1
|
Khan PM, Roy K. QSPR modelling for investigation of different properties of aminoglycoside-derived polymers using 2D descriptors. SAR AND QSAR IN ENVIRONMENTAL RESEARCH 2021; 32:595-614. [PMID: 34148451 DOI: 10.1080/1062936x.2021.1939150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 06/02/2021] [Indexed: 06/12/2023]
Abstract
The quantitative structure-property relationship (QSPR) method is commonly used to predict different physicochemical characteristics of interest of chemical compounds with an objective to accelerate the process of design and development of novel chemical compounds in the biotechnology and healthcare industries. In the present report, we have employed a QSPR approach to predict the different properties of the aminoglycoside-derived polymers (i.e. polymer DNA binding and aminoglycoside-derived polymers mediated transgene expression). The final QSPR models were obtained using the partial least squares (PLS) regression approach using only specific categories of two-dimensional descriptors and subsequently evaluated considering different internationally accepted validation metrics. The proposed models are robust and non-random, demonstrating excellent predictive ability using test set compounds. We have also developed different kinds of consensus models using several validated individual models to improve the prediction quality for external set compounds. The present findings provide new insight for exploring the design of an aminoglycoside-derived polymer library based on different identified physicochemical properties as well as predict their property before their synthesis.
Collapse
Affiliation(s)
- P M Khan
- Department of Pharmacoinformatics, National Institute of Pharmaceutical Educational and Research (NIPER), Kolkata, India
| | - K Roy
- Drug Theoretics and Cheminformatics Laboratory, Division of Medicinal and Pharmaceutical Chemistry, Department of Pharmaceutical Technology, Jadavpur University, Kolkata, India
| |
Collapse
|
2
|
Bravo-Anaya L, Garbay B, Nando-Rodríguez J, Carvajal Ramos F, Ibarboure E, Bathany K, Xia Y, Rosselgong J, Joucla G, Garanger E, Lecommandoux S. Nucleic acids complexation with cationic elastin-like polypeptides: Stoichiometry and stability of nano-assemblies. J Colloid Interface Sci 2019; 557:777-792. [DOI: 10.1016/j.jcis.2019.09.054] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Revised: 09/13/2019] [Accepted: 09/16/2019] [Indexed: 02/01/2023]
|
3
|
Guo R, Wang H, Cui J, Wang G, Li W, Hu JF. Inhibition of HIV-1 Viral Infection by an Engineered CRISPR Csy4 RNA Endoribonuclease. PLoS One 2015; 10:e0141335. [PMID: 26495836 PMCID: PMC4619743 DOI: 10.1371/journal.pone.0141335] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2015] [Accepted: 10/06/2015] [Indexed: 01/13/2023] Open
Abstract
The bacterial defense system CRISPR (clustered regularly interspaced short palindromic repeats) has been explored as a powerful tool to edit genomic elements. In this study, we test the potential of CRISPR Csy4 RNA endoribonuclease for targeting HIV-1. We fused human codon-optimized Csy4 endoribonuclease with VPR, a HIV-1 viral preintegration complex protein. An HIV-1 cell model was modified to allow quantitative detection of active virus production. We found that the trans-expressing VPR-Csy4 almost completely blocked viral infection in two target cell lines (SupT1, Ghost). In the MAGI cell assay, where the HIV-1 LTR β-galactosidase is expressed under the control of the tat gene from an integrated provirus, VPR-Csy4 significantly blocked the activity of the provirus-activated HIV-1 reporter. This proof-of-concept study demonstrates that Csy4 endoribonuclease is a promising tool that could be tailored further to target HIV-1.
