Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 87
Filter
1.
Mol Cell Endocrinol ; 288(1-2): 6-10, 2008 Jun 25.
Article in English | MEDLINE | ID: mdl-18400373

ABSTRACT

Hematopoietic stem cells (HSC) capable of both self-renewal and differentiation into all blood lineages reside within the bone marrow in specialized microenvironmental niches. While the precise location and composition of these niches largely remains unknown, it is now believed that osteoblasts at the endosteal surface play critical roles. Among the molecules demonstrated to influence the function of these niches are parathyroid hormone (PTH) and the insulin-like growth factors (IGF). Administration of PTH to both mice and men expands the number of bone marrow HSC, and an increase in the number of those cells in peripheral blood following treatment with mobilizing agents. Several molecules downstream of PTH are capable of signaling to HSC, including IGF that appear to regulate both the survival and expansion of hematopoietic stem and progenitor cells. As our current understanding of the role for PTH and IGF in hematopoietic niches is limited, we believe it is important that both their physiological importance and pharmacological potential be more fully investigated.


Subject(s)
Hematopoiesis/drug effects , Hematopoiesis/physiology , Parathyroid Hormone/pharmacology , Somatomedins/pharmacology , Animals , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Humans , Osteoblasts/cytology , Osteoblasts/drug effects , Phenotype
2.
Bone Marrow Transplant ; 35(9): 921-6, 2005 May.
Article in English | MEDLINE | ID: mdl-15765118

ABSTRACT

Nonmyeloablative stem cell transplantation (NST) harnesses the graft-versus-tumor effect while minimizing regimen-related toxicity, and can result in donor chimerism and remission. Acute graft-versus-host disease (GVHD) and infections are major complications after sibling NST. Toxicity of unrelated-donor (UD) NST and the most appropriate GVHD prophylaxis in this setting remain poorly defined. We describe 25 patients who received UD-NST conditioned with fludarabine and cyclophosphamide. The first six patients received cyclosporine (Cs) and mycophenolate mofetil (MMF) (n=5) or methotrexate (MTX) (n=1) as GVHD prophylaxis (group 1) and all developed grade III-IV acute GVHD. The next 19 patients received the same conditioning regimen with the addition of alemtuzumab, and all received Cs/MTX post-transplant. Engraftment and donor chimerism were achieved in all but one evaluable patient. In all, 15 patients died: five of six deaths in group 1 were attributable to acute GVHD, while deaths in group 2 were due to infection or progressive disease (P=0.05). The combination of Cs/MMF is inadequate GVHD prophylaxis for UD-NST. The use of Cs, MTX, and alemtuzumab eliminated severe acute GVHD; its impact on response merits further study.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation , Lymphoproliferative Disorders/therapy , Mycophenolic Acid/analogs & derivatives , Transplantation Conditioning , Vidarabine/analogs & derivatives , Adult , Alemtuzumab , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal, Humanized , Antibodies, Neoplasm/administration & dosage , Cyclophosphamide/administration & dosage , Female , Graft vs Host Disease/complications , Graft vs Host Disease/mortality , Humans , Lymphoproliferative Disorders/complications , Lymphoproliferative Disorders/mortality , Male , Middle Aged , Mycophenolic Acid/administration & dosage , Transplantation Chimera , Transplantation Conditioning/methods , Vidarabine/administration & dosage
3.
Bone Marrow Transplant ; 31(4): 241-52, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12621458

ABSTRACT

In the past decade, we have witnessed significant advances in ex vivo hematopoietic stem cell culture expansion, progressing to the point where clinical trials are being designed and conducted. Preclinical milestone investigations provided data to enable expansion of portions of hematopoietic grafts in a clinical setting, indicating safety and feasibility of this approach. Data derived from current clinical trials indicate successful reconstitution of hematopoiesis after myeloablative chemoradiotherapy using infusion of ex vivo-expanded perfusion cultures. Future avenues of exploration will focus upon refining preclinical and clinical studies in which cocktails of available cytokines, novel molecules and sophisticated expansion systems will explore expansion of blood, marrow and umbilical cord blood cells.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Mobilization/trends , Stem Cell Transplantation/methods , Cell Culture Techniques/methods , Hematopoietic Stem Cells/cytology , Humans , Stem Cell Transplantation/trends , Tissue Donors
4.
Blood ; 98(10): 3143-9, 2001 Nov 15.
Article in English | MEDLINE | ID: mdl-11698303

