Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
Add more filters










Publication year range
1.
Eur J Haematol ; 89(3): 198-205, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22639947

ABSTRACT

OBJECTIVES: Methods to expand hematopoietic stem cells (HSCs) ex vivo encompass an attractive approach that would substantially broaden the clinical applicability of HSCs derived from cord blood (CB). Recently, members of the angiopoietin-like (Angptl) family of growth factors were shown to expand both murine and human HSCs. Specifically, Angptl5 has been implicated in the expansion of human NOD/SCID-repopulating cells (SRCs) ex vivo. Here, we sought to evaluate the potential of additional Angptls to expand human SRCs from CB. Additionally, the purpose of this study was to evaluate the reproducibility of Angptl-mediated expansion of SRCs across independent experiments. METHODS: Human CD34(+) cells from CB were cultured in vitro for eleven or 8 d in the presence or absence of Angptls. The reconstitution capacity of expanded cells was subsequently measured in vivo by transplantation into NOD/SCID or NSG mice and compared with that of uncultured cells. RESULTS: We report here that Angptl4 functions to maintain SRC activity of CD34(+) CB-derived cells ex vivo as assayed in NOD/SCID and NSG mice. However, all Angptls tested, including Angptl1, Angptl4, and Angptl5, were associated with variation between experiments. CONCLUSION: Our findings indicate that Angptl4 and Angptl5 can lead to increased engraftment capacity of SRCs, but more frequently, these factors are associated with maintenance of SRC activity during ex vivo culture. Thus, Angptl-mediated expansion of SRCs ex vivo is associated with more interexperimental variation than previously thought. We conclude that Angptls would be useful in instances where there is a need to maintain HSCs ex vivo, such as during transduction for gene therapy applications.


Subject(s)
Angiopoietins/physiology , Antigens, CD34/immunology , Fetal Blood/cytology , Animals , Cell Transplantation , Cells, Cultured , Fetal Blood/immunology , Humans , Mice , Mice, Inbred NOD , Mice, SCID
2.
Exp Hematol ; 35(9): 1421-8, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17761289

ABSTRACT

OBJECTIVE: Members of the hox family of homeodomain-containing transcription factors, including hoxa9, hoxb3, and hoxb4 play an important role in the regulation of differentiation, proliferation and self-renewal of hematopoietic stem and progenitor cells. Lack-of-function studies using hoxa9, hoxb4, or hoxb3/hoxb4 null mice demonstrate that all these mutations compromise the repopulating ability of hematopoietic stem cells (HSC), implying similar functions of each of these genes in hematopoiesis. Because cross regulation and cooperativity are known features of hox proteins, we investigated mice with a compound deficiency in hoxa9, hoxb3 and hoxb4 (hoxa9/b3/b4) for evidence of synergy between these genes in hematopoiesis. MATERIALS AND METHODS: Hoxa9/b3/b4 were generated by mating the hoxb3/hoxb4 null mice with the hoxa9 null strain and HSC function was measured by competitive repopulating assay and by immunophenotype using fluorescence-activated cell sorting. RESULTS: Our findings demonstrate that the hoxa9/b3/b4 null mice are smaller in body weight, and display a marked reduction in spleen size and cellularity compared to control mice. The numbers of colony-forming unit (CFU)-granulocyte macrophage and CFU-spleen progenitor colonies were normal but hoxa9/b3/b4 null bone marrow contained increased numbers of immunophenotypic HSC (Lin(-), c-kit(+), Sca-1(+), CD150(+)). However the reconstitution defect in hoxa9 null HSC was not enhanced further in the hoxa9/b3/b4 null HSC. CONCLUSION: These findings demonstrate overlapping functions of hoxa9, hoxb3, and hoxb4 in hematopoietic cells, and emphasize an important role for these transcription factors for regulation of HSC proliferation. However, none of these hox proteins is absolutely essential for generation or maintenance of all major blood lineages.


