Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 105
Filter
1.
Ned Tijdschr Geneeskd ; 151(36): 1975-80, 2007 Sep 08.
Article in Dutch | MEDLINE | ID: mdl-17953170

ABSTRACT

Extensive research is ongoing worldwide on the clinical utility of gene therapy, particularly for the treatment of cancer and genetic disorders. Two gene therapy products have already been approved recently in China. Clinical experience with gene therapy has also been accumulating in the Netherlands: over 200 Dutch patients have now been treated in clinical trials. Published results indicate that gene therapy is generally safe. Gene therapy appears to be effective for some genetic disorders, such as severe combined immune deficiency and haemophilia B. The efficacy of gene therapy, particularly in the treatment of cancer, appears to be limited up till now.


Subject(s)
Gene Transfer Techniques , Genetic Therapy , Neoplasms/therapy , Clinical Trials as Topic , Forecasting , Genetic Vectors , Humans , Netherlands , Safety
2.
Leukemia ; 21(4): 754-63, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17268520

ABSTRACT

The occurrence of leukemia in a gene therapy trial for SCID-X1 has highlighted insertional mutagenesis as an adverse effect. Although retroviral integration near the T-cell acute lymphoblastic leukemia (T-ALL) oncogene LIM-only protein 2 (LMO2) appears to be a common event, it is unclear why LMO2 was preferentially targeted. We show that of classical T-ALL oncogenes, LMO2 is most highly transcribed in CD34+ progenitor cells. Upon stimulation with growth factors typically used in gene therapy protocols transcription of LMO2, LYL1, TAL1 and TAN1 is most prominent. Therefore, these oncogenes may be susceptible to viral integration. The interleukin-2 receptor gamma chain (IL2Rgamma), which is mutated in SCID-X1, has been proposed as a cooperating oncogene to LMO2. However, we found that overexpressing IL2Rgamma had no effect on T-cell development. In contrast, retroviral overexpression of LMO2 in CD34+ cells caused severe abnormalities in T-cell development, but B-cell and myeloid development remained unaffected. Our data help explain why LMO2 was preferentially targeted over many of the other known T-ALL oncogenes. Furthermore, during T-cell development retrovirus-mediated expression of IL2Rgamma may not be directly oncogenic. Instead, restoration of normal IL7-receptor signaling may allow progression of T-cell development to stages where ectopic LMO2 expression causes aberrant thymocyte growth.


Subject(s)
Antigens, CD34/immunology , DNA-Binding Proteins/genetics , Genetic Therapy/methods , Leukemia-Lymphoma, Adult T-Cell/genetics , Leukemia/genetics , Leukemia/therapy , Metalloproteins/genetics , Receptors, Interleukin-2/genetics , T-Lymphocytes/immunology , Adaptor Proteins, Signal Transducing , Antigens, CD/immunology , Growth Substances/pharmacology , Humans , LIM Domain Proteins , Leukemia-Lymphoma, Adult T-Cell/immunology , Leukemia-Lymphoma, Adult T-Cell/therapy , Mutagenesis, Insertional , Proto-Oncogene Proteins , Retroviridae
3.
Gene Ther ; 13(21): 1524-33, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16763662

ABSTRACT

Retroviral vectors with self-inactivating (SIN) long-terminal repeats not only increase the autonomy of the internal promoter but may also reduce the risk of insertional upregulation of neighboring alleles. However, gammaretroviral as opposed to lentiviral packaging systems produce suboptimal SIN vector titers, a major limitation for their clinical use. Northern blot data revealed that low SIN titers were associated with abundant transcription of internal rather than full-length transcripts in transfected packaging cells. When using the promoter of Rous sarcoma virus or a tetracycline-inducible promoter to generate full-length transcripts, we obtained a strong enhancement in titer (up to 4 x 10(7) transducing units per ml of unconcentrated supernatant). Dual fluorescence vectors and Northern blots revealed that promoter competition is a rate-limiting step of SIN vector production. SIN vector stocks pseudotyped with RD114 envelope protein had high transduction efficiency in human and non-human primate cells. This study introduces a new generation of efficient gammaretroviral SIN vectors as a platform for further optimizations of retroviral vector performance.


Subject(s)
Gammaretrovirus/genetics , Genetic Therapy/methods , Genetic Vectors/genetics , Promoter Regions, Genetic , Terminal Repeat Sequences , Animals , Antigens, CD34 , Cell Line , Flow Cytometry , Gene Expression Regulation , Genetic Engineering , Macaca mulatta , Male , Plasmids , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/virology , Transduction, Genetic/methods , Transfection , Virus Inactivation
4.
Radiat Res ; 155(3): 409-16, 2001 Mar.
Article in English | MEDLINE | ID: mdl-11182791