Collapse
Affiliation(s)
- Rui Guo
- Stem Cell and Cancer Center, First Hospital, Jilin University, Changchun, China
- Stanford University Medical School, Palo Alto, California, 94304, United States of America
| | - Hong Wang
- Stem Cell and Cancer Center, First Hospital, Jilin University, Changchun, China
- Stanford University Medical School, Palo Alto, California, 94304, United States of America
| | - Jiuwei Cui
- Stem Cell and Cancer Center, First Hospital, Jilin University, Changchun, China
| | - Guanjun Wang
- Stem Cell and Cancer Center, First Hospital, Jilin University, Changchun, China
| | - Wei Li
- Stem Cell and Cancer Center, First Hospital, Jilin University, Changchun, China
- * E-mail: (J-FH); (WL)
| | - Ji-Fan Hu
- Stem Cell and Cancer Center, First Hospital, Jilin University, Changchun, China
- Stanford University Medical School, Palo Alto, California, 94304, United States of America
- * E-mail: (J-FH); (WL)
| |
Collapse
|
4
|
|
5
|
Huelsmann PM, Hofmann AD, Knoepfel SA, Popp J, Rauch P, Di Giallonardo F, Danke C, Gueckel E, Schambach A, Wolff H, Metzner KJ, Berens C. A suicide gene approach using the human pro-apoptotic protein tBid inhibits HIV-1 replication. BMC Biotechnol 2011; 11:4. [PMID: 21223573 PMCID: PMC3224247 DOI: 10.1186/1472-6750-11-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2010] [Accepted: 01/11/2011] [Indexed: 12/15/2022] Open
Abstract
Background Regulated expression of suicide genes is a powerful tool to eliminate specific subsets of cells and will find widespread usage in both basic and applied science. A promising example is the specific elimination of human immunodeficiency virus type 1 (HIV-1) infected cells by LTR-driven suicide genes. The success of this approach, however, depends on a fast and effective suicide gene, which is expressed exclusively in HIV-1 infected cells. These preconditions have not yet been completely fulfilled and, thus, success of suicide approaches has been limited so far. We tested truncated Bid (tBid), a human pro-apoptotic protein that induces apoptosis very rapidly and efficiently, as suicide gene for gene therapy against HIV-1 infection. Results When tBid was introduced into the HIV-1 LTR-based, Tat- and Rev-dependent transgene expression vector pLRed(INS)2R, very efficient induction of apoptosis was observed within 24 hours, but only in the presence of both HIV-1 regulatory proteins Tat and Rev. Induction of apoptosis was not observed in their absence. Cells containing this vector rapidly died when transfected with plasmids containing full-length viral genomic DNA, completely eliminating the chance for HIV-1 replication. Viral replication was also strongly reduced when cells were infected with HIV-1 particles. Conclusions This suicide vector has the potential to establish a safe and effective gene therapy approach to exclusively eliminate HIV-1 infected cells before infectious virus particles are released.
Collapse
Affiliation(s)
- Peter M Huelsmann
- University of Erlangen-Nuremberg, Institute of Clinical and Molecular Virology, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Huang FW, Yang J, Feng J, Zhuo RX, Zhang XZ. Design of hepatocyte-targeted gene transfer vector and its in vitro transfer of tumor-suppressor p53 gene. ACTA ACUST UNITED AC 2011. [DOI: 10.1039/c0jm03618h] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
|
7
|
Buchholz F, Hauber J. In vitro evolution and analysis of HIV-1 LTR-specific recombinases. Methods 2010; 53:102-9. [PMID: 20600935 DOI: 10.1016/j.ymeth.2010.06.014] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2010] [Revised: 06/14/2010] [Accepted: 06/18/2010] [Indexed: 12/01/2022] Open
Abstract
Current antiretroviral therapies would greatly benefit from the concurrent removal of integrated HIV-1 proviral DNA from the patient's cells. In this review, we describe an experimental strategy that allowed the engineering and functional analysis of a HIV-1 LTR-specific recombinase (Tre-recombinase). We furthermore provide protocols that are utilized for the investigation of Tre's antiretroviral activity in infected tissue cultures as well as in infected humanized Rag2(-/-)γc(-/-) mice.
Collapse
Affiliation(s)
- Frank Buchholz
- Max-Planck-Institute for Molecular Cell Biology and Genetics, Pfotenhauerstrasse 108, 01307 Dresden, Germany
| | | |
Collapse
|
8
|
Oishi J, Jung J, Tsuchiya A, Toita R, Kang JH, Mori T, Niidome T, Tanizawa K, Kuroda S, Katayama Y. A gene-delivery system specific for hepatoma cells and an intracellular kinase signal based on human liver-specific bionanocapsules and signal-responsive artificial polymer. Int J Pharm 2010; 396:174-8. [PMID: 20558262 DOI: 10.1016/j.ijpharm.2010.06.012] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2010] [Revised: 05/28/2010] [Accepted: 06/09/2010] [Indexed: 01/16/2023]
Abstract
Recently, our group has proposed a novel gene-regulation system responding to cAMP-dependent protein kinase (PKA) that has been applied to living cells. In this study, human liver-specific bionanocapsules (BNCs) are used as a gene-delivery system to increase transfection efficiency and to target specific cell types. BNCs can efficiently deliver a target gene to human hepatocytes and hepatoma cells in vitro or in vivo. The combination of a signal-responsive gene-delivery system with BNCs led to an increase in the transfection efficiency and selectivity for hepatoma cells. Expression from the delivered gene was identified from PKA-activated hepatoma cells (HepG2), but not from colon tumor cells (WiDr). These results show that the combination of a gene-regulation system responding to an intracellular signal with BNC can be used for the selective treatment of human hepatoma cells.
