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










Publication year range
1.
Stem Cells Transl Med ; 4(9): 1064-72, 2015 Sep.
Article in English | MEDLINE | ID: mdl-26160960

ABSTRACT

UNLABELLED: Human cord blood (CB)-derived hematopoietic stem cells (HSCs) are an interesting source for HSC transplantation. However, the number of collected CB-HSCs is often too low for one transplantation; therefore, ex vivo expansion of CB-HSCs is desirable. Current expansion protocols are based on the use of cytokine combinations, including insulin-like growth factor-binding protein 2 (IGFBP2) and angiopoietin-like proteins, or combinations with "small molecules" such as stemregenin-1. The aim of our project was to compare the potential of different CB-HSC expansion strategies side-by-side by phenotypical analysis in vitro and serial engraftment properties in NOD/SCID/IL2rg-/- (NSG) immunodeficient mice. We further identified resveratrol, a naturally occurring polyphenol, as a new, alternative small molecule combined with cytokines to facilitate serum-free ex vivo expansion of human CB-HSCs. The cultivation in resveratrol preserved the CB-HSC phenotype in vitro most efficiently and was ∼2 times more potent than commonly used cytokine conditions (including stem cell factor, thrombopoietin, Fms-related tyrosine kinase 3 ligand, interleukin-6) and the recently established serum-free culture, including IGFBP2 and angiopoietin-like 5. Serial transplantation studies further confirmed resveratrol to support robust multilineage engraftment in primary and secondary NSG recipients. Therefore, our work proposes resveratrol as a new small molecule for improved ex vivo culture and modification of human HSCs based on an efficient ex vivo propagation of the HSC fate. SIGNIFICANCE: Human cord blood (CB)-derived hematopoietic stem cells (HSCs) are an important source for HSC transplantations but restricted in their usage because of their low numbers. In gene therapy, modifications of HSCs relies on their ex vivo modification without losing their stemness properties. Therefore, ex vivo cultivation and expansion of CB-HSCs is important for their effective application in HSC transplantation and gene therapy. Several promising protocols for serum-free cultivation of HSCs using different combinations of cytokines or so-called small molecules are described. A direct comparison was performed of three described serum-free cytokine conditions, demonstrating that the natural occurring polyphenol resveratrol is able to support ex vivo cultivation of CB-HSCs. The results show that resveratrol is an additional candidate for improving ex vivo cultures of HSCs for transplantation and gene therapeutic applications in the future.


Subject(s)
Antioxidants/pharmacology , Fetal Blood/drug effects , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/drug effects , Stilbenes/pharmacology , Angiopoietins/pharmacology , Animals , Fetal Blood/cytology , Fetal Blood/metabolism , Graft Survival , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , Insulin-Like Growth Factor Binding Protein 2/pharmacology , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Interleukin-6/pharmacology , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, SCID , Primary Cell Culture , Resveratrol , Stem Cell Factor/pharmacology , Thrombopoietin/pharmacology , Transplantation, Heterologous , Vascular Endothelial Growth Factor Receptor-1/pharmacology
2.
Cancer Res ; 75(13): 2641-52, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25904681

ABSTRACT

A key challenge in the field of T-cell immunotherapy for cancer is creating a suitable platform for promoting differentiation of effector cells while at the same time enabling self-renewal needed for long-term memory. Although transfer of less differentiated memory T cells increases efficacy through greater expansion and persistence in vivo, the capacity of such cells to sustain effector functions within immunosuppressive tumor microenvironments may still be limiting. We have therefore directly compared the impact of effector versus memory differentiation of therapeutic T cells in tumor-bearing mice by introducing molecular switches that regulate cell fate decisions via mTOR. Ectopic expression of RAS homolog enriched in brain (RHEB) increased mTORC1 signaling, promoted a switch to aerobic glycolysis, and increased expansion of effector T cells. By rapidly infiltrating tumors, RHEB-transduced T cells significantly reduced the emergence of immunoedited escape variants. In contrast, expression of proline-rich Akt substrate of 40 kDa (PRAS40) inhibited mTORC1, promoted quiescence, and blocked tumor infiltration. Fate mapping studies following transient expression of PRAS40 demonstrated that mTORC1(low) T cells made no contribution to initial tumor control but instead survived to become memory cells proficient in generating recall immunity. Our data support the design of translational strategies for generating heterogeneous T-cell immunity against cancer, with the appropriate balance between promoting effector differentiation and self-renewal. Unlike pharmacologic inhibitors, the genetic approach described here allows for upregulation as well as inhibition of the mTORC1 pathway and is highly selective for the therapeutic T cells without affecting systemic mTORC1 functions.


