1
|
Lindegger DJ. Advanced Therapies for Human Immunodeficiency Virus. Med Sci (Basel) 2024; 12:33. [PMID: 39051379 PMCID: PMC11270269 DOI: 10.3390/medsci12030033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 06/27/2024] [Accepted: 07/05/2024] [Indexed: 07/27/2024] Open
Abstract
Human Immunodeficiency Virus (HIV) remains a significant global health challenge with approximately 38 million people currently having the virus worldwide. Despite advances in treatment development, the virus persists in the human population and still leads to new infections. The virus has a powerful ability to mutate and hide from the human immune system in reservoirs of the body. Current standard treatment with antiretroviral therapy effectively controls viral replication but requires lifelong adherence and does not eradicate the virus. This review explores the potential of Advanced Therapy Medicinal Products as novel therapeutic approaches to HIV, including cell therapy, immunisation strategies and gene therapy. Cell therapy, particularly chimeric antigen receptor T cell therapy, shows promise in preclinical studies for targeting and eliminating HIV-infected cells. Immunisation therapies, such as broadly neutralising antibodies are being investigated to control viral replication and reduce reservoirs. Despite setbacks in recent trials, vaccines remain a promising avenue for HIV therapy development. Gene therapy using technologies like CRISPR/Cas9 aims to modify cells to resist HIV infection or eliminate infected cells. Challenges such as off-target effects, delivery efficiency and ethical considerations persist in gene therapy for HIV. Future directions require further research to assess the safety and efficacy of emerging therapies in clinical trials. Combined approaches may be necessary to achieve complete elimination of the HIV reservoir. Overall, advanced therapies offer new hope for advancing HIV treatment and moving closer to a cure.
Collapse
Affiliation(s)
- Daniel Josef Lindegger
- Independent Researcher, 6000 Lucerne, Switzerland;
- Independent Researcher, London SW1A2JR, UK
| |
Collapse
|
2
|
Senavirathne G, London J, Gardner A, Fishel R, Yoder KE. DNA strand breaks and gaps target retroviral intasome binding and integration. Nat Commun 2023; 14:7072. [PMID: 37923737 PMCID: PMC10624929 DOI: 10.1038/s41467-023-42641-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Accepted: 10/17/2023] [Indexed: 11/06/2023] Open
Abstract
Retrovirus integration into a host genome is essential for productive infections. The integration strand transfer reaction is catalyzed by a nucleoprotein complex (Intasome) containing the viral integrase (IN) and the reverse transcribed (RT) copy DNA (cDNA). Previous studies suggested that DNA target-site recognition limits intasome integration. Using single molecule Förster resonance energy transfer (smFRET), we show prototype foamy virus (PFV) intasomes specifically bind to DNA strand breaks and gaps. These break and gap DNA discontinuities mimic oxidative base excision repair (BER) lesion-processing intermediates that have been shown to affect retrovirus integration in vivo. The increased DNA binding events targeted strand transfer to the break/gap site without inducing substantial intasome conformational changes. The major oxidative BER substrate 8-oxo-guanine as well as a G/T mismatch or +T nucleotide insertion that typically introduce a bend or localized flexibility into the DNA, did not increase intasome binding or targeted integration. These results identify DNA breaks or gaps as modulators of dynamic intasome-target DNA interactions that encourage site-directed integration.
Collapse
Affiliation(s)
- Gayan Senavirathne
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - James London
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Anne Gardner
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH, 43210, USA
| | - Richard Fishel
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH, 43210, USA.
- Molecular Carcinogenesis and Chemoprevention Program, The James Comprehensive Cancer Center and Ohio State University, Columbus, OH, 43210, USA.
| | - Kristine E Yoder
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH, 43210, USA.
- Molecular Carcinogenesis and Chemoprevention Program, The James Comprehensive Cancer Center and Ohio State University, Columbus, OH, 43210, USA.
- Center for Retrovirus Research, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
3
|
Biomaterial-guided delivery of gene vectors for targeted articular cartilage repair. Nat Rev Rheumatol 2018; 15:18-29. [DOI: 10.1038/s41584-018-0125-2] [Citation(s) in RCA: 87] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
4
|
Lukashev AN, Zamyatnin AA. Viral Vectors for Gene Therapy: Current State and Clinical Perspectives. BIOCHEMISTRY (MOSCOW) 2017; 81:700-8. [PMID: 27449616 DOI: 10.1134/s0006297916070063] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Gene therapy is the straightforward approach for the application of recent advances in molecular biology into clinical practice. One of the major obstacles in the development of gene therapy is the delivery of the effector to and into the target cell. Unfortunately, most methods commonly used in laboratory practice are poorly suited for clinical use. Viral vectors are one of the most promising methods for gene therapy delivery. Millions of years of evolution of viruses have resulted in the development of various molecular mechanisms for entry into cells, long-term survival within cells, and activation, inhibition, or modification of the host defense mechanisms at all levels. The relatively simple organization of viruses, small genome size, and evolutionary plasticity allow modifying them to create effective instruments for gene therapy approaches. This review summarizes the latest trends in the development of gene therapy, in particular, various aspects and prospects of the development of clinical products based on viral delivery systems.
Collapse
Affiliation(s)
- A N Lukashev
- Chumakov Institute of Poliomyelitis and Viral Encephalitides, Moscow, 142782, Russia.
| | | |
Collapse
|
5
|
Kasala D, Yoon AR, Hong J, Kim SW, Yun CO. Evolving lessons on nanomaterial-coated viral vectors for local and systemic gene therapy. Nanomedicine (Lond) 2016; 11:1689-713. [PMID: 27348247 DOI: 10.2217/nnm-2016-0060] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Viral vectors are promising gene carriers for cancer therapy. However, virus-mediated gene therapies have demonstrated insufficient therapeutic efficacy in clinical trials due to rapid dissemination to nontarget tissues and to the immunogenicity of viral vectors, resulting in poor retention at the disease locus and induction of adverse inflammatory responses in patients. Further, the limited tropism of viral vectors prevents efficient gene delivery to target tissues. In this regard, modification of the viral surface with nanomaterials is a promising strategy to augment vector accumulation at the target tissue, circumvent the host immune response, and avoid nonspecific interactions with the reticuloendothelial system or serum complement. In the present review, we discuss various chemical modification strategies to enhance the therapeutic efficacy of viral vectors delivered either locally or systemically. We conclude by highlighting the salient features of various nanomaterial-coated viral vectors and their prospects and directions for future research.
Collapse
Affiliation(s)
- Dayananda Kasala
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Jinwoo Hong
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| | - Sung Wan Kim
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea.,Department of Pharmaceutics & Pharmaceutical Chemistry, Center for Controlled Chemical Delivery, University of Utah, Salt Lake City, UT 84112, USA
| | - Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, 222 Wangsimni-ro, Seongdong-gu, Seoul, Republic of Korea
| |
Collapse
|
6
|
Meng F, Chen C, Wan H, Zhou Q. [Advances of lentiviral vectors]. ZHONGGUO FEI AI ZA ZHI = CHINESE JOURNAL OF LUNG CANCER 2015; 17:870-6. [PMID: 25539614 PMCID: PMC6000409 DOI: 10.3779/j.issn.1009-3419.2014.12.09] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Lentiviral vectors are currently very effective tools in molecular and cell experiment. Lentiviral vector, a kind of retroviral vectors, has a number of unique advantages in target gene transferation, for example, the ability of transfection to the dividing or nondividing cells, its high efficiency of transfection and a capacity of large target gene fragments. This paper describes the sources of lentiviral vectors, molecular characteristics, research progress, etc.
