Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
Add more filters










Publication year range
3.
J Thromb Haemost ; 14(12): 2478-2492, 2016 12.
Article in English | MEDLINE | ID: mdl-27685947

ABSTRACT

Essentials B cells are attractive targets for gene therapy and particularly interesting for immunotherapy. A baboon envelope pseudotyped lentiviral vector (BaEV-LV) was tested for B-cell transduction. BaEV-LVs transduced mature and plasma human B cells with very high efficacy. BaEV-LVs allowed secretion of functional factor IX from B cells at therapeutic levels in vivo. SUMMARY: Background B cells are attractive targets for gene therapy for diseases associated with B-cell dysfunction and particularly interesting for immunotherapy. Moreover, B cells are potent protein-secreting cells and can be tolerogenic antigen-presenting cells. Objective Evaluation of human B cells for secretion of clotting factors such as factor IX (FIX) as a possible treatment for hemophilia. Methods We tested here for the first time our newly developed baboon envelope (BaEV) pseudotyped lentiviral vectors (LVs) for human (h) B-cell transduction following their adaptive transfer into an NOD/SCIDγc-/- (NSG) mouse. Results Upon B-cell receptor stimulation, BaEV-LVs transduced up to 80% of hB cells, whereas vesicular stomatitis virus G protein VSV-G-LV only reached 5%. Remarkably, BaEVTR-LVs permitted efficient transduction of 20% of resting naive and 40% of resting memory B cells. Importantly, BaEV-LVs reached up to 100% transduction of human plasmocytes ex vivo. Adoptive transfer of BaEV-LV-transduced mature B cells into NOD/SCID/γc-/- (NSG) [non-obese diabetic (NOD), severe combined immuno-deficiency (SCID)] mice allowed differentiation into plasmablasts and plasma B cells, confirming a sustained high-level gene marking in vivo. As proof of principle, we assessed BaEV-LV for transfer of human factor IX (hFIX) into B cells. BaEV-LVs encoding FIX efficiently transduced hB cells and their transfer into NSG mice demonstrated for the first time secretion of functional hFIX from hB cells at therapeutic levels in vivo. Conclusions The BaEV-LVs might represent a valuable tool for therapeutic protein secretion from autologous B cells in vivo in the treatment of hemophilia and other acquired or inherited diseases.


Subject(s)
B-Lymphocytes/cytology , Factor IX/metabolism , Genetic Vectors , Hemophilia A/blood , Lentivirus/genetics , Animals , Antigen-Presenting Cells/cytology , CD4-Positive T-Lymphocytes/cytology , Cell Proliferation , Gene Transfer Techniques , HEK293 Cells , Hemophilia A/therapy , Humans , Immunotherapy/methods , Membrane Glycoproteins/chemistry , Mice , Mice, Inbred NOD , Mice, SCID , Papio , Plasmids , Transduction, Genetic , Transgenes , Treatment Outcome , Viral Envelope Proteins/chemistry
4.
Leukemia ; 29(10): 1993-2002, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25925206

ABSTRACT

The BMI1 polycomb protein regulates self-renewal, proliferation and survival of cancer-initiating cells essentially through epigenetic repression of the CDKN2A tumor suppressor locus. We demonstrate here for the first time that BMI1 also prevents autophagy in chronic myeloid leukemia (CML) cell lines, to support their proliferation and clonogenic activity. Using chromatin immunoprecipitation, we identified CCNG2/cyclin G2 (CCNG2) as a direct BMI1 target. BMI1 downregulation in CD34+ CML cells by PTC-209 pharmacological treatment or shBMI1 transduction triggered CCNG2 expression and decreased clonogenic activity. Also, ectopic expression of CCNG2 in CD34+ CML cells strongly decreased their clonogenicity. CCNG2 was shown to act by disrupting the phosphatase 2A complex, which activates a PKCζ-AMPK-JNK-ERK pathway that engages autophagy. We observed that BMI1 and CCNG2 levels evolved inversely during the progression of CML towards an acute deadly phase, and therefore hypothesized that BMI1 could support acute transformation of CML through the silencing of a CCNG2-mediated tumor-suppressive autophagy response.