Collapse
Affiliation(s)
- Jun Oishi
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka, Nishi-Ku, Fukuoka 819-0395, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
9
|
Abstract
Therapeutic application of siRNA requires delivery to the correct
intracellular location, to interact with the RNAi machinery within the
target cell, within the target tissue responsible for the pathology. Each of
these levels of targeting poses a significant barrier. To overcome these
barriers several strategies have been developed, such as chemical
modifications of siRNA, viral nucleic acid delivery systems, and nonviral
nucleic acid delivery systems. Here, we discuss progress that has been made
to improve targeted delivery of siRNA in vivo for each of these strategies.
Collapse
Affiliation(s)
- Sabrina Oliveira
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Room Z735A,
PO Box 80082, 3508 Utrecht, The Netherlands
| | - Gert Storm
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Room Z735A,
PO Box 80082, 3508 Utrecht, The Netherlands
| | - Raymond M. Schiffelers
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Room Z735A,
PO Box 80082, 3508 Utrecht, The Netherlands
- * Raymond M. Schiffelers:
| |
Collapse
|
10
|
|
11
|
Abstract
Formation of small interfering RNA (siRNA) occurs in two steps involving binding of the RNA nucleases to a large double‐stranded RNA (dsRNA) and its cleavage into fragments called siRNA. In the second step, these siRNAs join a multinuclease complex, which degrades the homologous single‐stranded mRNAs. The delivery of siRNA involves viral‐ and non‐viral‐mediated delivery systems; the approaches for chemical modifications have also been developed. It has various therapeutic applications for disorders like cardiovascular diseases, central nervous system (CNS) disorders, cancer, human immunodeficiency virus (HIV), hepatic disorders, etc. The present review gives an overview of the applications of siRNA and their potential for treating many hitherto untreatable diseases.
Collapse
Affiliation(s)
- Bhoomika R Goyal
- Institute of Pharmacy, Nirma University of Science and Technology, Ahmedabad 382 481, Gujarat, India.
| | | | | | | |
Collapse
|
12
|
Asai D, Kuramoto M, Shoji Y, Kang JH, Kodama KB, Kawamura K, Mori T, Miyoshi H, Niidome T, Nakashima H, Katayama Y. Specific transgene expression in HIV-infected cells using protease-cleavable transcription regulator. J Control Release 2009; 141:52-61. [PMID: 19733602 DOI: 10.1016/j.jconrel.2009.08.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2009] [Revised: 08/18/2009] [Accepted: 08/23/2009] [Indexed: 11/28/2022]
Abstract
Gene therapy is a promising strategy for the treatment of HIV infection, but cell specificity remains an issue. Recently we have developed a new concept for a drug or gene delivery system responding to cellular signals (D-RECS) to achieve cell-specific transgene expression using a non-viral polymer-based vehicle. According to this concept, intracellular signaling enzymes, which are activated specifically in target cells, are used to trigger transgene expression. We previously applied this concept to HIV-1 protease and showed that the recombinant protease could act as a suitable signal. Here we further developed this system to achieve highly specific transgene expression in HIV-infected cells. We prepared a polymeric gene regulator grafted with a cationic peptide containing the HIV-Tat peptide via a specific substrate for HIV-1 protease. The regulator formed a stable polyplex with the transgene, suppressing its transcription. HIV-1 protease cleaved the peptide and released the transgene, which was consequently expressed specifically in activated HIV-infected cells, but remained unreleased and inactive in uninfected cells. The validity of this approach was further confirmed by applying it to the CVB1 2A protease of coxsackievirus (Picornaviridae family). This strategy should be widely applicable for specific expression of a variety of therapeutic genes in virus-infected cells.
Collapse
Affiliation(s)
- Daisuke Asai
- Department of Microbiology, St. Marianna University School of Medicine, Kawasaki 216-8511, Japan.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Kawamura K, Kuramoto M, Mori T, Toita R, Oishi J, Sato Y, Kang JH, Asai D, Niidome T, Katayama Y. Molecular mechanism of caspase-3-induced gene expression of polyplexes formed from polycations grafted with cationic substrate peptides. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2009; 20:967-80. [PMID: 19454163 DOI: 10.1163/156856209x444376] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
We previously reported a novel disease-site-specific gene targeting system that can release plasmid DNA (pDNA) from polymeric carriers responding to abnormally activated signal proteins in disease cells. In this study, the molecular mechanism of the gene targeting system responding to Caspase-3 activity was studied in detail. The polymeric carrier used was composed of a neutral main chain polymer and a grafted oligocationic peptide which contains the substrate sequence of Caspase-3. The polyplex formed from the polymeric carrier and pDNA was stable in physiological saline solution and protected from access of RNA polymerase and the transcriptional factors. These results indicate that the polyplex adopts a core-shell-like structure with a polyion complex core surrounded by neutral main chain polymers. In spite of the inert character of the polyplex to transcription, the polyplex afforded the access of Caspase-3 to the substrate peptide because the electrostatic interaction between each peptide and DNA is essentially weak. After the Caspase-3 reaction, the polyplex was weakened and then became available as a template for transcription.