Collapse
Affiliation(s)
- Fanrong Meng
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenviroment, Tianjin Lung Cancer Institute,
Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Chen Chen
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenviroment, Tianjin Lung Cancer Institute,
Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Haisu Wan
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenviroment, Tianjin Lung Cancer Institute,
Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Qinghua Zhou
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenviroment, Tianjin Lung Cancer Institute,
Tianjin Medical University General Hospital, Tianjin 300052, China
| |
Collapse
|
7
|
Ma Y, Chen B, He N, Chen G, Li L, Wu C. Revisiting Complexation between DNA and Polyethylenimine: Does the Disulfide Linkage Play a Critical Role in Promoting Gene Delivery? Macromol Biosci 2014; 14:1807-15. [DOI: 10.1002/mabi.201400253] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Revised: 08/17/2014] [Indexed: 01/23/2023]
Affiliation(s)
- Yongzheng Ma
- Department of Chemistry; The Chinese University of Hong Kong; Shatin N. T. Hong Kong
| | - Baizhu Chen
- Department of Chemistry; The Chinese University of Hong Kong; Shatin N. T. Hong Kong
| | - Ning He
- Department of Chemistry; The Chinese University of Hong Kong; Shatin N. T. Hong Kong
| | - Gaojian Chen
- Department of Chemistry; The Chinese University of Hong Kong; Shatin N. T. Hong Kong
| | - Lianwei Li
- The Hefei National Laboratory of Physical Science at Microscale, Department of Chemical Physics; University of Science and Technology of China; Hefei Anhui 230026 China
| | - Chi Wu
- Department of Chemistry; The Chinese University of Hong Kong; Shatin N. T. Hong Kong
- The Hefei National Laboratory of Physical Science at Microscale, Department of Chemical Physics; University of Science and Technology of China; Hefei Anhui 230026 China
| |
Collapse
|
8
|
Stornaiuolo A, Piovani BM, Bossi S, Zucchelli E, Corna S, Salvatori F, Mavilio F, Bordignon C, Rizzardi GP, Bovolenta C. RD2-MolPack-Chim3, a packaging cell line for stable production of lentiviral vectors for anti-HIV gene therapy. Hum Gene Ther Methods 2013; 24:228-40. [PMID: 23767932 DOI: 10.1089/hgtb.2012.190] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Over the last two decades, several attempts to generate packaging cells for lentiviral vectors (LV) have been made. Despite different technologies, no packaging clone is currently employed in clinical trials. We developed a new strategy for LV stable production based on the HEK-293T progenitor cells; the sequential insertion of the viral genes by integrating vectors; the constitutive expression of the viral components; and the RD114-TR envelope pseudotyping. We generated the intermediate clone PK-7 expressing constitutively gag/pol and rev genes and, by adding tat and rd114-tr genes, the stable packaging cell line RD2-MolPack, which can produce LV carrying any transfer vector (TV). Finally, we obtained the RD2-MolPack-Chim3 producer clone by transducing RD2-MolPack cells with the TV expressing the anti-HIV transgene Chim3. Remarkably, RD114-TR pseudovirions have much higher potency when produced by stable compared with transient technology. Most importantly, comparable transduction efficiency in hematopoietic stem cells (HSC) is obtained with 2-logs less physical particles respect to VSV-G pseudovirions produced by transient transfection. Altogether, RD2-MolPack technology should be considered a valid option for large-scale production of LV to be used in gene therapy protocols employing HSC, resulting in the possibility of downsizing the manufacturing scale by about 10-fold in respect to transient technology.
Collapse
|
9
|
Multiple Gag domains contribute to selective recruitment of murine leukemia virus (MLV) Env to MLV virions. J Virol 2012; 87:1518-27. [PMID: 23152533 DOI: 10.1128/jvi.02604-12] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Retroviruses, like all enveloped viruses, must incorporate viral glycoproteins to form infectious particles. Interactions between the glycoprotein cytoplasmic tail and the matrix domain of Gag are thought to direct recruitment of glycoproteins to native virions for many retroviruses. However, retroviruses can also incorporate glycoproteins from other viruses to form infectious virions known as pseudotyped particles. The glycoprotein murine leukemia virus (MLV) Env can readily form pseudotyped particles with many retroviruses, suggesting a generic mechanism for recruitment. Here, we sought to identify which components of Gag, particularly the matrix domain, contribute to recruitment of MLV Env into retroviral particles. Unexpectedly, we discovered that the matrix domain of HIV-1 Gag is dispensable for generic recruitment, since it could be replaced with a nonviral membrane-binding domain without blocking active incorporation of MLV Env into HIV virions. However, MLV Env preferentially assembles with MLV virions. When MLV and HIV particles are produced from the same cell, MLV Env is packaged almost exclusively by MLV particles, thus preventing incorporation into HIV particles. Surprisingly, the matrix domain of MLV Gag is not required for this selectivity, since MLV Gag containing the matrix domain from HIV is still able to outcompete HIV particles for MLV Env. Although MLV Gag is sufficient for selective incorporation to occur, no single Gag domain dictates the selectivity. Our findings indicate that Env recruitment is more complex than previously believed and that Gag assembly/budding sites have fundamental properties that affect glycoprotein incorporation.
Collapse
|
10
|
Preparation of vesicular stomatitis virus-G (VSV-G) conjugate and its use in gene transfer. Cold Spring Harb Protoc 2012; 2012:453-6. [PMID: 22474657 DOI: 10.1101/pdb.prot068528] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The fusiogenic envelope G glycoprotein of the vesicular stomatitis virus (VSV-G) that has been used to pseudotype retrovirus and lentivirus vectors can be used alone as an efficient vehicle for gene transfer. VSV-G protein is secreted into the culture medium as sendimentable vesicles from cells transfected with a VSV-G expression plasmid in the absence of other viral components. The VSV-G vesicles in the conditioned medium can be partially purified by pelleting through sucrose cushion ultracentrifugation. Protein-DNA complexes are formed by mixing the VSV-G vesicles with naked plasmid DNA. Such complexes show markedly enhanced transfection efficiency when added to the culture medium of recipient cells. The cell tropism of VSV-G-DNA complex-mediated gene transfer resembles that of VSV-G-pseudotyped retrovirus and lentivirus vectors, and the complex is therefore particularly useful for transfection of cells that are refractory to other methods. Still, some cells are refractory to VSV-G-mediated transfection. It should also be noted that overdose of VSV-G can be quite toxic to the recipient cells. The primitive complexes formed by mixing a viral fusiogenic envelope protein with naked DNA may represent a step toward fusing useful features of viral and nonviral vectors for safer and more efficient gene transfer. This protocol describes simple methods for preparation of VSV-G and for gene transfer with DNA-VSV-G complexes.
Collapse
|
11
|
The inside out of lentiviral vectors. Viruses 2011; 3:132-159. [PMID: 22049307 PMCID: PMC3206600 DOI: 10.3390/v3020132] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2010] [Revised: 01/25/2011] [Accepted: 02/08/2011] [Indexed: 11/30/2022] Open
Abstract
Lentiviruses induce a wide variety of pathologies in different animal species. A common feature of the replicative cycle of these viruses is their ability to target non-dividing cells, a property that constitutes an extremely attractive asset in gene therapy. In this review, we shall describe the main basic aspects of the virology of lentiviruses that were exploited to obtain efficient gene transfer vectors. In addition, we shall discuss some of the hurdles that oppose the efficient genetic modification mediated by lentiviral vectors and the strategies that are being developed to circumvent them.
Collapse
|
12
|
Chen F, Wang H, He H, Song L, Wu J, Gao Y, Liu X, He C, Yang H, Chen L, Wang L, Li G, Li Y, Kaplan DE, Zhong J. Short hairpin RNA-mediated silencing of bovine rotavirus NSP4 gene prevents diarrhoea in suckling mice. J Gen Virol 2010; 92:945-51. [PMID: 21148273 DOI: 10.1099/vir.0.027680-0] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
While RNA interference (RNAi) has been widely used to study rotavirus gene function in vitro, the potential therapeutic role for RNAi in vivo has not been explored. To this end, we constructed two recombinant lentiviral vectors containing short hairpin RNA (shRNA) against non-structural protein-4 (NSP4) of bovine rotavirus (BRV), RNAi-351 and RNAi-492. RNAi-351 and RNAi-492 strongly suppressed the transient expression of a FLAG-tagged NSP4 fusion protein in 293T cells. In BRV-susceptible MA104 cells, RNAi-492 more potently silenced NSP4 mRNA than RNAi-351 and combination of the two shRNAs almost completely silenced viral NSP4 gene expression. While 100% of suckling mice exposed to BRV and control shRNA developed severe diarrhoea, no suckling mice exposed to BRV in the presence of RNAi-492 or a combination of RNAi-492/RNAi-351 developed severe diarrhoea, and only 20 and 3.3% developed mild diarrhoea, respectively. In addition, RNAi-492 and RNAi-351 markedly abrogated rotaviral replication in MA104 cells and significantly inhibited BRV replication in mouse pups. These results indicated that shRNAs silencing NSP4 gene had substantial antiviral properties and inhibited replication of BRV in a sequence-specific manner that may have clinical application.
