Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters










Publication year range
1.
J Neurosci Methods ; 399: 109981, 2023 11 01.
Article in English | MEDLINE | ID: mdl-37783350

ABSTRACT

BACKGROUND: Adeno-associated viral vectors (AAVs) are a widely used gene transfer platform in neuroscience. Although naturally AAV serotypes can have preferences for certain tissues, selectivity for particular cell types in the CNS does not exist. Towards interneuron targeting, capsid engineering of AAV2 including display of the designed ankyrin repeat protein (DARPin) 2K19 specific for the glutamate receptor subunit 4 (GluA4) at the N-terminus of the VP2 capsid protein has been established. The resulting AAV-VP2N is highly specific for interneurons, but exhibits rather moderate transduction efficiencies. METHODS: Two alternative insertion sites for 2K19 in the GH2/GH3 loop of capsid proteins VP1 (AAV-VP1L) or VP2 (AAV-VP2L) were exploited to yield second generation GluA4-AAVs. Having packaged reporter genes under ubiquitous promoters, the vectors were characterized for biochemical properties as well as gene delivery into cell lines and rat hippocampal slice cultures. Electrophysiological recordings monitored the functional properties of transduced cells. RESULTS: Compared to AAV-VP2N, the second-generation vectors, especially AAV-VP1L, achieved about 2-fold higher genomic titers as well as a substantially improved GluA4 binding. Improvements in gene transfer activities were 18-fold on GluA4-overexpressing A549 cells and five-fold on rat hippocampal organotypic slice cultures reaching approximately 60 % of all parvalbumin positive interneurons upon a single administration. The spiking behaviour of transduced cells was unaltered and characteristic for a heterogeneous group of interneurons. CONCLUSION: The substantially improved gene transfer activity of the second generation GluA4-targeted AAV combined with low toxicity makes this vector an attractive tool for interneuron-directed gene transfer with unrestricted promotor and transgene choice.


Subject(s)
Dependovirus , Genetic Vectors , Rats , Animals , Dependovirus/genetics , Gene Transfer Techniques , Cell Line , Genetic Therapy/methods , Transduction, Genetic
2.
Oncoimmunology ; 11(1): 2127508, 2022.
Article in English | MEDLINE | ID: mdl-36249274

ABSTRACT

Glioblastoma (GB) is the most common primary brain tumor, which is characterized by low immunogenicity of tumor cells and prevalent immunosuppression in the tumor microenvironment (TME). Targeted local combination immunotherapy is a promising strategy to overcome these obstacles. Here, we evaluated tumor-cell specific delivery of an anti-PD-1 immunoadhesin (aPD-1) via a targeted adeno-associated viral vector (AAV) as well as HER2-specific NK-92/5.28.z (anti-HER2.CAR/NK-92) cells as components for a combination immunotherapy. In co-culture experiments, target-activated anti-HER2.CAR/NK-92 cells modified surrounding tumor cells and bystander immune cells by triggering the release of inflammatory cytokines and upregulation of PD-L1. Tumor cell-specific delivery of aPD-1 was achieved by displaying a HER2-specific designed ankyrin repeat protein (DARPin) on the AAV surface. HER2-AAV mediated gene transfer into GB cells correlated with HER2 expression levels, without inducing anti-viral responses in transduced cells. Furthermore, AAV-transduction did not interfere with anti-HER2.CAR/NK-92 cell-mediated tumor cell lysis. After selective transduction of HER2+ cells, aPD-1 expression was detected at the mRNA and protein level. The aPD-1 immunoadhesin was secreted in a time-dependent manner, bound its target on PD-1-expressing cells and was able to re-activate T cells by efficiently disrupting the PD-1/PD-L1 axis. Moreover, high intratumoral and low systemic aPD-1 concentrations were achieved following local injection of HER2-AAV into orthotopic tumor grafts in vivo. aPD-1 was selectively produced in tumor tissue and could be detected up to 10 days after a single HER2-AAV injection. In subcutaneous GL261-HER2 and Tu2449-HER2 immunocompetent mouse models, administration of the combination therapy significantly prolonged survival, including complete tumor control in several animals in the GL261-HER2 model. In summary, local therapy with aPD-1 encoding HER2-AAVs in combination with anti-HER2.CAR/NK-92 cells may be a promising novel strategy for GB immunotherapy with the potential to enhance efficacy and reduce systemic side effects of immune-checkpoint inhibitors.