Collapse
Affiliation(s)
- Kenji Kawamura
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, 744 Motooka Nishi-ku, Fukuoka 819-0395, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Tsen SWD, Wu CY, Meneshian A, Pai SI, Hung CF, Wu TC. Femtosecond laser treatment enhances DNA transfection efficiency in vivo. J Biomed Sci 2009; 16:36. [PMID: 19338665 PMCID: PMC2669473 DOI: 10.1186/1423-0127-16-36] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2008] [Accepted: 04/01/2009] [Indexed: 11/24/2022] Open
Abstract
Background Gene therapy with plasmid DNA is emerging as a promising strategy for the treatment of many diseases. One of the major obstacles to such therapy is the poor transfection efficiency of DNA in vivo. Methods In this report, we employed a very low power, near-infrared femtosecond laser technique to enhance the transfection efficiency of intradermally and intratumorally administered DNA plasmid. Results We found that femtosecond laser treatment can significantly enhance the delivery of DNA into the skin and into established tumors in mice. In addition, we found that both laser power density as well as duration of laser treatment are critical parameters for augmenting DNA transfection efficiency. The femtosecond laser technique employs a relatively unfocused laser beam that maximizes the transfected area, minimizes damage to tissue and simplifies its implementation. Conclusion This femtosecond new laser technology represents a safe and innovative technology for enhancing DNA gene transfer in vivo.
Collapse
Affiliation(s)
- Shaw-Wei D Tsen
- Department of Pathology, The Johns Hopkins School of Medicine, Baltimore, MD 21218, USA.
| | | | | | | | | | | |
Collapse
|
15
|
Nifontova IN, Sats NV, Surin VL, Svinareva DA, Gasparian ME, Drize NJ. Infection of stromal and hemopoietic precursor cells with lentivirus vector in vivo and in vitro. Bull Exp Biol Med 2008; 145:133-6. [PMID: 19024021 DOI: 10.1007/s10517-008-0030-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
We developed a method for gene transfer into mesenchymal stromal cells. Lentivirus vector containing green fluorescent protein gene for labeling stromal and hemopoietic precursor cells was obtained using two plasmid sets from different sources. The vector was injected into the femur of mice in vivo and added into culture medium for in vitro infection of the stromal sublayer of long-term bone marrow culture. From 25 to 80% hemopoietic stem cells forming colonies in the spleen were infected with lentivirus vector in vivo and in vitro. Fibroblast colony-forming cells from the femoral bones of mice injected with the lentivirus vector carried no marker gene. The marker gene was detected in differentiated descendants from mesenchymal stem cells (bone cavity cells from the focus of ectopic hemopoiesis formed after implantation of the femoral bone marrow cylinder infected with lentivirus vector under the renal capsule of syngeneic recipient). In in vitro experiments, the marker gene was detected in sublayers of long-term bone marrow cultures infected after preliminary 28-week culturing, when hemopoiesis was completely exhausted. The efficiency of infection of stromal precursor cells depended on the source of lentivirus. The possibility of transfering the target gene into hemopoietic precursor cells in vivo is demonstrated. Stromal precursor cells can incorporate the provirus in vivo and in vitro, but conditions and infection system for effective infection should be thoroughly selected.
Collapse
Affiliation(s)
- I N Nifontova
- Hematology Research Center, Russian Academy of Medical Sciences.