Subject(s)
CD8-Positive T-Lymphocytes/physiology , Immunologic Memory/genetics , Immunotherapy, Adoptive/methods , Neoplasms, Experimental/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/transplantation , Cell Line, Tumor , Humans , Immunologic Memory/immunology , Mechanistic Target of Rapamycin Complex 1 , Mice , Monomeric GTP-Binding Proteins/biosynthesis , Monomeric GTP-Binding Proteins/genetics , Multiprotein Complexes/immunology , Neoplasms, Experimental/prevention & control , Neuropeptides/biosynthesis , Neuropeptides/genetics , Phosphoproteins/biosynthesis , Phosphoproteins/genetics , Ras Homolog Enriched in Brain Protein , TOR Serine-Threonine Kinases/immunology , Transduction, Genetic
3.
Stem Cells Transl Med ; 1(8): 581-91, 2012 Aug.
Article in English | MEDLINE | ID: mdl-23197864

ABSTRACT

Induced pluripotent stem cells (iPSCs) may become a promising source for the generation of patient-specific hematopoietic stem cells (HSCs) in vitro. A crucial prerequisite will be the availability of reliable protocols for the directed and efficient differentiation toward HSCs. So far, the most robust strategy for generating HSCs from pluripotent cells in vitro has been established in the mouse model involving ectopic expression of the human transcription factor HOXB4. However, most differentiation protocols include coculture on a xenogenic stroma cell line and the use of animal serum. Involvement of any of both would pose a major barrier to the translation of those protocols to human autologous iPSCs intended for clinical use. Therefore, we asked whether long-term repopulating HSCs can, in principle, be generated from embryonic stem cells without stroma cells or serum. Here, we showed that long-term multilineage engraftment could be accomplished in immunocompetent mice when HSCs were generated in serum-free medium without stroma cell support and when hypoxic conditions were used. Under those conditions, HOXB4(+) embryonic stem cell-derived hematopoietic stem and progenitor cells were immunophenotypically similar to definitive bone marrow resident E-SLAM(+) (CD150(+)CD48(-)CD45(+)CD201(+)) HSCs. Thus, our findings may ease the development of definitive, adult-type HSCs from pluripotent stem cells, entirely in vitro.


Subject(s)
Cell Differentiation , Cell Lineage , Embryonic Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Hypoxia/metabolism , Pluripotent Stem Cells/cytology , Stromal Cells/cytology , Animals , Bone Marrow/metabolism , Cells, Cultured , Coculture Techniques , Culture Media, Serum-Free/pharmacology , Cytokines/pharmacology , Embryonic Stem Cells/metabolism , Flow Cytometry , Hematopoietic Stem Cells/metabolism , Mice , Mice, SCID , Pluripotent Stem Cells/metabolism , Regeneration/physiology , Stromal Cells/metabolism
4.
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
5.
Blood ; 117(11): 3053-64, 2011 Mar 17.
Article in English | MEDLINE | ID: mdl-21248062

ABSTRACT

Gene therapy has proven its potential to cure diseases of the hematopoietic system. However, severe adverse events observed in clinical trials have demanded improved gene-transfer conditions. Whereas progress has been made to reduce the genotoxicity of integrating gene vectors, the role of pretransplantation cultivation is less well investigated. We observed that the STIF (stem cell factor [SCF], thrombopoietin [TPO], insulin-like growth factor-2 [IGF-2], and fibroblast growth factor-1 [FGF-1]) cytokine cocktail developed to effectively expand murine hematopoietic stem cells (HSCs) also supports the expansion of leukemia-initiating insertional mutants caused by gammaretroviral gene transfer. We compared 4 protocols to examine the impact of prestimulation and posttransduction culture in STIF in the context of lentiviral gene transfer. Observing 56 transplanted mice for up to 9.5 months, we found consistent engraftment and gene-marking rates after prolonged ex vivo expansion. Although a lentiviral vector with a validated insertional-mutagenic potential was used, longitudinal analysis identifying > 7000 integration sites revealed polyclonal fluctuations, especially in "expanded" groups, with de novo detection of clones even at late time points. Posttransduction expansion in STIF did not enrich clones with insertions in proto-oncogenes but rather increased clonal diversity. Our data indicate that lentiviral transduction in optimized media mediates intact polyclonal hematopoiesis without selection for growth-promoting hits by posttransduction expansion.