Subject(s)
Glioblastoma , Adenoviridae/genetics , Animals , B7-H1 Antigen/genetics , Cell Line, Tumor , Cytokines , Glioblastoma/genetics , Glioblastoma/therapy , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Killer Cells, Natural/metabolism , Killer Cells, Natural/transplantation , Mice , RNA, Messenger , Receptor, ErbB-2/metabolism , Therapies, Investigational , Tumor Microenvironment
3.
Cell Death Dis ; 5: e1391, 2014 Aug 28.
Article in English | MEDLINE | ID: mdl-25165877

ABSTRACT

Accumulating evidence indicates that loss of physiologic amyloid precursor protein (APP) function leads to reduced neuronal plasticity, diminished synaptic signaling and enhanced susceptibility of neurons to cellular stress during brain aging. Here we investigated the neuroprotective function of the soluble APP ectodomain sAPPα (soluble APPα), which is generated by cleavage of APP by α-secretase along the non-amyloidogenic pathway. Recombinant sAPPα protected primary hippocampal neurons and SH-SY5Y neuroblastoma cells from cell death induced by trophic factor deprivation. We show that this protective effect is abrogated in neurons from APP-knockout animals and APP-depleted SH-SY5Y cells, but not in APP-like protein 1- and 2- (APLP1 and APLP2) depleted cells, indicating that expression of membrane-bound holo-APP is required for sAPPα-dependent neuroprotection. Trophic factor deprivation diminished the activity of the Akt survival pathway. Strikingly, both recombinant sAPPα and the APP-E1 domain were able to stimulate Akt activity in wild-type (wt) fibroblasts, SH-SY5Y cells and neurons, but failed to rescue in APP-deficient neurons or fibroblasts. The ADAM10 (a disintegrin and metalloproteinase domain-containing protein 10) inhibitor GI254023X exacerbated neuron death in organotypic (hippocampal) slice cultures of wt mice subjected to trophic factor and glucose deprivation. This cell death-enhancing effect of GI254023X could be completely rescued by applying exogenous sAPPα. Interestingly, sAPPα-dependent Akt induction was unaffected in neurons of APP-ΔCT15 mice that lack the C-terminal YENPTY motif of the APP intracellular region. In contrast, sAPPα-dependent rescue of Akt activation was completely abolished in APP mutant cells lacking the G-protein interaction motif located in the APP C-terminus and by blocking G-protein-dependent signaling with pertussis toxin. Collectively, our data provide new mechanistic insights into the physiologic role of APP in antagonizing neurotoxic stress: they suggest that cell surface APP mediates sAPPα-induced neuroprotection via G-protein-coupled activation of the Akt pathway.


Subject(s)
Amyloid beta-Protein Precursor/metabolism , Proto-Oncogene Proteins c-akt/metabolism , ADAM Proteins/antagonists & inhibitors , ADAM Proteins/metabolism , ADAM10 Protein , Amino Acid Motifs , Amyloid Precursor Protein Secretases/antagonists & inhibitors , Amyloid Precursor Protein Secretases/metabolism , Amyloid beta-Protein Precursor/deficiency , Amyloid beta-Protein Precursor/genetics , Animals , Cell Line , Cell Survival/drug effects , Dipeptides/pharmacology , Hippocampus/metabolism , Hippocampus/pathology , Humans , Hydroxamic Acids/pharmacology , In Vitro Techniques , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Pertussis Toxin/toxicity , Phosphatidylinositol 3-Kinases/metabolism , Phosphoinositide-3 Kinase Inhibitors , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/pharmacology , Signal Transduction/drug effects
4.
Gene Ther ; 20(1): 16-23, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22218301

ABSTRACT

Lentiviral vectors are vectors of choice for many gene therapy applications. Recently, efficient targeting of lentiviral vectors pseudotyped with the Measles virus (MV) glycoproteins has been reported. However, MV antibodies in patients might limit the clinical use of these vectors. We demonstrate here that lentiviral vectors can also be pseudotyped with the glycoproteins of Tupaia paramyxovirus (TPMV), the hemagglutinin (H) and fusion (F) protein. As this animal paramyxovirus has no known close relatives in humans, we do not expect TPMV antibodies in patients. Because TPMV normally does not infect human cells, 'detargeting' from natural receptors is unnecessary. Similar to the MV system, TPMV glycoproteins can mediate targeted cell entry by displaying different single-chain antibodies (scAb) directed against surface molecules on target cells on the viral hemagglutinin. We generated a panel of H and F proteins with truncated cytoplasmic tails and determined the variants that efficiently pseudotyped lentiviral vectors. The B-cell marker CD20 was used as a model antigen, and CD20-targeted TPMV vectors selectively transduced CD20-positive cells, including quiescent primary human B-cells. Lentiviral vectors pseudotyped with targeted TPMV envelope proteins might be a valuable vector choice when systemic application of targeted lentiviral vectors in humans is required.