ABSTRACT

The Chernobyl nuclear power plant accident on April 26, 1986 was the largest in the history of the peaceful use of nuclear energy. Of the 237 individuals initially suspected to have been significantly exposed to radiation during or in the immediate aftermath of the accident, the diagnosis of acute radiation sickness (ARS) could be confirmed in 134 cases on the basis of clinical symptoms. Of these, 54 patients suffered from cutaneous radiation syndrome (CRS) to varying degrees. Among the 28 patients who died from the immediate consequences of accidental radiation exposure, acute hemopoietic syndrome due to bone marrow failure was the primary cause of death only in a minority. In 16 of these 28 deaths, the primary cause was attributed to CRS. This report describes the characteristic cutaneous sequelae as well as associated clinical symptoms and diseases of 15 survivors of the Chernobyl accident with severe localized exposure who were systematically followed up by our groups between 1991 and 2000. All patients presented with CRS of varying severity, showing xerosis, cutaneous telangiectasias and subungual splinter hemorrhages, hemangiomas and lymphangiomas, epidermal atrophy, disseminated keratoses, extensive dermal and subcutaneous fibrosis with partial ulcerations, and pigmentary changes including radiation lentigo. Surprisingly, no cutaneous malignancies have been detected so far in those areas that received large radiation exposures and that developed keratoses; however, two patients first presented in 1999 with basal cell carcinomas on the nape of the neck and the right lower eyelid, areas that received lower exposures. During the follow-up period, two patients were lost due to death from myelodysplastic syndrome in 1995 and acute myelogenous leukemia in 1998, respectively. Other radiation-induced diseases such as dry eye syndrome (3/15), radiation cataract (5/15), xerostomia (4/15) and increased FSH levels (7/15) indicating impaired fertility were also documented. This study, which analyzes 14 years in the clinical course of a cohort of patients with a unique exposure pattern, corroborates the requirement for long-term, if not life-long, follow-up not only in atomic bomb survivors, but also after predominantly local radiation exposure.


Subject(s)
Power Plants , Radiation Injuries/physiopathology , Radioactive Hazard Release , Cohort Studies , Follow-Up Studies , Humans , Skin/injuries , Skin/radiation effects , Ukraine
5.
Exp Hematol ; 29(1): 30-40, 2001 Jan.
Article in English | MEDLINE | ID: mdl-11164103

ABSTRACT

A sufficiently high dose of thrombopoietin to overcome initial c-mpl-mediated clearance stimulates hematopoietic reconstitution following myelosuppressive treatment. We studied the efficacy of thrombopoietin on survival after supralethal total body irradiation (9 Gy) of C57BL6/J mice and the occurrence of infectious and thrombotic complications in comparison with a bone marrow graft or prophylactic antibiotic treatment. Administration of 0.3 microg thrombopoietin, 2 hours after irradiation, protected 62% of the mice as opposed to no survival in placebo controls. A graft with a supraoptimal number of syngeneic bone marrow cells (10(6) cells) fully prevented mortality, whereas antibiotic treatment was ineffective. Blood cell recovery was observed in the thrombopoietin-treated mice but not in the placebo or antibiotic-treated group. Bone marrow and spleen cellularity as well as colony-forming unit granulocyte-macrophage and burst-forming unit erythroid were considerably increased in thrombopoietin-treated mice relative to controls. Histologic examination at day 11 revealed numerous petechiae and vascular obstructions within the brain microvasculature of placebo-treated mice, which was correlated with hypercoagulation and hypofibrinolysis. Thrombopoietin treatment prevented coagulation/fibrinolysis disorder and vascular thrombosis. High fibrinogen levels were related to bacterial infections in 67% of placebo-treated mice and predicted mortality, whereas the majority of the thrombopoietin-treated mice did not show high fibrinogen levels and endotoxin was not detectable in plasma. We conclude that thrombopoietin administration prevents mortality in mice subjected to 9-Gy total body irradiation both by interfering in the cascade leading to thrombotic complications and by amelioration of neutrophil and platelet recovery and thus protects against infections and hemorrhages.


Subject(s)
Bacterial Infections/prevention & control , Radiation Injuries, Experimental/drug therapy , Thrombopoietin/therapeutic use , Thrombosis/prevention & control , Animals , Bacterial Infections/etiology , Biomarkers , Blood Coagulation Disorders/etiology , Bone Marrow/drug effects , Bone Marrow Diseases/complications , Bone Marrow Diseases/etiology , Disease Susceptibility , Drug Evaluation, Preclinical , Endotoxemia/etiology , Endotoxemia/prevention & control , Fibrinogen/analysis , Fibrinogen/biosynthesis , Fibrinogen/genetics , Fibrinolysis/drug effects , Hemorrhage/etiology , Hemorrhage/prevention & control , Immunologic Deficiency Syndromes/etiology , Leukocyte Count , Male , Mice , Mice, Inbred C57BL , Neutrophils , Platelet Activation/drug effects , Platelet Count , RNA, Messenger/biosynthesis , Radiation Injuries, Experimental/blood , Radiation Injuries, Experimental/complications , Radiation Injuries, Experimental/immunology , Recombinant Proteins/pharmacology , Recombinant Proteins/therapeutic use , Thrombopoietin/pharmacology , Thrombosis/etiology , Whole-Body Irradiation/adverse effects
6.
Hum Gene Ther ; 11(15): 2129-41, 2000 Oct 10.
Article in English | MEDLINE | ID: mdl-11044914

ABSTRACT

The effects of cytokine stimulation during retroviral transduction on in vivo reconstitution of mouse hematopoietic stem cells was tested in a murine competitive repopulation assay with alpha-thalassemia as a marker to distinguish donor and recipient red blood cells (RBCs) and the enhanced green fluorescent protein (EGFP) as a marker for gene transfer. After transplantation, EGFP was detected in up to 90% of circulating RBCs, platelets, and leukocytes, and in primitive progenitors in bone marrow (BM), spleen, and thymus of individual transplanted mice for observation periods of more than 6 months. Large quantitative differences in reconstitution were observed after transplantation with graded numbers (1000-30, 000) of EGFP(+) cells preconditioned with various combinations of Kit ligand (KL), FLT-3 ligand (FL), thrombopoietin (TPO), interleukin 3 (IL-3), and IL-11. Relative to nonmanipulated BM cells, repopulation of EGFP(+) cells was maintained by KL/FL/TPO stimulation, but approximately 30-fold reduced after KL/FL/TPO/IL-3, or KL/FL/IL-3/IL-11. These differences were not caused by changes in the ability of immature hematopoietic cells to home to the BM, which was only moderately reduced. In conclusion, these quantitative transplantation studies of mice demonstrate the importance of optimal ex vivo cytokine stimulation for gene transfer to stem cells with retention of their in vivo hematopoietic potential, and also emphasize that overall in vitro transduction frequency does not predict gene transfer to repopulating stem cells.