Subject(s)
Autophagy , Cell Proliferation , Cyclin G2/metabolism , Gene Expression Regulation, Leukemic , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology , Polycomb Repressive Complex 1/metabolism , Apoptosis , Blotting, Western , Chromatin Immunoprecipitation , Cyclin G2/genetics , Humans , Leukemia, Myelogenous, Chronic, BCR-ABL Positive/genetics , Polycomb Repressive Complex 1/antagonists & inhibitors , Polycomb Repressive Complex 1/genetics , RNA, Small Interfering/genetics , Signal Transduction , Tumor Cells, Cultured
5.
Leukemia ; 29(5): 1163-76, 2015 May.
Article in English | MEDLINE | ID: mdl-25394713

ABSTRACT

Deregulated expression of glycolytic enzymes contributes not only to the increased energy demands of transformed cells but also has non-glycolytic roles in tumors. However, the contribution of these non-glycolytic functions in tumor progression remains poorly defined. Here, we show that elevated expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), but not of other glycolytic enzymes tested, increased aggressiveness and vascularization of non-Hodgkin's lymphoma. Elevated GAPDH expression was found to promote nuclear factor-κB (NF-κB) activation via binding to tumor necrosis factor receptor-associated factor-2 (TRAF2), enhancing the transcription and the activity of hypoxia-inducing factor-1α (HIF-1α). Consistent with this, inactive mutants of GAPDH failed to bind TRAF2, enhance HIF-1 activity or promote lymphomagenesis. Furthermore, elevated expression of gapdh mRNA in biopsies from diffuse large B-cell non-Hodgkin's lymphoma patients correlated with high levels of hif-1α, vegf-a, nfkbia mRNA and CD31 staining. Collectively, these data indicate that deregulated GAPDH expression promotes NF-κB-dependent induction of HIF-1α and has a key role in lymphoma vascularization and aggressiveness.


Subject(s)
Gene Expression Regulation, Enzymologic , Gene Expression Regulation, Neoplastic , Glyceraldehyde 3-Phosphate Dehydrogenase (NADP+)/metabolism , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Lymphoma, Non-Hodgkin/metabolism , NF-kappa B p50 Subunit/metabolism , Animals , Biopsy , Cell Line, Tumor , Enzyme Inhibitors/chemistry , HeLa Cells , Humans , Lymphoma/metabolism , Mice , Mice, Transgenic , Phenotype , Proto-Oncogene Proteins c-myc/metabolism , TNF Receptor-Associated Factor 2/metabolism , Vascular Endothelial Growth Factor A/metabolism
6.
Cell Death Dis ; 5: e1451, 2014 Oct 09.
Article in English | MEDLINE | ID: mdl-25299776

ABSTRACT

Chronic lymphoid leukemia (CLL) is characterized by the accumulation of functionally defective CD5-positive B lymphocytes. The clinical course of CLL is highly variable, ranging from a long-lasting indolent disease to an unpredictable and rapidly progressing leukemia requiring treatment. It is thus important to identify novel factors that reflect disease progression or contribute to its assessment. Here, we report on a novel STAT3-mediated pathway that characterizes CLL B cells-extended viability and oxidative stress control. We observed that leukemic but not normal B cells from CLL patients exhibit constitutive activation of an atypical form of the STAT3 signaling factor, phosphorylated on serine 727 (Ser(727)) in the absence of detectable canonical tyrosine 705 (Tyr705)-dependent activation in vivo. The Ser(727)-phosphorylated STAT3 molecule (pSTAT3Ser(727)) is localized to the mitochondria and associates with complex I of the respiratory chain. This pSer(727) modification is further controlled by glutathione-dependent antioxidant pathway(s) that mediate stromal protection of the leukemic B cells and regulate their viability. Importantly, pSTAT3Ser(727), but neither Tyr705-phosphorylated STAT3 nor total STAT3, levels correlate with prolonged in vivo CLL B cells survival. Furthermore, STAT3 activity contributes to the resistance to apoptosis of CLL, but not normal B cells, in vitro. These data reveal that mitochondrial (Mt) pSTAT3Ser(727) overactivity is part of the antioxidant defense pathway of CLL B cells that regulates their viability. Mt pSTAT3Ser(727) appears to be a newly identified cell-protective signal involved in CLL cells survival. Targeting pSTAT3Ser(727) could be a promising new therapeutic approach.