Subject(s)
Genetic Vectors/genetics , Lentivirus/genetics , Paramyxoviridae/genetics , Viral Fusion Proteins/genetics , Amino Acid Sequence , Animals , Antigens, CD20/immunology , B-Lymphocytes/immunology , B-Lymphocytes/virology , Hemagglutinins, Viral/genetics , Hemagglutinins, Viral/immunology , Humans , Molecular Sequence Data , Protein Structure, Tertiary , Transformation, Genetic , Tupaia/virology , Viral Fusion Proteins/chemistry , Viral Fusion Proteins/immunology
5.
Gene Ther ; 18(10): 953-60, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21472010

ABSTRACT

RNAi represents a powerful technology to specifically downregulate the expression of target genes. For cancer research and therapy, an efficient in vivo delivery system is supposed to distribute RNAi to all tumour cells upon systemic administration. We present replication-competent murine leukaemia virus (MLV) vectors, which deliver RNAi to tumour tissue upon tail vein injection. In HT1080 cells stably expressing GFP or luciferase, GFP expression was suppressed by more than 80% and luciferase (luc) activity by more than 90%, even when only 0.1% of the cells were initially infected with reporter gene specific vectors. To demonstrate its potential, PLK1- and MMP14-specific small hairpin RNA expression cassettes were applied in the system. Upon infection, PLK1 and MMP14 levels were reduced on mRNA and protein level. MLV-shPLK1-infected cells were arrested in the G2-phase and underwent apoptosis. MLV-shMMP14-infected cells showed reduced MMP2 activity, as well as substantially reduced invasion and tumour growth. In vivo, MLV-shLuc silenced luc expression in HT1080-luc tumour tissue by more than 80% and MLV-shPLK1 reduced tumour growth substantially, demonstrating the therapeutic relevance of this system. This RNAi vector system allows long-term downregulation of target gene expression as well as efficient delivery to and distribution throughout tumour tissue in vivo.


Subject(s)
G2 Phase Cell Cycle Checkpoints/genetics , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Leukemia Virus, Murine/genetics , Neoplasms/metabolism , Neoplasms/therapy , RNA Interference , Animals , Blotting, Western , Cell Cycle Proteins/genetics , Cell Line, Tumor , DNA Primers/genetics , Female , Flow Cytometry , Green Fluorescent Proteins/metabolism , Humans , Luciferases/metabolism , Matrix Metalloproteinase 14/genetics , Mice , Mice, SCID , Protein Serine-Threonine Kinases/genetics , Proto-Oncogene Proteins/genetics , Real-Time Polymerase Chain Reaction , Polo-Like Kinase 1
6.
Article in English | MEDLINE | ID: mdl-19940966

ABSTRACT

Many gene therapy medicinal products and also some vaccines consist of, or contain, genetically modified organisms (GMOs), which require specific consideration in the environmental risk assessment (ERA) before marketing authorisation or clinical trial applications. The ERA is performed in order to identify the potential risks for public health and the environment, which may arise due to the clinical use of these medicinal products. If such environmental risks are identified and considered as not acceptable, the ERA should go on to propose appropriate risk management strategies capable to reduce these risks. This article will provide an overview of the legal basis and requirements for the ERA of GMO-containing medicinal products in the context of marketing authorisation in the EU and clinical trials in Germany. Furthermore, the scientific principles and methodology that generally need to be followed when preparing an ERA for GMOs are discussed.


Subject(s)
Biological Therapy/adverse effects , Cell Transplantation/legislation & jurisprudence , Conservation of Natural Resources/legislation & jurisprudence , Genetic Engineering/legislation & jurisprudence , Genetic Therapy/legislation & jurisprudence , Organisms, Genetically Modified , Clinical Trials as Topic/legislation & jurisprudence , Genetic Therapy/adverse effects , Marketing of Health Services/legislation & jurisprudence , Risk Assessment
7.
Gene Ther ; 16(5): 700-5, 2009 May.
Article in English | MEDLINE | ID: mdl-19212424

ABSTRACT

We pseudotyped HIV-1 vectors with cytoplasmic tail-truncated envelope glycoproteins of a wild-type (WT) measles virus (MV). The particles entered the lymphatic cells exclusively through the signaling lymphocyte activation molecule (SLAM, CD150), whereas particles pseudotyped with the MV vaccine strain glycoproteins also recognized the ubiquitous membrane cofactor protein (CD46) as receptor and had less specific cell entry. MV(WT)-HIV vectors reached titers of 10(8) t.u. ml(-1), which were up to 10-fold higher than those of MV(Vac)-HIV vectors, and discriminated between SLAM-positive and SLAM-negative cells, also in mixed cell cultures. As these vectors transduce primary human cells more efficiently than vesicular stomatitis virus-G pseudotyped vectors do, they are promising candidates for gene transfer to human lymphocytes and certain epithelial cells.