Subject(s)
Bone Marrow Cells/metabolism , Gene Transfer Techniques , Interleukin-11/genetics , Interleukin-3/genetics , Membrane Proteins/genetics , Retroviridae/genetics , Stem Cell Factor/genetics , Thrombopoietin/genetics , 3T3 Cells , Animals , Blood Platelets/metabolism , Cell Separation , Erythrocytes/metabolism , Female , Flow Cytometry , Genetic Vectors , Green Fluorescent Proteins , Immunophenotyping , Interleukin-11/metabolism , Interleukin-3/metabolism , Light , Luminescent Proteins/genetics , Male , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Scattering, Radiation , Stem Cell Factor/metabolism , Stem Cells/immunology , Thrombopoietin/metabolism , Time Factors , Transduction, Genetic , alpha-Thalassemia/genetics
7.
Exp Dermatol ; 9(4): 252-7, 2000 Aug.
Article in English | MEDLINE | ID: mdl-10949546

ABSTRACT

Keratinocytes have great promise as targets for gene therapy involving both skin as well as for systemic disorders due to their availability and potential long life span. Improvement of gene transfer into keratinocytes will be greatly facilitated by markers that will allow both rapid detection and efficient selection of transduced cells. For these purposes, a recombinant version of the Aequorea victoria green fluorescent protein that is enhanced for high-level expression in mammalian cells (EGFP) was placed into a replication-deficient retroviral vector. High-titer retrovirus was used to transduce both primary cultures of neonatal foreskin-derived human keratinocytes (HK) as well as the immortalized keratinocyte-derived cell line HaCaT. Both cell types stably expressed the EGFP, and this marker allowed rapid purification of transduced cells by fluorescence-activated cell sorting. EGFP expression was seen in HaCaT keratinocytes for at least 40 passages, and the presence of this construct did not effect cell growth, or apoptosis in response to UVB or etoposide. Transduced populations of HK were grafted into SCID mice, resulting in a functional epidermis. EGFP expression was readily seen in vivo by exposing the xenografts to an ultraviolet light source. These studies demonstrate the feasibility of using EGFP as a convenient and rapid marker to monitor keratinocyte gene transfer both in vitro and in vivo.


Subject(s)
Genetic Therapy/methods , Keratinocytes/metabolism , Luminescent Proteins/genetics , Animals , Apoptosis , Cell Line , Cells, Cultured , Gene Expression , Genetic Vectors , Green Fluorescent Proteins , Humans , Keratinocytes/cytology , Keratinocytes/transplantation , Mice , Mice, SCID , Recombinant Proteins/genetics , Retroviridae/genetics , Transduction, Genetic , Transplantation, Heterologous
8.
Exp Hematol ; 28(7): 753-9, 2000 Jul.
Article in English | MEDLINE | ID: mdl-10907636

ABSTRACT

OBJECTIVE: If administered in a sufficiently high dose to overcome receptor-mediated clearance and in a well-scheduled manner, thrombopoietin (TPO) prominently stimulates hematopoietic reconstitution following myelosuppressive treatment and potentiates the efficacy of both granulocyte-macrophage colony-stimulating factor (GM-CSF) and granulocyte colony-stimulating factor (G-CSF). However, TPO alone is not effective after bone marrow transplantation. Based on results of GM-CSF and TPO treatment after myelosuppression that resulted in augmented thrombocyte, reticulocyte, and leukocyte regeneration, we evaluated TPO/GM-CSF treatment after lethal irradiation followed by autologous bone marrow transplantation. MATERIALS AND METHODS: Young adult Rhesus monkeys were subjected to 8-Gy total body irradiation (TBI) (x-rays) followed by transplantation of 10(7)/kg unfractionated bone marrow cells. TPO 5 microg/kg was administered intravenously at day 0 to obtain rapidly high levels. Animals then were treated with 5 microg/kg Rhesus TPO and 25 microg/kg GM-CSF given SC on days 1 to 14 after TBI. RESULTS: The grafts shortened the profound pancytopenia induced by 8-Gy TBI from 5-6 weeks to 3 weeks. The combination of TPO and GM-CSF did not significantly influence the recovery patterns of thrombocytes (p = 0.39), reticulocytes (p = 0.08), white blood cells (p = 0.08), or bone marrow progenitors compared to TPO alone. CONCLUSIONS: The present study demonstrates that, after high-dose TBI and transplantation of a limited number of unfractionated bone marrow cells, simultaneous administration of TPO and GM-CSF after TBI is ineffective in preventing pancytopenia. This result contrasts sharply with the prominent stimulation observed in a 5-Gy TBI myelosuppression model, despite a similar level of pancytopenia in the 8-Gy model of the present study. The discordant results of this growth factor combination in these two models may imply codependence of the hematopoietic response to TPO and/or GM-CSF on other factors or cytokines.