Subject(s)
Genetic Vectors/genetics , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Lentivirus/genetics , Transduction, Genetic , Animals , Cell Proliferation/drug effects , Cell Survival/drug effects , Cells, Cultured , Chimerism , Clone Cells , Culture Media/pharmacology , Cytokines/pharmacology , Gene Dosage/genetics , Hematopoietic Stem Cells/drug effects , High-Throughput Nucleotide Sequencing , Lentivirus/drug effects , Leukemia/pathology , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional/drug effects , Oncogenes/genetics , Phenotype , Polymerase Chain Reaction , Time Factors
6.
J Mol Biol ; 407(2): 193-221, 2011 Mar 25.
Article in English | MEDLINE | ID: mdl-21241707

ABSTRACT

Traditional DNA transduction routes used for the modification of cellular genomes are subject to unpredictable alterations, as the cell-intrinsic repair machinery may affect both the integrity of the transgene and the recipient locus. These problems are overcome by recombinase-mediated cassette exchange (RMCE) approaches enabling predictable expression patterns by the nondisruptive insertion of a gene cassette at a pre-characterized genomic locus. The destination is marked by a "tag" consisting of two heterospecific recombination target sites (RTs) at the flanks of a selection marker. Provided on a circular donor vector, an analogous cassette encoding the gene of interest can cleanly replace the resident cassette under the influence of a site-specific recombinase. RMCE was first based on the yeast integrase Flp but had to give way to the originally more active phage-derived Cre enzyme. To be effective, both Tyr-recombinases have to be applied at a considerable concentration, which, in the case of Cre, triggers endonucleolytic activities and therefore cellular toxicity. This review addresses the particularities of both recombination routes depending on the structure of the synaptic complex and on improved integrase and RT variants. While the performance of Flp-RMCE can now firmly rely on optimized Flp variants and multiple sets of functional target sites (FRTs), the Cre system suffers from the promiscuity of its RT mutants, which is explained in molecular terms. At present, RMCE enters applications in the stem cell field. Remarkable efforts are noted in the framework of various mouse mutagenesis programs, which, in their first phase, have targeted virtually all genes and now start to shift their emphasis from gene trapping to gene modification.


Subject(s)
DNA Nucleotidyltransferases/genetics , Gene Transfer Techniques , Genetic Engineering/methods , Animals , Gene Targeting , Mice , Transgenes
7.
Hum Gene Ther ; 22(2): 166-76, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20825282

ABSTRACT

The regulated expression of therapeutic genes may become crucial in gene therapy when their constitutive expression interferes with cell fate in vivo. The efficient regulation of transgene expression requires tightly controlled inducible promoters, as shown for the tetracycline regulatory system (tet-system). However, its application requires the introduction of two components into the target cell genome: the tet-responsive transactivator and the regulated expression cassette. In order to facilitate the usage of the tet-system for approaches in gene therapy, both components have to be transferred by a single vector, thus eliminating the preselection of transactivator positive cells. Published "all-in-one" vectors for regulated transgene expression display a relatively low signal-to-noise ratio, resulting in regulatory windows of around 500-fold even in selected clones. In this study, we show that a modified vector architecture combined with the introduction of new tet-responsive promoters, Ptet, improved the dynamic range of such all-in-one vectors to levels up to 14,000-fold for viral and 25,000-fold for nonviral transfer vectors in nonclonal human cell lines, and up to 2,800-fold in murine hematopoietic cell lines. This improved regulation was the result of a strong reduction of background expression in the off-state, even if cells were transduced at high multiplicity of infection, while induction remained at high levels. In addition, the results indicated that successful regulation of gene expression in different target cells depended on vector architecture as well as the choice of the Ptet-promoter.