Subject(s)
Hematopoietic System/abnormalities , Homeodomain Proteins/physiology , Transcription Factors/physiology , Animals , Body Weight , Cell Lineage , Hematopoietic System/pathology , Homeodomain Proteins/genetics , Leukocytes, Mononuclear/pathology , Mice , Mice, Knockout , Mutation , Organ Size , Spleen/anatomy & histology , Spleen/cytology , Transcription Factors/genetics
3.
Blood ; 109(12): 5178-85, 2007 Jun 15.
Article in English | MEDLINE | ID: mdl-17332244

ABSTRACT

Infantile malignant osteopetrosis (IMO) is a fatal disease caused by lack of functional osteoclasts, and the only available treatment is hematopoietic stem cell (HSC) transplantation. In the majority of patients, the TCIRG1 gene, coding for a subunit of a proton pump essential for bone resorption, is mutated. Oc/oc mice have a deletion in the homologue gene (tcirg1) and die at 3 to 4 weeks, but can be rescued by neonatal transplantation of HSCs. Here, HSC-targeted gene therapy of osteopetrosis in the oc/oc mouse model was developed. Oc/oc fetal liver cells depleted of Ter119-expressing erythroid cells were transduced with a retroviral vector expressing tcirg1 and GFP, and subsequently transplanted intraperitoneally to irradiated neonatal oc/oc mice. Eight of 15 mice survived past the normal life span of oc/oc mice. In vitro osteoclastogenesis revealed formation of GFP-positive osteoclasts and bone resorption, albeit at a lower level than from wild-type cells. The skeletal phenotype was analyzed by X-ray and histopathology and showed partial correction at 8 weeks and almost normalization after 18 weeks. In summary, osteopetrosis in oc/oc mice can be reversed by neonatal transplantation of gene-modified HSCs leading to long-term survival. This represents a significant step toward the development of gene therapy for osteopetrosis.


Subject(s)
Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Osteopetrosis/therapy , Vacuolar Proton-Translocating ATPases/administration & dosage , Animals , Animals, Newborn , Disease Progression , Mice , Mice, Mutant Strains , Sequence Deletion , Survival Rate , Vacuolar Proton-Translocating ATPases/genetics
4.
Exp Hematol ; 35(3): 343-9, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17309814

ABSTRACT

The proliferation and differentiation of primitive hematopoietic cells is tightly controlled by a number of signaling pathways. Transient blockage or enhancement of these signaling pathways may provide a new approach to manipulate the proliferation and differentiation of primitive hematopoietic cells. Adenoviral vectors have in recent years emerged as powerful tools for transient gene expression in human primitive hematopoietic cells. Important advantageous properties of adenoviral vectors include: feasible production of high-titer vector preparations, high efficiency in transducing both quiescent and actively dividing cells, high levels of transient gene expression, and a lack of mutagenic properties associated with integrating vectors. Progress in adenoviral fiber retargeting was recently demonstrated to enable high gene transfer efficiency into nondividing human CD34(+) cells and nonobese diabetic/severe combined immunodeficient mouse bone marrow repopulating cells (SRCs), via the ubiquitously expressed CD46 as a cellular receptor. Importantly, fiber-retargeted adenoviral vectors can be engineered to report gene expression in single living CD34(+) cells, thereby facilitating the isolation and characterization of SRCs and its downstream progenitors based on intrinsic signaling pathways. This review focuses on the current progress and the potential future applications of adenoviral gene transfer into human primitive hematopoietic cells and leukemic cells.


Subject(s)
Adenoviridae/genetics , Gene Expression , Genetic Vectors , Hematopoietic Stem Cells/metabolism , Animals , Gene Transfer Techniques , Genes, Reporter , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/virology , Humans
5.
Blood ; 109(9): 3687-96, 2007 May 01.
Article in English | MEDLINE | ID: mdl-17234739

ABSTRACT

The Homeobox (Hox) transcription factors are important regulators of normal and malignant hematopoiesis because they control proliferation, differentiation, and self-renewal of hematopoietic cells at different levels of the hematopoietic hierarchy. In transgenic mice we show that the expression of HOXA10 is tightly regulated by doxycycline. Intermediate concentrations of HOXA10 induced a 15-fold increase in the repopulating capacity of hematopoietic stem cells (HSCs) after 13 days of in vitro culture. Notably, the proliferation induction of HSC by HOXA10 was dependent on the HOXA10 concentration, because high levels of HOXA10 had no effect on HSC proliferation. Furthermore, high levels of HOXA10 blocked erythroid and megakaryocyte development, demonstrating that tight regulation of HOXA10 is critical for normal development of the erythroid and megakaryocytic lineages. The HOXA10-mediated effects on hematopoietic cells were associated with altered expression of genes that govern stem-cell self-renewal and lineage commitment (eg, hepatic leukemia factor [HlF], Dickkopf-1 [Dkk-1], growth factor independent-1 [Gfi-1], and Gata-1). Interestingly, binding sites for HOXA10 were found in HLF, Dkk-1, and Gata-1, and Dkk-1 and Gfi-1 were transcriptionally activated by HOXA10. These findings reveal novel molecular pathways that act downstream of HOXA10 and identify HOXA10 as a master regulator of postnatal hematopoietic development.