Collapse
Affiliation(s)
- Fangyuan Chen
- Dairy Cattle Research Center, Shandong Academy of Agricultural Sciences, Jinan 250100, PR China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Human immunodeficiency virus type 1 protease inhibitor drug-resistant mutants give discordant results when compared in single-cycle and multiple-cycle fitness assays. J Clin Microbiol 2010; 48:4035-43. [PMID: 20826651 DOI: 10.1128/jcm.00605-10] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
The replication fitness of HIV-1 drug-resistant mutants has been measured using either multiple-cycle or single-cycle assays (MCAs or SCAs); these assays have not been systematically compared. We developed an MCA and an SCA that utilized either intact or env-deleted recombinant viral vectors, respectively, in which virus-infected cells were detected by flow cytometry of a reporter gene product. Fitness was measured using each assay for 11 protease mutants, 9 reverse transcriptase mutants, and two mutants with mutations in gag p6, which is important for the release of virus particles from the cell membrane. In the SCA, fitness (replication capacity [RC]) was defined as the proportion of cells infected by the mutant compared to the wild type 40 h after infection. MCA fitness (1+s) was determined by comparing the changes in the relative proportions of cells infected by the mutant and the wild type between 3 and 5 days after infection. Five protease mutants showed statistically different fitness values by the MCA versus the SCA: the D30N, G48V, I50V, I54L, and I54M mutants. When all the mutants were ranked in order from most to least fit for both assays, 4 protease mutants moved more than 5 positions in rank: the D30N, I54L, I54M, and V82A mutants. There were no significant differences in fitness for the gag p6 or reverse transcriptase mutants. We propose that discordant results in the MCA and SCA are due to alterations in late events in the virus life cycle that are not captured in an SCA, such as burst size, cell-to-cell transmission, or infected-cell life span.
Collapse
|
14
|
Abstract
Lentiviral transgenesis is a promising alternative to direct microinjection of DNA into pronuclei, which is by and large restricted to certain mouse strains. Lentiviruses are complex retroviruses that integrate their genome into the host chromosome. Vectors derived from lentiviruses can efficiently transfer transgenes in oocytes and early embryos, which is the basis for the use of these vectors in transgenesis. Lentivirus transgenesis has been used in many different species, including mouse, rat, pig, bovine, monkeys, and even birds. Here we present a protocol for generating transgenic animals by lentiviral transduction of early embryos as well as for analyzing viral integrants in transgenic animals.
Collapse
Affiliation(s)
- Alexander Pfeifer
- Institute of Pharmacology and Toxicology, Biomedical Center (BMZ), University of Bonn, Germany
| | | | | |
Collapse
|
15
|
|
16
|
Young J, Tang Z, Yu Q, Yu D, Wu Y. Selective killing of HIV-1-positive macrophages and T cells by the Rev-dependent lentivirus carrying anthrolysin O from Bacillus anthracis. Retrovirology 2008; 5:36. [PMID: 18439272 PMCID: PMC2391154 DOI: 10.1186/1742-4690-5-36] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2007] [Accepted: 04/25/2008] [Indexed: 11/17/2022] Open
Abstract
Background The ability of Human Immunodeficiency Virus (HIV) to persist in the body has proven to be a long-standing challenge to virus eradication. Current antiretroviral therapy cannot selectively destroy infected cells; it only halts active viral replication. With therapeutic cessation or interruption, viral rebound occurs, and invariably, viral loads return to pre-treatment levels. The natural reservoirs harboring replication-competent HIV-1 include CD4 T cells and macrophages. In particular, cells from the macrophage lineage resist HIV-1-mediated killing and support sustained viral production. To develop a complementary strategy to target persistently infected cells, this proof-of-concept study explores an HIV-1 Rev-dependent lentiviral vector carrying a bacterial hemolysin, anthrolysin O (anlO) from Bacillus anthracis, to achieve selective killing of HIV-1- infected cells. Results We demonstrate that in the Rev-dependent lentiviral vector, anlO expression is exclusively dependent on Rev, a unique HIV-1 protein present only in infected cells. Intracellular expression and oligomerization of AnlO result in membrane pore formation and cytolysis. We have further overcome a technical hurdle in producing a Revdependent AnlO lentivirus, through the use of β-cyclodextrin derivatives to inhibit direct killing of producer cells by AnlO. Using HIV-1-infected macrophages and T cells as a model, we demonstrate that this Rev-dependent AnlO lentivirus diminishes HIV-1- positive cells. Conclusion The Rev-dependent lentiviral vector has demonstrated its specificity in targeting persistently infected cells. The choice of anlO as the first suicidal gene tested in this vector is based on its cytolytic activity in macrophages and T cells. We conclude that Rev-regulated expression of suicidal genes in HIV-1-positive cells is possible, although future in vivo delivery of this system needs to address numerous safety issues.
Collapse
Affiliation(s)
- Jessica Young
- Department of Molecular and Microbiology, George Mason University, Manassas, VA 20110, USA.
| | | | | | | | | |
Collapse
|
17
|
Abstract
In the last years, different non-biological and biological carrier systems have been developed for anti-HIV1 therapy. Liposomes are excellent potential anti-HIV1 carriers that have been tested with drugs, antisense oligonucleotides, ribozymes and therapeutic genes. Nanoparticles and low-density lipoproteins (LDLs) are cell-specific transporters of drugs against macrophage-specific infections such as HIV1. Through a process of protein transduction, cell-permeable peptides of natural origin or designed artificially allow the delivery of drugs and genetic material inside the cell. Erythrocyte ghosts and bacterial ghosts are a promising delivery system for therapeutic peptides and HIV vaccines. Of interest are the advances made in the field of HIV gene therapy by the use of autologous haematopoietic stem cells and viral vectors for HIV vaccines. Although important milestones have been reached in the development of carrier systems for the treatment of HIV, especially in the field of gene therapy, further clinical trials are required so that the efficiency and safety of these new systems can be guaranteed in HIV patients.
Collapse
Affiliation(s)
- José M Lanao
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Salamanca, Salamanca, Spain.
| | | | | |
Collapse
|
18
|
Masurier C, Boutin S, Veron P, Bernard J, Danos O, Davoust J. Enhanced Lentiviral Transduction of Monocyte-Derived Dendritic Cells in The Presence of Conditioned Medium from Dying Monocytes. Hum Gene Ther 2007; 18:161-70. [PMID: 17326725 DOI: 10.1089/hum.2006.106] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Lentiviral vectors (LVs) are attractive vehicles for the transduction of human dendritic cells (DCs) in order to mobilize their endogenous antigen presentation pathways. We analyzed here how to improve the efficiency of LV transduction, which we performed at the initial stages of the differentiation of purified monocytes into dendritic cells (Mo-DCs). Using LVs pseudotyped with the vesicular stomatitis virus envelope G glycoprotein (VSV-G), we found that a conditioned medium derived from dying monocytes (MCM) improved by 2- to 10- fold the proportion of transduced Mo-DCs. This enhanced transduction efficiency requires the presence of MCM during the initial stage of LV transduction and does not affect the phenotype and antigen presentation function of terminally differentiated Mo-DCs. Importantly, we found that MCM derived from a human acute monocytic leukemia cell line, THP-1, was equally effective. The MCM activity was heat stable (56 degrees C) and was present in the soluble fraction after high-speed centrifugation. Altogether our results show that a soluble factor present in dying monocyte cultures can replace advantageously facilitating agents such as Polybrene, to achieve high LV transductions levels. This protocol can be performed with autologous monocytes and is therefore applicable in clinical settings.
Collapse
Affiliation(s)
- C Masurier
- Laboratoire d'Immunologie, Généthon, CNRS UMR 8115, 91002 Evry Cedex, France.
| | | | | | | | | | | |
Collapse
|
19
|
Abstract
Considerable progress has been made in methods for production of transgenic livestock; beginning with pronuclear microinjection over 20 years ago. New methods, including the use of viral vectors, sperm-mediated gene transfer and somatic cell cloning, have overcome many of the limitations of pronuclear microinjection. It is now possible to not only readily make simple insertional genetic modifications, but also to accomplish, more complex, homozygous gene targeting and artificial chromosome transfer in livestock.