| | | | | | | | | | | |
Collapse
|
16
|
Jin Q, Marsh J, Cornetta K, Alkhatib G. Resistance to human immunodeficiency virus type 1 (HIV-1) generated by lentivirus vector-mediated delivery of the CCR5{Delta}32 gene despite detectable expression of the HIV-1 co-receptors. J Gen Virol 2008; 89:2611-2621. [PMID: 18796731 DOI: 10.1099/vir.0.2008/003624-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
It has previously been demonstrated that there are two distinct mechanisms for genetic resistance to human immunodeficiency virus type 1 (HIV-1) conferred by the CCR5Delta32 gene: the loss of wild-type CCR5 surface expression and the generation of CCR5Delta32 protein, which interacts with CXCR4. To analyse the protective effects of long-term expression of the CCR5Delta32 protein, recombinant lentiviral vectors were used to deliver the CCR5Delta32 gene into human cell lines and primary peripheral blood mononuclear cells that had been immortalized by human T-cell leukemia virus type 1. Blasticidin S-resistant cell lines expressing the lentivirus-encoded CCR5Delta32 showed a significant reduction in HIV-1 Env-mediated fusion assays. It was shown that CD4(+) T lymphocytes expressing the lentivirus-encoded CCR5Delta32 gene were highly resistant to infection by a primary but not by a laboratory-adapted X4 strain, suggesting different infectivity requirements. In contrast to previous studies that analysed the CCR5Delta32 protective effects in a transient expression system, this study showed that long-term expression of CCR5Delta32 conferred resistance to HIV-1 despite cell-surface expression of the HIV co-receptors. The results suggest an additional unknown mechanism for generating the CCR5Delta32 resistance phenotype and support the hypothesis that the CCR5Delta32 protein acts as an HIV-suppressive factor by altering the stoichiometry of the molecules involved in HIV-1 entry. The lentiviral-CCR5Delta32 vectors offer a method of generating HIV-resistant cells by delivery of the CCR5Delta32 gene that may be useful for stem cell- or T-cell-based gene therapy for HIV-1 infection.
Collapse
Affiliation(s)
- Qingwen Jin
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Jon Marsh
- Indiana University Vector Production Facility, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kenneth Cornetta
- Indiana University Vector Production Facility, Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Ghalib Alkhatib
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
17
|
Acid-degradable cationic methacrylamide polymerized in the presence of plasmid DNA as tunable non-viral gene carrier. Biomaterials 2008; 29:3872-81. [DOI: 10.1016/j.biomaterials.2008.06.003] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2008] [Accepted: 06/07/2008] [Indexed: 11/24/2022]
|
18
|
Evaluation of safety and efficacy of RNAi against HIV-1 in the human immune system (Rag-2-/-γc-/-) mouse model. Gene Ther 2008; 16:148-53. [DOI: 10.1038/gt.2008.124] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
19
|
ter Brake O, 't Hooft K, Liu YP, Centlivre M, von Eije KJ, Berkhout B. Lentiviral vector design for multiple shRNA expression and durable HIV-1 inhibition. Mol Ther 2008; 16:557-64. [PMID: 18180777 PMCID: PMC7172400 DOI: 10.1038/sj.mt.6300382] [Citation(s) in RCA: 189] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2006] [Accepted: 11/15/2007] [Indexed: 01/07/2023] Open
Abstract
Human immunodeficiency virus type 1 (HIV-1) replication in T cells can be inhibited by RNA interference (RNAi) through short hairpin RNA (shRNA) expression from a lentiviral vector. However, for the development of a durable RNAi-based gene therapy against HIV-1, multiple shRNAs need to be expressed simultaneously in order to avoid viral escape. In this study, we tested a multiple shRNA expression strategy for different shRNAs using repeated promoters in a lentiviral vector. Although highly effective in co-transfection experiments, a markedly reduced activity of each expressed shRNA was observed in transduced cells. We found that this reduced activity was due to recombination of the expression cassette repeat sequences during the transduction of the lentiviral vector, which resulted in deletions of one or multiple cassettes. To avoid recombination, we tested different promoters for multiple shRNA expression. We compared the activity of the human polymerase III promoters U6, H1, and 7SK and the polymerase II U1 promoter. Activities of these promoters were similar, irrespective of which shRNA was expressed. We showed that these four expression cassettes can be combined in a single lentiviral vector without causing recombination. Moreover, whereas HIV-1 could escape from a single shRNA, we now show that HIV-1 escape can be prevented when four shRNAs are simultaneously expressed in a cell.