Subject(s)
Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Mitochondria/metabolism , STAT3 Transcription Factor/chemistry , STAT3 Transcription Factor/metabolism , Serine/metabolism , Apoptosis , B-Lymphocytes/cytology , B-Lymphocytes/metabolism , Cell Survival , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/physiopathology , Mitochondria/chemistry , Mitochondria/genetics , Phosphorylation , STAT3 Transcription Factor/genetics , Serine/genetics , Signal Transduction
7.
Leukemia ; 26(7): 1663-70, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22318450

ABSTRACT

A lot of genes deregulated in malignant plasma cells (PCs) involved in multiple myeloma have been reported these last years. The expression of some of these genes is associated with poor survival. A critical step is to elucidate the biological mechanisms triggered by these gene products. Such studies are hampered by the difficulty to obtain malignant PCs and to genetically modify them. Usual lentiviral vectors (LVs) pseudotyped with vesicular stomatitis virus envelope glycoprotein poorly transduced healthy and malignant PCs. Here, we report that LVs pseudotyped with the hemagglutinin and fusion glycoproteins from the measles Edmonston strain (H/F-LVs) can efficiently and stably transduce healthy and primary malignant PCs, without modifying their main phenotypic characteristics. Both LV pseudotypes efficiently transduced human myeloma cell lines. Importantly, both healthy and malignant PCs expressed CD46 and SLAMF1/CD150 membrane proteins, which are critical receptors for binding and productive genetic modification by H/F-LVs. The ability to efficiently introduce and express a given gene into PCs opens the possibility to study in detail PC biology.


Subject(s)
Genetic Vectors , Glycoproteins/genetics , Lentivirus/genetics , Measles virus/genetics , Multiple Myeloma/genetics , Multiple Myeloma/therapy , Plasma Cells/metabolism , Antigens, CD , Cells, Cultured , Flow Cytometry , Fluorescent Antibody Technique , Gene Transfer Techniques , Genetic Therapy , Humans , Immunophenotyping , Membrane Cofactor Protein , Plasma Cells/cytology , Plasma Cells/virology , Receptors, Cell Surface , Signaling Lymphocytic Activation Molecule Family Member 1 , Transduction, Genetic , Viral Envelope Proteins/genetics
8.
J Virol ; 86(9): 5192-203, 2012 May.
Article in English | MEDLINE | ID: mdl-22345444

ABSTRACT

Dendritic cells (DCs) are potent antigen-presenting cells capable of promoting or regulating innate and adaptive immune responses against non-self antigens. To better understand the DC biology or to use them for immune intervention, a tremendous effort has been made to improve gene transfer in these cells. Lentiviral vectors (LVs) have conferred a huge advantage in that they can transduce nondividing cells such as human monocyte-derived DCs (MDDCs) but required high amounts of viral particles and/or accessory proteins such as Vpx or Vpr to achieve sufficient transduction rates. As a consequence, these LVs have been shown to cause dramatic functional modifications, such as the activation or maturation of transduced MDDCs. Taking advantage of new pseudotyped LVs, i.e., with envelope glycoproteins from the measles virus (MV), we demonstrate that MDDCs are transduced very efficiently with these new LVs compared to the classically used vesicular stomatitis virus G-pseudotyped LVs and thus allowed to achieve high transduction rates at relatively low multiplicities of infection. Moreover, in this experimental setting, no activation or maturation markers were upregulated, while MV-LV-transduced cells remained able to mature after an appropriate Toll-like receptor stimulation. We then demonstrate that our MV-pseudotyped LVs use DC-SIGN, CD46, and CD150/SLAM as receptors to transduce MDDCs. Altogether, our results show that MV-pseudotyped LVs provide the most accurate and simple viral method for efficiently transferring genes into MDDCs without affecting their activation and/or maturation status.