Subject(s)
Genetic Vectors/genetics , HIV-1/genetics , Lentivirus/genetics , Measles virus/genetics , Viral Envelope Proteins/genetics , B-Lymphocytes/virology , Cell Line , Epithelial Cells/virology , Gene Targeting/methods , HIV-1/physiology , Humans , Intracellular Signaling Peptides and Proteins/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , Transfection , Viral Tropism/genetics , Virus Internalization
8.
Gene Ther ; 15(22): 1500-10, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18509380

ABSTRACT

Virotherapy is currently being developed for many different types of viruses including replication-competent murine leukaemia virus (MLV) as a novel tool in cancer therapy. However, there is the risk of insertional mutagenesis associated with this virus, making careful preclinical studies necessary before its first application in man. We have previously generated conditionally replication-competent MLV variants that require activation by tumour-associated proteases to become infectious. Here we analysed in a comparative study the spreading of non-targeted and of such tumour-targeted MLV variants to tumour and extratumoural organs in immunodeficient mice. Both virus types were able to efficiently infect tumour cells after systemic administration. The non-targeted virus, however, also infected extratumoural organs like bone marrow, spleen and liver efficiently. In contrast, the targeted viruses revealed in a quantitative analysis of virus spreading an up to 500-fold more selective infection of tumour tissue than the non-targeted virus. The data raise serious doubts about a safe clinical use of non-targeted MLV. Engineering the virus to become activatable by tumour-associated proteases can significantly improve the safety of MLV.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Neoplasms/therapy , Oncolytic Virotherapy/methods , Retroviridae/genetics , Animals , Bone Marrow/virology , Cell Line , Cell Line, Tumor , Female , Humans , Injections, Intravenous , Liver/virology , Matrix Metalloproteinases/metabolism , Mice , Mice, SCID , Neoplasms/enzymology , Neoplasms/virology , Spleen/virology , Virus Activation , Virus Integration , Virus Replication , Xenograft Model Antitumor Assays
9.
Gene Ther ; 12 Suppl 1: S146-52, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16231047

ABSTRACT

We report here the topics discussed during the round table of the 2nd European Conference & Practical Course: Towards Clinical Gene Therapy: Preclinical Gene Transfer Assessment, held in Bellaterra (Spain), 1-14 February, 2004. First, how to predict the risk of pathologies generated by changes of the gene expression after proviral genome integration. In the light of the scientific information that emerged after the SAEs occurred in three X-SCID patients treated in France, (a) it is necessary to take into the account the dose of vector used in transduction protocols, in order to minimize the risk to target potentially pathogenic loci. Namely, low vector doses are recommended to minimize the number of vector genomes inserted per cell. (b) The potency of vector elements (ie promoter and transgene), in terms of activation of undesired cell function(s), should be elucidated to devise safe transduction protocols. (c) Target cells should be better characterized before and after transduction to avoid reinfusion into patients' cells, with proviral integration that may be pathogenic. (d) The possibility of replacing onco-retroviruses with other vector systems should be envisaged, for example, nonintegrative gene correction strategies. Second, adequate animal models are required in preclinical experimentation before going to clinics. Although animal models are not yet predictive for risk assessment of proviral insertion, they allow validation of the proof of principle of gene therapy strategies and pharmacological characterization of gene transfer products. Third, a dialogue between researchers and members of regulatory agencies is necessary to implement the regulatory frame where gene therapy products are to be used as new bio-pharmaceuticals. This will implement the whole gene therapy process development at both preclinic (research, development and clinical designs) and postclinic (follow-up of patients) stages. Hence, a European cooperation between professionals (researchers, physicians, industries, patients' associations, investors, etc) will allow implementation of gene therapy regulation in Eastern European countries.


Subject(s)
Biomedical Research/standards , Biopharmaceutics/standards , Biotechnology/standards , Genetic Therapy/standards , Safety Management , Animals , Clinical Trials as Topic , Ethics, Research , Genetic Vectors/adverse effects , Humans , Models, Animal , Transgenes , Vaccines, DNA/adverse effects
10.
Gene Ther ; 12(11): 918-26, 2005 Jun.
Article in English | MEDLINE | ID: mdl-15716977

ABSTRACT

Viruses conditionally replicating in cancer cells form an attractive novel class of antitumoral agents. To engineer such viruses infectivity can be coupled with proteolytic activity of the target cell by modifying the envelope (Env) protein of murine leukaemia virus (MLV) with blocking domains that prevent cell entry unless they are cleaved off by tumour-associated proteases like the matrix metalloproteases (MMP). Here we show that MLV variants selectively spreading through MMP-positive cells can be evolved from virus libraries, in which a standard MMP-2 substrate peptide connecting the blocking domain CD40L with the Env protein was diversified. Passaging the virus library on human fibrosarcoma or glioma cell lines resulted in the selection of about 10 virus clones, of which the three most frequent ones were shown to become activated by MMPs and to be replication competent on MMP-positive cells only. On these cells, the selected linker peptides improved the spreading by several orders of magnitude in vitro, as well as in tumour xenografts in vivo, approaching the kinetic of the unmodified wild-type virus. The data suggest that retroviral protease substrate libraries form a potent tool for the engineering of viruses conditionally replicating in a given cancer cell type of interest.