Subject(s)
Gene Expression Regulation, Viral , Genetic Vectors , Retroelements , Retroviridae/genetics , Transgenes , Animals , Cell Line , Genetic Therapy , Humans , Mice , Promoter Regions, Genetic , Tetracycline/metabolism , Trans-Activators/genetics , Transduction, Genetic
9.
Mol Ther ; 17(9): 1537-47, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19532134

ABSTRACT

In gene therapeutic approaches targeting hematopoietic cells, insertional mutagenesis may provoke clonal dominance with potential progress to overt leukemia. To investigate the contribution of cell-intrinsic features and determine the frequency of insertional proto-oncogene activation, we sorted hematopoietic subpopulations before transduction with replication-deficient gamma-retroviral vectors and studied the clonal repertoire in transplanted C57BL/6J mice. Progressive clonal dominance only developed in the progeny of populations with intrinsic stem cell potential, where expanding clones with insertional upregulation of proto-oncogenes such as Evi1 were retrieved with a frequency of approximately 10(-4). Longitudinal studies by high-throughput sequencing and locus-specific quantitative PCR showed clones with >50-fold expansion between weeks 5 and 31 after transplantation. In contrast, insertional events in proto-oncogenes did not endow the progeny of multipotent or myeloid-restricted progenitors with the potential for clonal dominance (risk <10(-6)). Transducing sorted hematopoietic stem cells (HSCs) with self-inactivating (SIN) lentiviral vectors in short-term cultures improved chimerism, and although clonal dominance developed, there was no evidence for insertional events in the vicinity of proto-oncogenes as the underlying cause. We conclude that cell-intrinsic properties cooperate with vector-related features to determine the incidence and consequences of insertional mutagenesis. Furthermore, our study offers perspectives for refinement of animal experiments in the assessment of vector-related genotoxicity.


Subject(s)
Genetic Vectors/genetics , Mutagenesis, Insertional/methods , Animals , Female , Hematopoietic Stem Cell Transplantation , Humans , Lentivirus/genetics , Mice , Mice, Inbred C57BL , Mutagenesis, Insertional/genetics , Proto-Oncogene Mas , Retroviridae/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transduction, Genetic
10.
Blood ; 113(21): 5111-20, 2009 May 21.
Article in English | MEDLINE | ID: mdl-19270262

ABSTRACT

Ectopic delivery of HOXB4 elicits the expansion of engrafting hematopoietic stem cells (HSCs). We hypothesized that inhibition of tumor necrosis factor-alpha (TNF-alpha) signaling may be central to the self-renewal signature of HOXB4. Because HSCs derived from Fanconi anemia (FA) knockout mice are hypersensitive to TNF-alpha, we studied Fancc(-/-) HSCs to determine the physiologic effects of HOXB4 on TNF-alpha sensitivity and the relationship of these effects to the engraftment defect of FA HSCs. Overexpression of HOXB4 reversed the in vitro hypersensitivity to TNF-alpha of Fancc(-/-) HSCs and progenitors (P) and partially rescued the engraftment defect of these cells. Coexpression of HOXB4 and the correcting FA-C protein resulted in full correction compared with wild-type (WT) HSCs. Ectopic expression of HOXB4 resulted in a reduction in both apoptosis and reactive oxygen species in Fancc(-/-) but not WT HSC/P. HOXB4 overexpression was also associated with a significant reduction in surface expression of TNF-alpha receptors on Fancc(-/-) HSC/P. Finally, enhanced engraftment was seen even when HOXB4 was expressed in a time-limited fashion during in vivo reconstitution. Thus, the HOXB4 engraftment signature may be related to its effects on TNF-alpha signaling, and this pathway may be a molecular target for timed pharmacologic manipulation of HSC during reconstitution.


Subject(s)
Fanconi Anemia/pathology , Hematopoietic Stem Cells/pathology , Homeodomain Proteins/pharmacology , Transcription Factors/pharmacology , Tumor Necrosis Factor-alpha/pharmacology , Animals , Apoptosis/drug effects , Fanconi Anemia Complementation Group C Protein/deficiency , Graft Survival , Hematopoietic Stem Cell Transplantation , Homeodomain Proteins/administration & dosage , Mice , Mice, Knockout , Reactive Oxygen Species , Receptors, Tumor Necrosis Factor/analysis , Transcription Factors/administration & dosage
11.
Methods Mol Biol ; 506: 23-31, 2009.
Article in English | MEDLINE | ID: mdl-19110617

