p62(dok)-(/)- mice, that the loss of p62(dok) results in increased cell proliferation upon growth factor treatment. Moreover, Ras and mitogen-activated protein kinase (MAPK) activation is markedly sustained in p62(dok)-(/)- cells after the removal of growth factor. However, p62(dok) inactivation does not affect DNA damage and growth factor deprivation-induced apoptosis. Furthermore, p62(dok) inactivation causes a significant shortening in the latency of the fatal myeloproliferative disease induced by retroviral-mediated transduction of p210(bcr-abl) in bone marrow cells. These data indicate that p62(dok) acts as a negative regulator of growth factor-induced cell proliferation, at least in part through downregulating Ras/MAPK signaling pathway, and that p62(dok) can oppose leukemogenesis by p210(bcr-abl).


Subject(s)
DNA-Binding Proteins , Fusion Proteins, bcr-abl/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/prevention & control , Mitogen-Activated Protein Kinases/metabolism , Phosphoproteins/metabolism , RNA-Binding Proteins , ras Proteins/metabolism , Animals , Cell Division , Cells, Cultured , Enzyme Activation , Gene Targeting , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mice, Knockout , Phosphoproteins/genetics , Signal Transduction
5.
J Immunol ; 167(3): 1387-92, 2001 Aug 01.
Article in English | MEDLINE | ID: mdl-11466357

ABSTRACT

B cells and dendritic cells (DCs) each develop from poorly described progenitor cells in the bone marrow (BM). Although a subset of DCs has been proposed to arise from lymphoid progenitors, a common developmental pathway for B cells and BM-derived DCs has not been clearly identified. To address this possibility, we performed a comprehensive analysis of DC differentiative potential among lymphoid and B lymphoid progenitor populations in adult mouse BM. We found that both the common lymphoid progenitors (CLPs), shown here and elsewhere to give rise exclusively to lymphocytes, and a down-stream early B-lineage precursor population devoid of T and NK cell precursor potential each give rise to DCs when exposed to the appropriate cytokines. This result contrasts with more mature B-lineage precursors, all of which failed to give rise to detectable numbers of DCs. Significantly, both CLP and early B-lineage-derived DCs acquired several surface markers associated with functional DCs, and CLP-derived DCs readily induced proliferation of allogeneic CD4(+) T cells. Surprisingly, however, DC differentiation from both lymphoid-restricted progenitors was accompanied by up-regulation of CD11b expression, a cell surface molecule normally restricted to myeloid lineage cells including putative myeloid DCs. Together, these data demonstrate that loss of DC developmental potential is the final step in B-lineage commitment and thus reveals a previously unrecognized link between early B cell and DC ontogeny.


Subject(s)
B-Lymphocyte Subsets/cytology , Dendritic Cells/cytology , Hyaluronan Receptors , Membrane Glycoproteins , Aging/immunology , Animals , B-Lymphocyte Subsets/immunology , B-Lymphocyte Subsets/metabolism , Bone Marrow Cells/cytology , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , CD4 Antigens/biosynthesis , Cell Differentiation/immunology , Cell Lineage/immunology , Cells, Cultured , Dendritic Cells/immunology , Dendritic Cells/metabolism , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Immunophenotyping , Leukocyte Common Antigens/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitochondrial Proteins , Receptors, Complement/biosynthesis , Receptors, Interleukin-7/biosynthesis
6.
J Exp Med ; 194(1): 99-106, 2001 Jul 02.
Article in English | MEDLINE | ID: mdl-11435476