Collapse
Affiliation(s)
- J M Robl
- Hematech Inc, Sioux Falls, SD 57106, USA.
| | | | | | | |
Collapse
|
20
|
Yang G, Zhong Q, Huang W, Reiser J, Schwarzenberger P. Retrovirus molecular conjugates: a versatile and efficient gene transfer vector system for primitive human hematopoietic progenitor cells. Cancer Gene Ther 2006; 13:460-8. [PMID: 16282988 DOI: 10.1038/sj.cgt.7700911] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In principle, transient nongenetic modification of a noninfectious gene transfer virus enabling a one time infection and transduction of human cells could eliminate the risk of formation of replication competent virus. Formation of a molecular conjugate vector by conjugation of noninfective ecotropic murine Moloney leukemia virus to polylysine (eMMLV-PL) enabled high-efficiency transduction of human HPC using in vitro and in vivo assays. Xenotransplanted NOD-SCID mice durably expressed the transgene in human leukocytes and human progenitor cells with eMMLV-PL achieving three-fold increased transduction efficiency when directly compared to optimized amphotropic MMLV (aMMLV) transduction. Both aMMLV and eMMLV assembled conjugate vectors showed similar transduction efficiency indicating predominant polylysine-mediated uptake. Integration of retroviral sequences was determined from individual human HPC recovered from eMMLV-PL-xenotransplanted animals. This simple and versatile concept of conjugate gene transfer vectors has the potential to enhance transduction efficiency as well as to improve certain safety aspects of human gene therapy. Moreover, because it permits effective cellular internalization of particles, this concept of molecular conjugates can be used as research tool to investigate the interactions of otherwise noninfectious viruses or modified viral particles at the genomic level.
Collapse
Affiliation(s)
- G Yang
- Department of Genetics, Louisiana State University Health Sciences Center, New Orleans, LA 70112, USA
| | | | | | | | | |
Collapse
|
21
|
Abstract
Lentiviral vectors portend a promising system to deliver antiviral genes for treating viral infections such as HIV-1 as they are capable of stably transducing both dividing and nondividing cells. Recently, small interfering RNAs (siRNAs) have been shown to be quite efficacious in silencing target genes. RNA interference is a natural mechanism, conserved in nature from Yeast to Humans, by which siRNAs operate to specifically and potently down regulate the expression of a target gene either transcriptionally (targeted to DNA) or post-transcriptionally (targeted to mRNA). The specificity and relative simplicity of siRNA design insinuate that siRNAs will prove to be favorable therapeutic agents. Since siRNAs are a small nucleic acid reagents, they are unlikely to elicit an immune response and genes encoding these siRNAs can be easily manipulated and delivered by lentiviral vectors to target cells. As such, lentiviral vectors expressing siRNAs represent a potential therapeutic approach for the treatment of viral infections such as HIV-1. This review will focus on the development, lentiviral based delivery, and the potential therapeutic use of siRNAs in treating viral infections.
Collapse
Affiliation(s)
- K V Morris
- Department of Molecular and Experimental Medicine, Division of Rheumatology, The Scripps Research Institute, La Jolla, CA USA
| | - J J Rossi
- Division of Molecular Biology, Beckman Research Institute of the City of Hope, Duarte, CA USA
| |
Collapse
|
22
|
Morris K, Castanotto D, Al-Kadhimi Z, Jensen M, Rossi J, Cooper LJN. Enhancing siRNA effects in T cells for adoptive immunotherapy. ACTA ACUST UNITED AC 2006; 10:461-7. [PMID: 16321811 DOI: 10.1080/10245330500233569] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Genetically manipulated T cells can be endowed with novel functions to obtain desired in vivo effects after adoptive transfer. This genetic approach is being used to introduce genes such as chimeric immunoreceptors and tumor-specific T cells are being evaluated in early phase clinic trials. However, the ability to alter the genetic programming of T cells also presents opportunities to remove unwanted T-cell functions in order to augment an anti-tumor effect or endow resistance such as to HIV infection. Specifically, the use of RNA interference (RNAi) to disrupt gene expression by targeting either the mRNA or the promoter, provides investigators with many new opportunities to genetically modify T cells that should prove useful in future applications of adoptive immunotherapy.
Collapse
Affiliation(s)
- Kevin Morris
- Division of Molecular Biology, Beckman Research Institute and City of Hope National Medical Center, Duarte, CA 90010-3000, USA
| | | | | | | | | | | |
Collapse
|
23
|
Morris KV, Rossi JJ. Lentivirus-Mediated RNA Interference Therapy for Human Immunodeficiency Virus Type 1 Infection. Hum Gene Ther 2006. [DOI: 10.1089/hum.2006.17.ft-209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
|
24
|
Morris KV, Rossi JJ. Lentivirus-Mediated RNA Interference Therapy for Human Immunodeficiency Virus Type 1 Infection. Hum Gene Ther 2006; 17:479-86. [PMID: 16716105 DOI: 10.1089/hum.2006.17.479] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
RNA interference (RNAi) is a natural mechanism by which small interfering RNAs (siRNAs) operate to specifically and potently downregulate the expression of a target gene. This downregulation has been demonstrated by targeting siRNAs to the mRNA (posttranscriptional gene silencing) as well as to the gene promoter, regulating gene expression epigenetically by transcriptional gene silencing. These observations significantly broaden the role RNA plays in the cell and suggest that siRNAs could prove to be a potent future therapeutic for the treatment of diseases such as human immunodeficiency virus type 1 (HIV-1) infection. The specificity and simplicity of design and the ability to express siRNAs from mammalian promoters make the use of siRNAs to target and suppress virtually any gene or gene promoter of interest a soon-to-be-realized technology. However, the delivery and stable expression of siRNAs to target cells remain an enigma that could be surmounted, at least regarding the treatment of HIV-1 infection, by the application of lentiviral vectors to deliver and express anti-HIV-1 siRNAs in target cells. This review focuses on the development, delivery, and potential therapeutic use of antiviral siRNAs in treating HIV-1.
Collapse
Affiliation(s)
- Kevin V Morris
- Department of Molecular and Experimental Medicine, Scripps Research Institute, La Jolla, CA 92037, USA
| | | |
Collapse
|
25
|
|
26
|
Erdmann V, Barciszewski J, Brosius J. Antiviral Applications of RNAi. Handb Exp Pharmacol 2006:105-16. [PMID: 16594613 PMCID: PMC7122589 DOI: 10.1007/3-540-27262-3_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
RNA interference is a natural mechanism by which small interfering (si)RNA operates to specifically and potently down-regulate the expression of a target gene. This down-regulation has been thought to predominantly function at the level of the messenger (m)RNA, post-transcriptional gene silencing (PTGS). Recently, the discovery that siRNAs can function to suppress a gene's expression at the level of transcription, i.e., transcriptional gene silencing (TGS), has created a major paradigm shift in mammalian RNAi. These recent findings significantly broaden the role RNA, specifically siRNAs and potentially microRNAs, plays in the regulation of gene expression as well as the breadth of potential siRNA target sites. Indeed, the specificity and simplicity of design makes the use of siRNAs to target and suppress virtually any gene or gene promoter of interest a realized technology. Furthermore, since siRNAs are a small nucleic acid reagent, they are unlikely to elicit an immune response, making them a theoretically good future therapeutic. This review will focus on the development, delivery, and potential therapeutic use of antiviral siRNAs in treating viral infections as well as emerging viral threats.
Collapse
Affiliation(s)
- Volker Erdmann
- Institute of Chemistry/Biochemistry, Free University Berlin, Thielallee 63, 14195 Berlin, Germany
| | - Jan Barciszewski
- Institute of Bioorganic Chemistry, Polish Academy of Scienes, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Jürgen Brosius
- Institute of Experimental Pathology, Molecular Neurobiology (ZMBE), University of Münster, Von-Esmarch-Str. 56, 48149 Münster, Germany
| |
Collapse
|
27
|
Galois L, Freyria AM, Herbage D, Mainard D. Ingénierie tissulaire du cartilage : état des lieux et perspectives. ACTA ACUST UNITED AC 2005; 53:590-8. [PMID: 16364811 DOI: 10.1016/j.patbio.2004.12.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2004] [Accepted: 12/09/2004] [Indexed: 11/24/2022]
Abstract
Lesions of the articular cartilage have a large variety of causes among which traumatic damage, osteoarthritis and osteochondritis dissecans are the most frequent. Returning damaged cartilage in articular joints back to a functionally normal state has been a major challenge for orthopaedic surgeons. This interest results in large part because cartilage defects cannot adequately heal themselves. Current techniques used in orthopaedic practice to repair cartilage give variable and unpredictable results. Bone marrow stimulation techniques such as abrasion arthroplasty, drilling and microfracture produce mostly fibrocartilage. Autologous osteochondral transplant systems (mosaicplasty) have shown encouraging results. Autologous chondrocyte transplantation has led to a hyaline articular cartilage repair but little is known about the predictability and reliability of the procedure. The rapidly emerging field of tissue engineering promises creation of viable substitutes for failing cartilage tissue. Current tissue engineering approaches are mainly focused on the restoration of pathologically altered tissue structure based on the transplantation of cells in combination with supportive matrices and molecules. Among natural and synthetic matrices, collagen and polysaccharidic biomaterials have been extensively used with promising results. Recently, interest has switched to the use of mesenchymal stem cells instead of chondrocytes. Tissue engineering offers the possibility to treat localised cartilage lesions. Genetic engineering techniques using genetically modified chondrocytes offer also the opportunity to treat diffuse cartilage lesions occurring in osteoarthritis or inflammatory joint diseases. Electroporation is specially a reliable and inexpensive technique that shares with electrochemotherapy an ability to target the chondrocytes despite the barrier effect of the extracellular matrix without viral vectors. The authors review recent research achievements and highlight the potential clinical applications of new technologies in the treatment of patients with cartilage injuries.