ABSTRACT

Hematopoietic stem cells (HSCs) represent an important target cell population in bone marrow transplantation and gene therapy applications. Their progeny cells carry the genetic information of the HSCs and replenish the blood and immune system. Therefore, in the setting of inherited diseases, transduction of HSCs with retroviral vectors (including gammaretro- and lentiviral vectors) offers the possibility to correct the phenotype in all blood lineages as demonstrated in clinical trials for immunodeficiencies (e.g., X-SCID). In the process of developing gene therapy strategies for patient applications, suitable mouse models for the human gene therapy are important to validate the concept. Stem-cell-enriched populations such as lineage negative cells as the functional equivalent of human CD34(+) cells can be isolated from murine bone marrow and efficiently transduced using retroviral vectors. This chapter provides a step-by-step protocol for retroviral transduction of murine lineage negative cells.


Subject(s)
Genetic Vectors , Hematopoietic Stem Cells/metabolism , Retroviridae/genetics , Transduction, Genetic , Animals , Mice
12.
Mol Ther ; 16(3): 590-8, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18180772

ABSTRACT

Gene therapy for X-linked severe combined immunodeficiency (SCID-X1) has proven highly effective for long-term restoration of immunity in human subjects. However, the development of lymphoproliferative complications due to dysregulated proto-oncogene expression has underlined the necessity for developing safer vector systems. To reduce the potential for insertional mutagenesis, we have evaluated the efficacy of self-inactivating (SIN) gammaretroviral vectors in cellular and in vivo models of SCID-X1. Vectors incorporating an internal human elongation factor-1alpha regulatory element were capable of fully restoring the lymphoid differentiation potential of gammac-deficient lineage negative cells. Multilineage lymphoid reconstitution of a murine model was achieved at a similar level to that achieved by a conventional long-terminal repeat (LTR)-regulated vector used in previous clinical trials. Functional proliferative responses to mitogenic stimuli were also restored, and serum immunoglobulin levels were normalized. The reduced mutagenic potential conferred by SIN vector configurations and alternative non-LTR-based regulatory elements, together with proven efficacy in correction of cellular defects provides an important platform for development of the next phase of clinical trials for SCID-X1.


Subject(s)
Gammaretrovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , X-Linked Combined Immunodeficiency Diseases/therapy , Animals , B-Lymphocytes/cytology , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Blotting, Northern , Cell Differentiation , Cell Line , Cells, Cultured , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Humans , Killer Cells, Natural/cytology , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Mice , Mice, Inbred C57BL , Models, Genetic , Polymerase Chain Reaction , Proto-Oncogene Mas , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , X-Linked Combined Immunodeficiency Diseases/genetics , X-Linked Combined Immunodeficiency Diseases/immunology
13.
Proc Natl Acad Sci U S A ; 104(43): 16952-7, 2007 Oct 23.
Article in English | MEDLINE | ID: mdl-17940039

ABSTRACT

Homeodomain-containing transcription factors are important regulators of stem cell behavior. HOXB4 mediates expansion of adult and embryo-derived hematopoietic stem cells (HSCs) when expressed ectopically. To define the underlying molecular mechanisms, we performed gene expression profiling in combination with subsequent functional analysis with enriched adult HSCs and embryonic derivatives expressing inducible HOXB4. Thereby, we identified a set of overlapping genes that likely represent "universal" targets of HOXB4. A substantial number of loci are involved in signaling pathways important for controlling self-renewal, maintenance, and differentiation of stem cells. Functional assays performed on selected pathways confirmed the biological coherence of the array results. HOXB4 activity protected adult HSCs from the detrimental effects mediated by the proinflammatory cytokine TNF-alpha. This protection likely contributes to the competitive repopulation advantage of HOXB4-expressing HSCs observed in vivo. The concept of TNF-alpha inhibition may also prove beneficial for patients undergoing bone marrow transplantation. Furthermore, we demonstrate that HOXB4 activity and FGF signaling are intertwined. HOXB4-mediated expansion of adult and ES cell-derived HSCs was enhanced by specific and complete inhibition of FGF receptors. In contrast, the expanding activity of HOXB4 on hematopoietic progenitors in day 4-6 embryoid bodies was blunted in the presence of basic FGF (FGF2), indicating a dominant negative effect of FGF signaling on the earliest hematopoietic cells. In summary, our results strongly suggest that HOXB4 modulates the response of HSCs to multiple extrinsic signals in a concerted manner, thereby shifting the balance toward stem cell self-renewal.