ABSTRACT

Because human CD34+ and murine Sca-1+ hematopoietic stem-progenitor cells (HSPCs) express platelet-binding sialomucin P-selectin (CD162) and integrin Mac-1 (CD11b-CD18) antigen, it was inferred that these cells might interact with platelets. As a result of this interaction, microparticles derived from platelets (PMPs) may transfer many platelet antigens (CD41, CD61, CD62, CXCR4, PAR-1) to the surfaces of HSPCs. To determine the biologic significance of the presence of PMPs on human CD34+ and murine Sca-1+ cells, their expressions on mobilized peripheral blood (mPB) and on nonmobilized PB- and bone marrow (BM)-derived CD34+ cells were compared. In addition, the effects of PMPs on the proliferation of CD34+ and Sca-1+ cells and on adhesion of HSPCs to endothelium and immobilized SDF-1 were studied. Finally, the hematopoietic reconstitution of lethally irradiated mice receiving transplanted BM mononuclear cells covered or not covered with PMPs was examined. It was found that PMPs are more numerous on mPB than on BM CD34+ cells, do not affect the clonogenicity of human and murine HSPCs, and increase adhesion of these cells to endothelium and immobilized SDF-1. Moreover, murine BM cells covered with PMPs engrafted lethally irradiated mice significantly faster than those not covered, indicating that PMPs play an important role in the homing of HSPCs. This could explain why in a clinical setting human mPB HSPCs (densely covered with PMPs) engraft more rapidly than BM HSPCs (covered with fewer PMPs). These findings indicate a new role for PMPs in stem cell transplantation and may have clinical implications for the optimization of transplantations.


Subject(s)
Blood Platelets/metabolism , Graft Survival , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/metabolism , Animals , Antigens, CD34/analysis , Antigens, Human Platelet/metabolism , Antigens, Ly/analysis , Blood Platelets/ultrastructure , Bone Marrow Cells/metabolism , Cell Adhesion , Cell Division , Cell Membrane/metabolism , Cells, Cultured , Chemokine CXCL12 , Chemokines, CXC/metabolism , Colony-Forming Units Assay , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Female , HL-60 Cells , Hematopoietic Stem Cell Mobilization , Humans , Membrane Glycoproteins/analysis , Membrane Proteins/analysis , Mice , Mice, Inbred C57BL , Platelet Activation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Radiation Chimera , Time Factors , Umbilical Veins
6.
Blood ; 98(12): 3367-75, 2001 Dec 01.
Article in English | MEDLINE | ID: mdl-11719376

ABSTRACT

Graft-versus-host disease (GVHD) is a major cause of morbidity and mortality of allogeneic stem cell transplantation. Strategies to control GVHD while maintaining graft versus leukemia (GVL) include herpes simplex virus thymidine kinase (HSV-tk) gene transduction of donor T cells followed by treatment with ganciclovir (GCV). Alternatively, GVHD and GVL may be mediated by distinct processes. In this regard, whether cytokine polarization occurs and to what degrees various subsets of cytokine-producing T cells mediate GVHD or GVL has been an active area of research using cytokine or cytokine antibody infusion or genetically deficient mice. This study takes a different approach that allows simultaneous investigation into both the mechanisms underlying GVHD reactions and the efficacy of HSV-tk suicide gene-based T-cell deletion. A source of donor T cells, splenocytes from mice transgenic for HSV-tk controlled by elements of either the interleukin-2 (IL-2) or IL-4 promoters (IL-2-tk and IL-4-tk, respectively) was used, thus allowing investigation into the roles of T1 and T2 cells in ongoing GVHD reactions. To assess treatment rather than prevention of GVHD, GCV was started at peak disease. Remarkably, treatment at this late time point rescued mice from the clinical effects of GVHD caused by T cells expressing either transgene. Thus, both T1 and T2 cells play an important role in clinical GVHD in a minor histocompatibility antigen-mismatched setting. In addition, because clinical disease was reversible even at its maximum, these observations provide controlled evidence that this strategy of treating ongoing GVHD could be effective clinically.