Collapse
Affiliation(s)
- L Galois
- Département de chirurgie orthopédique et traumatologique, hôpital central, 29, avenue Maréchal-de-Lattre-de-Tassigny, 54035 Nancy cedex, France.
| | | | | | | |
Collapse
|
28
|
Goila R, Banerjea AC. Sequence-specific cleavage of hepatitis X RNA in cis and trans by novel monotarget and multitarget hammerhead motif-containing ribozymes. Oligonucleotides 2005; 14:249-62. [PMID: 15665593 DOI: 10.1089/oli.2004.14.249] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
We constructed two monoribozymes and a diribozyme against the conserved region of the X RNA of hepatitis B virus (HBV). All the ribozymes (Rzs) possessed sequence-specific cleavage activities under standard and simulated physiologic conditions. Specific cleavage was also obtained when the same Rzs were placed in cis configuration with respect to X gene in multiple combinations. Rz-expressing cells were able to specifically interfere with the functional expression of X RNA and protein production in a liver-specific cell line HepG2. Potential applications of these novel Rzs are discussed.
Collapse
Affiliation(s)
- Ritu Goila
- Laboratory of Virology, National Institute of Immunology, New Delhi-110067, India
| | | |
Collapse
|
29
|
Abstract
The ability of small interfering RNA (siRNA) to mediate gene-specific post-transcriptional silencing in mammalian cells will undoubtedly revolutionise functional genomics, as well as drug target identification and validation. Furthermore, there is widespread excitement that siRNA itself might prove useful in the clinical setting. For those wishing to develop siRNA as a therapeutic agent, the most difficult obstacle to overcome will be delivery. Recently, several breakthroughs have highlighted viruses as excellent vehicles for siRNA delivery. Retroviruses, the transgene-delivery vector of choice for many experimental gene therapy studies, have been engineered to deliver and stably express therapeutic siRNA within cells, both in vitro and in vivo. These findings are important milestones for the development of siRNA as a gene therapy for treatment of viral infections, cancer, autoimmune syndromes and numerous genetic disorders. This review describes the development of retroviral siRNA vectors, highlights proof-of-concept experiments demonstrating their therapeutic efficacy and explores therapeutic targets particularly suitable for retroviral-mediated gene silencing.
Collapse
Affiliation(s)
- Eric Devroe
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School/Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| | | |
Collapse
|
30
|
|
31
|
Abstract
Nucleic acid delivery has many applications in basic science, biotechnology, agriculture, and medicine. One of the main applications is DNA or RNA delivery for gene therapy purposes. Gene therapy, an approach for treatment or prevention of diseases associated with defective gene expression, involves the insertion of a therapeutic gene into cells, followed by expression and production of the required proteins. This approach enables replacement of damaged genes or expression inhibition of undesired genes. Following two decades of research, there are two major methods for delivery of genes. The first method, considered the dominant approach, utilizes viral vectors and is generally an efficient tool of transfection. Attempts, however, to resolve drawbacks related with viral vectors (e.g., high risk of mutagenicity, immunogenicity, low production yield, limited gene size, etc.), led to the development of an alternative method, which makes use of non-viral vectors. This review describes non-viral gene delivery vectors, termed "self-assembled" systems, and are based on cationic molecules, which form spontaneous complexes with negatively charged nucleic acids. It introduces the most important cationic polymers used for gene delivery. A transition from in vitro to in vivo gene delivery is also presented, with an emphasis on the obstacles to achieve successful transfection in vivo.
Collapse
Affiliation(s)
- H. Eliyahu
- Department of Medicinal Chemistry and Natural Products, School of Pharmacy, Jerusalem, Israel
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University – Hadassah Medical School, Jerusalem, Israel
| | - Y. Barenholz
- Laboratory of Membrane and Liposome Research, Department of Biochemistry, The Hebrew University – Hadassah Medical School, Jerusalem, Israel
| | - A. J. Domb
- Department of Medicinal Chemistry and Natural Products, School of Pharmacy, Jerusalem, Israel
| |
Collapse
|
32
|
Conklin LD, McAninch RE, Schulz D, Kaluza GL, LeMaire SA, Coselli JS, Raizner AE, Sutton RE. HIV-based vectors and angiogenesis following rabbit hindlimb ischemia1. J Surg Res 2005; 123:55-66. [PMID: 15652951 DOI: 10.1016/j.jss.2004.06.010] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2004] [Indexed: 11/24/2022]
Abstract
BACKGROUND Numerous medical and surgical options exist for the treatment of vessel ischemia, which some patients fail or cannot tolerate. These investigations were designed to determine the effects of lentiviral-delivered vascular endothelial-derived growth factor (VEGF) and angiopoietin-2 (Ang-2) on collateralization in a rabbit model of hindlimb ischemia. MATERIALS AND METHODS Self-inactivating human immunodeficiency virus (HIV)-based vectors were constructed encoding VEGF or Ang-2, co-transfected with vesicular stomatitis virus glycoprotein (VSV G) into 293T cells, and vector supernatants (1 x 10(8) IU/ml after concentration) were harvested. New Zealand white rabbits had ligation of either the right or left external iliac artery and excision of the ipsilateral femoral artery. Ten days later, empty, VEGF, or VEGF+Ang-2 vector supernatant was injected intramuscularly (IM) into the ipsilateral thigh. Ankle systolic blood pressure (SBP) ratios were recorded and venous blood samples collected on postoperative days (POD) 10, 25, and 40. On POD 40, run-off angiography was performed to measure vessel collateralization. Capillary density was determined by thin sectioning of muscle. RESULTS A significant increase was noted in SBP in the VEGF-treated animals over time. Capillary density was not elevated despite significantly increased large vessel collateralization in rabbits receiving VEGF, which was counteracted by Ang-2. Antibodies against vector components were detected in exposed serum. CONCLUSIONS Arterial collateralization and SBP increased significantly following VEGF vector administration, which was reversed by the Ang-2 vector. Development of antibody against VSV G can limit repeated injections of vector. Future experiments will involve the addition of other pro-angiogenic factors, repeated vector administration, and alternative routes of vector delivery.
Collapse
Affiliation(s)
- Lori D Conklin
- The Michael E. DeBakey Department of Surgery, Division of Cardiothoracic Surgery, Baylor College of Medicine, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Pfeifer A, Hofmann A, Kessler B, Wolf E. Response to Whitelaw: Lentiviral transgenesis in livestock. Trends Biotechnol 2004. [DOI: 10.1016/j.tibtech.2004.02.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
34
|
Abstract
Genetic modification strategies have the potential to improve outcome following cell/organ transplantation. A unique opportunity in transplantation is that gene therapies need not be restricted to in vivo approaches and that ex vivo genetic modification of cell and/or organs can be of value. Improvements in vector design, production, and delivery should enhance transfection efficiency and optimize gene expression. Herein, we discuss potential modes of gene therapy, focusing on viral, liposome, or naked DNA-based systems for gene delivery. We suggest gene therapy targets taking into consideration the essential constituents of anti-allograft repertory. In addition to strategies that may have salutary effects in mitigating the threat of acute rejection, we suggest genetic strategies for minimizing ischemia/reperfusion injury as well as for the perennial problem of progressive functional loss of the transplanted organ. Data from pre-clinical transplant models support the idea that gene therapy may improve allograft function and survival. We are optimistic that gene therapy will be of clinical value in the near future in the management of recipients of allografts; we believe that genetic strategies would be essential for successful breaching of the formidable challenge of xenotransplantation.