Collapse
Affiliation(s)
- Olivier ter Brake
- Laboratory of Experimental Virology, Department of Medical Microbiology, Center for Infection and Immunity Amsterdam (CINIMA), Academic Medical Center of University of Amsterdam, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
20
|
Giacca M. Gene therapy to induce cellular resistance to HIV-1 infection: lessons from clinical trials. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2008; 56:297-325. [PMID: 18086416 DOI: 10.1016/s1054-3589(07)56010-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Affiliation(s)
- Mauro Giacca
- Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology (ICGEB), Padriciano 99, 34012 Trieste, Italy
| |
Collapse
|
21
|
Awong G, La Motte-Mohs RN, Zúñiga-Pflücker JC. Generation of pro-T cells in vitro: potential for immune reconstitution. Semin Immunol 2007; 19:341-9. [PMID: 17997108 DOI: 10.1016/j.smim.2007.10.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/22/2007] [Accepted: 10/02/2007] [Indexed: 10/22/2022]
Abstract
Immunodeficient individuals are susceptible to opportunistic infection. While stem cell transplantation can restore a functional immune system, T cells are slow to recover and limited in eliciting adaptive immune responses. Approaches to selectively enhance T cell function have focused on boosting thymopoiesis to generate new T cells or expanding existing T cells. By taking advantage of the role of Notch signaling in T cell development, we have developed an in vitro system able to generate large numbers of progenitor T cells from human hematopoietic stem cells. Here, we discuss this in vitro system and its implications for the potential treatment of T cell immunodeficiency.
Collapse
Affiliation(s)
- Génève Awong
- Department of Immunology, University of Toronto, 2075 Bayview Avenue, Toronto, Ontario M4N 3M5, Canada
| | | | | |
Collapse
|
22
|
Abstract
Adoptive transfer of antigen-specific T cells is a promising approach for preventing progressive viral infections in immunosuppressed hosts. By contrast, effective T-cell therapy of malignant disease has proven to be much more difficult to achieve. This, in part, reflects the difficulty of isolating high avidity T cells specific for tumor-associated antigens, many of which are self-antigens that have induced some level of tolerance in the host. Even when tumor-reactive T cells can be isolated, the ability of these cells to survive in vivo and traffic to tumor sites is often impaired. Additionally, most tumors employ multiple mechanisms to escape T-cell recognition, including interference in antigen presentation, secretion of inhibitory factors and recruitment of regulatory or immunosuppressive cells. The genetic modification of T cells prior to transfer provides a potential means to overcome many of these obstacles and enhance the efficacy of T-cell therapy. This review article discusses the rationale for genetic modification of T cells, the critical steps involved in gene transfer, and potential advantages and disadvantages of strategies that are now being examined to engineer improved effector T cells for the treatment of human infectious and malignant disease.
Collapse
Affiliation(s)
- Carolina Berger
- Fred Hutchinson Cancer Research Center, Program in Immunology, Seattle, WA 98109-1024, USA.
| | | | | | | |
Collapse
|
23
|
Abstract
Aptamers are oligonucleotides evolved in vitro or in nature to bind target ligands with high affinity and specificity. They are emerging as powerful tools in the fields of therapeutics, drug development, target validation and diagnostics. Aptamers are attractive alternatives to antibody- and small-molecule-based therapeutics owing to their stability, low toxicity, low immunogenicity and improved safety. With the recent approval of the first aptamer drug Macugen by the US FDA, there is great impetus to develop therapeutic aptamers that can target a wide array of disease states. The recent demonstration that aptamer activity can be reversed by the administration of a simple antidote greatly enhances the potential value of aptamers as therapeutic agents.
Collapse
Affiliation(s)
- N S Que-Gewirth
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | |
Collapse
|
24
|
Nagasaki T, Shinkai S. The concept of molecular machinery is useful for design of stimuli-responsive gene delivery systems in the mammalian cell. J INCL PHENOM MACRO 2007. [DOI: 10.1007/s10847-007-9303-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
25
|
Rupp B, Ruzsics Z, Buser C, Adler B, Walther P, Koszinowski UH. Random screening for dominant-negative mutants of the cytomegalovirus nuclear egress protein M50. J Virol 2007; 81:5508-17. [PMID: 17376929 PMCID: PMC1900260 DOI: 10.1128/jvi.02796-06] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
Inactivation of gene products by dominant-negative (DN) mutants is a powerful tool to assign functions to proteins. Here, we present a two-step procedure to establish a random screen for DN alleles, using the essential murine cytomegalovirus gene M50 as an example. First, loss-of-function mutants from a linker-scanning library were tested for inhibition of virus reconstitution with the help of FLP-mediated ectopic insertion of the mutants into the viral genome. Second, DN candidates were confirmed by conditional expression of the inhibitory proteins in the virus context. This allowed the quantification of the inhibitory effect, the identification of the morphogenesis block, and the construction of DN mutants with improved activity. Based on these observations a DN mutant of the homologous gene (UL50) in human cytomegalovirus was predicted and constructed. Our data suggest that a proline-rich sequence motif in the variable region of M50/UL50 represents a new functional site which is essential for nuclear egress of cytomegalovirus capsids.