Subject(s)
Disease Models, Animal , Graft vs Host Disease/immunology , Lymphocyte Subsets/immunology , T-Lymphocytes/immunology , Animals , Cell Death/genetics , Cytokines/biosynthesis , Cytokines/metabolism , Flow Cytometry , Ganciclovir/therapeutic use , Graft vs Host Disease/drug therapy , Graft vs Host Disease/pathology , Graft vs Leukemia Effect/immunology , Hematopoiesis , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Interleukin-2/genetics , Interleukin-4/genetics , Male , Mice , Mice, Transgenic , Promoter Regions, Genetic , Simplexvirus/enzymology , Spleen/cytology , Thymidine Kinase/genetics , Thymus Gland/cytology , Weight Gain
7.
Blood ; 97(10): 3075-85, 2001 May 15.
Article in English | MEDLINE | ID: mdl-11342433

ABSTRACT

The aim of this study was to explore further the hypothesis that early stages of normal human hematopoiesis might be coregulated by autocrine/paracrine regulatory loops and by cross-talk among early hematopoietic cells. Highly purified normal human CD34(+) cells and ex vivo expanded early colony-forming unit-granulocyte-macrophage (CFU-GM)-derived, burst forming unit-erythroid (BFU-E)-derived, and CFU-megakaryocyte (CFU-Meg)-derived cells were phenotyped for messenger RNA expression and protein secretion of various growth factors, cytokines, and chemokines to determine the biological significance of this secretion. Transcripts were found for numerous growth factors (kit ligand [KL], FLT3 ligand, fibroblast growth factor-2 [FGF-2], vascular endothelial growth factor [VEGF], hepatocyte growth factor [HGF], insulinlike growth factor-1 [IGF-1], and thrombopoietin [TPO]); cytokines (tumor necrosis factor-alpha, Fas ligand, interferon alpha, interleukin 1 [IL-1], and IL-16); and chemokines (macrophage inflammatory protein-1alpha [MIP-1alpha], MIP-1beta, regulated upon activation, normal T cell expressed and secreted [RANTES], monocyte chemotactic protein-3 [MCP-3], MCP-4, IL-8, interferon-inducible protein-10, macrophage-derived chemokine [MDC], and platelet factor-4 [PF-4]) to be expressed by CD34(+) cells. More importantly, the regulatory proteins VEGF, HGF, FGF-2, KL, FLT3 ligand, TPO, IL-16, IGF-1, transforming growth factor-beta1 (TGF-beta1), TGF-beta2, RANTES, MIP-1alpha, MIP-1beta, IL-8, and PF-4 were identified in media conditioned by these cells. Moreover, media conditioned by CD34(+) cells were found to inhibit apoptosis and slightly stimulate the proliferation of other freshly isolated CD34(+) cells; chemo-attract CFU-GM- and CFU-Meg-derived cells as well as other CD34(+) cells; and, finally, stimulate the proliferation of human endothelial cells. It was also demonstrated that these various hematopoietic growth factors, cytokines, and chemokines are expressed and secreted by CFU-GM-, CFU-Meg-, and BFU-E-derived cells. It is concluded that normal human CD34(+) cells and hematopoietic precursors secrete numerous regulatory molecules that form the basis of intercellular cross-talk networks and regulate in an autocrine and/or a paracrine manner the various stages of normal human hematopoiesis.