Subject(s)
Dendritic Cells/metabolism , Genetic Vectors/genetics , Lentivirus/genetics , Measles virus/genetics , Transduction, Genetic , Vesicular stomatitis Indiana virus/genetics , Viral Envelope Proteins/genetics , Cell Adhesion Molecules/genetics , Cell Differentiation , Dendritic Cells/cytology , Dendritic Cells/virology , Gene Expression , Gene Transfer Techniques , Humans , Lectins, C-Type/genetics , Membrane Cofactor Protein/genetics , Receptors, Cell Surface/genetics , Resting Phase, Cell Cycle , Toll-Like Receptor 3/agonists
10.
Leukemia ; 23(11): 2063-74, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19641520

ABSTRACT

Toll-like receptors (TLRs) constitute a family of nonpolymorphic receptors that are devoted to pathogen recognition. In this work, we have explored the impact of TLR ligands (TLR-L) on human hematopoietic stem cells (HSCs) and hematopoietic progenitor cells (HPCs). We show that HSCs and HPCs have a comparable pattern of expression of TLR transcripts characterized by the predominance of TLR1, -2, -3, -4 and -6. In long-term cultures of HSCs, HPCs and stromal cells, most TLR-L profoundly inhibited B-cell development while preserving or enhancing the production of myeloid cells. In short-term cultures, the TLR1/2 ligand PAM(3)CSK(4) induced a large proportion of HPCs to express markers of the myelomonocytic lineage. PAM(3)CSK(4) induced only marginal expression of myeloid lineage markers on HSCs but promoted their myeloid commitment as revealed by their acquisition of the phenotype of multi- and bipotential myeloid progenitors and by upregulation of the transcription factors PU.1, C/EBPalpha and GATA-1. Our results suggest that TLR agonists can bias the lineage commitment of human HSCs and shift the differentiation of lineage-committed progenitors to favor myelopoiesis at the expense of lymphoid B-cell development.


Subject(s)
Hematopoietic Stem Cells/cytology , Hematopoietic Stem Cells/drug effects , Lipopeptides/pharmacology , Myeloid Cells/cytology , Toll-Like Receptor 1/agonists , Toll-Like Receptor 2/agonists , Animals , Antigens, CD34/metabolism , B-Lymphocytes/cytology , B-Lymphocytes/drug effects , Cell Differentiation/drug effects , Cell Line , Cell Lineage/drug effects , DNA-Binding Proteins/genetics , Fetal Blood/cytology , Humans , Lymphopoiesis/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Stromal Cells/cytology , Toll-Like Receptor 1/genetics , Toll-Like Receptor 2/genetics , Transcription, Genetic/drug effects
11.
Methods Mol Biol ; 434: 99-112, 2008.
Article in English | MEDLINE | ID: mdl-18470641

ABSTRACT

A major limitation of current lentiviral vectors (LVs) is their inability to govern efficient gene transfer into quiescent cells, such as human CD34+ cells that reside in the G0 phase of the cell cycle and that are highly enriched in hematopoietic stem cells. This hampers their application for gene therapy of hematopoietic cells. We describe here novel LVs that overcome this restriction by displaying early-acting cytokines on their surface. Display of thrombopoietin, stem cell factor or both cytokines on LV surface allows high transfer into quiescent cord blood CD34+ cells. Moreover, these surface-engineered LVs preferentially transduce and promote survival of resting CD34+ cells rather than cycling cells. These novel LVs allow superior gene transfer in the most immature CD34+ cells compared to conventional LVs, even in the presence of recombinant cytokines. This is demonstrated by their capacity to promote selective transduction in long-term culture initiating cell colonies (LTC-ICs) and of long-term non-obese diabetic/severe combined immunodeficient (NOD/SCID) repopulating cells (SRCs). Here we describe the production of these "early acting cytokine" displaying vectors and the methodology to confirm the capacity of these vectors to promote selective transduction of HSCs.