Collapse
Affiliation(s)
- Dolca Thomas
- Division of Nephrology, Department of Medicine, Weill Medical College of Cornell University, New York Weill Cornell Center, 525 East 68th Street, New York, NY 10021, USA
| | | |
Collapse
|
35
|
Goujon C, Jarrosson-Wuilleme L, Bernaud J, Rigal D, Darlix JL, Cimarelli A. Heterologous human immunodeficiency virus type 1 lentiviral vectors packaging a simian immunodeficiency virus-derived genome display a specific postentry transduction defect in dendritic cells. J Virol 2003; 77:9295-304. [PMID: 12915545 PMCID: PMC187397 DOI: 10.1128/jvi.77.17.9295-9304.2003] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Heterologous lentiviral vectors (LVs) represent a way to address safety concerns in the field of gene therapy by decreasing the possibility of genetic recombination between vector and packaging constructs and the generation of replication-competent viruses. Using described LVs based on human immunodeficiency virus type 1 (HIV-1) and simian immunodeficiency virus MAC251 (SIV(MAC251)), we asked whether heterologous virion particles in which trans-acting factors belonged to HIV-1 and cis elements belonged to SIV(MAC251) (HIV-siv) would behave as parental homologous vectors in all cell types. To our surprise, we found that although the heterologous HIV-siv vector was as infectious as its homologous counterpart in most human cells, it was defective in the transduction of dendritic cells (DCs) and, to a lesser extent, macrophages. In DCs, the main postentry defect was observed in the formation of two-long-terminal-repeat circles, despite the fact that full-length proviral DNA was being synthesized and was associated with the nucleus. Taken together, our data suggest that heterologous HIV-siv vectors display a cell-dependent infectivity defect, most probably at a post-nuclear entry migration step. As homologous HIV and SIV vectors do transduce DCs, we believe that these results underscore the importance of a conserved interaction between cis elements and trans-acting viral factors that is lost or suboptimal in heterologous vectors and essential only in the transduction of certain cell types. For gene therapy purposes, these findings indicate that the cellular tropism of LVs can be modulated not only through the use of distinct envelope proteins or tissue-specific promoters but also through the specific combinatorial use of packaging and transfer vector constructs.
Collapse
Affiliation(s)
- Caroline Goujon
- INSERM U412, Ecole Normale Supérieure de Lyon. Etablissement Français du Sang, Lyon, France
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
OBJECTIVE Gene therapy is a rapidly evolving novel treatment for human disease. This review discusses the latest development in gene transfer technology and its potential use in the female reproductive tract. METHODS A comprehensive search using the MEDLINE database was performed to review current, innovative trends in gene transfer technology. In addition, articles on reproductive tract gene transfer were reviewed. CONCLUSION(S) Recent developments, such as the Human Genome Project, have generated great interest in the genetic basis of human health and disease. Gene therapy is a rapidly evolving field that uses gene transfer to treat disease. Ongoing research in the field focuses on improving vector technology to enable efficient in vivo gene transfer. Although multiple techniques for gene transfer have been described, no single technique can be used in all instances. The human female reproductive tract is easily accessible and can be readily transfected. In vivo gene transfer has resulted in successful alteration of implantation rates and has demonstrated potential for use in treatment of ovarian cancer.
Collapse
Affiliation(s)
- Gaurang S Daftary
- Department of Obstetrics and Gynecology, Yale University School of Medicine, New Haven, Connecticut 06520, USA
| | | |
Collapse
|
37
|
Segall HI, Yoo E, Sutton RE. Characterization and detection of artificial replication-competent lentivirus of altered host range. Mol Ther 2003; 8:118-29. [PMID: 12842435 DOI: 10.1016/s1525-0016(03)00134-5] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
Abstract
Replication-competent lentivirus (RCL) may be generated during the production phase or subsequently after introduction of a lentiviral vector into target cells, potentially by homologous or nonhomologous recombination. Because most gene transfer of HIV-based vectors involves the use of high-titer vesicular stomatitis virus (VSV) G-pseudotyped particles, one particular concern would be the generation of an RCL of altered host range, i.e., one that has incorporated the VSV G envelope in cis configuration. We report here on the artificial generation and properties of such a virus, including its detection after biological amplification. Viral spread, beginning with a very low inoculum, takes several weeks in culture and is characterized by "autoinfection," resulting in multiple proviral copies per cell, higher levels of viral gene expression, and eventual cell death. After this initial amplification step, the RCL is easily detectable by standard p24 assay or by "marker-rescue" assay. For the latter, a 293T-based cell line that has an integrated replication-defective provirus encoding alkaline phosphatase (AP) was used and mobilization of AP-containing virus was detected by transduction of naïve cells. Replication-defective virus was not amplified nor detected, demonstrating assay specificity. These results suggest that these artificial RCLs of broad host range have slightly different biological properties compared to wild-type HIV but still spread and are readily detectable.
Collapse
Affiliation(s)
- Harry I Segall
- Center for Cell and Gene Therapy, Department of Molecular Virology and Microbiology, Baylor College of Medicine, One Baylor Plaza, Houston, Texas 77030, USA
| | | | | |
Collapse
|
38
|
Baillou C, Simon A, Leclercq V, Azar N, Rosenzwajg M, Herson S, Klatzmann D, Lemoine FM. Highly active antiretroviral therapy corrects hematopoiesis in HIV-1 infected patients: interest for peripheral blood stem cell-based gene therapy. AIDS 2003; 17:563-74. [PMID: 12598777 DOI: 10.1097/00002030-200303070-00012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES To study, in asymptomatic HIV-1-infected (HIV+) patients, whether peripheral blood hematopoietic progenitor/stem cells (PBPC) mobilized by granulocyte colony stimulating factor (G-CSF), can be used as a source of cells for retroviral gene therapy. DESIGN PBPC from two groups of HIV+ patients (treated or untreated by highly active antiretroviral therapy) and from seronegative donors were mobilized with G-CSF. METHODS PBPC collected by leukapheresis were enriched for CD34 cells, immunophenotypically and functionally characterized, cultured and infected with retroviral vectors. HIV proviral integration was studied on fresh and cultured cells. RESULTS G-CSF moderately and transiently increased the viral load in untreated patients only, and induced in both groups of HIV+ patients mobilization of percentages and numbers of CD34 cells comparable to those of seronegative volunteers. The most immature CD34 cell subset, the clonogenic progenitor and long-term culture initiating cells were significantly decreased in leukapheresis products and CD34-enriched fractions from untreated HIV+ patients but not in those from treated HIV+ patients. Cell cycle activation and growth factor responses of CD34 cells from both groups of HIV+ patients were not different from those of the control group. Culture and retroviral infection of CD34 cells from HIV+ patients did not enhance HIV replication, and yielded transduction levels similar to those obtained using CD34 cells from seronegative donors. CONCLUSIONS G-CSF-mobilized PBPC can be safely used for HIV retroviral gene therapy in asymptomatic treated patients while highly active antiretroviral therapy would control the G-CSF-induced increase in viral load and correct the defective hematopoiesis observed in untreated patients, without inhibiting the retroviral transduction of PBPC.
Collapse
Affiliation(s)
- Claude Baillou
- Biologie et Thérapeutique des Pathologies Immunitaires, UMR CNRS Hospital Pitié Salpetriére, Paris, France
| | | | | | | | | | | | | | | |
Collapse
|
39
|
Bovia F, Salmon P, Matthes T, Kvell K, Nguyen TH, Werner-Favre C, Barnet M, Nagy M, Leuba F, Arrighi JF, Piguet V, Trono D, Zubler RH. Efficient transduction of primary human B lymphocytes and nondividing myeloma B cells with HIV-1-derived lentiviral vectors. Blood 2003; 101:1727-33. [PMID: 12406892 DOI: 10.1182/blood-2001-12-0249] [Citation(s) in RCA: 60] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
We studied the transduction of primary human B lymphocytes and myeloma cells with lentiviral vectors. In peripheral blood B cells that had been activated with helper T cells (murine thymoma EL-4 B5) and cytokines, multiply attenuated HIV-1-derived vectors pseudotyped with vesicular stomatitis virus (VSV) G-envelope protein achieved the expression of green fluorescence protein (GFP) in 27% +/- 12% (mean +/- 1 SD; median, 27%) of B cells in different experiments. When compared in parallel cultures, the transducibility of B cells from different donors exhibited little variation. The human cytomegalovirus (CMV) promoter gave 4- to 6-fold higher GFP expression than did the human elongation factor-1alpha promoter. A murine retroviral vector pseudotyped with VSV G protein proved inefficient even in mitotically active primary B cells. B cells freshly stimulated with Epstein-Barr virus were also transducible by HIV vectors (24% +/- 9%), but B cells activated with CD40 ligand and cytokines resisted transduction. Thus, different culture systems gave different results. Freshly isolated, nondividing myeloma cells were efficiently transduced by HIV vectors; for 6 myelomas the range was 14% to 77% (median, 28%) GFP(+) cells. HIV vectors with a mutant integrase led to no significant GFP signal in primary B or myeloma cells, suggesting that vector integration was required for high transduction. In conclusion, HIV vectors are promising tools for studies of gene functions in primary human B cells and myeloma cells for the purposes of research and the development of gene therapies.