Subject(s)
Bone Marrow Transplantation , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Thrombopoietin/therapeutic use , Whole-Body Irradiation , Animals , Cell Separation , Drug Therapy, Combination , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Lymphocyte Subsets/cytology , Macaca mulatta , Male , Thrombopoietin/administration & dosage
9.
Leukemia ; 13(4): 605-13, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10214869

ABSTRACT

The feasibility of using the enhanced green fluorescent protein (EGFP) as a selectable reporter molecule of retroviral-mediated gene transfer in immature rhesus monkey and human CD34+ hematopoietic cells was examined. Retroviral transduction with the MFG-EGFP retroviral vector resulted in readily detectable EGFP expression in 27% of human and 11-35% of rhesus monkey bone marrow cells, and in 17-38% of rhesus monkey peripheral blood cells mobilized with FLT3 ligand (FL) and granulocyte colony-stimulating factor (G-CSF). In addition, we used the human CD34+ KG1A cell line as a model to study viability and growth of successfully transduced cells. Cultures of mock- and EGFP-transduced KG1A cells generated equal viable cell numbers for at least 1 month, indicating the absence of a cytotoxic effect of EGFP expression in these cells. FACS selection on the basis of EGFP and CD34 expression resulted in enriched subsets (> or = 87%) of CD34+ EGFP-negative and CD34+ EGFP-positive KG1A, rhesus monkey and human bone marrow cells, demonstrating the potential of obtaining almost pure populations of transduced immature hematopoietic cells. EGFP expression was also readily demonstrated in erythroid and granulocyte/macrophage colonies derived from the CD34+ EGFP-positive rhesus monkey and human bone marrow cells by either inverted fluorescence microscopy or flow cytometry. Using four-color flow cytometry, EGFP expression could also be demonstrated in viable and phenotypically defined immature subpopulations of the CD34+ cells, ie those expressing little or no HLA-DR (rhesus monkey) or CD38 (human) antigens at the cell surface. These results demonstrate that EGFP is a very useful marker to monitor gene transfer efficiency in phenotypically defined immature rhesus monkey and human hematopoietic cell types and to select for these cells by multicolor flow cytometry prior to transplantation.


Subject(s)
Cell Separation/methods , Flow Cytometry/methods , Hematopoietic Stem Cell Mobilization , Hematopoietic Stem Cells/cytology , Luminescent Proteins/analysis , Recombinant Fusion Proteins/analysis , Transfection , Animals , Antigens, CD34/analysis , Biomarkers , Bone Marrow Cells , Cell Line , Cell Lineage , Colony-Forming Units Assay , Feasibility Studies , Gene Expression , Genes, Reporter , Genetic Vectors/genetics , Green Fluorescent Proteins , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/metabolism , Humans , Immunophenotyping , Luminescent Proteins/biosynthesis , Luminescent Proteins/genetics , Macaca mulatta , Male , Membrane Proteins/pharmacology , Recombinant Fusion Proteins/biosynthesis , Retroviridae/genetics
10.
Leukemia ; 13(4): 618-28, 1999 Apr.
Article in English | MEDLINE | ID: mdl-10214871

ABSTRACT

In this study the ability of malignant and normal progenitors in peripheral blood (PB) and bone marrow (BM) of CML patients in chronic phase to proliferate and produce mature progeny after transplantation into hereditary immunodeficient (SCID and NOD/SCID) mice was examined. Engraftment in NOD/SCID mice preconditioned by total body irradiation (TBI) alone was 10-fold higher than in SCID mice preconditioned by macrophage depletion and TBI, demonstrating that NOD/SCID mice are more suitable for engraftment of chronic phase CML cells. Low-density cells at cell doses of 10-30 x 10(6) and purified CD34+ cells at doses of approximately 0.2 x 10(6) engrafted NOD/SCID mice, with levels of 2 to 20% CD45+ cells with production of monocytes, granulocytes, erythroid cells, B-lymphocytes, CD34+ cells and variable frequencies of erythroid and myeloid colony-forming cells. As demonstrated by fluorescent in situ hybridization (FISH) analysis, purified human myeloid, B-lymphoid, erythroid and CD34+ cells from chimeric mouse BM contained Philadelphia-chromosome (Ph)-positive cells and Ph- cells in similar frequencies as primary cells from the CML patients. These results demonstrate that production of mature normal as well as malignant cells of multiple lineages were supported with similar efficiency. In contrast, all human erythroid and myeloid clonogenic cells detected in the mice were Ph-, which can be attributed to less efficient maintenance or more rapid differentiation of immature Ph+ cells in the mouse microenvironment. CML blast crisis cells also grew well in NOD/SCID mice, with 80-90% of human cells produced containing the Ph- chromosome. The availability of an in vivo assay that supports outgrowth of normal and malignant stem cells from chronic phase and blast crisis CML patients will facilitate examination of differential effects of growth factors, inhibitory cytokines and cytotoxic drugs on survival of normal and malignant stem cells in vivo and on progression of chronic phase CML towards blast crisis.


Subject(s)
Hematopoietic Stem Cells/pathology , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Neoplastic Stem Cells/pathology , Animals , Blast Crisis/pathology , Cell Differentiation , Cell Lineage , Graft Survival , Humans , Immunocompromised Host , In Situ Hybridization, Fluorescence , Leukemia, Myeloid, Chronic-Phase/pathology , Mice , Mice, Inbred NOD , Mice, SCID , Neoplasm Transplantation , Philadelphia Chromosome , Radiation Chimera , Transplantation, Heterologous , Tumor Stem Cell Assay
11.
J Virol ; 73(5): 3960-7, 1999 May.
Article in English | MEDLINE | ID: mdl-10196291

