Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
1.
Am J Primatol ; 68(8): 753-64, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16847969

ABSTRACT

Natural killer (NK) cells from nonhuman primates have not been completely characterized, and methods for expanding nonhuman primates NK cells in vitro have been described only in rhesus species. The purpose of this report was to characterize NK cells in pigtail macaques (Macaca nemestrina), a species that is frequently used in studies of transplantation biology/immunology, virology, vaccine development, and reproductive biology. NK cells from Macaca nemestrina peripheral blood were best defined by the expression of CD16 and CD8alpha, and the absence of CD3. Subsets of these cells express CD56, NKp30, and NKp46. An enhanced ability to kill K562 cells was not present in fluorescence activated cell sorted (FACS)-purified CD16-/CD3+ and CD16-/CD56+ cells isolated from fresh peripheral blood. However, FACS-purified CD16+/CD3- and CD16+/CD56- cells were highly efficient killers of K562 cells. Macaca nemestrina NK cells can be expanded by in vitro culturing of FACS-purified CD16+/CD2-/CD3-/CD56- cells, or from peripheral blood cells depleted of cells expressing CD3, CD4, and HLA-DR. Cells in these cultures expand 70-fold after 21 days of culturing. After culturing, these cells express high levels of natural cytotoxicity receptors (NCRs) NKp30 and NKp46. NK cell populations obtained from FACS-purified CD16+/CD3-, CD16+/CD56- cells and CD3/CD4/HLA-DR-depleted cells were highly efficient killers of K562 cells. These data suggest that a population of highly enriched cytolytic NK cells can be obtained from purified CD16+/CD3- and CD16+/CD56- cells obtained from peripheral blood, as well as from cells that have been cultured and expanded from peripheral blood that is depleted of CD3/CD4/HLA-DR-expressing cells.


Subject(s)
Killer Cells, Natural/immunology , Macaca nemestrina/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Cell Culture Techniques , Cells, Cultured , Cytotoxicity Tests, Immunologic , Humans , K562 Cells , Killer Cells, Natural/classification , Lymphocyte Activation , Macaca nemestrina/blood , Phenotype , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism
2.
J Med Primatol ; 34(4): 201-8, 2005 Aug.
Article in English | MEDLINE | ID: mdl-16053498

ABSTRACT

In utero hematopoietic stem cell transplantation is a therapeutic procedure that could potentially cure many developmental diseases affecting the immune and hematopoietic systems. In most clinical and experimental settings of fetal hematopoietic transplantation the level of donor cell engraftment has been low, suggesting that even in the fetus there are significant barriers to donor cell engraftment. In postnatal hematopoietic transplantation donor cells obtained from mobilized peripheral blood engraft more rapidly than cells derived from marrow. We tested the hypothesis that use of donor hematopoietic/stem cells obtained from mobilized peripheral blood would improve engraftment and the level of chimerism after in utero transplantation in non-human primates. Despite the potential competitive advantage from the use of CD 34(+) from mobilized peripheral blood, the level of chimerism was not appreciably different from a group of animals receiving marrow-derived CD 34(+) donor cells. Based on these results, it is unlikely that this single change in cell source will influence the clinical outcome of fetal hematopoietic transplantation.


Subject(s)
Antigens, CD34/immunology , Fetal Therapies/methods , Hematopoiesis/physiology , Hematopoietic Stem Cell Transplantation/methods , Macaca nemestrina/physiology , Transplantation Chimera/physiology , Animals , Blood Component Removal/veterinary , Bone Marrow Cells/immunology , Bone Marrow Cells/physiology , Chimerism/veterinary , Female , Graft vs Host Disease/immunology , Hematopoiesis/immunology , Macaca nemestrina/embryology , Macaca nemestrina/immunology , Polymerase Chain Reaction/veterinary , Pregnancy , Transplantation Chimera/immunology
3.
Blood ; 105(9): 3749-56, 2005 May 01.
Article in English | MEDLINE | ID: mdl-15637143