Subject(s)
Embryonic Stem Cells/cytology , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Cell Differentiation/drug effects , Cell Proliferation/drug effects , Embryonic Stem Cells/drug effects , Embryonic Stem Cells/metabolism , Fibroblast Growth Factors/pharmacology , Gene Expression Regulation, Developmental/drug effects , Gene Regulatory Networks , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Homeodomain Proteins/genetics , Mice , Mice, Inbred C57BL , Oligonucleotide Array Sequence Analysis , Reproducibility of Results , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction/drug effects , Time Factors , Transcription Factors/genetics , Tumor Necrosis Factor-alpha/pharmacology
14.
Growth Factors ; 24(2): 97-105, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16801129

ABSTRACT

Key players in self-renewal of hemopoietic stem cells are homeobox (HOX) transcription factors. In murine cells, overexpression of HOXB4 results in expansion of hematopoietic stem- and committed progenitor cells in vitro without obvious hematopoietic alterations. In vivo, HOXB4 induced HSC expansion continued until stem cell regeneration reached pretransplantation levels. HOXB4 is thus an attractive candidate for amplification of stem cells provided that human HOXB4 overexpressing cells can also be restricted to normal growth in vivo. The stromal microenvironment provides the regulatory mechanisms controlling the balance of stem cell self-renewal and differentiation. Here, we compared the response of HOXB4- and GFP-control vector transduced human CD34(+) cells to stroma encoded signals in vitro. In serum-sustained cocultures MS-5 stroma contact reduced the output of late CD34- HOXB4(+) cells in relation to GFP-controls 9-fold whereas the expansion of early CD34(+)HOXB4(+) cells remained unchanged as compared to liquid cultures. In presence of insulin HOXB4 overexpressing cells do not react to stroma encoded growth-restricting signals. Our results show that ectopic expression of HOXB4 in combination with MS-5 stroma exerts different effects in early and late human cord blood CD34(+) cells resulting in an enhanced proliferation of early CD34(+) cells in absence or presence of MS-5 stroma and an impaired output of late committed CD34(+) cells on MS-5 stroma.


Subject(s)
Antigens, CD34/metabolism , Cell Differentiation/physiology , Cell Proliferation , Hematopoietic Stem Cells/physiology , Homeodomain Proteins/physiology , Transcription Factors/physiology , Animals , Bone Marrow Cells/physiology , Cells, Cultured , Fetal Blood/cytology , Gene Expression , Homeodomain Proteins/metabolism , Humans , Insulin/physiology , Mice , Serum/physiology , Transcription Factors/metabolism
15.
Cell Cycle ; 5(1): 14-22, 2006 Jan.
Article in English | MEDLINE | ID: mdl-16357528

ABSTRACT

Ectopic expression of the homeodomain transcription factor HOXB4 expands hematopoietic stem and progenitor cells in vivo and in vitro, making HOXB4 a highly interesting candidate for therapeutic stem cell expansion. However, when expressed at high levels, HOXB4 concomitantly perturbs differentiation and thus likely predisposes the manipulated cells for leukemogenesis. We therefore asked whether the expression level of HOXB4 may be a critical parameter that influences the growth and transformation properties of transduced cells. Using a set of retroviral vectors which covered a 40-fold range of expression levels, we studied the consequences of HOXB4 expression at different levels in the well established Rat-1 fibroblast cell system. HOXB4 transformed Rat-1 fibroblasts beyond a certain threshold level of expression. Further escalation of HOXB4 expression, however, did not enhance transformation. Instead, HOXB4 mediated a dose dependent anti-proliferative effect on Rat-1 and NIH3T3 fibroblasts. This effect was aggravated under reduced serum concentrations and was, at least partially, due to an enhanced sensitivity of HOXB4 overexpressing cells to induction of apoptosis. Based on these results we propose that HOXB4 affects cell growth in a dose-dependent manner by sensitizing cells towards extrinsic signals.