Subject(s)
Antigens, Differentiation/biosynthesis , Cell Differentiation/physiology , Cell Proliferation , Homeodomain Proteins/metabolism , Megakaryocytes/metabolism , Signal Transduction/physiology , Animals , Anti-Bacterial Agents/pharmacology , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Gene Expression Regulation/drug effects , Gene Expression Regulation/physiology , Homeobox A10 Proteins , Homeodomain Proteins/genetics , Megakaryocytes/cytology , Mice , Mice, Transgenic , Signal Transduction/drug effects , Tetracycline/pharmacology
6.
Stem Cells ; 24(3): 653-61, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16210402

ABSTRACT

Enforced expression of the HOXB4 transcription factor and downregulation of p21(Cip1/Waf) (p21) can each independently increase proliferation of murine hematopoietic stem cells (HSCs). We asked whether the increase in HSC self-renewal generated by overexpression of HOXB4 is enhanced in p21-deficient HSCs. HOXB4 was overexpressed in hematopoietic cells from wild-type (wt) and p21-/- mice. Bone marrow (BM) cells were transduced with a retroviral vector expressing HOXB4 together with GFP (MIGB4), or a control vector containing GFP alone (MIG) and maintained in liquid culture for up to 11 days. At day 11 of the expansion culture, the number of primary CFU-GM (colony-forming unit granulocyte-macrophage) colonies and the repopulating ability were significantly increased in MIGB4 p21-/- BM (p21B4) cells compared with MIGB4-transduced wt BM (wtB4) cells. To test proliferation of HSCs in vivo, we performed competitive repopulation experiments and obtained significantly higher long-term engraftment of expanded p21B4 cells compared with wtB4 cells. The 5-day expansion of p21B4 HSCs generated 100-fold higher numbers of competitive repopulating units compared with wtMIG and threefold higher numbers compared with wtB4. The findings demonstrate that increased expression of HOXB4, in combination with suppression of p21 expression, could be a useful strategy for effective and robust expansion of HSCs.


Subject(s)
Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Granulocyte Precursor Cells/metabolism , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/deficiency , Cyclin-Dependent Kinase Inhibitor p21/genetics , Gene Expression Regulation/genetics , Graft Survival/genetics , Granulocyte Precursor Cells/cytology , Hematopoietic Stem Cell Transplantation , Mice , Mice, Knockout , Transduction, Genetic/methods
7.
Blood ; 103(11): 4126-33, 2004 Jun 01.
Article in English | MEDLINE | ID: mdl-14962901

ABSTRACT

Enforced expression of Hoxb4 dramatically increases the regeneration of murine hematopoietic stem cells (HSCs) after transplantation and enhances the repopulation ability of human severe combined immunodeficiency (SCID) repopulating cells. Therefore, we asked what physiologic role Hoxb4 has in hematopoiesis. A novel mouse model lacking the entire Hoxb4 gene exhibits significantly reduced cellularity in spleen and bone marrow (BM) and a subtle reduction in red blood cell counts and hemoglobin values. A mild reduction was observed in the numbers of primitive progenitors and stem cells in adult BM and fetal liver, whereas lineage distribution was normal. Although the cell cycle kinetics of primitive progenitors was normal during endogenous hematopoiesis, defects in proliferative responses of BM Lin(-) Sca1(+) c-kit(+) stem and progenitor cells were observed in culture and in vivo after the transplantation of BM and fetal liver HSCs. Quantitative analysis of mRNA from fetal liver revealed that a deficiency of Hoxb4 alone changed the expression levels of several other Hox genes and of genes involved in cell cycle regulation. In summary, the deficiency of Hoxb4 leads to hypocellularity in hematopoietic organs and impaired proliferative capacity. However, Hoxb4 is not required for the generation of HSCs or the maintenance of steady state hematopoiesis.