Collapse
MESH Headings
- B-Lymphocytes/drug effects
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- B-Lymphocytes/virology
- CD40 Ligand/pharmacology
- Cells, Cultured/drug effects
- Cells, Cultured/immunology
- Cells, Cultured/metabolism
- Cells, Cultured/virology
- Cytokines/pharmacology
- Cytomegalovirus/genetics
- Defective Viruses/genetics
- Defective Viruses/physiology
- Genes, Reporter
- Genes, gag
- Genes, pol
- Genes, rev
- Genes, tat
- Genetic Vectors/genetics
- Genetic Vectors/physiology
- Green Fluorescent Proteins
- HIV Integrase/deficiency
- HIV Integrase/genetics
- HIV Integrase/physiology
- HIV-1/genetics
- HIV-1/physiology
- Herpesvirus 4, Human/physiology
- Humans
- Leukemia Virus, Murine/genetics
- Leukemia Virus, Murine/physiology
- Luminescent Proteins/biosynthesis
- Luminescent Proteins/genetics
- Lymphocyte Activation
- Multiple Myeloma/pathology
- Neoplastic Stem Cells/drug effects
- Neoplastic Stem Cells/metabolism
- Neoplastic Stem Cells/virology
- Peptide Elongation Factor 1/genetics
- Peptide Elongation Factor 1/physiology
- Promoter Regions, Genetic
- Recombinant Fusion Proteins/biosynthesis
- T-Lymphocytes, Helper-Inducer/immunology
- Tumor Cells, Cultured/drug effects
- Tumor Cells, Cultured/immunology
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/virology
- Vesicular stomatitis Indiana virus/genetics
Collapse
Affiliation(s)
- Fabrice Bovia
- Division of Hematology, Department of Medicine, University Hospital, Geneva, Switzerland
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Virus-based vectors for gene expression in mammalian cells: Lentiviruses. ACTA ACUST UNITED AC 2003. [DOI: 10.1016/s0167-7306(03)38015-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
|
41
|
Roesler J, Brenner S, Bukovsky AA, Whiting-Theobald N, Dull T, Kelly M, Civin CI, Malech HL. Third-generation, self-inactivating gp91(phox) lentivector corrects the oxidase defect in NOD/SCID mouse-repopulating peripheral blood-mobilized CD34+ cells from patients with X-linked chronic granulomatous disease. Blood 2002; 100:4381-90. [PMID: 12393624 DOI: 10.1182/blood-2001-12-0165] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
HIV-1-derived lentivectors are promising for gene transfer into hematopoietic stem cells but require preclinical in vivo evaluation relevant to specific human diseases. Nonobese diabetic/severe combined immunodeficient (NOD/SCID) mice accept human hematopoietic stem cell grafts, providing a unique opportunity for in vivo evaluation of therapies targeting human hematopoietic diseases. We demonstrate for the first time that hematopoietic stem cells from patients with X-linked chronic granulomatous disease (X-CGD) give rise to X-CGD-phenotype neutrophils in the NOD/SCID model that can be corrected using VSV-G-pseudotyped, 3rd-generation, self-inactivating (SIN) lentivector encoding gp91(phox). We transduced X-CGD patient-mobilized CD34(+) peripheral blood stem cells (CD34(+)PBSCs) with lentivector-gp91(phox) or amphotropic oncoretrovirus MFGS-gp91(phox) and evaluated correction ex vivo and in vivo in NOD/SCID mice. Only lentivector transduced CD34(+)PBSCs under ex vivo conditions nonpermissive for cell division, but both vectors performed best under conditions permissive for proliferation (multiple growth factors). Under the latter conditions, lentivector and MFGS achieved significant ex vivo correction of X-CGD CD34(+)PBSCs (18% and 54% of cells expressing gp91(phox), associated with 53% and 163% of normal superoxide production, respectively). However, lentivector, but not MFGS, achieved significant correction of human X-CGD neutrophils arising in vivo in NOD/SCID mice that underwent transplantation (20% and 2.4%, respectively). Thus, 3rd-generation SIN lentivector-gp91(phox) performs well as assessed in human X-CGD neutrophils differentiating in vivo, and our studies suggest that the NOD/SCID model is generally applicable for in vivo study of therapies evaluated in human blood cells expressing a specific disease phenotype.
Collapse
Affiliation(s)
- Joachim Roesler
- Laboratory of Host Defenses, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892-1886, USA
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Liang C, Hu J, Russell RS, Wainberg MA. Translation of Pr55(gag) augments packaging of human immunodeficiency virus type 1 RNA in a cis-acting manner. AIDS Res Hum Retroviruses 2002; 18:1117-26. [PMID: 12396450 DOI: 10.1089/088922202320567851] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The full-length RNA of human immunodeficiency virus type 1 (HIV-1) serves both as a messenger (mRNA) to direct the translation of Pr55(gag) proteins and as genomic or viral particle RNA (vpRNA) to be packaged into virions. In this study, we have assessed a putative cis-acting effect of Pr55(gag) translation on HIV-1 RNA packaging. To pursue this subject, we have measured the relative competence of two distinct types of HIV-1 RNA for being packaged by virus particles under conditions in which only one of them is permissive for production of Pr55(gag). Not surprisingly, wild-type BH10 RNA was packaged at far higher efficiency than that associated with mutant viral RNA that was deleted of RNA packaging signals and incapable of Pr55(gag) production. However, when production of Pr55(gag) was eliminated from the wild-type BH10 viral RNA by insertion of stop codons either in matrix (MA) or in capsid (CA) sequences, regardless of retention of wild-type RNA packaging signals, these Pr55(gag)-deficient viral RNAs were packaged at low levels similar to those observed with viral RNA species that lack RNA packaging signals and are capable of Pr55(gag) generation. Moreover, loss of Pr55(gag) production did not affect stability of the relevant viral RNA; this observation rules out the possibility that lowered packaging efficiency associated with Pr55(gag)-deficient HIV-1 RNA is a result of reduced RNA stability. Taken together, our data demonstrate that cis translation of Pr55(gag) is needed for efficient packaging of HIV-1 RNA.
Collapse
Affiliation(s)
- Chen Liang
- McGill AIDS Centre, Lady Davis Institute, Jewish General Hospital, Montreal, Quebec, Canada H3T 1E2.
| | | | | | | |
Collapse
|
43
|
Alesci S, Chrousos GP, Pacak K. Genomic medicine: exploring the basis of a new approach to endocrine hypertension. Ann N Y Acad Sci 2002; 970:177-92. [PMID: 12381553 DOI: 10.1111/j.1749-6632.2002.tb04424.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Recent improvements in defining the molecular basis of disease have encouraged scientists worldwide to develop new therapeutic strategies based on engineered genes and cells. Genomic medicine has the potential to revolutionize diagnosis and therapy of a variety of human diseases, including endocrine disorders. Hypertension is the presenting feature of some of these disorders, such as congenital adrenal diseases, and adrenal and pituitary tumors. Preclinical data indicate that gene transfer to both the adrenal gland and the pituitary is not only feasible but also quite efficient. Research in this field is only in its infancy, but with the ever-increasing advances in DNA technologies, genomic therapies for endocrine hypertension may become available within the next few decades.