ABSTRACT

The role of transplantation in infants with acute lymphoblastic leukemia (ALL) is not defined. We analyzed results of 40 infants diagnosed before age 12 months who received a hematopoietic cell transplant (HCT) between July 1982 and February 2003 in first complete remission (CR1; n = 17), CR2/3 (n = 7), or relapse (n = 16). Patients were conditioned with cyclophosphamide with total body irradiation (n = 39) or busulfan (n = 1). Donors were matched related (n = 8), mismatched related (n = 16), or unrelated (n = 16). Graft-versus-host disease (GVHD) prophylaxis was methotrexate or cyclosporine (n = 7) or methotrexate plus cyclosporine (n = 33). Thirty-nine patients engrafted, 20 developed acute GVHD, and 7 developed chronic GVHD. Sixteen patients relapsed and 7 died of other causes. Patients in CR1 had disease-free survival (DFS) of 76% compared with 45% for CR2/CR3 and 8% for relapse (P < .001). Of 33 patients with cytogenetic data, 26 (79%) had MLL gene rearrangement. Fourteen of these 26 were in CR1 and 11 survive in remission. Outcome was associated with phase of disease, but having the MLL gene was not a factor predictive of outcome. Late effects included growth and other hormone deficiencies. These data demonstrate that infants with ALL and MLL gene have excellent DFS when they received transplants in CR1, and consideration for transplantation in CR1 is warranted.


Subject(s)
Hematopoietic Stem Cell Transplantation/methods , Precursor Cell Lymphoblastic Leukemia-Lymphoma/therapy , Cause of Death , Female , Follow-Up Studies , Graft Survival , Graft vs Host Disease , Hematopoietic Stem Cell Transplantation/adverse effects , Hematopoietic Stem Cell Transplantation/mortality , Humans , Infant , Infant, Newborn , Male , Precursor Cell Lymphoblastic Leukemia-Lymphoma/complications , Precursor Cell Lymphoblastic Leukemia-Lymphoma/mortality , Remission Induction , Survival Analysis , Transplantation, Homologous , Treatment Outcome
4.
Clin Immunol ; 114(1): 30-41, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15596407

ABSTRACT

We evaluated whether long-term (2 months) administration of interleukin-7 (IL7) hastens immune recovery in baboons rendered severely lymphopenic by total body irradiation and antithymocyte globulin (ATG). Four baboons were treated with recombinant baboon IL7 and three baboons with placebo. Median CD4 T cell count at the end of IL7/placebo treatment was higher in the IL7-treated animals (2262 vs. 618/microl, P = 0.03). This appeared to be a result of peripheral expansion rather than de novo generation. Median cytomegalovirus (CMV)-specific IFNgamma-producing CD4 T cell count at the end of IL7/placebo treatment was higher in the IL7-treated animals (122 vs. 1/microl, P = 0.03). All animals were pretransplant cytomegalovirus-seropositive. One animal died at the end of IL7 treatment; necropsy showed extensive T cell infiltration of kidneys and lungs. In conclusion, IL7 stimulates the expansion of CD4 T cells, including functional antiviral cells. Clinical risk-benefit ratio needs to be evaluated.


Subject(s)
CD4-Positive T-Lymphocytes/drug effects , Interleukin-7/therapeutic use , Lymphopenia/drug therapy , Animals , CD4 Lymphocyte Count , CD4-Positive T-Lymphocytes/immunology , Cytomegalovirus/immunology , Cytomegalovirus Infections/immunology , Drug Administration Schedule , Interleukin-7/adverse effects , Interleukin-7/blood , Interleukin-7/immunology , Lymphopenia/immunology , Male , Papio
5.
Blood ; 105(6): 2410-4, 2005 Mar 15.
Article in English | MEDLINE | ID: mdl-15479725

ABSTRACT

T cells recognizing self-peptides are typically deleted in the thymus by negative selection. It is not known whether T cells against persistent viruses (eg, herpesviruses) are generated by the thymus (de novo) after the onset of the infection. Peptides from such viruses might be considered by the thymus as self-peptides, and T cells specific for these peptides might be deleted (negatively selected). Here we demonstrate in baboons infected with baboon cytomegalovirus and baboon lymphocryptovirus (Epstein-Barr virus-like virus) that after autologous transplantation of yellow fluorescent protein (YFP)-marked hematopoietic cells, YFP+ CD4 T cells against these viruses were generated de novo. Thus the thymus generates CD4 T cells against not only pathogens absent from the host but also pathogens present in the host. This finding provides a strong rationale to improve thymopoiesis in recipients of hematopoietic cell transplants and, perhaps, in other persons lacking de novo-generated CD4 T cells, such as AIDS patients and elderly persons.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Cytomegalovirus Infections/immunology , Epstein-Barr Virus Infections/immunology , Hematopoietic Stem Cells , Lymphopoiesis/immunology , Recovery of Function/immunology , Acquired Immunodeficiency Syndrome/immunology , Acquired Immunodeficiency Syndrome/therapy , Age Factors , Animals , Autoantigens/immunology , CD4-Positive T-Lymphocytes/virology , Cytomegalovirus/immunology , Herpesvirus 4, Human/immunology , Humans , Immune Tolerance , Papio anubis , Papio cynocephalus , Transplantation, Autologous
6.
Stem Cells ; 22(5): 759-69, 2004.
Article in English | MEDLINE | ID: mdl-15342940