Collapse
Affiliation(s)
- Brigitte Rupp
- Max von Pettenkofer Institut für Virologie, Ludwig-Maximilians-Universität München, Pettenkoferstrasse 9a, 80336 Munich, Germany
| | | | | | | | | | | |
Collapse
|
26
|
Kawamura K, Oishi J, Sakakihara S, Niidome T, Katayama Y. Intracellular signal-responsive artificial gene regulation. J Drug Target 2007; 14:456-64. [PMID: 17062392 DOI: 10.1080/10611860600845470] [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] [Indexed: 10/24/2022]
Abstract
In gene therapy, in order to avoid serious side effects due to the unexpected expression of the transgene in non-target cells, transgenes have to be delivered only to the target cells. In response to this issue, many researchers have aimed at developing target cell-selective gene carriers using active targeting strategies. However, such methodology does not always work, because an ideal molecular marker, which is specific to the target disease cells, is not always available. In this study, we introduce a new concept regarding target disease cell-selective gene therapy (D-RECS). Here, we use intracellular signals, which are activated to an extraordinary degree only in the target disease cells, as a trigger for transgene expression using polymer-peptide conjugates. This strategy could actually activate gene expression in the target signal-activated cells only. Hyper-activation of certain intracellular signals has been reported in many diseases. Thus, this new strategy is expected to provide a powerful methodology for future gene therapy. In this review, the basic concept, some examples, and the molecular design of D-RECS carriers are introduced.
Collapse
Affiliation(s)
- Kenji Kawamura
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Fukuoka, 819-0395, Japan
| | | | | | | | | |
Collapse
|
27
|
Oishi J, Ijuin M, Sonoda T, Kang JH, Kawamura K, Mori T, Niidome T, Katayama Y. A protein kinase signal-responsive gene carrier modified RGD peptide. Bioorg Med Chem Lett 2006; 16:5740-3. [PMID: 16971118 DOI: 10.1016/j.bmcl.2006.08.096] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2006] [Revised: 08/21/2006] [Accepted: 08/23/2006] [Indexed: 11/20/2022]
Abstract
We have previously reported artificial gene-regulation systems responding to cyclic AMP-dependent protein kinase (PKA) using a cationic polymer. However, this polymer alone cannot deliver any gene into living cells. In the present work, we modified the signal-responsive polymer to the RGD peptide for the introduction of a polymer/DNA complex into living cells and succeeded in regulating the gene expression responding to intracellular PKA activation.
Collapse
Affiliation(s)
- Jun Oishi
- Graduate School of Systems Life Sciences, Kyushu University, Motooka 744, Fukuoka 819-0395, Japan
| | | | | | | | | | | | | | | |
Collapse
|
28
|
von Laer D, Hasselmann S, Hasselmann K. Gene therapy for HIV infection: what does it need to make it work? J Gene Med 2006; 8:658-67. [PMID: 16598816 DOI: 10.1002/jgm.908] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
The efficacy of antiviral drug therapy for HIV infection is limited by toxicity and viral resistance. Thus, alternative therapies need to be explored. Several gene therapeutic strategies for HIV infection have been developed, but in clinical testing therapeutically effective levels of the transgene product were not achieved. This review focuses on the determinants of therapeutic efficacy and discusses the potential and also the limits of current gene therapy approaches for HIV infection.
Collapse
Affiliation(s)
- Dorothee von Laer
- Georg-Speyer-Haus, Paul-Ehrlich-Strasse 42, Frankfurt a.M., Germany.
| | | | | |
Collapse
|
29
|
Modlich U, Bohne J, Schmidt M, von Kalle C, Knöss S, Schambach A, Baum C. Cell-culture assays reveal the importance of retroviral vector design for insertional genotoxicity. Blood 2006; 108:2545-53. [PMID: 16825499 PMCID: PMC1895590 DOI: 10.1182/blood-2005-08-024976] [Citation(s) in RCA: 263] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Retroviral vectors with long terminal repeats (LTRs), which contain strong enhancer/promoter sequences at both ends of their genome, are widely used for stable gene transfer into hematopoietic cells. However, recent clinical data and mouse models point to insertional activation of cellular proto-oncogenes as a dose-limiting side effect of retroviral gene delivery that potentially induces leukemia. Self-inactivating (SIN) retroviral vectors do not contain the terminal repetition of the enhancer/promoter, theoretically attenuating the interaction with neighboring cellular genes. With a new assay based on in vitro expansion of primary murine hematopoietic cells and selection in limiting dilution, we showed that SIN vectors using a strong internal retroviral enhancer/promoter may also transform cells by insertional mutagenesis. Most transformed clones, including those obtained after dose escalation of SIN vectors, showed insertions upstream of the third exon of Evi1 and in reverse orientation to its transcriptional orientation. Normalizing for the vector copy number, we found the transforming capacity of SIN vectors to be significantly reduced when compared with corresponding LTR vectors. Additional modifications of SIN vectors may further increase safety. Improved cell-culture assays will likely play an important role in the evaluation of insertional mutagenesis.