Subject(s)
Hematopoiesis/physiology , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Homeodomain Proteins/genetics , Transcription Factors/genetics , Animals , Antimetabolites/pharmacology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/physiology , Cell Count , Cell Division/physiology , Female , Fetal Tissue Transplantation , Fluorouracil/pharmacology , Gene Expression Regulation, Developmental , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Hepatocytes/cytology , Hepatocytes/transplantation , Liver/cytology , Liver/embryology , Liver/physiology , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Animal , Pregnancy
8.
Mol Ther ; 8(4): 618-28, 2003 Oct.
Article in English | MEDLINE | ID: mdl-14529835

ABSTRACT

Retroviral overexpression of the transcription factor HOXB4 results in a rapid increase in proliferation of murine hematopoietic stem cells both in vivo and in vitro. Therefore, we asked whether transient overexpression of HOXB4 would increase proliferation of human primitive hematopoietic progenitors. Transient overexpression of HOXB4 was generated in umbilical cord blood (CB) CD34(+) cells by a recombinant adenovirus (AdHOXB4) expressing HOXB4 together with the enhanced green fluorescent protein (GFP). Transduced, GFP(+) cells were cultured in serum-free medium containing cytokines that primarily support the growth of primitive hematopoietic progenitors. In contrast to previous findings using retroviral overexpression of HOXB4, we did not observe any increase in proliferation of primitive progenitors or increased colony formation of clonogenic progenitors, including progenitor progeny from long-term culture-initiating cells following adenoviral vector overexpression of HOXB4 in CB CD34(+) cells. However, enforced expression of HOXB4 by the adenoviral vector significantly increased myeloid differentiation of primitive hematopoietic progenitors. Since retroviral vectors generate low and continuous levels of transgene expression in contrast to the high, transient levels generated by the adenoviral vector, our findings suggest that the high levels of HOXB4 expression generated by AdHOXB4 in human CB CD34(+) cells direct the cells toward a myeloid differentiation program rather than increased proliferation.


Subject(s)
Antigens, CD34/metabolism , Cell Differentiation/physiology , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Myeloid Cells/metabolism , Transcription Factors/genetics , Adenoviridae , Cell Differentiation/genetics , Cell Division/physiology , Fetal Blood/cytology , Fetal Blood/metabolism , Gene Transfer Techniques , Genetic Vectors , Homeodomain Proteins/metabolism , Humans , Transcription Factors/metabolism
9.
Mol Cell Biol ; 23(11): 3872-83, 2003 Jun.
Article in English | MEDLINE | ID: mdl-12748289

ABSTRACT

Several homeobox transcription factors, such as HOXB3 and HOXB4, have been implicated in regulation of hematopoiesis. In support of this, studies show that overexpression of HOXB4 strongly enhances hematopoietic stem cell regeneration. Here we find that mice deficient in both Hoxb3 and Hoxb4 have defects in endogenous hematopoiesis with reduced cellularity in hematopoietic organs and diminished number of hematopoietic progenitors without perturbing lineage commitment. Analysis of embryonic day 14.5 fetal livers revealed a significant reduction in the hematopoietic stem cell pool, suggesting that the reduction in cellularity observed postnatally is due to insufficient expansion during fetal development. Primitive Lin(-) ScaI(+) c-kit(+) hematopoietic progenitors lacking Hoxb3 and Hoxb4 displayed impaired proliferative capacity in vitro. Similarly, in vivo repopulating studies of Hoxb3/Hoxb4-deficient hematopoietic cells resulted in lower repopulating capability compared to normal littermates. Since no defects in homing were observed, these results suggest a slower regeneration of mutant HSC. Furthermore, treatment with cytostatic drugs demonstrated slower cell cycle kinetics of hematopoietic stem cells deficient in Hoxb3 and Hoxb4, resulting in increased tolerance to antimitotic drugs. Collectively, these data suggest a direct physiological role of Hoxb4 and Hoxb3 in regulating stem cell regeneration and that these genes are required for maximal proliferative response.