Subject(s)
Homeodomain Proteins/metabolism , Signal Transduction/drug effects , Transcription Factors/metabolism , Animals , Cell Line , Cell Proliferation , Dose-Response Relationship, Drug , Gene Expression Regulation , Genetic Vectors/genetics , Homeodomain Proteins/genetics , Humans , Mice , Proto-Oncogene Proteins c-myc/metabolism , Rats , Transcription Factors/genetics
16.
Proc Natl Acad Sci U S A ; 102(34): 12101-6, 2005 Aug 23.
Article in English | MEDLINE | ID: mdl-16093308

ABSTRACT

Genetic manipulation of hematopoietic stem and progenitor cells is an important tool for experimental and clinical applied hematology. However, techniques that allow for gene targeting, subsequent in vitro selection, and expansion of genetically defined clones are available only for ES cells. Such molecularly defined and, hence, "safe" clones would be highly desirable for somatic gene therapy. Here, we demonstrate that in vitro differentiated ES cells completely recapitulate the growth and differentiation properties of adult bone marrow cells, in vitro and in vivo, when ectopically expressing HOXB4. Myeloid development was enforced and (T) lymphoid development suppressed over a wide range of expression levels, whereas only high expression levels of the transcription factor were detrimental for erythroid development. This indicates a close association between the amounts of ectopic HOXB4 present within a progenitor cell and and the decision to self renew or differentiate. Because HOXB4 mediates similar fates of ES-derived and bone marrow hematopoietic stem cells, the primitive embryonic cells can be considered a promising alternative for investigating hematopoietic reconstitution, in vivo, based on well defined clones. Provided that HOXB4 levels are kept within a certain therapeutic window, ES cells also carry the potential of efficient and safe somatic gene therapy.


Subject(s)
Bone Marrow Cells/cytology , Cell Differentiation/physiology , Gene Expression , Hematopoietic Stem Cells/cytology , Homeodomain Proteins/metabolism , Transcription Factors/metabolism , Animals , Bone Marrow Cells/metabolism , Flow Cytometry , Genetic Vectors , Hematopoietic Stem Cells/metabolism , Mice , Retroviridae
17.
Ann N Y Acad Sci ; 1044: 6-15, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15958692

ABSTRACT

The homeodomain transcription factor HOXB4 is one of the most attractive tools to expand hematopoietic stem cells in vitro and in vivo and to promote the formation of hematopoietic cells from in vitro differentiated embryonic stem cells. However, the expression levels compatible with the favorable effect of enhanced self-renewal without perturbing differentiation, in vivo, remain to be determined. In this paper, we discuss the necessity to define the "therapeutic width" of HOXB4 expression, based on observations from our lab and others that demonstrate that ectopic HOXB4 expression leads to a concentration-dependent perturbation of lineage differentiation of mouse and human hematopoietic cells. In summary, the combined results argue in favor of the existence of certain threshold levels for HOXB4 activity that control the differentiation and self-renewal behavior of hematopoietic stem and progenitor cells. Indeed, existing evidence suggests that dosage effects of ectopically expressed transcription factors may be more the rule than an exception.


Subject(s)
Cell Differentiation/physiology , Genes, Homeobox , Hematopoiesis , Hematopoietic Stem Cells , Homeodomain Proteins/genetics , Animals , Cell Lineage , Humans , In Vitro Techniques , Models, Biological
18.
Blood ; 101(6): 2191-8, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12424203

ABSTRACT

Increasing use of hematopoietic stem cells for retroviral vector-mediated gene therapy and recent reports on insertional mutagenesis in mice and humans have created intense interest to characterize vector integrations on a genomic level. We studied retrovirally transduced human peripheral blood progenitor cells with bone marrow-repopulating ability in immune-deficient mice. By using a highly sensitive and specific ligation-mediated polymerase chain reaction (PCR) followed by sequencing of vector integration sites, we found a multitude of simultaneously active human stem cell clones 8 weeks after transplantation. Vector integrations occurred with significantly increased frequency into chromosomes 17 and 19 and into specific regions of chromosomes 6, 13, and 16, although most of the chromosomes were targeted. Preferred genomic target sites have previously only been reported for wild-type retroviruses. Our findings reveal for the first time that retroviral vector integration into human marrow-repopulating cells can be nonrandom (P =.000 37).