Subject(s)
Gene Library , Matrix Metalloproteinases/metabolism , Neoplasms/virology , Retroviridae/physiology , Animals , Blotting, Western , Gene Targeting/methods , Gene Transfer Techniques , Genetic Vectors , Humans , Mice , Mice, SCID , Neoplasm Transplantation , Neoplasms/enzymology , Plasmids/genetics , Retroviridae/genetics , Tropism , Tumor Cells, Cultured , Virus Activation/physiology , Virus Replication/physiology
11.
Gene Ther ; 10(16): 1370-80, 2003 Aug.
Article in English | MEDLINE | ID: mdl-12883534

ABSTRACT

Protease-activatable retroviral vectors offer the possibility of targeted gene transfer into cancer cells expressing a unique set of proteases as, for example, the matrix metalloproteases (MMPs). However, it is difficult to predict which substrate sequence will be optimally cleaved by a given tumour cell type. Therefore, we developed a novel approach that allows the selection of MMP-activatable retroviruses from libraries of viruses displaying combinatorially diversified protease substrates. Starting from a virus harbouring a standard MMP-2 substrate motif, after only two consecutive cycles of diversification and in vivo selection, MMP-activatable viruses were recovered. Biochemical characterization of the selected viruses revealed that their linker peptides showed a considerably increased sensitivity for MMP-2 cleavage, and interestingly also improved the particle incorporation rate of the Env protein. Owing to the optimized linker peptide, the selected viruses exhibited a greatly enhanced spreading efficiency through human fibrosarcoma cells, while having retained the dependency on MMP activation. Moreover, cell entry efficiency and virus titres were considerably improved as compared to the parental virus displaying the standard MMP-2 substrate. The results presented imply that retroviral protease substrate libraries allow the definition of MMP substrate specificities under in vivo conditions as well as the generation of optimally adapted tumour-specific viruses.


Subject(s)
Evolution, Molecular , Genetic Therapy/methods , Matrix Metalloproteinases/metabolism , Neoplasms/therapy , Retroviridae/genetics , Cell Line , Genetic Engineering/methods , Humans , Peptide Library , Virus Activation
12.
Gene Ther ; 8(15): 1202-6, 2001 Aug.
Article in English | MEDLINE | ID: mdl-11509952

ABSTRACT

In contrast to murine leukaemia virus (MLV)-derived vector systems, vector particles derived from the avian spleen necrosis virus (SNV) have been successfully targeted to subsets of human cells by envelope modification with antibody fragments (scFv). However, an in vivo application of the SNV vector system in gene transfer protocols is hampered by its lack of resistance against human complement. To overcome this limitation we established pseudotyping of MLV vector particles produced in human packaging cell lines with the SNV envelope (Env) protein. Three variants of SNV Env proteins differing in the length of their cytoplasmic domains were all efficiently incorporated into MLV core particles. These pseudotype particles infected the SNV permissive cell line D17 at titers of up to 10(5) IU/ml. A stable packaging cell line (MS4) of human origin released MLV(SNV) pseudotype vectors that were resistant against human complement inactivation. To redirect their tropism to human T cells, MS4 cells were transfected with the expression gene encoding the scFv 7A5 in fusion with the transmembrane domain (TM) of the SNV Env protein, previously shown to retarget SNV vector particles to human lymphocytes. MLV(SNV-7A5)-vector particles released from these cells were selectively infectious for human T cell lines. The data provide a proof of principle for targeting MLV-derived vectors to subpopulations of human cells through pseudotyping with SNV targeting envelopes.


Subject(s)
Genetic Therapy/methods , Genetic Vectors/administration & dosage , Retroviridae/genetics , Transfection/methods , Animals , Cell Line , Dogs , Genetic Engineering/methods , Humans , Leukemia Virus, Murine/genetics
13.
Virology ; 273(1): 16-20, 2000 Jul 20.
Article in English | MEDLINE | ID: mdl-10891403

ABSTRACT

Lentiviral vectors pseudotyped with the envelope glycoproteins (Env) of amphotropic murine leukemia virus (MLV) and the G protein of vesicular stomatitis virus (VSV-G) have been successfully used in recent preclinical gene therapy studies. We report here the generation of infectious HIV-1-derived vector particles pseudotyped with the Env of the molecular clone 10A1 of MLV and with chimeric envelope glycoprotein variants derived from gibbon ape leukemia virus (GaLV) and MLV. Formation of infectious HIV-1 (GaLV) pseudotype vectors was only possible with the substitution of the cytoplasmic tail of GaLV Env with that of MLV. The lentiviral vectors exhibited a host cell range identical with that of MLV(GaLV) and MLV(10A1) vectors, which are known to enter cells either via the GaLV-receptor Glvr-1 (Pit-1) or via the amphotropic receptor Ram-1 (Pit-2) in addition to Glvr-1, respectively. Thus, HIV-1(GaLV) and HIV-1(10A1) pseudotype vectors may be useful for efficient gene transfer into a variety of human tissues like primary human hematopoietic cells.