Subject(s)
Cell Division/physiology , Hematopoietic Stem Cells/physiology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Xenopus Proteins/metabolism , Animals , Antimetabolites/pharmacology , Bone Marrow Transplantation , Fluorouracil/pharmacology , Gene Targeting , Hematopoiesis/physiology , Hematopoietic Stem Cells/drug effects , Homeodomain Proteins/genetics , Liver/physiology , Mice , Mice, Knockout , Transcription Factors/genetics , Xenopus Proteins/genetics
10.
Mol Ther ; 7(5 Pt 1): 613-22, 2003 May.
Article in English | MEDLINE | ID: mdl-12718904

ABSTRACT

Diamond-Blackfan anemia (DBA) is a congenital bone marrow failure syndrome characterized by a specific deficiency in erythroid progenitors. Since some patients with DBA develop a reduction in thrombocytes and granulocytes with age, we asked whether multipotent hematopoietic progenitors from DBA patients had normal proliferative capacity in liquid expansion cultures. CD34(+) cells derived from DBA patients showed deficient proliferation in liquid culture containing IL-3, IL-6, and SCF. Single CD34(+) CD38(-) cells from DBA patients exhibited deficient proliferation recruitment in a limiting dilution assay containing IL-3, IL-6, SCF, Tpo, FL, and G-CSF or containing IL-3, IL-6, and SCF. Our findings suggest that the underlying hematopoietic defect in DBA may not be limited to the erythroid lineage. Since a fraction of DBA patients have a deficiency in ribosomal protein S19 (RPS19), we constructed lentiviral vectors containing the RPS19 gene for overexpression in hematopoietic progenitors from RPS19-deficient DBA patients. Enforced expression of the RPS19 transgene improved the proliferation of CD34(+) cells from DBA patients with RPS19 mutation. Similarly, enforced expression of RPS19 improved erythroid development of RPS19-deficient hematopoietic progenitors as determined by colony assays and erythroid differentiation cultures. These findings suggest that gene therapy for RPS19-deficient DBA is feasible.


Subject(s)
Anemia, Diamond-Blackfan/therapy , Gene Expression Regulation/physiology , Genetic Therapy , Hematopoietic Stem Cells/metabolism , Ribosomal Proteins/genetics , Anemia, Diamond-Blackfan/metabolism , Antigens, CD/analysis , Bone Marrow/pathology , Case-Control Studies , Cell Division/physiology , Cell Line , Colony-Forming Units Assay , Cytokines/metabolism , Gene Transfer Techniques , Genetic Vectors , Hematopoietic Stem Cells/pathology , Humans , Lentivirus/genetics , Ribosomal Proteins/deficiency , Transfection
11.
Mol Ther ; 6(3): 400-6, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12231177

ABSTRACT

Retroviral transduction of CD34(+) cells on Retronectin using gibbon ape leukemia virus (GALV) pseudotyped vectors is inhibited by high concentrations of vector containing medium (VCM). Furthermore, this inhibitory activity is stable for at least 48 hours at 37 degrees C and partially blocks a second hit with a GALV pseudotyped vector. We hypothesized that this inhibition was due to interference at the receptor level between infectious and noninfectious vector particles and that it might be possible to overcome it by increasing receptor expression on target cells. Activation of protein kinase C in CD34(+) cells with the phorbol ester PMA (phorbol 12-myristate 13-acetate) increased the mRNA level of the GALV receptor (GLVR1) and the transduction efficiency (TE), and fully reversed the inhibition of transduction seen with high-titer GALV VCM. A murine stem cell virus (MSCV) vector with the GLVR1 receptor and green fluorescent protein cDNAs (MGLIG) was used to transduce fibroblasts, and clones expressing different levels of GLVR1 were isolated. The TE of these cells using a GALV vector correlated with the level of GLVR1 expression. When CD34(+) cells or K562 cells were first transduced with MGLIG and then with high-titer GALV VCM, no inhibition of transduction was seen. The low level of GLVR1 expression limits gene transfer to K562 and CD34(+) cells using GALV pseudotyped vectors, especially in the presence of high-titer VCMs.