ABSTRACT

In utero hematopoietic stem cell transplantation could potentially be used to treat many genetic diseases but rarely has been successful except in severe immunodeficiency syndromes. We explored two ways to potentially increase chimerism in a nonhuman primate model: (a) fetal immune suppression at the time of transplantation and (b) postnatal donor stem cell infusion. Fetal Macaca nemestrina treated with a combination of the corticosteroid betamethasone (0.9 mg/kg) and rabbit thymoglobulin (ATG; 50 mg/kg) were given haploidentical, marrow-derived, CD34+ -enriched donor cells. Animals treated postnatally received either donor-derived T cell-depleted or CD34+ -enriched marrow cells. Chimerism was determined by traditional and real-time polymerase chain reaction from marrow, marrow progenitors, peripheral blood, and mature peripheral blood progeny. After birth, the level of chimerism in the progenitor population was higher in the immune-suppressed animals relative to controls (11.3% +/- 2.7% and 5.1% +/- 1.5%, respectively; p = .057). Chimerism remained significantly elevated in both marrow (p = .02) and fluorescence-activated cell sorted and purified CD34+ cells (p = .01) relative to control animals at > or = 14 months of age. Peripheral blood chimerism, both at birth and long term, was similar in immune-suppressed and control animals. In the animals receiving postnatal donor cell infusions, there was an initial increase in progenitor chimerism; however, at 6-month follow-up, the level of chimerism was unchanged from the preinfusion values. Although fetal immune suppression was associated with an increase in the level of progenitor and marrow chimerism, the total contribution to marrow and the levels of mature donor progeny in the peripheral blood remained low. The level of long-term chimerism also was not improved with postnatal donor cell infusion.


Subject(s)
Blood Transfusion, Intrauterine/methods , Fetus/immunology , Hematopoietic Stem Cell Transplantation/methods , Immunosuppression Therapy/methods , Transplantation Chimera/immunology , Animals , Animals, Newborn , Antigens, CD34/immunology , Blood Donors , Bone Marrow/drug effects , Bone Marrow/immunology , Bone Marrow Cells/cytology , Bone Marrow Cells/drug effects , Bone Marrow Cells/immunology , Female , Hematopoietic Stem Cells/immunology , Immunosuppressive Agents/pharmacology , Macaca nemestrina , Male , Models, Animal , Pregnancy , Treatment Outcome , Up-Regulation/drug effects , Up-Regulation/immunology
7.
Mol Ther ; 9(3): 389-95, 2004 Mar.
Article in English | MEDLINE | ID: mdl-15006605

ABSTRACT

There has been significant progress toward clinically relevant levels of retroviral gene transfer into hematopoietic stem cells (HSC), and the therapeutic potential of HSC-based gene transfer has been convincingly demonstrated in children with severe combined immunodeficiency syndrome (SCID). However, the subsequent development of leukemia in two children with X-linked SCID who were apparently cured after transplantation of retrovirally corrected CD34+ cells has raised concerns regarding the safety of gene therapy approaches utilizing integrating vectors. Nonhuman primates and dogs represent the best available models for gene transfer safety and efficacy and are particularly valuable for evaluation of long-term effects. We have followed 42 rhesus macaques, 23 baboons, and 17 dogs with significant levels of gene transfer for a median of 3.5 years (range 1-7) after infusion of CD34+ cells transduced with retroviral vectors expressing marker or drug-resistance genes. None developed abnormal hematopoiesis or leukemia. Integration site analysis confirmed stable, polyclonal retrovirally marked hematopoiesis, without progression toward mono- or oligoclonality over time. These results suggest that retroviral integrations using replication-incompetent vectors, at copy numbers achieved using standard protocols, are unlikely to result in leukemogenesis and that patient- or transgene-specific factors most likely contributed to the occurrence of leukemia in the X-SCID gene therapy trial.