Subject(s)
Chemokines/metabolism , Cytokines/metabolism , Growth Substances/metabolism , Hematopoiesis , Hematopoietic Stem Cells/physiology , Homeostasis , Antigens, CD34/analysis , Cell Division , Cell Separation , Cell Survival , Cells, Cultured , Chemokines/genetics , Chemotaxis , Culture Media, Conditioned , Cytokines/genetics , Erythroblasts/physiology , Flow Cytometry , Gene Expression , Granulocytes/physiology , Growth Substances/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/immunology , Humans , Megakaryocytes/physiology , RNA, Messenger/analysis , Rh-Hr Blood-Group System/physiology
8.
Stem Cells ; 19(3): 236-46, 2001.
Article in English | MEDLINE | ID: mdl-11359949

ABSTRACT

HIV-based lentiviral vectors can transduce nondividing cells, an important advantage over murine leukemia virus (MLV)-based vectors when transducing slowly dividing hematopoietic stem cells. However, we find that in human CD34(+) hematopoietic cells, the HIV-based vectors with an internal cytomegalovirus (CMV) promoter express transgenes 100- to 1,000-fold less than the MLV-based retroviral vector murine stem cell virus (MSCV). To increase the expression of the integrated lentivirus, we replaced CMV promoter with that of the Rous sarcoma virus or MSCV and obtained a modest augmentation in expression. A more dramatic effect was seen when the CMV enhancer/promoter was removed and the HIV long-terminal repeat (LTR) was replaced by a novel HIV/MSCV hybrid LTR. This vector retains the ability to transduce nondividing cells but now expresses its transgene (enhanced green fluorescent protein) 10- to 100-fold greater than the original HIV-based vector. When compared under identical conditions, the HIV vector with the hybrid LTR transduced a higher percentage of CD34(+) cells than the MSCV-based retroviral vector (19.4% versus 2.4%). The number of transduced cells and level of transgene expression remain constant over 5-8 weeks as determined by long-term culture-initiating cells, fluoresence-activated cell sorting, and nonobese diabetic/severe combined immunodeficiency repopulation assay.


Subject(s)
Antigens, CD34/biosynthesis , Gene Transfer Techniques , Genetic Vectors , HIV/genetics , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Leukemia Virus, Murine/genetics , Transgenes , Animals , Avian Sarcoma Viruses/genetics , Bone Marrow Cells/cytology , Cell Line , Cell Separation , Cells, Cultured , Fetal Blood/cytology , Flow Cytometry , Green Fluorescent Proteins , HIV Long Terminal Repeat/genetics , HeLa Cells , Humans , K562 Cells , Luminescent Proteins/metabolism , Mice , Mice, SCID , Models, Genetic , Polymerase Chain Reaction , Promoter Regions, Genetic , Recombinant Fusion Proteins/metabolism , Time Factors
9.
J Exp Med ; 192(10): 1479-90, 2000 Nov 20.
Article in English | MEDLINE | ID: mdl-11085749

ABSTRACT

The homeobox genes encode a family of transcription factors that regulate development and postnatal tissue homeostasis. Since HOXB4 plays a key role in regulating the balance between hematopoietic stem cell renewal and differentiation, we studied the molecular regulation of HOXB4 expression in human hematopoietic stem cells. HOXB4 expression in K562 cells is regulated at the level of transcription, and transient transfection defines primary HOXB4 regulatory sequences within a 99-bp 5' promoter. Culture of highly purified human CD34(+) bone marrow cells in thrombopoietin/Flt-3 ligand/stem cell factor induced HOXB4 3-10-fold, whereas culture in granulocyte/macrophage colony-stimulating factor, only increased HOXB4/luciferase expression 20-50%. Mutations within the HOXB4 promoter identified a potential E box binding site (HOX response element [HXRE]-2) as the most critical regulatory sequence, and yeast one hybrid assays evaluating bone marrow and K562 libraries for HXRE-2 interaction identified upstream stimulating factor (USF)-2 and micropthalmia transcription factor (MITF). Electrophoretic mobility shift assay with K562 extracts confirmed that these proteins, along with USF-1, bind to the HOXB4 promoter in vitro. Cotransfection assays in both K562 and CD34(+) cells showed that USF-1 and USF-2, but not MITF, induce the HOXB4 promoter in response to signals stimulating stem cell self-renewal, through activation of the mitogen-activated protein kinase pathway. Thus hematopoietic expression of the human HOXB4 gene is regulated by the binding of USF-1 and USF-2, and this process may be favored by cytokines promoting stem cell self-renewal versus differentiation.