ABSTRACT

Notch1 signaling is required for T cell development. We have previously demonstrated that expression of a dominant active Notch1 (ICN1) transgene in hematopoietic stem cells (HSCs) leads to thymic-independent development of CD4(+)CD8(+) double-positive (DP) T cells in the bone marrow (BM). To understand the function of Notch1 in early stages of T cell development, we assessed the ability of ICN1 to induce extrathymic T lineage commitment in BM progenitors from mice that varied in their capacity to form a functional pre-T cell receptor (TCR). Whereas mice repopulated with ICN1 transduced HSCs from either recombinase deficient (Rag-2(-/)-) or Src homology 2 domain--containing leukocyte protein of 76 kD (SLP-76)(-/)- mice failed to develop DP BM cells, recipients of ICN1-transduced Rag-2(-/)- progenitors contained two novel BM cell populations indicative of pre-DP T cell development. These novel BM populations are characterized by their expression of CD3 epsilon and pre-T alpha mRNA and the surface proteins CD44 and CD25. In contrast, complementation of Rag-2(-/)- mice with a TCR beta transgene restored ICN1-induced DP development in the BM within 3 wk after BM transfer (BMT). At later time points, this population selectively and consistently gave rise to T cell leukemia. These findings demonstrate that Notch signaling directs T lineage commitment from multipotent progenitor cells; however, both expansion and leukemic transformation of this population are dependent on T cell-specific signals associated with development of DP thymocytes.


Subject(s)
DNA-Binding Proteins/genetics , Membrane Proteins/genetics , Membrane Proteins/metabolism , Receptors, Cell Surface , T-Lymphocytes/physiology , Transcription Factors , Animals , Bone Marrow/physiology , Cell Lineage , DNA-Binding Proteins/metabolism , Hematopoietic Stem Cells/physiology , Hyaluronan Receptors/genetics , Hyaluronan Receptors/metabolism , Leukemia, T-Cell/genetics , Mice , Mice, Transgenic , Receptor, Notch1 , Receptors, Antigen, T-Cell, alpha-beta/genetics , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Interleukin-2/genetics , Receptors, Interleukin-2/metabolism , Signal Transduction , Thymus Gland/cytology
7.
Immunity ; 14(3): 253-64, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11290335

ABSTRACT

Notch signaling regulates cell fate decisions in multiple lineages. We demonstrate in this report that retroviral expression of activated Notch1 in mouse thymocytes abrogates differentiation of immature CD4+CD8+ thymocytes into both CD4 and CD8 mature single-positive T cells. The ability of Notch1 to inhibit T cell development was observed in vitro and in vivo with both normal and TCR transgenic thymocytes. Notch1-mediated developmental arrest was dose dependent and was associated with impaired thymocyte responses to TCR stimulation. Notch1 also inhibited TCR-mediated signaling in Jurkat T cells. These data indicate that constitutively active Notch1 abrogates CD4+ and CD8+ maturation by interfering with TCR signal strength and provide an explanation for the physiological regulation of Notch expression during thymocyte development.


Subject(s)
CD4-Positive T-Lymphocytes/cytology , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Membrane Proteins/metabolism , Nuclear Proteins , Receptors, Antigen, T-Cell/metabolism , Receptors, Cell Surface , Animals , Antigens, CD/metabolism , Antigens, Differentiation, T-Lymphocyte/metabolism , CD4-Positive T-Lymphocytes/immunology , CD5 Antigens/metabolism , CD8-Positive T-Lymphocytes/immunology , DNA-Binding Proteins/metabolism , Flow Cytometry , Gene Expression Regulation , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Jurkat Cells , Lectins, C-Type , Liver/cytology , Liver/embryology , Membrane Proteins/genetics , Mice , Mice, Transgenic , NFATC Transcription Factors , Promoter Regions, Genetic/genetics , Receptor, Notch1 , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/immunology , Response Elements/genetics , Signal Transduction , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism , Transcription Factor AP-1/metabolism , Transcription Factors/metabolism
8.
Blood ; 97(1): 277-87, 2001 Jan 01.
Article in English | MEDLINE | ID: mdl-11133772

ABSTRACT

Bcr-Abl plays a critical role in the pathogenesis of chronic myelogenous leukemia (CML). It was previously shown that expression of Bcr-Abl in bone marrow cells by retroviral transduction efficiently induces a myeloproliferative disorder (MPD) in mice resembling human CML. This in vivo experimental system allows the direct determination of the effect of specific domains of Bcr-Abl, or specific signaling pathways, on the complex in vivo pathogenesis of CML. In this report, the function of the SH2 domain of Bcr-Abl in the pathogenesis of CML is examined using this murine model. It was found that the Bcr-Abl SH2 mutants retain the ability to induce a fatal MPD but with an extended latency compared with wild type (wt) Bcr-Abl. Interestingly, in contrast to wt Bcr-Abl-induced disease, which is rapid and monophasic, the disease caused by the Bcr-Abl SH2 mutants is biphasic, consisting of an initial B-lymphocyte expansion followed by a fatal myeloid proliferation. The B-lymphoid expansion was diminished in mixing experiments with bcr-abl/DeltaSH2 and wt bcr-abl cells, suggesting that the Bcr-Abl-induced MPD suppresses B-lymphoid expansion.