Subject(s)
Hematopoietic Stem Cells/cytology , Leukemia/pathology , Retroviridae/genetics , Animals , Antibodies/chemistry , Antigens, CD34/biosynthesis , Antigens, CD34/chemistry , Antigens, CD34/metabolism , Disease Models, Animal , Dogs , Follow-Up Studies , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors , Hematopoietic Stem Cells/pathology , Leukemia/etiology , Macaca , Papio , Severe Combined Immunodeficiency/genetics , Severe Combined Immunodeficiency/therapy , Stem Cells/cytology , Time Factors
8.
Blood ; 102(13): 4329-35, 2003 Dec 15.
Article in English | MEDLINE | ID: mdl-12816869

ABSTRACT

The nonobese diabetic/severe combined immune-deficient (NOD/SCID) mouse xenotransplantation assay is the most commonly used surrogate assay for the study of human candidate stem cells. In contrast to large animal and human studies, however, it is limited by the short life span of the recipients, the limited proliferative demand placed on the transplanted cells, and the inability to support differentiation into all hematopoietic lineages. In the present study, we directly compared hematopoietic repopulation in NOD/SCID mice with autologous reconstitution in the baboon, a well-established preclinical large animal model for stem cell transplantation. Baboon CD34-enriched marrow cells were retrovirally marked and infused into the irradiated baboon and the NOD/SCID mice. Although the percentage of gene-marked cells was high and remained stable in NOD/SCID mice up to 12 weeks and in those that underwent secondary transplantation, we observed a considerable decline and overall a significantly (10-fold) lower percentage of gene-marked cells in the baboons. In addition, clonal integration site analysis revealed common proviral vector integrants in NOD/SCID repopulating cells and in the baboon at 6 weeks but not at 6 months after transplantation. These results suggest that distinct hematopoietic stem/progenitor cells are responsible for hematopoietic reconstitution in NOD/SCID mice compared with nonhuman primates.


Subject(s)
Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/classification , Mice, Inbred NOD/physiology , Mice, SCID/physiology , Papio/physiology , Animals , Bacterial Proteins/genetics , Cell Lineage , Colony-Forming Units Assay , Genetic Vectors , Graft Survival , Green Fluorescent Proteins , Hematopoietic Stem Cells/cytology , Luminescent Proteins/genetics , Mice , Radiation Chimera , Species Specificity , Transplantation, Autologous , Virus Integration
9.
Stem Cells ; 21(3): 304-14, 2003.
Article in English | MEDLINE | ID: mdl-12743325

ABSTRACT

In utero transplantation of hematopoietic stem cells is a promising treatment for immune and hematologic diseases of fetuses and newborns. Unfortunately, there are limited data from nonhuman primates and humans describing optimal transplantation conditions. The purpose of this investigation was to determine the effect of T-cell number on engraftment and the level of chimerism after in utero transplantation in nonhuman primates. CD34(+) allogeneic adult bone marrow cells, obtained from the sire after G-CSF and stem cell factor administration, were transplanted into female fetal recipients. The average CD34(+) cell dose was 3.0 x 10(9)/kg (range, 9.9 x 10(8) to 4.4 x 10(9)) and the T-cell dose ranged from 2.6 x 10(5) to 1.1 x 10(8)/kg. Chimerism was determined in peripheral blood subsets (CD2, CD13, and CD20) and in progenitor cell populations by using polymerase chain reaction. Chimerism was noted in seven of eight live-born animals. The level of chimerism in the progenitor population was related to the fetal T-cell dose (r = 0.64, p < 0.02). At the lowest T-cell dose (2.6 x 10(5)/kg), no chimerism was detected. As the T-cell dose increased to 10(6-7)/kg, the level of chimerism increased. Adjusting the T-cell dose to 1.1 x 10(8)/kg resulted in fatal graft-versus-host disease (GVHD). The results of this study emphasize the importance of T cells in facilitating donor cell engraftment and in producing GVHD in fetal nonhuman primates. Some animals achieved levels of chimerism in the marrow hematopoietic progenitor cell population that would likely have clinical relevance. However, the levels of chimerism in peripheral blood were too low for therapeutic benefit. Further studies are needed to test methods that are likely to enhance donor cell engraftment and peripheral blood levels of donor cells.