Subject(s)
Gene Transfer Techniques , Genetic Vectors , Hematopoietic Stem Cells/physiology , Lentivirus/physiology , Stem Cell Factor/metabolism , Thrombopoietin/metabolism , Animals , Antigens, CD34/metabolism , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Transduction, Genetic
12.
Gene Ther ; 15(12): 930-41, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18323794

ABSTRACT

Wiskott-Aldrich syndrome (WAS) gene therapy requires highly efficient and well-controlled vectors. Here we studied the performance of a lentiviral vector (LV) harbouring a 500-bp fragment of the WAS proximal promoter (WW), which we previously characterized as haematopoietic-specific and capable of restoring WAS phenotype in patients' T cells. We used an LV (WE) expressing eGFP to evaluate whether this promoter was following the expression pattern of endogenous WASp. Transgene expression was analysed in WE-transduced hCD34+ population and its progeny after in vitro and in vivo differentiation in the Rag2-/-, gammac-/- humanized mouse. We revealed very poor expression from the WE internal promoter in macrophages and erythroid cells. Therefore, we designed a novel LV including a fragment of the alternative WAS promoter in WE vector (AWE). This new vector sustained high transgene levels along the whole lymphoid lineage in vivo. Most importantly, the performance of AWE vector was highly superior to WE vector since AWE clearly improved transgene levels in in vitro and in vivo hCD34+-derived macrophages, erythroid cells, megakaryocytes and B cells while supporting a high expression in human T cells. This emphasizes that it is a suitable LV backbone for gene therapy of haematopoietic diseases such as WAS.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Lentivirus/genetics , Promoter Regions, Genetic , Wiskott-Aldrich Syndrome Protein/genetics , Wiskott-Aldrich Syndrome/therapy , Animals , Electrophoresis, Polyacrylamide Gel , Flow Cytometry , Gene Expression , Genetic Engineering , Genetic Vectors/genetics , Green Fluorescent Proteins/genetics , Hematopoiesis , Humans , Mice , Mice, Inbred BALB C , Mice, Knockout , Reverse Transcriptase Polymerase Chain Reaction/methods , Transduction, Genetic/methods , Transgenes , Wiskott-Aldrich Syndrome Protein/metabolism
13.
Mol Ther ; 14(2): 285-92, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16697259

ABSTRACT

We developed a new strategy that provides well-defined high-titer producer cells for recombinant retroviruses in a minimum amount of time. The strategy involves the targeted integration of the retroviral vector into a chromosomal locus with favorable properties. For proof of concept we established a novel HEK293-based retroviral producer cell line, called Flp293A, with a single-copy retroviral vector integrated at a selected chromosomal locus. The vector was flanked by noninteracting Flp-recombinase recognition sites and was exchanged for different retroviral vectors via Flp-mediated cassette exchange. All analyzed cell clones showed correct integration and identical titers for each of the vectors, confirming that the expression characteristics from the parental cell were preserved. Titers up to 2.5 x 10(7) infectious particles/10(6) cells were obtained. Also, high-titer producer cells for a therapeutic vector that encodes the 8.9-kb collagen VII cDNA in a marker-free cassette were obtained within 3 weeks without screening. Thus, we provide evidence that the precise integration of viral vectors into a favorable chromosomal locus leads to high and predictable virus production. This method is compatible with other retroviral vectors, including self-inactivating vectors and marker-free vectors. Further, it provides a tool for evaluation of different retroviral vector designs.


Subject(s)
Gene Targeting/methods , Genetic Vectors , Retroviridae/genetics , Virus Integration , Animals , Cell Line , Collagen Type VII , DNA Nucleotidyltransferases/metabolism , Humans , Mice , Retroviridae/physiology , Transfection
14.
Osteoarthritis Cartilage ; 10(2): 109-18, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11869070