Subject(s)
Fusion Proteins, bcr-abl/pharmacology , Myeloproliferative Disorders/etiology , src Homology Domains/physiology , 3T3 Cells , Animals , Bone Marrow Transplantation , Disease Models, Animal , Fusion Proteins, bcr-abl/chemistry , Fusion Proteins, bcr-abl/genetics , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Interleukin-3/biosynthesis , Leukemia, B-Cell/chemically induced , Leukemia, B-Cell/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/chemically induced , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/etiology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Mice , Mutagenesis, Site-Directed , Myeloproliferative Disorders/chemically induced , Myeloproliferative Disorders/metabolism , Neoplasm Transplantation/methods , Retroviridae , Transduction, Genetic , src Homology Domains/genetics
9.
Nat Med ; 6(11): 1278-81, 2000 Nov.
Article in English | MEDLINE | ID: mdl-11062542

ABSTRACT

Hematopoietic stem cells give rise to progeny that either self-renew in an undifferentiated state or lose self-renewal capabilities and commit to lymphoid or myeloid lineages. Here we evaluated whether hematopoietic stem cell self-renewal is affected by the Notch pathway. Notch signaling controls cell fate choices in both invertebrates and vertebrates by inhibiting certain differentiation pathways, thereby permitting cells to either differentiate along an alternative pathway or to self-renew. Notch receptors are present in hematopoietic precursors and Notch signaling enhances the in vitro generation of human and mouse hematopoietic precursors, determines T- or B-cell lineage specification from a common lymphoid precursor and promotes expansion of CD8(+) cells. Here, we demonstrate that constitutive Notch1 signaling in hematopoietic cells established immortalized, cytokine-dependent cell lines that generated progeny with either lymphoid or myeloid characteristics both in vitro and in vivo. These data support a role for Notch signaling in regulating hematopoietic stem cell self-renewal. Furthermore, the establishment of clonal, pluripotent cell lines provides the opportunity to assess mechanisms regulating stem cell commitment and demonstrates a general method for immortalizing stem cell populations for further analysis.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Membrane Proteins/physiology , Receptors, Cell Surface , Signal Transduction , Transcription Factors , Animals , B-Lymphocytes/immunology , Bone Marrow Cells/cytology , Cell Line, Transformed , Cells, Cultured , Cytokines/pharmacology , Gamma Rays , Humans , Hypoxanthine Phosphoribosyltransferase/genetics , Interleukin-11/pharmacology , Leukopoiesis , Mice , Mice, Inbred C57BL , Receptor, Notch1 , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/immunology , Thymus Gland/immunology , Transfection
10.
Proc Natl Acad Sci U S A ; 97(22): 12056-61, 2000 Oct 24.
Article in English | MEDLINE | ID: mdl-11050236

ABSTRACT

Mice deficient in the hematopoietic cell-specific adapter protein SLP-76 demonstrate a failure of T cell development and fetal hemorrhage. Although SLP-76-deficient platelets manifest defective collagen receptor signaling, this alone may not explain the observed bleeding diathesis. Because alpha IIb beta 3, the platelet fibrinogen receptor, is required for normal hemostasis, we explored a potential role for SLP-76 in alpha IIb beta 3 signaling. Interaction of soluble or immobilized fibrinogen with normal human or murine platelets triggers rapid tyrosine phosphorylation of SLP-76. Moreover, platelet adhesion to fibrinogen stimulates actin rearrangements, filopodial and lamellipodial extension, and localization of tyrosine phosphorylated proteins to the cell periphery. In contrast, SLP-76-deficient murine platelets bind fibrinogen normally, but spread poorly and exhibit reduced levels of phosphotyrosine. The in vivo bleeding diathesis as well as the defects in platelet responses to fibrinogen and collagen are reversed by retroviral transduction of SLP-76 into bone marrow derived from SLP-76-deficient mice. These studies establish that SLP-76 functions downstream of alpha IIb beta 3 and collagen receptors in platelets. Furthermore, expression of SLP-76 in hematopoietic cells, including platelets, plays a necessary role in hemostasis.