Subject(s)
Fetus/surgery , Hematopoietic Stem Cell Transplantation/methods , T-Lymphocytes/transplantation , Transplantation Chimera/immunology , Animals , Antigens, CD34/immunology , Antigens, Surface/immunology , Female , Fetus/immunology , Graft Survival/physiology , Graft vs Host Disease/immunology , Graft vs Host Disease/physiopathology , Graft vs Host Disease/prevention & control , Hematopoietic Stem Cell Transplantation/trends , Lymphocyte Count , Macaca nemestrina , Male , Papio , T-Lymphocytes/immunology , Uterus/surgery
10.
Blood ; 101(10): 4209-18, 2003 May 15.
Article in English | MEDLINE | ID: mdl-12543864

ABSTRACT

In mice, interleukin-7 (IL-7) hastens T-cell reconstitution and might cause autoimmune diseases, lymphoma, and osteoporosis. We assessed the effect of IL-7 on T-cell reconstitution and toxicity in baboons that underwent total body irradiation followed by autologous transplantation of marrow CD34 cells. Three baboons received placebo and 3 baboons received recombinant human IL-7 (rhIL-7, 75 microg/kg twice a day subcutaneously) between 6 and 10 weeks after transplantation. The mean increase in blood absolute CD4 T-cell counts was 0.9-fold in the placebo-treated animals versus 9.0-fold in those treated with IL-7 (P =.02). The increase observed in the IL-7-treated animals appeared attributable to peripheral expansion rather than de novo generation. The IL-7-treated animals had greater mean increases in the volumes of the spleen (2.0-fold with placebo versus 4.5-fold with IL-7, P =.02) and lymph nodes (1.8-fold with placebo versus 4.1-fold with IL-7, P =.10) but not the thymus (3.4-fold with placebo versus 1.1-fold with IL-7, P =.18). Side effects of IL-7 included thrombocytopenia and possibly neutropenia and hemolytic anemia. One IL-7-treated animal failed to thrive due to a disease resembling graft-versus-host disease. No animals developed lymphoma. Bone density was not decreased. In conclusion, IL-7 raises CD4 T-cell counts in irradiated primates. It remains to be determined whether this is associated with clinical benefit.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Hematopoietic Stem Cells/immunology , Interleukin-7/therapeutic use , Stem Cell Transplantation , Transplantation, Autologous/immunology , Animals , Antigens, CD34/analysis , Base Sequence , CD4-Positive T-Lymphocytes/drug effects , DNA Primers , Filgrastim , Granulocyte Colony-Stimulating Factor/therapeutic use , Interleukin-7/immunology , Major Histocompatibility Complex , Papio , Polymerase Chain Reaction , Recombinant Proteins , Stem Cell Factor/therapeutic use
11.
Blood ; 100(8): 2737-43, 2002 Oct 15.
Article in English | MEDLINE | ID: mdl-12351380

ABSTRACT

Hematopoietic bone marrow stem cells generate differentiated blood cells and, when transplanted, may contribute to other organs, such as the brain, heart, and liver. An understanding of in vivo clonal behavior of stem cells will have important implications for cellular and gene therapy. For the first time, we have directly demonstrated the derivation of circulating peripheral blood cells from individual stem cell clones. We analyzed the clonal composition of retrovirus-marked peripheral blood leukocyte populations in 2 different primate models by a novel direct genomic sequencing technique allowing the identification of vector insertion sites. More than 80 contributing long-term hematopoietic clones were identified in individual rhesus macaque peripheral blood transplant recipients and more than 25 different clones in a baboon marrow transplant recipient. Up to 5 insertion sequences from each animal were used to trace the long-term contribution of stem cell clones in these primate models. Continuous and mostly pluripotent contributions of peripheral blood leukocytes from each of the traced clones could be detected for the entire follow-up period of 23 to 33 months. Our study provides direct molecular evidence for a polyclonal, multilineage, and sustained contribution of individual stem cells to primate hematopoiesis.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/physiology , Animals , Base Sequence , Bone Marrow Cells/cytology , Bone Marrow Cells/physiology , Clone Cells , DNA Primers , Genetic Vectors , Models, Animal , Papio , Polymerase Chain Reaction
12.
J Hematother Stem Cell Res ; 11(4): 617-31, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12201950

ABSTRACT

Successful in utero hematopoietic stem cell transplantation will likely represent a major step forward in the management of patients with congenital hematological, metabolic, and immunological disorders. We review the naturally occurring models of hematopoietic chimerism in animals and humans, as well as available experimental animal data and human clinical attempts of fetal transplantation. Data available from naturally occurring models and experimental models of fetal transplantation suggest that this technique should be translatable to the human fetus. However, to date, the success of human fetal hematopoietic stem cell therapy has been limited to fetuses with severe immunologic defects. Evaluation of successful attempts of human transplantation, the ontogeny of fetal immune development, and data available from animals provide insights into innovative approaches to fetal therapy that may bring the reality of successful fetal transplantation closer.