Subject(s)
DNA-Binding Proteins , Hematopoietic Stem Cells , Homeodomain Proteins/genetics , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Base Sequence , Bone Marrow Cells , Genomic Library , Humans , K562 Cells , Mitogen-Activated Protein Kinases , Molecular Sequence Data , Nuclear Proteins/metabolism , Promoter Regions, Genetic , Protein Binding , Signal Transduction , Transcriptional Activation , Upstream Stimulatory Factors , ras Proteins
11.
Exp Hematol ; 28(6): 716-25, 2000 Jun.
Article in English | MEDLINE | ID: mdl-10880758

ABSTRACT

By virtue of its high expression in both developing hematopoietic tissues and many myeloid leukemia cells lines, the embryonic tyrosine kinase receptor ETK2 (also known as Tyro3, Sky, and Rse) has been postulated to play a role in early hematopoiesis. To investigate this role, we expressed murine ETK2 in the interleukin 3 (IL-3) dependent myeloid progenitor cell line FDC-P1 and examined its effect on growth factor dependence.ETK2 cDNAs encoding full-length or kinase domain-deleted receptor were retrovirally transduced into murine FDC-P1 cells. Survival, cell cycle status, and proliferative responses of ETK2 expressing clones were studied at normal and reduced growth factor concentrations. ETK2 was expressed as a functional tyrosine kinase of 110 and 150 kDa. This proto-oncogene altered the growth of FDC-P1 cells, allowing survival at reduced growth factor concentrations and delaying apoptosis after IL-3 withdrawal. ETK2-expressing clones contained a higher fraction of cells in the S/G2/M phases of the cell cycle, both after cytokine withdrawal and in the presence of IL-3. Furthermore, these cells had a modestly enhanced proliferative response to IL-3 and granulocyte-macrophage colony-stimulating factor, suggesting that ETK2 intracellular signaling may converge with that of hematopoietic growth factors. The effects of ETK2 expression on viability and proliferation were largely dependent on a functional intracellular tyrosine kinase domain. These results support a role for ETK2 in the survival and/or expansion of primitive hematopoietic cells and suggest that this tyrosine kinase may be implicated in myeloid leukemogenesis as well.


Subject(s)
Hematopoietic Stem Cells/cytology , Receptor Protein-Tyrosine Kinases/physiology , Animals , Apoptosis/drug effects , Cell Cycle , Cell Survival , Cell Transformation, Neoplastic , Cells, Cultured/drug effects , Culture Media/pharmacology , Culture Media, Serum-Free/pharmacology , Genes, Synthetic , Genetic Vectors/genetics , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Hematopoietic Stem Cells/drug effects , Interleukin-3/pharmacology , Leukemia/etiology , Mice , Phosphorylation , Protein Processing, Post-Translational , Protein Structure, Tertiary , Proto-Oncogene Mas , Receptor Protein-Tyrosine Kinases/chemistry , Receptor Protein-Tyrosine Kinases/genetics , Recombinant Fusion Proteins/physiology , Retroviridae/genetics , Sequence Deletion , Signal Transduction/drug effects , Transfection
12.
Science ; 285(5426): 412-5, 1999 Jul 16.
Article in English | MEDLINE | ID: mdl-10411505

ABSTRACT

Graft versus host disease, an alloimmune attack on host tissues mounted by donor T cells, is the most important toxicity of allogeneic bone marrow transplantation. The mechanism by which allogeneic T cells are initially stimulated is unknown. In a murine allogeneic bone marrow transplantation model it was found that, despite the presence of numerous donor antigen-presenting cells, only host-derived antigen-presenting cells initiated graft versus host disease. Thus, strategies for preventing graft versus host disease could be developed that are based on inactivating host antigen-presenting cells. Such strategies could expand the safety and application of allogeneic bone marrow transplantation in treatment of common genetic and neoplastic diseases.