Subject(s)
Bone Marrow Cells/cytology , Genetic Vectors , Retroviridae/genetics , Animals , Chromosomes, Human, Pair 13 , Chromosomes, Human, Pair 16 , Chromosomes, Human, Pair 17 , Chromosomes, Human, Pair 19 , Chromosomes, Human, Pair 6 , Cloning, Molecular , Female , Hematopoiesis , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/metabolism , Humans , In Situ Hybridization, Fluorescence , Mice , Mice, Inbred NOD , Mice, SCID , Polymerase Chain Reaction/methods , Transfection , Virus Integration
19.
Blood ; 101(5): 1759-68, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12406897

ABSTRACT

Ectopic retroviral expression of homeobox B4 (HOXB4) causes an accelerated and enhanced regeneration of murine hematopoietic stem cells (HSCs) and is not known to compromise any program of lineage differentiation. However, HOXB4 expression levels for expansion of human stem cells have still to be established. To test the proposed hypothesis that HOXB4 could become a prime tool for in vivo expansion of genetically modified human HSCs, we retrovirally overexpressed HOXB4 in purified cord blood (CB) CD34+ cells together with green fluorescent protein (GFP) as a reporter protein, and evaluated the impact of ectopic HOXB4 expression on proliferation and differentiation in vitro and in vivo. When injected separately into nonobese diabetic-severe combined immunodeficient (NOD/SCID) mice or in competition with control vector-transduced cells, HOXB4-overexpressing cord blood CD34+ cells had a selective growth advantage in vivo, which resulted in a marked enhancement of the primitive CD34+ subpopulation (P =.01). However, high HOXB4 expression substantially impaired the myeloerythroid differentiation program, and this was reflected in a severe reduction of erythroid and myeloid progenitors in vitro (P <.03) and in vivo (P =.01). Furthermore, HOXB4 overexpression also significantly reduced B-cell output (P <.01). These results show for the first time unwanted side effects of ectopic HOXB4 expression and therefore underscore the need to carefully determine the therapeutic window of HOXB4 expression levels before initializing clinical trials.


Subject(s)
Hematopoiesis/physiology , Homeodomain Proteins/physiology , Lymphocytes/cytology , Myeloid Cells/cytology , Transcription Factors/physiology , Animals , Endogenous Retroviruses/genetics , Fetal Blood/cytology , Gene Expression Regulation , Genetic Vectors/genetics , Homeodomain Proteins/genetics , Homeodomain Proteins/toxicity , Humans , Interleukin-3/pharmacology , Interleukin-6/pharmacology , K562 Cells/cytology , Leukemia Virus, Murine/genetics , Mice , Mice, Inbred NOD , Mice, SCID , Recombinant Fusion Proteins/physiology , Recombinant Fusion Proteins/toxicity , Recombinant Proteins/pharmacology , Transcription Factors/genetics , Transcription Factors/toxicity , Transduction, Genetic
20.
Nucleic Acids Res ; 30(12): e59, 2002 Jun 15.
Article in English | MEDLINE | ID: mdl-12060697

ABSTRACT

Site-specific recombination in genetically modified cells can be achieved by the activity of Cre recombinase from bacteriophage P1. Commonly an expression vector encoding Cre is introduced into cells; however, this can lead to undesired side-effects. Therefore, we tested whether cell-permeable Cre fusion proteins can be directly used for lox-specific recombination in a cell line tailored to shift from red to green fluorescence after loxP-specific recombination. Comparison of purified recombinant Cre proteins with and without a heterologous 'protein transduction domain' surprisingly showed that the unmodified Cre recombinase already possesses an intrinsic ability to cross the membrane border. Addition of purified recombinant Cre enyzme to primary bone marrow cells isolated from transgenic C/EBPalpha(fl/fl) mice also led to excision of the 'floxed' C/EBPalpha gene, thus demonstrating its potential for in vivo applications. We conclude that Cre enyzme itself or its intrinsic membrane-permeating moiety are attractive tools for direct manipulation of mammalian cells.


Subject(s)
Gene Targeting/methods , Integrases/metabolism , Recombination, Genetic , Viral Proteins/metabolism , Animals , Bone Marrow Cells/cytology , Bone Marrow Cells/metabolism , Cell Line , Cell Membrane/enzymology , Cells, Cultured , Genes, Reporter , Integrases/genetics , Mice , Mice, Transgenic , Protein Transport , Recombinant Fusion Proteins/isolation & purification , Recombinant Fusion Proteins/metabolism , Viral Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...