ABSTRACT

OBJECTIVE: One of the main open questions in chondrocyte transplantation is the fate of the implanted cells in vivo. We intended to establish prerequisites for such studies in animal models and to show the feasibility of this approach in rabbits. Isolated articular chondrocytes were retrovirally marked using green fluorescence protein (GFP) as a cell-specific marker in order to allow an in vivo follow-up of these cells. METHODS: Chondrocytes from rabbits, sheep, cattle and humans were isolated and infected with murine leukemia virus-derived retroviruses carrying the GFP gene. The influence of the host range of three packaging cell lines (PA317, PT67, PG13), start cell concentrations, number of cell passages and number of infection cycles on the efficiency of infection was investigated. Stability of GFP expression was followed by FACS analysis, confocal imaging and fluorescence microscopy. For in vivo follow-up of GFP expression we used marked allogeneic chondrocyte populations grown on scaffold material and implanted them into full-thickness defects in knee joints of rabbits. RESULTS: Retroviruses from all three packaging cell lines were able to infect rabbit and human chondrocytes, whereas only retroviruses released from PG13 cells were able to infect sheep and bovine chondrocytes efficiently. Optimization of the infection with these viruses resulted in efficiencies of 60-90% GFP-expressing chondrocytes. Populations of 100% marked chondrocytes were obtained by cell sorting. GFP expression stability of such marked chondrocyte populations was followed in monolayer culture and in 3-D culture on different scaffold materials. The expression of GFP was stable on all tested materials for at least 4 weeks. In monolayer culture GFP expression was stable for more than 8 months. In vivo, we observed stable GFP expression in the transplants during a four-week time course. CONCLUSION: Retroviral GFP gene transfer led to long-term expression in chondrocytes from rabbits, sheep, cattle and humans. Transgene expression and the number of implanted chondrocytes remain stable for at least 4 weeks in vivo. This method permits a rapid monitoring of chondrocytes and provides a basis for following the fate of these cells in vivo.


Subject(s)
Cartilage, Articular/cytology , Chondrocytes/physiology , Leukemia Virus, Murine/genetics , Luminescent Proteins/genetics , Animals , Cattle , Cell Separation , Cells, Cultured , Chondrocytes/transplantation , Feasibility Studies , Flow Cytometry , Gene Expression , Genetic Markers , Green Fluorescent Proteins , Humans , Microscopy, Confocal , Microscopy, Fluorescence , Rabbits , Reverse Transcriptase Polymerase Chain Reaction , Sheep
15.
Hum Gene Ther ; 12(8): 933-44, 2001 May 20.
Article in English | MEDLINE | ID: mdl-11387058

ABSTRACT

Successful retroviral vector construction is still empirical. Test systems for vector efficiency are based on statistical comparison of numerous infectants with single proviral integrates, since their expression depends on the chromosomal surroundings. More reliable data would be obtained if different vector constructs were studied in an identical chromosomal context. Here, we demonstrate the use of a new method, in which chromosomal sites are provirally tagged in such a way that they can be targeted with other expression cassettes. The original tagging integrate is replaced in one step by the targeting element. This permits a reliable comparison of different retroviral vector configurations, eliminating the influence of neighboring chromosomal elements. We compared different retroviral vector types for coexpression of two genes: a vector containing an internal promoter and a vector with an internal ribosome entry site (IRES) element. In contrast to bicistronic retroviral vectors, dual-promoter proviruses exhibited rapid inactivation of the long terminal repeat (LTR)-driven gene expression. Targeted exchange of the dual-promoter provirus with a bicistronic retroviral cassette resulted in gain of expression stability. The reverse experiment confirmed this promoter interaction phenomenon since initial expression stability from a single-promoter bicistronic provirus was lost by targeted exchange with a dual-promoter cassette. In addition, targeting exchange of the dual-promoter provirus, replacing the LTR with an artificial (Tet) promoter restored expression stability. These observations, valid for various integration sites, prove the strong interaction between the LTR and the internal promoter. Our results have implications for retroviral vector design and suggest that retroviral coexpression of two genes is more predictable in the bicistronic configuration.