Subject(s)
Antigen-Presenting Cells/immunology , Bone Marrow Transplantation/immunology , CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/prevention & control , Animals , Bone Marrow Transplantation/adverse effects , Dendritic Cells/immunology , Graft vs Host Disease/immunology , H-2 Antigens/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Inbred Strains , Mice, Knockout , Minor Histocompatibility Antigens/immunology , Spleen/immunology , Transplantation Chimera
14.
Hematology ; 4(5): 421-6, 1999.
Article in English | MEDLINE | ID: mdl-27426847

ABSTRACT

Cells of the bone marrow are chiefly dedicated to two processes; the production of blood, and the production of bone that houses the hematopoietic organ. The majority of our understanding of these processes comes from data focused on one of these functions. Yet, in vivo the processes are intermixed. Our recent data demonstrate that human osteoblast-like cells have important accessory roles in hematopoiesis. These data include the demonstration that human osteoblast-like cells; support the growth of primitive human hematopoietic progenitors (CD34(+) cells) in short and long term cultures and, synthesize multiple cytokines believed to regulate hematopoiesis. Based upon anatomic and developmental findings characterizing hematopoietic cells in close approximation with endosteal cells, and these findings, we hypothesize that osteoblasts play a critical role in hematopoietic cells development in vivo.

15.
J Leukoc Biol ; 64(2): 147-55, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9715252

ABSTRACT

Stem cell transplantation (SCT) has achieved significant therapeutic success over the last 10 years, providing a viable treatment option for many previously incurable diseases. However, several inherent limitations of the procedure have restricted its widespread use. These include: lack of sufficient donors for all recipients, a period of bone marrow (BM) aplasia leading to severe, prolonged neutropenia and thrombocytopenia, and the potential for tumor contamination in autologous SCT. Umbilical cord blood (UCB) provides a unique, and potentially more successful, approach to alleviating these limitations. Ex vivo manipulation of hematopoietic stem (HSCs) and progenitor cells (HPCs) derived from UCB using a liquid culture system has revealed that the primitive HSCs from UCB are not identical to their BM counterparts. In fact, these cells may derive from a more primitive stem cell compartment. Ultimately, successful engraftment of UCB HSCs manipulated in an ex vivo environment may lead to a larger number of these life-saving procedures being performed and the full potential of SCT realized.


Subject(s)
Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Neutropenia/therapy , Thrombocytopenia/therapy , Humans
16.
Hematol Oncol Clin North Am ; 12(3): 631-48, 1998 Jun.
Article in English | MEDLINE | ID: mdl-9684102

ABSTRACT

Gene therapy of malignant diseases can be divided into four basic approaches: gene interference, gene insertion, immunopotentiation, and suicide gene approaches. This article reviews the application of these approaches in the therapy of leukemias and lymphomas.


Subject(s)
Genetic Therapy , Leukemia/therapy , Lymphoma/therapy , Animals , Genetic Therapy/methods , Humans , Leukemia/genetics , Lymphoma/genetics
18.
Blood ; 91(8): 2935-47, 1998 Apr 15.
Article in English | MEDLINE | ID: mdl-9531604

ABSTRACT

Mice lacking interleukin-2 (IL-2) developed a severe hematopoietic disorder characterized by the abnormal development of myeloid cells and neutropenia. Analysis of the bone marrow of IL-2-deficient (IL-2(-/-)) mice showed that the number of mature polymorphonuclear cells was decreased by 65% to 75%, and granulocyte/macrophage precursor cells were reduced by 50%. Bone marrow cells from IL-2(-/-) mice were unable to sustain myelopoiesis in lethally irradiated mice and in long-term bone marrow cultures (LTBMC). The addition of exogenous IL-2 to LTBMC of IL-2(-/-) cells partially restored hematopoietic progenitor activity. In the bone marrow of wild-type mice, immature (Mac-1(lo)) myeloid cells, including myeloblasts and promyelocytes, constitutively expressed the beta-chain of the IL-2R, and the number of Mac-1(lo)IL-2Rbeta+ cells was increased by twofold to threefold in IL-2(-/-) mice. During culture in the presence of IL-2 and the absence of stromal cells, Mac-1(lo)IL-2Rbeta+ immature myeloid cells proliferated and gave rise to mature granulocytes and macrophages. Collectively, these observations indicate that defective myelopoiesis in IL-2(-/-) mice is at least in part a consequence of their direct dependency on IL-2, and by regulating the growth of immature myeloid cells, IL-2 plays an important role in the homeostatic regulation of myelocytic cell generation.