Collapse
Affiliation(s)
- Salvatore Alesci
- Pediatric and Reproductive Endocrinology Branch, National Institute of Child Health and Human Development/NIH, Building 10, Room 9D42, 10 Center Drive MSC 1583, Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
44
|
Evans ME, Lesnaw JA. Infection control for gene therapy: a busy physician's primer. Clin Infect Dis 2002; 35:597-605. [PMID: 12173136 DOI: 10.1086/342194] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2002] [Revised: 05/16/2002] [Indexed: 01/26/2023] Open
Abstract
Gene therapy is being studied for the treatment of a wide variety of acquired and inherited disorders. Retroviruses, adenoviruses, poxviruses, adeno-associated virus, herpesviruses, and others are being engineered to serve as gene therapy vectors and are being administered to patients in a clinical setting. Infection control professionals will be asked to evaluate the use and safety of these agents in their clinics and hospitals. This review summarizes key aspects of the biotechnology and the vectors involved in gene therapy and makes recommendations for infection control.
Collapse
Affiliation(s)
- Martin E Evans
- Division of Infectious Diseases, Department of Internal Medicine, University of Kentucky, Lexington, USA.
| | | |
Collapse
|
45
|
Ijichi S, Ijichi N, Osame M, Hall WW. In vivo induction of human immunodeficiency virus type 1 entry into nucleus-free cells by CD4 gene transfer to hematopoietic stem cells: a hypothetical possible strategy for therapeutic intervention. Med Hypotheses 2002; 59:24-34. [PMID: 12160677 DOI: 10.1016/s0306-9877(02)00194-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
As a useful alternative to employing soluble CD4 to inhibit binding of human immunodeficiency virus type 1 (HIV-1) to target cells, the introduction of CD4-bearing erythrocyte has been proposed by two study groups (see Refs. (5,6)). Prominently, Nicolau and colleagues demonstrated that the electroinserted CD4 molecules in the membranes of erythrocytes are capable of mediating HIV-1 entry. The implications of the studies are that inactivation of the integration-dependent retrovirus by the facilitation of entry into the nucleus-free cells, referred to as 'fake host trap' or 'host cell decoy', may be a possible therapeutic approach. Here we expand this concept to include genetic modification of autologous hematopoietic stem cells and review the relevant theoretical basis. Effective application of molecular technologies to induce partial replacement of hematopoiesis may be critical for this strategy.
Collapse
Affiliation(s)
- S Ijichi
- Nagahama Shinryojyo, Shimokoshiki-mura, Satsuma-gun, Kagoshima, Japan.
| | | | | | | |
Collapse
|
46
|
Affiliation(s)
- L E Ailles
- Laboratory for Gene Transfer and Therapy, Institute for Cancer Research and Treatment, University of Torino Medical School, Candiolo, Torino, Italy
| | | |
Collapse
|
47
|
Abstract
OBJECTIVE To review the literature on the principles of gene therapy and its potential application in reproductive medicine. DESIGN Literature review. SETTING Gene therapy involves transfer of genetic material to target cells using a delivery system, or vector. Attention has primarily focused on viral vectors. Significant problems remain to be overcome including low efficacy of gene transfer, the transient expression of some vectors, safety issues with modified adenoviruses and retroviruses, and ethical concerns. If these issues can be resolved, gene therapy will be applicable to an increasing spectrum of single and multiple gene disorders, as the Human Genome Project data are analyzed, and the genetic component of human disease becomes better understood. Gynecologic gene therapy has advanced to human clinical trials for ovarian carcinoma, and shows potential for the treatment of uterine leiomyomata. Obstetric applications of gene therapy, including fetal gene therapy, remain more distant goals. CONCLUSION(S) Concerns about the safety of human gene therapy research are being actively addressed, and remarkable progress in improving DNA transfer has been made. The first treatment success for a genetic disease (severe combined immunodeficiency disease) has been achieved, and ongoing research efforts will eventually yield clinical applications in many spheres of reproductive medicine.
Collapse
Affiliation(s)
- John M Stribley
- Department of Obstetrics and Gynecology, Reproductive Sciences Program, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | |
Collapse
|
48
|
Abstract
Gene therapy can be broadly defined as the transfer of genetic material to cure a disease or at least to improve the clinical status of a patient. One of the basic concepts of gene therapy is to transform viruses into genetic shuttles, which will deliver the gene of interest into the target cells. Based on the nature of the viral genome, these gene therapy vectors can be divided into RNA and DNA viral vectors. The majority of RNA virus-based vectors have been derived from simple retroviruses like murine leukemia virus. A major shortcoming of these vectors is that they are not able to transduce nondividing cells. This problem may be overcome by the use of novel retroviral vectors derived from lentiviruses, such as human immunodeficiency virus (HIV). The most commonly used DNA virus vectors are based on adenoviruses and adeno-associated viruses. Although the available vector systems are able to deliver genes in vivo into cells, the ideal delivery vehicle has not been found. Thus, the present viral vectors should be used only with great caution in human beings and further progress in vector development is necessary.
Collapse
Affiliation(s)
- A Pfeifer
- The Salk Institute, La Jolla, California 92037, USA.
| | | |
Collapse
|
49
|
Mautino MR, Morgan RA. Gene therapy of HIV-1 infection using lentiviral vectors expressing anti-HIV-1 genes. AIDS Patient Care STDS 2002; 16:11-26. [PMID: 11839215 DOI: 10.1089/108729102753429361] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The use of vectors based on primate lentiviruses for gene therapy of human immunodeficiency virus type 1 (HIV-1) infection has many potential advantages over the previous murine retroviral vectors used for delivery of genes that inhibit replication of HIV-1. First, lentiviral vectors have the ability to transduce dividing and nondividing cells that constitute the targets of HIV-1 infection such as resting T cells, dendritic cells, and macrophages. Lentiviral vectors can also transfer genes to hematopoietic stem cells with a superior gene transfer efficiency and without affecting the repopulating capacity of these cells. Second, these vectors could be potentially mobilized in vivo by the wild-type virus to secondary target cells, thus expanding the protection to previously untransduced cells. And finally, lentiviral vector backbones have the ability to block HIV-1 replication by several mechanisms that include sequestration of the regulatory proteins Tat and Rev, competition for packaging into virions, and by inhibition of reverse transcription in heterodimeric virions with possible generation of nonfunctional recombinants between the vector and viral genomes. The inhibitory ability of lentiviral vectors can be further increased by expression of anti-HIV-1 genes. In this case, the lentiviral vector packaging system has to be modified to become resistant to the anti-HIV-1 genes expressed by the vector in order to avoid self-inhibition of the vector packaging system during vector production. This review focuses on the use of lentiviral vectors as the main agents to mediate inhibition of HIV-1 replication and discusses the different genetic intervention strategies for gene therapy of HIV-1 infection.
Collapse
Affiliation(s)
- Mario R Mautino
- Medical Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland, USA
| | | |
Collapse
|
50
|
Okimoto T, Friedmann T, Miyanohara A. VSV-G envelope glycoprotein forms complexes with plasmid DNA and MLV retrovirus-like particles in cell-free conditions and enhances DNA transfection. Mol Ther 2001; 4:232-8. [PMID: 11545614 DOI: 10.1006/mthe.2001.0443] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We have previously shown that vesicles containing the spike glycoprotein of the vesicular stomatitis virus (VSV-G) can associate efficiently with immature, non-infectious, envelope-deficient retrovirus-like particles assembled by packaging cells to produce infectious, pseudotyped viruses in cell-free conditions in vitro. We have also previously reported that VSV-G can enhance DNA lipofection efficiency by interacting with liposomes to form fusogenic, serum-stable liposomes with enhanced transfection properties. Here, we report that VSV-G can form a complex directly with naked plasmid DNA in the absence of a lipofection reagent and can thereby enhance the transfection efficiency of the naked plasmid vector. Sucrose gradient sedimentation analysis demonstrated that VSV-G can also associate with plasmid DNA and murine leukemia virus (MLV) gag-pol particles to form ternary complexes that co-sediment with high DNA transfecting activity. The increased transfection efficiency with VSV-G was dependent on the presence of the polycation (Polybrene) in the culture medium during transfection. Enhanced transfection was abolished by a neutralizing antibody to VSV-G. These results may be useful in the study of retrovirus assembly, in the further design of hybrid DNA-based retrovirus-like vectors, and in the full in vitro, cell-free assembly of infectious virus-like particles from component parts.
Collapse
Affiliation(s)
- T Okimoto
- Program in Human Gene Therapy, Center for Molecular Genetics, UCSD School of Medicine, La Jolla, CA 92093-0692, USA
| | | | | |
Collapse
|