Subject(s)
Blood Platelets/physiology , Hematopoiesis , Hemostasis/physiology , Integrins/physiology , Phosphoproteins/physiology , Platelet Glycoprotein GPIIb-IIIa Complex/physiology , Signal Transduction/physiology , Adaptor Proteins, Signal Transducing , Animals , Fibrinogen/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Phosphorylation , Protein Binding , Receptors, Collagen , Tyrosine/metabolism
11.
Mol Cell Biol ; 20(20): 7505-15, 2000 Oct.
Article in English | MEDLINE | ID: mdl-11003647

ABSTRACT

Notch receptors participate in a conserved signaling pathway that controls the development of diverse tissues and cell types, including lymphoid cells. Signaling is normally initiated through one or more ligand-mediated proteolytic cleavages that permit nuclear translocation of the intracellular portion of the Notch receptor (ICN), which then binds and activates transcription factors of the Su(H)/CBF1 family. Several mammalian Notch receptors are oncogenic when constitutively active, including Notch1, a gene initially identified based on its involvement in a (7;9) chromosomal translocation found in sporadic T-cell lymphoblastic leukemias and lymphomas (T-ALL). To investigate which portions of ICN1 contribute to transformation, we performed a structure-transformation analysis using a robust murine bone marrow reconstitution assay. Both the ankyrin repeat and C-terminal transactivation domains were required for T-cell leukemogenesis, whereas the N-terminal RAM domain and a C-terminal domain that includes a PEST sequence were nonessential. Induction of T-ALL correlated with the transactivation activity of each Notch1 polypeptide when fused to the DNA-binding domain of GAL4, with the exception of polypeptides deleted of the ankyrin repeats, which lacked transforming activity while retaining strong transactivation activity. Transforming polypeptides also demonstrated moderate to strong activation of the Su(H)/CBF1-sensitive HES-1 promoter, while polypeptides with weak or absent activity on this promoter failed to cause leukemia. These experiments define a minimal transforming region for Notch1 in T-cell progenitors and suggest that leukemogenic signaling involves recruitment of transcriptional coactivators to ICN1 nuclear complexes.


Subject(s)
Ankyrin Repeat , Cell Transformation, Neoplastic/pathology , Leukemia, T-Cell/pathology , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Receptors, Cell Surface , Transcription Factors , Transcriptional Activation , Animals , Bone Marrow Transplantation , Cell Transformation, Neoplastic/metabolism , Clone Cells , Female , Gene Expression Regulation, Neoplastic , Humans , Immunophenotyping , Leukemia, T-Cell/genetics , Leukemia, T-Cell/metabolism , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Peptide Fragments/genetics , Protein Structure, Tertiary , Receptor, Notch1 , Recombinant Fusion Proteins , Retroviridae/genetics , Sequence Deletion , Transfection
12.
Blood ; 95(9): 2913-21, 2000 May 01.
Article in English | MEDLINE | ID: mdl-10779439

ABSTRACT

The reciprocal translocation between chromosomes 9 and 22 that fuses coding sequences of the Bcr and Abl genes is responsible for a remarkably diverse group of hematologic malignancies. A newly described 230-kd form of Bcr-Abl has been associated with an indolent myeloproliferative syndrome referred to as chronic neutrophilic leukemia. We have cloned the corresponding gene and examined the biologic and biochemical properties of p230 Bcr-Abl after retroviral-mediated gene transfer into hematopoietic cell lines and primary bone marrow cells. p230 Bcr-Abl-expressing 32D myeloid cells were fully growth factor-independent and activated similar signal transduction pathways as the well-characterized p210 and p185 forms of Bcr-Abl. In contrast, primary mouse bone marrow cells expressing p230 required exogenous hematopoietic growth factors for optimal growth, whereas p185- and p210-expressing cells were independent of growth factors. The 3 Bcr-Abl proteins exerted different effects on differentiation of bone marrow cells. p185 induced outgrowth of lymphoid precursors capable of tumor formation in immunodeficient mice. In contrast, p210- and p230-expressing bone marrow cells caused limited outgrowth of lymphoid precursors that failed to form tumors in immunodeficient mice. Removal of cytokines and autologous stroma from Bcr-Abl-expressing bone marrow cultures produced the expansion of distinct lineages by the various Bcr-Abl proteins. p185 drove expansion of cytokine-independent lymphoid progenitors, while p210 and p230 generated cytokine-independent monocyte/myeloid cells. These findings suggest that the different Bcr-Abl fusion proteins drive the expansion of different hematopoietic populations, which may explain the association of the various Bcr-Abl oncoproteins with different spectra of human leukemias. (Blood. 2000;95:2913-2921)