ABSTRACT

Several factors are thought to limit the efficiency of retroviral transduction in clinical gene therapy protocols that target hematopoietic stem cells. For example, the level of expression of the amphotropic receptor Pit-2, a phosphate symporter, appears to be low in human and murine hematopoietic stem cells. We have previously demonstrated that transduction of hematopoietic cells in the presence of the fibronectin (FN) fragment CH-296 is extremely efficient (H. Hanenberg, X. L. Xiao, D. Dilloo, K. Hashino, I. Kato, and D. A. Williams, Nat. Med. 2:876-882, 1996). To examine functionally whether the retrovirus receptor is a limiting factor in transduction of hematopoietic cells, we performed competition experiments in the presence of FN CH-296 with retrovirus vectors pseudotyped with the same or a different envelope protein. We demonstrate in both human erythroleukemia (HEL) cells and primary human CD34(+) hematopoietic cells inhibition of efficient infection due to receptor interference when two vectors targeting the amphotropic receptor are used simultaneously. Receptor interference lasted up to 24 h. No interference was demonstrated when vectors targeting the amphotropic receptor and the gibbon ape leukemia virus (GALV) receptor Pit-1 were used concurrently. In contrast, simultaneous infection with vectors targeting both Pit-1 and Pit-2 yielded transduction efficiencies consistently higher than with either vector alone in both HEL cells and human CD34(+) hematopoietic cells. These data demonstrate that the use of FN CH-296 leads to amphotropic receptor saturation in these cells. Simultaneous infection with vectors targeting both amphotropic and GALV receptors may prove to be of additional benefit in the design of gene therapy protocols.


Subject(s)
Fibronectins/metabolism , Receptors, Virus/metabolism , Retroviridae Proteins, Oncogenic/metabolism , Retroviridae/metabolism , Viral Envelope Proteins/metabolism , Adult , Cell Transformation, Viral , Humans , Time Factors , Tumor Cells, Cultured
12.
Int J Radiat Biol ; 75(2): 183-91, 1999 Feb.
Article in English | MEDLINE | ID: mdl-10072179

ABSTRACT

PURPOSE: To investigate the long-term effects of X-irradiation on different aspects of gastrointestinal function in the non-human primate (Macaca mulatta). MATERIALS AND METHODS: Animals were exposed to X-radiation (5 or 6 Gy) or not (sham) and gastrointestinal function was investigated 4-6 years after exposure. Basal and agonist-stimulated short circuit current (Isc) responses were measured in isolated jejunum. Intestinal tissue was taken for histological analysis as well as for determination of mucosal marker enzyme activities and gastrointestinal regulatory peptide levels. Vasoactive intestinal peptide receptor characteristics were determined as well as VIP-stimulated Isc responses. GI peptides were also measured in plasma. RESULTS: Few differences were seen in basal electrical parameters or tissue morphology but there was a tendency for reduced basolateral membrane enzyme activity. VIP-stimulated Isc responses were reduced in irradiated animals as were VIP-stimulated adenylate cyclase responses. Plasma and tissue (ileal and colonic muscle layers) gastrin releasing peptide levels were increased in irradiated animals. In contrast circulating gastrin levels were lower. CONCLUSIONS: Late effects of total-body irradiation on GI function in monkeys showed altered circulating and tissue levels of some GI peptides. In addition the biological effects of vasoactive intestinal peptide were modified.


Subject(s)
Digestive System/metabolism , Digestive System/radiation effects , Gastrin-Releasing Peptide/radiation effects , Vasoactive Intestinal Peptide/radiation effects , Adenylyl Cyclases/blood , Adenylyl Cyclases/metabolism , Adenylyl Cyclases/radiation effects , Animals , Carbachol/pharmacology , Cell Membrane/enzymology , Cell Membrane/radiation effects , Digestive System Physiological Phenomena , Enzyme Activation/radiation effects , Gastrin-Releasing Peptide/blood , Gastrin-Releasing Peptide/metabolism , Iodine Radioisotopes , Macaca mulatta , Membrane Potentials/radiation effects , Muscarinic Agonists/pharmacology , Time Factors , Vasoactive Intestinal Peptide/metabolism , Vasoactive Intestinal Peptide/physiology , Whole-Body Irradiation , X-Rays
13.
Int J Radiat Oncol Biol Phys ; 43(4): 867-75, 1999 Mar 01.
Article in English | MEDLINE | ID: mdl-10098443

ABSTRACT

PURPOSE: The therapeutic potential of thrombopoietin (TPO), the major regulator of platelet production, was evaluated for hematopoietic recovery and survival in mice following lethal and supralethal total body irradiation (TBI). METHODS AND MATERIALS: Hematopoietic recovery was studied in C57BL6/J mice after 8 Gy TBI (gamma-rays). Survival experiments were performed with C57BL6/J and BCBA F1 mice. Two protocols of TPO administration were evaluated: treatment for 7 consecutive days (7 x 0.3 microg/mice) beginning 2 h after exposure, or a single dose (0.3 microg/mice) administered 2 h after irradiation. RESULTS: TPO improved the platelet nadir and accelerated the platelet reconstitution of irradiated mice in comparison to placebo-treated mice. Recovery of neutrophils and erythrocytes was stimulated as well. TPO induced an accelerated recovery of hematopoietic progenitors and immature multilineage progenitors in bone marrow and spleen. In addition, TPO administration induced approximately 90% survival of 8 Gy irradiated C57BL6/J mice, a TBI dose which resulted in 100% mortality within 30 days for placebo-treated mice. Single TPO administration was as effective as repeated injections for hematopoietic recovery and prevention of mortality. Dose-effect survival experiments were performed in BCBA F1 mice and demonstrated that TPO shifted the LD50/30 from approximately 9.5 Gy to 10.5 Gy TBI given as a single dose, and from 14 Gy to as high as 17 Gy when TBI was given in three equal doses, each separated by 24 h. CONCLUSION: These results demonstrate that the multilineage hematopoietic effects of TPO may be advantageously used to protect against lethal bone marrow failure following high dose TBI.