Subject(s)
Gene Products, env/metabolism , HIV-1/metabolism , Leukemia Virus, Gibbon Ape/genetics , Leukemia Virus, Murine/genetics , AIDS Vaccines/chemistry , AIDS Vaccines/genetics , Amino Acid Sequence , Animals , Cell Line , Dogs , Flow Cytometry , Gene Products, env/chemistry , Gene Products, env/genetics , Genetic Vectors/genetics , HIV-1/genetics , HIV-1/physiology , Humans , Mice , Molecular Sequence Data , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Transfection , Vaccines, Synthetic/chemistry , Vaccines, Synthetic/genetics , Viral Plaque Assay
14.
Hum Gene Ther ; 11(2): 293-303, 2000 Jan 20.
Article in English | MEDLINE | ID: mdl-10680843

ABSTRACT

To generate T cell-specific retroviral vectors an scFv phage display library derived from immunized mice was selected for binding to the human T cell line Molt-4/8. The scFv cDNAs recovered from the selected phages were transiently expressed as an N-terminal fusion of the spleen necrosis virus (SNV) transmembrane protein (TM) subunit of the viral envelope protein (Env) in the cell line DSH-cxl, which packages the beta-galactosidase gene into SNV particles. Screening of supernatants from about 150 transfections resulted in the identification of 5 scFvs that mediated efficient transduction of Molt-4/8 cells. Using stable packaging cell lines vector preparations with titers greater than 10(4) EFU/ml on human T cells were obtained. The scFv 7A5 in particular was able to mediate selective transduction of human T cells with high efficiency. Titers of up to 106 EFU/ml were reached on Molt-4/8, Jurkat, and A301 cells, while titers on HeLa cells, TE671 cells, 293T cells, and HT1080 cells were below 102 EFU/ml. Transduction of stimulated primary human peripheral blood cells, which consisted mainly of T cells, was about fivefold more efficient than transduction of B cells. Western blot analysis of supernatant from the 7A5 packaging cells demonstrated incorporation of 7A5-TM into vector particles and indicated proteolytic processing of the coexpressed unmodified TM during particle formation. Binding of bacterially expressed 7A5-scFv to a panel of cell lines correlated well with the transduction results. These data provide the first proof of concept that a general approach can be taken to obtain scFvs able to mediate selective gene transfer into target cells.


Subject(s)
Antibodies, Monoclonal/chemistry , Retroviridae/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/genetics , B-Lymphocytes/immunology , Blotting, Western , Cell Line , Flow Cytometry , Gene Transfer Techniques , Genetic Vectors , HeLa Cells , Humans , Immunoglobulin Fragments/metabolism , Leukocytes, Mononuclear/immunology , Mice , Mice, Inbred BALB C , Molecular Sequence Data , Peptide Library , Radioimmunoprecipitation Assay , Retroviridae/genetics
15.
Virology ; 267(2): 229-36, 2000 Feb 15.
Article in English | MEDLINE | ID: mdl-10662618

ABSTRACT

Retroviral vectors derived from amphotropic murine leukemia viruses (MLV) mediate gene transfer into almost all human cells and are thus not suitable for in vivo applications in gene therapy in which cell-specific gene delivery is required. We and others recently reported the generation of MLV-derived vectors pseudotyped by variants of the envelope glycoproteins (Env) of human immunodeficiency virus type 1 (HIV-1), thus displaying the CD4-dependent tropism of the parental lentivirus (Mammano et al., 1997, J. Virol. 71, 3341-3345; Schnierle et al., 1997, Proc. Natl. Acad. Sci. USA 76, 8640-8645). However, because of their HIV-1-derived envelopes these vectors are neutralized by HIV-specific antibodies present in some infected patients. To circumvent this problem, we pseudotyped MLV capsid particles with variants of Env proteins derived from the apathogenic simian immunodeficiency virus (SIVagm) of African green monkeys (AGM; Chlorocebus pygerythrus). Truncation of the C-terminal domain of the transmembrane protein was found to be necessary to allow formation of infectious pseudotype vectors. These [MLV(SIVagm)] vectors efficiently transduced various human CD4-expressing cell lines using the coreceptors CCR5 and Bonzo to enter target cells. Moreover, they were resistant to neutralization by antibodies directed against HIV-1. Therefore, [MLV(SIVagm)] vectors will be useful to study the mechanisms of SIVagm cell entry and for the selective gene transfer into CD4+ T-cells of AIDS patients.