Subject(s)
Bone Marrow Cells/physiology , Fusion Proteins, bcr-abl/genetics , Fusion Proteins, bcr-abl/metabolism , Hematopoietic Stem Cells/physiology , Protein-Tyrosine Kinases/metabolism , Animals , Antigens, Differentiation/analysis , Bone Marrow Cells/cytology , Cell Cycle , Cell Line , Cells, Cultured , Cloning, Molecular , Genes, abl , Green Fluorescent Proteins , Hematopoietic Stem Cells/cytology , Humans , Luminescent Proteins/genetics , Mice , Oncogenes , Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins/biosynthesis , Transfection , Tumor Cells, Cultured
14.
Immunity ; 11(3): 299-308, 1999 Sep.
Article in English | MEDLINE | ID: mdl-10514008

ABSTRACT

Notch receptors regulate fate decisions in many cells. One outcome of Notch signaling is differentiation of bipotential precursors into one cell type versus another. To investigate consequences of Notch1 expression in hematolymphoid progenitors, mice were reconstituted with bone marrow (BM) transduced with retroviruses encoding a constitutively active form of Notch1. Although neither granulocyte or monocyte differentiation were appreciably affected, lymphopoiesis was dramatically altered. As early as 3 weeks following transplantation, mice receiving activated Notch1-transduced BM contained immature CD4+ CD8+ T cells in the BM and exhibited a simultaneous block in early B cell lymphopoiesis. These results suggest that Notch1 provides a key regulatory signal in determining T lymphoid versus B lymphoid lineage decisions, possibly by influencing lineage commitment from a common lymphoid progenitor cell.


Subject(s)
B-Lymphocytes/cytology , Membrane Proteins/metabolism , Receptors, Cell Surface , T-Lymphocytes/cytology , Transcription Factors , Animals , Bone Marrow Cells/cytology , Cell Differentiation , Cell Lineage , Female , Gene Expression , Granulocytes/cytology , Humans , Leukopoiesis , Macrophages/cytology , Membrane Proteins/genetics , Mice , Mice, Inbred BALB C , Receptor, Notch1 , Transcriptional Activation
15.
J Immunol ; 162(2): 635-8, 1999 Jan 15.
Article in English | MEDLINE | ID: mdl-9916679

ABSTRACT

The Notch receptor protein was originally identified in Drosophila and is known to mediate cell to cell communication and influence cell fate decisions. Members of this family have been isolated from invertebrates as well as vertebrates. We isolated mouse Notch-1 in a yeast two-hybrid screen with Nur77, which is a protein that has been shown previously to be required for apoptosis in T cell lines. The data presented below indicate that Notch-1 expression provides significant protection to T cell lines from TCR-mediated apoptosis. These data demonstrate a new antiapoptotic role for Notch-1, providing evidence that, in addition to regulating cell fate decisions, Notch-1 can play a critical role in controlling levels of cell death in T cells.