Subject(s)
Hematopoiesis/drug effects , Hematopoiesis/radiation effects , Thrombopoietin/therapeutic use , Whole-Body Irradiation , Animals , Blood Cell Count/drug effects , Blood Cell Count/radiation effects , Dose-Response Relationship, Drug , Drug Administration Schedule , Female , Hematopoietic Stem Cells/drug effects , Hematopoietic Stem Cells/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Whole-Body Irradiation/mortality
14.
Blood ; 92(11): 4013-22, 1998 Dec 01.
Article in English | MEDLINE | ID: mdl-9834203

ABSTRACT

Purified CD34(+) and CD34(+)CD38(-) human umbilical cord blood (UCB) cells were transduced with the recombinant variant of Moloney murine leukemia virus (MoMLV) MFG-EGFP or with SF-EGFP, in which EGFP expression is driven by a hybrid promoter of the spleen focus-forming virus (SFFV) and the murine embryonic stem cell virus (MESV). Infectious MFG-EGFP virus was produced by an amphotropic virus producer cell line (GP+envAm12). SF-EGFP was produced in the PG13 cell line pseudotyped for the gibbon ape leukemia virus (GaLV) envelope proteins. Using a 2-day growth factor prestimulation, followed by a 2-day, fibronectin fragment CH-296-supported transduction, CD34(+) and CD34(+)CD38(-) UCB subsets were efficiently transduced using either vector. The use of the SF-EGFP/PG13 retroviral packaging cell combination consistently resulted in twofold higher levels of EGFP-expressing cells than the MFG-EGFP/Am12 combination. Transplantation of 10(5) input equivalent transduced CD34(+) or 5 x 10(3) input equivalent CD34(+)CD38(-) UCB cells in nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice resulted in median engraftment percentages of 8% and 5%, respectively, which showed that the in vivo repopulating ability of the cells had been retained. In addition, mice engrafted after transplantation of transduced CD34(+) cells using the MFG-EGFP/Am12 or the SF-EGFP/PG13 combination expressed EGFP with median values of 2% and 23% of human CD45(+) cells, respectively, which showed that the NOD/SCID repopulating cells were successfully transduced. EGFP+ cells were found in all human hematopoietic lineages produced in NOD/SCID mice including human progenitors with in vitro clonogenic ability. EGFP-expressing cells were also detected in the human cobblestone area-forming cell (CAFC) assay at 2 to 6 weeks of culture on the murine stromal cell line FBMD-1. During the transduction procedure the absolute numbers of CAFC week 6 increased 5- to 10-fold. The transduction efficiency of this progenitor cell subset was similar to the fraction of EGFP+ human cells in the bone marrow of the NOD/SCID mice transplanted with MFG-EGFP/Am12 or SF-EGFP/PG13 transduced CD34(+) cells, ie, 6% and 27%, respectively. The study thus shows that purified CD34(+) and highly purified CD34(+)CD38(-) UCB cells can be transduced efficiently with preservation of repopulating ability. The SF-EGFP/PG13 vector/packaging cell combination was much more effective in transducing repopulating cells than the MFG-EGFP/Am12 combination.


Subject(s)
Gene Transfer Techniques , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/physiology , Luminescent Proteins/genetics , Animals , Cell Differentiation , Cells, Cultured , Colony-Forming Units Assay/methods , Fetal Blood/cytology , Graft Survival , Green Fluorescent Proteins , Hematopoietic Stem Cells/cytology , Humans , Mice , Mice, SCID
15.
Stem Cells ; 16(6): 375-86, 1998.
Article in English | MEDLINE | ID: mdl-9831863

ABSTRACT

Radiation-induced pancytopenia proved to be a suitable model system in mice and rhesus monkeys for studying thrombopoietin (TPO) target cell range and efficacy. TPO was highly effective in rhesus monkeys exposed to the mid-lethal dose of 5 Gy (300 kV x-rays) TBI, a model in which it alleviated thrombocytopenia, promoted red cell reconstitution, accelerated reconstitution of immature CD34+ bone marrow cells, and potentiated the response to growth factors such as GM-CSF and G-CSF. In contrast to the results in the 5 Gy TBI model, TPO was ineffective following transplantation of limited numbers of autologous bone marrow or highly purified stem cells in monkeys conditioned with 8 Gy TBI. In the 5 Gy model, a single dose of TPO augmented by GM-CSF 24 h after TBI was effective in preventing thrombocytopenia. The strong erythropoietic stimulation may result in iron depletion, and TPO treatment should be accompanied by monitoring of iron status. This preclinical evaluation thus identified TPO as a potential major therapeutic agent for counteracting radiation-induced pancytopenia and demonstrated pronounced stimulatory effects on the reconstitution of immature CD34+ hemopoietic cells with multilineage potential. The latter observation explains the potentiation of the hematopoietic responses to G-CSF and GM-CSF when administered concomitantly. It also predicts the effective use of TPO to accelerate reconstitution of immature hematopoietic cells as well as possible synergistic effects in vivo with various other growth factors acting on immature stem cells and their direct lineage-committed progeny. The finding that a single dose of TPO might be sufficient for a clinically significant response emphasizes its potency and is of practical relevance. The heterogeneity of the TPO response encountered in the various models used for evaluation points to multiple mechanisms operating on the TPO response and heterogeneity of its target cells. Mechanistic mouse studies made apparent that the response of multilineage cells shortly after TBI to a single administration of TPO is quantitatively more important for optimal efficacy than the lineage-restricted response obtained at later intervals after TBI and emphasized the importance of a relatively high dose of TPO to overcome initial c-mpl-mediated clearance. Further elucidation of mechanisms determining efficacy might very well result in a further improvement, e.g., following transplantation of limited numbers of stem cells. Adverse effects of TPO administration to myelosuppressed or stem cell transplanted experimental animals were not observed.