Subject(s)
CD4-Positive T-Lymphocytes/metabolism , Genetic Vectors/genetics , HIV Infections/blood , Immune Sera/immunology , Leukemia Virus, Murine/genetics , Receptors, G-Protein-Coupled , Amino Acid Sequence , Animals , CD4-Positive T-Lymphocytes/virology , Cell Line , Chlorocebus aethiops , DNA, Recombinant , DNA, Viral/genetics , Gene Expression Regulation , Genes, env/genetics , Genetic Variation , Genetic Vectors/immunology , Giant Cells/virology , HeLa Cells , Humans , Jurkat Cells , Leukemia Virus, Murine/immunology , Mice , Molecular Sequence Data , Neutralization Tests , Receptors, CCR5/physiology , Receptors, CXCR6 , Receptors, Chemokine , Receptors, Cytokine/physiology , Receptors, Virus/physiology , Retroviridae/genetics , Retroviridae/immunology , Simian Immunodeficiency Virus/genetics , Tumor Cells, Cultured
16.
J Virol ; 73(7): 5265-73, 1999 Jul.
Article in English | MEDLINE | ID: mdl-10364272

ABSTRACT

CD46, which serves as a receptor for measles virus (MV; strain Edmonston), is rapidly downregulated from the cell surface after contact with viral particles or infected cells. We show here that the same two CD46 complement control protein (CCP) domains responsible for primary MV attachment mediate its downregulation. Optimal downregulation efficiency was obtained with CD46 recombinants containing CCP domains 1 and 2, whereas CCP 1, alone and duplicated, induced a slight downregulation. Using persistently infected monocytic/promyelocytic U937 cells which release very small amounts of infectious virus, and uninfected HeLa cells as contact partners, we then showed that during contact the formation of CD46-containing patches and caps precedes CD46 internalization. Nevertheless, neither substances inhibiting capping nor the fusion-inhibiting peptide Z-D-Phe-L-Phe-Gly-OH (FIP) blocked CD46 downregulation. Thus, CD46 downregulation can be uncoupled from fusion and subsequent virus uptake. Interestingly, in that system cell-cell contacts lead to a remarkably efficient infection of the target cells which is only partially inhibited by FIP. The finding that the contact of an infected with uninfected cells results in transfer of infectious viral material without significant (complete) fusion of the donor with the recipient cell suggests that microfusion events and/or FIP-independent mechanisms may mediate the transfer of MV infectivity from cell to cell.


Subject(s)
Antigens, CD/metabolism , Measles virus/metabolism , Membrane Glycoproteins/metabolism , Receptors, Virus/metabolism , Animals , Antigens, CD/genetics , CHO Cells , Cell Communication , Cricetinae , Down-Regulation , HeLa Cells , Hemagglutinins, Viral/metabolism , Humans , Measles virus/physiology , Membrane Cofactor Protein , Membrane Fusion , Membrane Glycoproteins/genetics , Nucleocapsid Proteins , Nucleoproteins/metabolism , Receptors, Virus/genetics , U937 Cells , Viral Proteins/metabolism , Virus Latency
17.
Curr Opin Mol Ther ; 1(5): 613-21, 1999 Oct.
Article in English | MEDLINE | ID: mdl-11249668

ABSTRACT

The availability of cell targeting vectors is an unalterable requirement for in vivo gene therapy trials. This review will describe the different strategies developed over the past few years in order to target retroviral vectors to preselected human cell types by genetic modification of the envelope (Env) proteins. Current targeting concepts include the substitution of the complete Env protein as well as the incorporation of new receptor binding domains into the Env protein. These approaches are aimed at altering the host range of vectors with a natural tropism for non-human cells to specific human cell types, or achieving tissue-specificity for vectors that would naturally infect a wide spectrum of human cell types. Targeting concepts and efficient targeting vectors with potential for clinical trials will be described, and their advantages and disadvantages will be discussed.