Subject(s)
Interleukin-2/deficiency , Leukopoiesis , Animals , Cell Differentiation/drug effects , Cells, Cultured , Granulocytes/cytology , Granulocytes/physiology , Interleukin-2/genetics , Interleukin-2/pharmacology , Leukopoiesis/drug effects , Mice , Mice, Mutant Strains , Neutrophils/cytology , Neutrophils/physiology
19.
J Immunol ; 160(1): 385-94, 1998 Jan 01.
Article in English | MEDLINE | ID: mdl-9551995

ABSTRACT

IL-2-deficient (IL-2(-/-)) mice develop disorders of the hemopoietic and immune systems characterized by anemia, lymphocytic hyperplasia, and colitis. The mechanisms responsible for these abnormalities remain unclear. To investigate the underlying basis of autoimmunity, the particular role of commensal gut flora in the initiation of colitis, and the role of IL-2 in the development of intestinal intraepithelial lymphocytes (iIEL), we evaluated IL-2(-/-) mice reared and maintained under gnotobiotic (germfree) conditions. By 8 wk of age, 80% (20 of 25) of germfree IL-2(-/-) mice show signs of disease, including anemia, disturbances in bone marrow hemopoietic cells, lymphocytic hyperplasia, and generalized autoimmunity, similar to those seen in specific pathogen-free (SPF) IL-2(-/-) mice. In striking contrast to SPF IL-2(-/-) mice, germfree IL-2(-/-) mice do not develop colitis. However, the numbers of gammadelta+ and TCR alphabeta+ CD8 alphaalpha+ iIELs are reduced, and in lethally irradiated SPF IL-2(+/+) mice, reconstituted with IL-2(-/-) bone marrow TCR gammadelta+ iIELs fail to develop, consistent with an important role of IL-2/IL-2R signaling in the development of gammadelta iIELs. Consequently, our findings demonstrate that the colitis seen in SPF IL-2(-/-) mice depends upon the presence of intestinal bacterial flora and that environmental Ags are not responsible for the anemia and extraintestinal lymphoid hyperplasia that occur in IL-2(-/-) mice. Thus, germfree IL-2(-/-) mice represent a unique system in which the role of IL-2 deficiency in hemopoietic and immune system disorders can be investigated in dissociation from complications that may arise due to colitis.


Subject(s)
Autoimmunity , Germ-Free Life , Interleukin-2/deficiency , Age Factors , Animals , Bone Marrow Cells/immunology , Colitis/immunology , Flow Cytometry , Hematopoiesis , Immune Tolerance , Immunity, Mucosal , Liver/cytology , Lymphocyte Subsets/immunology , Mice , Mice, Knockout , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Spleen/cytology
20.
Stem Cells ; 16(1): 7-15, 1998.
Article in English | MEDLINE | ID: mdl-9474743

ABSTRACT

Hematopoietic stem cell differentiation occurs in direct proximity to osteoblasts within the bone marrow cavity. Despite this striking affiliation, surprisingly little is known about the precise cellular and molecular impact of osteoblasts on the bone marrow microenvironment. Recently, it has been proposed that human osteoblasts support the growth of primitive human hematopoietic cells in vitro and possibly in vivo. Evidence to support this hypothesis is reviewed as follows: the influence of osteoblasts on osteoclast development; the participation of osteoblasts in long-term bone marrow cultures; the production of positive hematopoietic regulatory molecules by osteoblasts; the production of cell-cycle inhibitory factors by osteoblasts, and cell-cell interactions between early hematopoietic cells and osteoblasts.


Subject(s)
Hematopoiesis/physiology , Osteoblasts/physiology , Bone Marrow Cells/cytology , Cell Communication , Hematopoietic Stem Cells/cytology , Humans
SELECTION OF CITATIONS
SEARCH DETAIL
...