Subject(s)
Antigens, CD34/metabolism , Gene Transfer Techniques , Genetic Vectors , Receptors, Virus/genetics , Fetal Blood , Humans , K562 Cells , Leukemia Virus, Gibbon Ape , Leukocytes/drug effects , Leukocytes/metabolism , Protein Kinase C/metabolism , Receptors, Virus/biosynthesis , Receptors, Virus/metabolism , Tetradecanoylphorbol Acetate/pharmacology , Transduction, Genetic
12.
Blood ; 100(8): 2724-31, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12351378

ABSTRACT

Diamond-Blackfan anemia (DBA) is a congenital bone marrow failure syndrome characterized by a specific deficiency in erythroid progenitors. Forty percent of the patients are blood transfusion-dependent. Recent reports show that the ribosomal protein S19 (RPS19) gene is mutated in 25% of all patients with DBA. We constructed oncoretroviral vectors containing the RPS19 gene to develop gene therapy for RPS19-deficient DBA. These vectors were used to introduce the RPS19 gene into CD34(+) bone marrow (BM) cells from 4 patients with DBA with RPS19 gene mutations. Overexpression of the RPS19 transgene increased the number of erythroid colonies by almost 3-fold. High expression levels of the RPS19 transgene improved erythroid colony-forming ability substantially whereas low expression levels had no effect. Overexpression of RPS19 had no detrimental effect on granulocyte-macrophage colony formation. Therefore, these findings suggest that gene therapy for RPS19-deficient patients with DBA using viral vectors that express the RPS19 gene is feasible.


Subject(s)
Anemia, Diamond-Blackfan/genetics , Anemia, Diamond-Blackfan/therapy , Bone Marrow Cells/physiology , Bone Marrow Transplantation/methods , Hematopoietic Stem Cells/physiology , Ribosomal Proteins/deficiency , Ribosomal Proteins/genetics , Adult , Antigens, CD/analysis , Antigens, CD34/analysis , Bone Marrow Cells/cytology , Cell Line , Child , Colony-Forming Units Assay , Female , Gene Transfer Techniques , Genetic Vectors , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/pathology , Humans , Male , Reverse Transcriptase Polymerase Chain Reaction , Transfection
13.
J Immunol ; 168(2): 755-62, 2002 Jan 15.
Article in English | MEDLINE | ID: mdl-11777969

ABSTRACT

Hemopoietic stem cells (HSCs) are maintained at relative quiescence by the balance between the positive and negative regulatory factors that stimulate or inhibit their proliferation. Blocking the action of negative regulatory factors may provide a new approach for inducing HSCs into proliferation. A variety of studies have suggested that TGF-beta negatively regulates cell cycle progression of HSCs. In this study, a dominant negatively acting mutant of TGF-beta type II receptor (TbetaRIIDN) was transiently expressed in HSCs by using adenoviral vector-mediated gene delivery, such that the effects of disrupting the autocrine TGF-beta signaling in HSCs can be directly examined at a single cell level. Adenoviral vectors allowing the expression of TbetaRIIDN and green fluorescence protein in the same CD34(+)CD38(-)Lin(-) cells were constructed. Overexpression of TbetaRIIDN specifically disrupted TGF-beta-mediated signaling. Autocrine TGF-beta signaling in CD34(+)CD38(-)Lin(-) cells was studied in single cell assays under serum-free conditions. Transient blockage of autocrine TGF-beta signaling in CD34(+)CD38(-)Lin(-) cells enhanced their survival. Furthermore, the overall proliferation potential and proliferation kinetics in these cells were significantly enhanced compared with the CD34(+)CD38(-)Lin(-) cells expressing green fluorescence protein alone. Therefore, we have successfully blocked the autocrine TGF-beta-negative regulatory loop of primitive hemopoietic progenitor cells.


Subject(s)
Antigens, CD , Hematopoietic Stem Cells/cytology , Signal Transduction/genetics , Transforming Growth Factor beta/genetics , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Adenoviridae/genetics , Animals , Antigens, CD34/biosynthesis , Antigens, Differentiation/biosynthesis , Autocrine Communication/genetics , Cattle , Cell Cycle/genetics , Cell Division/genetics , Cell Lineage/genetics , Cell Survival/genetics , Cells, Cultured , Culture Media, Serum-Free , Genetic Vectors/biosynthesis , HeLa Cells , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , K562 Cells , Membrane Glycoproteins , NAD+ Nucleosidase/biosynthesis , Signal Transduction/physiology , Transduction, Genetic , Transforming Growth Factor beta/antagonists & inhibitors , Transforming Growth Factor beta/pharmacology , Transforming Growth Factor beta/physiology
SELECTION OF CITATIONS
SEARCH DETAIL
...