Subject(s)
Apoptosis/immunology , Membrane Proteins/physiology , Receptors, Antigen, T-Cell/physiology , Receptors, Cell Surface , Animals , Cell Death/genetics , Cell Death/immunology , Cell Line , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/physiology , Humans , Hybrid Cells , Lymphoma, T-Cell , Mice , Nuclear Receptor Subfamily 4, Group A, Member 1 , Receptor, Notch1 , Receptors, Cytoplasmic and Nuclear , Receptors, Steroid , Retroviridae/genetics , Saccharomyces cerevisiae/genetics , T-Lymphocytes/metabolism , Transcription Factors/biosynthesis , Transcription Factors/genetics , Transcription Factors/physiology , Tumor Cells, Cultured
16.
Blood ; 92(10): 3780-92, 1998 Nov 15.
Article in English | MEDLINE | ID: mdl-9808572

ABSTRACT

Expression of the 210-kD bcr/abl fusion oncoprotein can cause a chronic myelogenous leukemia (CML)-like disease in mice receiving bone marrow cells transduced by bcr/abl-encoding retroviruses. However, previous methods failed to yield this disease at a frequency sufficient enough to allow for its use in the study of CML pathogenesis. To overcome this limitation, we have developed an efficient and reproducible method for inducing a CML-like disease in mice receiving P210 bcr/abl-transduced bone marrow cells. All mice receiving P210 bcr/abl-transduced bone marrow cells succumb to a myeloproliferative disease between 3 and 5 weeks after bone marrow transplantation. The myeloproliferative disease recapitulates many of the hallmarks of human CML and is characterized by high white blood cell counts and extensive extramedullary hematopoiesis in the spleen, liver, bone marrow, and lungs. Use of a retroviral vector coexpressing P210 bcr/abl and green fluorescent protein shows that the vast majority of bcr/abl-expressing cells are myeloid. Analysis of the proviral integration pattern shows that, in some mice, the myeloproliferative disease is clonal. In multiple mice, the CML-like disease has been transplantable, inducing a similar myeloproliferative syndrome within 1 month of transfer to sublethally irradiated syngeneic recipients. The disease in many of these mice has progressed to the development of acute lymphoma/leukemia resembling blast crisis. These results demonstrate that murine CML recapitulates important features of human CML. As such, it should be an excellent model for addressing specific issues relating to the pathogenesis and treatment of this disease.


Subject(s)
Bone Marrow Transplantation , Disease Models, Animal , Fusion Proteins, bcr-abl/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Myeloproliferative Disorders/etiology , Oncogenes , Animals , Blast Crisis/pathology , Bone Marrow Cells/virology , Cells, Cultured/transplantation , Clone Cells/pathology , Fusion Proteins, bcr-abl/analysis , Genes, abl , Genetic Vectors/genetics , Green Fluorescent Proteins , Hematopoiesis, Extramedullary , Humans , Luminescent Proteins/genetics , Mice , Mice, Inbred BALB C , Myeloproliferative Disorders/genetics , Myeloproliferative Disorders/pathology , Neoplasm Transplantation , Proviruses/genetics , Radiation Chimera , Recombinant Fusion Proteins/analysis , Reproducibility of Results , Retroviridae/genetics , Splenomegaly/pathology , Transfection , Virus Integration
17.
Methods Mol Med ; 7: 41-57, 1997.
Article in English | MEDLINE | ID: mdl-24493417

ABSTRACT

Retroviral gene transfer is presently one of the most powerful techniques for introducing stably heritable genetic material into mammalian cells (reviewed in ref. 1). One serious drawback of this technique, however, has been the difficulty in readily producing high-titer recombinant retroviruses. For many applications, such as infecting rare target cells or the majority of cells in tissue culture, the recombinant virus titer must be at least 10(6) infectious units/mL. Although one can usually obtain high-titer mixtures of recombinant and replication-competent retroviruses in a relatively short time, many applications such as cell marking studies or studying genes in vivo demand freedom from replication-competent virus.

18.
J Exp Med ; 183(5): 2283-91, 1996 May 01.
Article in English | MEDLINE | ID: mdl-8642337

ABSTRACT

Notch is a highly conserved transmembrane protein that is involved in cell fate decisions and is found in organisms ranging from Drosophila to humans. A human homologue of Notch, TAN1, was initially identified at the chromosomal breakpoint of a subset of T-cell lymphoblastic leukemias/lymphomas containing a t(7;9) chromosomal translocation; however, its role in oncogenesis has been unclear. Using a bone marrow reconstitution assay with cells containing retrovirally transduced TAN1 alleles, we analyzed the oncogenic potential of both nuclear and extranuclear forms of truncated TAN1 in hematopoietic cells. Although the Moloney leukemia virus long terminal repeat drives expression in most hematopoietic cell types, retroviruses encoding either form of the TAN1 protein induced clonal leukemias of exclusively immature T cell phenotypes in approximately 50% of transplanted animals. All tumors overexpressed truncated TAN1 of the size and subcellular localization predicted from the structure of the gene. These results show that TAN1 is an oncoprotein and suggest that truncation and overexpression are important determinants of transforming activity. Moreover, the murine tumors caused by TAN1 in the bone marrow transplant model are very similar to the TAN1-associated human tumors and suggest that TAN1 may be specifically oncotropic for T cells.