Subject(s)
Genetic Vectors , Retroviridae/genetics , ErbB Receptors/genetics , Gene Products, env/chemistry , Gene Products, env/genetics , Gene Targeting , Gene Transfer Techniques , Genetic Therapy/methods , Humans
18.
Nat Biotechnol ; 16(10): 951-4, 1998 Oct.
Article in English | MEDLINE | ID: mdl-9788352

ABSTRACT

Phage display libraries are widely used for selection and optimization of polypeptide ligands or protease substrates. Because they are expressed and amplified in bacterial hosts, phage are not ideal for displaying eukaryotic polypeptides or for probing mammalian cells. As retroviruses do not suffer from these limitations we constructed plasmids encoding replication-competent murine leukemia viruses displaying a virally encoded epidermal growth factor (EGF) domain at the N-terminus of the envelope glycoprotein. The EGF-displaying viruses replicated freely on EGF receptor-poor cells without deleting the displayed EGF domain but did not propagate on EGF receptor-rich cells because they were sequestered by the EGF receptors. A retrovirus display library was then generated by diversifying the seven-residue linker between the envelope glycoprotein and the displayed EGF domain. Selective pressure for loss of EGF receptor-binding activity was applied to the library by serial passage on EGF receptor-rich HT1080 cells. The selected viruses propagated on these cells with wild-type efficiencies, a phenotype that was conferred by intracellular cleavage of their displayed linker sequences. The selected linker sequences invariably presented arginine-rich motifs matching the consensus cleavage signal for furin-like proteases. Retrovirus display libraries can be used for the selection of polypeptides interacting with components of living mammalian cells.


Subject(s)
Endopeptidases/metabolism , Leukemia Virus, Murine/genetics , 3T3 Cells , Amino Acid Sequence , Animals , Base Sequence , Cloning, Molecular , DNA Primers , Epidermal Growth Factor/genetics , Epidermal Growth Factor/metabolism , ErbB Receptors/metabolism , Hydrolysis , Mice , Molecular Sequence Data , Tumor Cells, Cultured
19.
J Virol ; 72(9): 7420-7, 1998 Sep.
Article in English | MEDLINE | ID: mdl-9696838

ABSTRACT

Attenuated Edmonston measles virus (MV-Edm) is not pathogenic in standard mice. We show here that MV-Edm inoculated via the natural respiratory route has a limited propagation in the lungs of mice with a targeted mutation inactivating the alpha/beta interferon receptor. A high dose of MV-Edm administered intracerebrally is lethal for about half of these mice. To study the consequences of the availability of a high-affinity receptor for MV propagation, we generated alpha/beta interferon-defective mice expressing human CD46 with human-like tissue specificity. Intranasal infection of these mice with MV-Edm resulted in enhanced spread to the lungs and more prominent inflammatory response. Virus replication was also detected in peripheral blood mononuclear cells, the spleen, and the liver. Moreover, intracerebral inoculation of adult animals with low MV-Edm doses caused encephalitis with almost inevitably lethal outcome. We conclude that in mice alpha/beta interferon controls MV infection and that a high-affinity receptor facilitates, but is not strictly required for, MV spread and pathogenesis.


Subject(s)
Antigens, CD/metabolism , Measles virus/pathogenicity , Membrane Glycoproteins/metabolism , Animals , Antigens, CD/genetics , Brain/pathology , Brain/virology , Gene Deletion , Humans , Injections , Interferon-alpha/genetics , Interferon-alpha/physiology , Interferon-beta/genetics , Interferon-beta/physiology , Lung/pathology , Lung/virology , Measles virus/physiology , Membrane Cofactor Protein , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Virus Replication
20.
J Biol Chem ; 272(35): 22072-9, 1997 Aug 29.
Article in English | MEDLINE | ID: mdl-9268348

ABSTRACT

The measles virus (MV) hemagglutinin binds to the complement control protein (CCP) CD46 primarily through the two external modules, CCP-I and -II. To define the residues involved in binding, 40 amino acids predicted to be solvent-exposed on the CCP-I-II module surface were changed to either alanine or serine. Altered proteins were expressed on the cell surface, and their abilities to bind purified MV particles, a soluble form of hemagglutinin (sH) and nine CD46-specific antibodies competing to different levels with sH attachment, were measured. All proteins retained, at least in part, MV and sH binding, but some completely lost binding to certain antibodies. Amino acids essential for binding of antibodies weakly or moderately competing with sH attachment are situated in the membrane-distal tip of CCP-I, whereas residues involved in binding of strongly sH competing antibodies cluster in the center of CCP-I (Arg-25, Asp-27) or in CCP-II (Arg-69, Asp-70). Both clusters face the same side of CCP-I-II and map close to amino acid exchanges impairing sH binding (E11A, R29A, P39A, and D70A) or MV binding (D70A and E84A) and to a six-amino acid loop, previously shown to be necessary for sH binding.


Subject(s)
Antigens, CD/chemistry , Complement Inactivator Proteins/chemistry , Measles virus/chemistry , Membrane Glycoproteins/chemistry , Receptors, Virus/chemistry , Amino Acid Sequence , Animals , Antibodies, Monoclonal/metabolism , Binding Sites , Epitope Mapping , Hemagglutinins/metabolism , Membrane Cofactor Protein , Mice , Mice, Inbred BALB C , Models, Molecular , Molecular Sequence Data , Protein Binding , Protein Structure, Tertiary
SELECTION OF CITATIONS
SEARCH DETAIL
...