Subject(s)
Fetal Tissue Transplantation/physiology , Hematologic Diseases/therapy , Stem Cell Transplantation/methods , Animals , Fetal Tissue Transplantation/methods , Fetal Tissue Transplantation/trends , Humans , Immune System Diseases/therapy , Metabolic Diseases/therapy , Models, Animal , Stem Cell Transplantation/trends
13.
Mol Ther ; 6(1): 83-90, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12095307

ABSTRACT

Downregulation and loss of proviral expression have been demonstrated to occur in a variety of in vitro studies and in mouse models. Here we evaluated the kinetics of proviral expression after transplantation in a competitive repopulating model in the baboon. Transgene persistence and green fluorescent protein (GFP) expression in peripheral blood leukocytes (PBL) were analyzed in four animals by semiquantitative PCR and flow cytometry for up to 80 weeks (range 17-80). All animals were transplanted with cells transduced with EGFP or EYFP reporters driven by Moloney murine leukemia virus (MoMuLV) or a modified promoter/enhancer, (MND) respectively. Simultaneous dual-color analysis of fluorescence levels in granulocyte and lymphocyte subsets following hematopoietic reconstitution demonstrated progressive loss of fluorescence intensity occurring predominantly early after transplant in cells transduced with both retrovirus backbones and at serial time points. In addition, we carried out PCR analysis of DNA extracted from sorted EGFP(-)/EYFP(-) cells and confirmed the presence of cells genetically marked by either vector in this population, indicating the persistence of cells that have downregulated or lost retroviral gene expression. In comparison to mouse studies, however, we did not detect substantial differences between MND and MoMuLV backbones.


Subject(s)
Gene Expression , Genetic Vectors , Luminescent Proteins/metabolism , Retroviridae/genetics , Animals , Flow Cytometry , Green Fluorescent Proteins , Kinetics , Moloney murine leukemia virus/genetics , Papio , Promoter Regions, Genetic
14.
Hum Gene Ther ; 13(8): 891-9, 2002 May 20.
Article in English | MEDLINE | ID: mdl-12031122

ABSTRACT

In an effort to improve hematopoietic stem cell gene transfer rates using gibbon ape leukemia virus (GALV)-pseudotype retroviral vectors in baboons, we have studied preselection of transduced green fluorescent protein (GFP)-expressing CD34-enriched marrow cells. Three animals were transplanted with GFP-selected (GS) CD34-enriched marrow. To ensure engraftment, preselected GFP-positive cells were infused together with unselected neo-transduced cells. After transduction on fibronectin, cells were cultured for an additional 2 days to allow for expression of GFP. GFP-expressing cells were enriched by fluorescence-activated cell sorting and infused together with cells from the unselected fractions after myeloablative irradiation of the recipient. Three other animals were transplanted with GFP-transduced CD34-enriched cells without prior GFP selection (GU). At 4 weeks after transplant, the percentage of GFP-expressing white blood cells was significantly higher in the GS group (6.6%) than in the GU group (1.3%) (p < 0.002). The higher gene transfer levels in the animals transplanted with GS cells gradually declined, and by day 100 after transplant, gene transfer levels were similar in both groups. PCR analysis performed on genomic DNA isolated from peripheral blood cells demonstrated that the decline in GFP-positive cells was due to the loss of gene-marked cells and not due to loss of expression. These results show that transplantation of CD34-positive marrow cells selected for GFP-positive cells after transduction provides high levels of transduced granulocytes in the short term. However, using this experimental design with concomitant infusion of unselected cells and the use of oncoretroviral vectors, preenrichment of vector-expressing, transduced CD34-enriched cells does not improve long-term persistence and expression.


Subject(s)
Antigens, CD34/genetics , Bone Marrow Cells/metabolism , Genetic Vectors/metabolism , Leukemia Virus, Gibbon Ape/metabolism , Luminescent Proteins/genetics , Transduction, Genetic , Animals , Bone Marrow Transplantation , Flow Cytometry , Gene Transfer Techniques , Genetic Therapy/methods , Genetic Vectors/genetics , Green Fluorescent Proteins , Kinetics , Leukemia Virus, Gibbon Ape/genetics , Luminescent Proteins/blood , Mice , Microscopy, Fluorescence , Papio
SELECTION OF CITATIONS
SEARCH DETAIL
...