Subject(s)
Hematopoietic Stem Cell Transplantation , Thrombocytopenia/prevention & control , Thrombopoietin/pharmacology , Animals , Disease Models, Animal , Humans , Immunity/radiation effects , Mice , Primates , Recombinant Proteins/pharmacology , Thrombocytopenia/drug therapy , Thrombocytopenia/etiology
16.
Vox Sang ; 74 Suppl 2: 463-6, 1998.
Article in English | MEDLINE | ID: mdl-9704482

ABSTRACT

Expansion of hemopoietic stem cells occurs in vivo following transplantation of limited numbers of bone marrow cells or of highly purified stem cells. Stem cell expansion can in principle be achieved in vitro and also be promoted in vivo by growth factor treatment, notably with thrombopoietin. Advances in identification of stromal elements, growth factors and culture conditions that stimulate immature hemopoietic stem cell proliferation may result in effective stem cell expansion protocols and contribute to efficient retrovirally mediated gene transfer. In vivo expansion of immature cells by growth factor treatment may both be a valid alternative and an adjuvant to in vitro expansion.


Subject(s)
Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cell Transplantation/methods , Hematopoietic Stem Cells/cytology , Animals , Bone Marrow Cells/physiology , Cell Division/drug effects , Cells, Cultured/transplantation , Coculture Techniques , Hematopoiesis/drug effects , Hematopoietic Cell Growth Factors/pharmacology , Hematopoietic Stem Cells/drug effects , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Radiation Chimera , Stromal Cells/physiology , Thrombopoietin/pharmacology
17.
Int J Radiat Biol ; 74(2): 265-72, 1998 Aug.
Article in English | MEDLINE | ID: mdl-9712556

ABSTRACT

PURPOSE: To obtain more accurate information on the dose distribution in rhesus monkeys for total body irradiation with orthovoltage X-rays. MATERIALS AND METHODS: Dose measurements were performed with an ionization chamber inside homogeneous cylindrical and rectangular phantoms of various dimensions and in phantoms containing lung-equivalent material. The irradiations were carried out with reference to a monitor ionization chamber placed alongside the phantom or the irradiation cage. RESULTS: Correction factors for mass and lung dose relative to the average dose in a homogeneous reference phantom, showed linear relationships with the effective diameter of the monkey. The lung dose correction factor relative to the homogeneous phantom was about 1.12 for a 3.5 kg monkey. The stated values for the average absorbed dose in the animal of standard weight should be multiplied by a factor of 0.93 for experiments performed before 1983. All publications on total body irradiations of monkeys at TNO after 1983 contain the corrected dose values. CONCLUSION: Dose distributions are reported for phantoms of different diameters and of cylindrical or rectangular shape. The new dosimetry has also resulted in a revised statement of the LD50 for the occurrence of bone marrow syndrome after X-irradiation; 4.9 Gy instead of 5.3 Gy.


Subject(s)
Macaca mulatta , Radiometry/methods , X-Rays , Animals , Body Constitution , Body Weight , Bone Marrow/radiation effects , Gamma Rays , Lung/radiation effects , Polymethyl Methacrylate/radiation effects , Whole-Body Irradiation
18.
Blood ; 92(5): 1586-97, 1998 Sep 01.
Article in English | MEDLINE | ID: mdl-9716586

ABSTRACT

Thrombopoietin (TPO) has been used in preclinical myelosuppression models to evaluate the effect on hematopoietic reconstitution. Here we report the importance of dose and dose scheduling for multilineage reconstitution after myelosuppressive total body irradiation (TBI) in mice. After 6 Gy TBI, a dose of 0.3 microgram TPO/mouse (12 microgram/kg) intraperitoneally (IP), 0 to 4 hours after TBI, prevented the severe thrombopenia observed in control mice, and in addition stimulated red and white blood cell regeneration. Time course studies showed a gradual decline in efficacy after an optimum within the first hours after TBI, accompanied by a replacement of the multilineage effects by lineage dominant thrombopoietic stimulation. Pharmacokinetic data showed that IP injection resulted in maximum plasma levels 2 hours after administration. On the basis of the data, we inferred that a substantial level of TPO was required at a critical time interval after TBI to induce multilineage stimulation of residual bone marrow cells. A more precise estimate of the effect of dose and dose timing was provided by intravenous administration of TPO, which showed an optimum immediately after TBI and a sharp decline in efficacy between a dose of 0.1 microgram/mouse (4 microgram/kg; plasma level 60 ng/mL), which was fully effective, and a dose of 0.03 microgram/mouse (1.2 microgram/kg; plasma level 20 ng/mL), which was largely ineffective. This is consistent with a threshold level of TPO required to overcome initial c-mpl-mediated clearance and to reach sufficient plasma levels for a maximum hematopoietic response. In mice exposed to fractionated TBI (3 x 3 Gy, 24 hours apart), IP administration of 0. 3 microgram TPO 2 hours after each fraction completely prevented the severe thrombopenia and anemia that occurred in control mice. Using short-term transplantation assays, ie, colony-forming unit-spleen (CFU-S) day 13 (CFU-S-13) and the more immature cells with marrow repopulating ability (MRA), it could be shown that TPO promoted CFU-S-13 and transiently depleted MRA. The initial depletion of MRA in response to TPO was replenished during long-term reconstitution followed for a period of 3 months. Apart from demonstrating again that MRA cells and CFU-S-13 are separate functional entities, the data thus showed that TPO promotes short-term multilineage repopulating cells at the expense of more immature ancestral cells, thereby preventing pancytopenia. The short time interval available after TBI to exert these effects shows that TPO is able to intervene in mechanisms that result in functional depletion of its multilineage target cells shortly after TBI and emphasizes the requirement of dose scheduling of TPO in keeping with these mechanisms to obtain optimal clinical efficacy.