Subject(s)
Chromosomes , DNA Nucleotidyltransferases/genetics , Gene Transfer Techniques , Genetic Vectors , Retroviridae/genetics , 3T3 Cells , Animals , Genes , Lac Operon/genetics , Luciferases/metabolism , Mice , Models, Genetic , Plasmids/metabolism , Promoter Regions, Genetic , Recombination, Genetic
16.
Transplantation ; 72(12): 1898-906, 2001 Dec 27.
Article in English | MEDLINE | ID: mdl-11773886

ABSTRACT

The critical shortage of human donor organs has generated growing interest for porcine to human xenotransplantation. The major immunological barrier to xenotransplantation is the hyperacute rejection (HAR) response that is mediated by preformed xenoreactive antibodies and complement. A promising strategy to control the complement activation, is the expression of human complement regulatory proteins in transgenic animals. We have used the human early cytomegalovirus (CMV) promoter to drive expression of the human complement regulatory protein CD59 (hCD59) in transgenic pigs. A total of eight live transgenic founder animals was born from which five transgenic lines could be established. mRNA analysis and Western blotting revealed high expression of hCD59 in heart, kidney, skeletal muscle, and skin in animals of lines 1 and 5, as well as in the pancreas of four lines. This pattern of expression was confirmed by immunhistological staining. A cell-specific expression in heart and kidney tissue of transgenic lines 1 and 5 was determined. Primary fibroblasts and endothelial cell cultures derived from the aorta of transgenic pigs showed a significantly diminished sensitivity against the challenge with xenoreactive human antibodies and complement whereas non-transgenic control cells were highly susceptible to complement mediated lysis. Ex vivo perfusion of kidneys with pooled human blood revealed a significant protective effect of hCD59 against HAR. The average survival of transgenic kidneys was significantly extended (P<0.05) over nontransgenic controls (207.5+/-54.6 vs. 57.5+/-64.5 min). These data support the concept that hCD59 protects nonprimate cells against human complement mediated lysis and suggest that donor pigs transgenic for hCD59 could play a crucial role in clinical xenotransplantation. Two of five hCD59 transgenic lines showed strong hCD59 expression in several organs relevant for xenotransplantation and a protective effect against HAR. This indicates that the use of the CMV-promoter can facilitate the selection process for optimized transgene expression.


Subject(s)
CD59 Antigens/genetics , Cytomegalovirus/genetics , Gene Expression/physiology , Graft Rejection/prevention & control , Organ Transplantation , Promoter Regions, Genetic/physiology , Swine/genetics , 3T3 Cells , Acute Disease , Animals , Animals, Genetically Modified/genetics , Animals, Genetically Modified/metabolism , Blood Physiological Phenomena , Cell Death/physiology , Cell Membrane/metabolism , Complement System Proteins/physiology , Endothelium, Vascular/cytology , Endothelium, Vascular/physiology , Fibroblasts/physiology , Humans , Immunohistochemistry , Kidney , Mice , Perfusion
17.
Cells Tissues Organs ; 167(2-3): 75-80, 2000.
Article in English | MEDLINE | ID: mdl-10971031

ABSTRACT

A number of hurdles have to be overcome for efficient and specific gene therapy approaches. Here, we report on two different strategies that should lead to an improvement of current protocols. A strategy is presented to tag unique chromosomal integration sites by means of retroviral infection, which can be reused for exchange with the gene of interest by action of site-specific recombinases. Targeting exchange is achieved in one step with 100% efficiency by a stringent positive selection, which makes further screening superfluous. With this strategy a predictable gene expression is obtained for foreign genes integrated into a predefined chromatin structure. A second approach aims at the stabilization of mouse retroviruses towards human serum which is a prerequisite for in vivo gene therapy protocols. To stabilize murine leukemia virus-based retroviruses against human serum, complement regulatory proteins were fused to the retroviral ENV proteins. This resulted in infectious and human complement-protected particles.


Subject(s)
Gene Targeting , Genetic Therapy/trends , 3T3 Cells , Animals , CD55 Antigens/genetics , Chromosomes/genetics , Complement System Proteins/genetics , Gene Expression , Humans , Leukemia Virus, Murine/genetics , Mice , Receptors, Complement/genetics , Retroviridae/genetics , Transformation, Genetic , Viral Fusion Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...