Subject(s)
Bone Marrow Transplantation , Bone Marrow/pathology , Leukemia, T-Cell/pathology , Lymphoma, T-Cell/pathology , Membrane Proteins/biosynthesis , Receptors, Cell Surface , Transcription Factors , Animals , Bone Marrow Cells , Chromosomes, Human, Pair 7 , Chromosomes, Human, Pair 9 , Drosophila , Drosophila Proteins , Gene Rearrangement, beta-Chain T-Cell Antigen Receptor , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Leukemia, T-Cell/genetics , Lymphoma, T-Cell/genetics , Mice , Moloney murine leukemia virus/genetics , Receptor, Notch1 , Receptors, Notch , Repetitive Sequences, Nucleic Acid , Sequence Deletion , Translocation, Genetic , Transplantation, Isogeneic , Virus Integration
19.
Immunity ; 3(2): 175-86, 1995 Aug.
Article in English | MEDLINE | ID: mdl-7648391

ABSTRACT

An immature v-abl-transformed mast cell line (V3-MC) was derived from a mouse that developed systemic mastocytosis after transplantation of v-abl-infected bone marrow cells. V3-MCs injected intravenously into adult BALB/c mice infiltrated the liver, spleen, and intestine by day 6 and underwent progressive differentiation and maturation, eventually resembling indigenous mast cells. In terms of their protease content, the V3-MCs that localized in the liver and spleen differed from those in the intestine, and both differed from the cultured V3-MCs. The acquired expression of certain proteases and the loss of expression of other proteases in these tissue V3-MCs defines particular phenotypes and indicates that the differentiation and maturation of mast cell-committed progenitor cells are primarily regulated by factors in the different tissue microenvironments.


Subject(s)
Genes, abl , Mast Cells/cytology , Mastocytosis/pathology , Oncogene Proteins, Viral/physiology , Animals , Base Sequence , Bone Marrow Cells , Cell Differentiation , DNA Primers/chemistry , Gene Expression , Immunohistochemistry , Mice , Mice, Inbred BALB C , Molecular Sequence Data , RNA, Messenger/genetics , Serine Endopeptidases/genetics , Serine Endopeptidases/metabolism , Tissue Distribution
20.
Proc Natl Acad Sci U S A ; 90(18): 8392-6, 1993 Sep 15.
Article in English | MEDLINE | ID: mdl-7690960

ABSTRACT

The generation of high-titer, helper-free retroviruses by transient transfection has been achieved by using the highly transfectable 293T cell line into which are stably introduced constructs that express retroviral packaging functions. The resulting ecotropic virus packaging cell line BOSC 23 produces infectious retrovirus at > 10(6) infectious units/ml of supernatant within 72 hr after CaPO4-mediated transfection. A stringent assay for replication-competent virus showed that no helper virus was present. The system can produce high titers of retroviral vectors expressing genes that are extremely difficult to propagate at high titer in stable producer lines. This method should facilitate and extend the use of helper-free retroviral gene transfer, as well as be useful for gene therapy.


Subject(s)
Helper Viruses/physiology , Retroviridae/physiology , Transfection , Virus Replication , 3T3 Cells , Animals , Bone Marrow Transplantation/physiology , Cell Line , Cell Transformation, Neoplastic , Chloroquine/pharmacology , Female , Genes, abl , Humans , Male , Mice , Mice, Inbred C57BL , Plasminogen Activators/biosynthesis , RNA-Directed DNA Polymerase/analysis , Repetitive Sequences, Nucleic Acid , Retroviridae/drug effects , Retroviridae/genetics , Virus Replication/drug effects , beta-Galactosidase/biosynthesis , beta-Galactosidase/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...