Subject(s)
Bone Marrow Cells/cytology , Hematopoietic Stem Cells/cytology , Pancytopenia/prevention & control , Spleen/cytology , Thrombopoietin/therapeutic use , Whole-Body Irradiation/adverse effects , Animals , Cell Count , Colony-Forming Units Assay , Female , Kinetics , Mice , Mice, Inbred C57BL , Mice, Inbred CBA , Pancytopenia/etiology , Recombinant Proteins/therapeutic use , Thrombopoietin/administration & dosage , Thrombopoietin/pharmacokinetics
19.
Blood ; 91(6): 1966-76, 1998 Mar 15.
Article in English | MEDLINE | ID: mdl-9490679

ABSTRACT

In vivo expansion and multilineage outgrowth of human immature hematopoietic cell subsets from umbilical cord blood (UCB) were studied by transplantation into hereditary immunodeficient (SCID) mice. The mice were preconditioned with Cl2MDP-liposomes to deplete macrophages and 3.5 Gy total body irradiation (TBI). As measured by immunophenotyping, this procedure resulted in high levels of human CD45(+) cells in SCID mouse bone marrow (BM) 5 weeks after transplantation, similar to the levels of human cells observed in NOD/SCID mice preconditioned with TBI. Grafts containing approximately 10(7) unfractionated cells, approximately 10(5) purified CD34+ cells, or 5 x 10(3) purified CD34+CD38- cells yielded equivalent numbers of human CD45+ cells in the SCID mouse BM, which contained human CD34+ cells, monocytes, granulocytes, erythroid cells, and B lymphocytes at different stages of maturation. Low numbers of human GpA+ erythroid cells and CD41+ platelets were observed in the peripheral blood of engrafted mice. CD34+CD38+ cells (5 x 10(4)/mouse) failed to engraft, whereas CD34- cells (10(7)/mouse) displayed only low levels of chimerism, mainly due to mature T lymphocytes. Transplantation of graded numbers of UCB cells resulted in a proportional increase of the percentages of CD45+ and CD34+ cells produced in SCID mouse BM. In contrast, the number of immature, CD34+CD38- cells produced in vivo showed a second-order relation to CD34+ graft size, and mice engrafted with purified CD34+CD38- grafts produced 10-fold fewer CD34+ cells without detectable CD34+CD38- cells than mice transplanted with equivalent numbers of unfractionated or purified CD34+ cells. These results indicate that SCID repopulating CD34+CD38- cells require CD34+CD38+ accessory cell support for survival and expansion of immature cells, but not for production of mature multilineage progeny in SCID mouse BM. These accessory cells are present in the purified, nonrepopulating CD34+CD38+ subset as was directly proven by the ability of this fraction to restore the maintenance and expansion of immature CD34+CD38- cells in vivo when cotransplanted with purified CD34+CD38- grafts. The possibility to distinguish between maintenance and outgrowth of immature repopulating cells in SCID mice will facilitate further studies on the regulatory functions of accessory cells, growth factors, and other stimuli. Such information will be essential to design efficient stem cell expansion procedures for clinical use.


Subject(s)
Antigens, CD , Fetal Blood/cytology , Hematopoietic Stem Cell Transplantation , Hematopoietic Stem Cells/cytology , Macrophages , ADP-ribosyl Cyclase , ADP-ribosyl Cyclase 1 , Animals , Antigens, CD34/analysis , Antigens, Differentiation/analysis , Cell Lineage , Clodronic Acid/pharmacology , Female , Graft Survival , Hematopoiesis , Hematopoietic Stem Cells/classification , Humans , Macrophages/drug effects , Membrane Glycoproteins , Mice , Mice, Inbred NOD , Mice, SCID , NAD+ Nucleosidase/analysis , Radiation Chimera , Specific Pathogen-Free Organisms , Transplantation Conditioning , Transplantation, Heterologous
20.
Stem Cells ; 16 Suppl 1: 185-91, 1998.
Article in English | MEDLINE | ID: mdl-11012161

ABSTRACT

Under conditions of steady-state hemopoiesis, a small fraction of immature hemopoietic cells, including stem cells, circulates in peripheral blood (PB). In rhesus monkeys, a median number of 1.2 x 10(7)/l CD34+ cells was observed as opposed to a median number of 1.5 x 10(9)/l in aspirated bone marrow (BM). The concentration of circulating CD34+ cells is therefore approximately two logs less than that in BM. Since a 4-kg rhesus monkey has an estimated number of 3 x 10(10) BM cells and approximately 300 ml of blood, the fraction of CD34+ cells that circulates can be estimated at approximately 0.4% of the total pool of CD34+ cells. During hemopoietic reconstitution following a cytotoxic insult such as results from a midlethal dose of TBI, PB CD34+ cell numbers appeared to be correlated to those of BM, suggesting that PB CD34+ cells may reflect reconstitution of BM CD34+ cells. Reconstitution of BM immature cells can be accelerated by treatment with pharmacological doses of growth factors, resulting in largely expanded immature cell populations within a few weeks after TBI. Growth factors observed to exert such an effect included, notably, thrombopoietin. Such an acceleration can be monitored by daily assessment of circulating CD34+ cells. Expansion of immature circulating cells indicates expansion of similar cells in the bone marrow rather than growth factor-induced selective mobilization of immature cells.


Subject(s)
Bone Marrow Cells/cytology , Hematopoiesis/physiology , Hematopoietic Stem Cell Mobilization/methods , Hematopoietic Stem Cells/cytology , Animals , Antigens, CD34/analysis , Bone Marrow Cells/radiation effects , Growth Substances/physiology , Hematopoiesis/radiation effects , Hematopoietic Stem Cells/radiation effects , Macaca mulatta , Male , Whole-Body Irradiation
SELECTION OF CITATIONS
SEARCH DETAIL
...