Collapse
Affiliation(s)
- Ute Modlich
- Department of Experimental Hematology, Hannover Medical School, Carl-Neuberg-Strasse 1, D-30625 Hannover, Germany
| | | | | | | | | | | | | |
Collapse
|
30
|
Al‐Gharabli SI, Shah STA, Weik S, Schmidt MF, Mesters JR, Kuhn D, Klebe G, Hilgenfeld R, Rademann J. An efficient method for the synthesis of peptide aldehyde libraries employed in the discovery of reversible SARS coronavirus main protease (SARS-CoV Mpro) inhibitors. Chembiochem 2006; 7:1048-55. [PMID: 16688706 PMCID: PMC7417790 DOI: 10.1002/cbic.200500533] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2005] [Indexed: 11/10/2022]
Abstract
A method for the parallel solid-phase synthesis of peptide aldehydes has been developed. Protected amino acid aldehydes obtained by the racemization-free oxidation of amino alcohols with Dess-Martin periodinane were immobilized on threonyl resins as oxazolidines. Following Boc protection of the ring nitrogen to yield the N-protected oxazolidine linker, peptide synthesis was performed efficiently on this resin. A peptide aldehyde library was designed for targeting the SARS coronavirus main protease, SARS-CoV M(pro)(also known as 3CL(pro)), on the basis of three different reported binding modes and supported by virtual screening. A set of 25 peptide aldehydes was prepared by this method and investigated in inhibition assays against SARS-CoV M(pro). Several potent inhibitors were found with IC(50) values in the low micromolar range. An IC(50) of 7.5 muM was found for AcNSTSQ-H and AcESTLQ-H. Interestingly, the most potent inhibitors seem to bind to SARS-CoV M(pro) in a noncanonical binding mode.
Collapse
Affiliation(s)
- Samer I. Al‐Gharabli
- Leibniz Institut für Molekulare Pharmakologie (FMP), Department for Medicinal Chemistry, Robert‐Rössle‐Straße 10, 13125 Berlin, Germany, Fax: (+49) 30‐94793‐280
- Current address: Chemical and Pharmaceutical Engineering, German–Jordanian University, 35247 Amman 11180, Jordan
| | - Syed T. Ali Shah
- Leibniz Institut für Molekulare Pharmakologie (FMP), Department for Medicinal Chemistry, Robert‐Rössle‐Straße 10, 13125 Berlin, Germany, Fax: (+49) 30‐94793‐280
| | - Steffen Weik
- Leibniz Institut für Molekulare Pharmakologie (FMP), Department for Medicinal Chemistry, Robert‐Rössle‐Straße 10, 13125 Berlin, Germany, Fax: (+49) 30‐94793‐280
| | - Marco F. Schmidt
- Leibniz Institut für Molekulare Pharmakologie (FMP), Department for Medicinal Chemistry, Robert‐Rössle‐Straße 10, 13125 Berlin, Germany, Fax: (+49) 30‐94793‐280
- Freie Universität Berlin, Institut für Chemie und Biochemie, Takustrasse 3, 14195 Berlin, Germany
| | - Jeroen R. Mesters
- Institut für Biochemie, Center for Structural and Cell Biology in Medicine, Universität Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Daniel Kuhn
- Institut für Pharmazeutische Chemie, Philipps‐Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Gerhard Klebe
- Institut für Pharmazeutische Chemie, Philipps‐Universität Marburg, Marbacher Weg 6, 35032 Marburg, Germany
| | - Rolf Hilgenfeld
- Institut für Biochemie, Center for Structural and Cell Biology in Medicine, Universität Lübeck, Ratzeburger Allee 160, 23538 Lübeck, Germany
| | - Jörg Rademann
- Leibniz Institut für Molekulare Pharmakologie (FMP), Department for Medicinal Chemistry, Robert‐Rössle‐Straße 10, 13125 Berlin, Germany, Fax: (+49) 30‐94793‐280
- Freie Universität Berlin, Institut für Chemie und Biochemie, Takustrasse 3, 14195 Berlin, Germany
| |
Collapse
|
31
|
Chou CF, Mulky A, Maitra S, Lin WJ, Gherzi R, Kappes J, Chen CY. Tethering KSRP, a decay-promoting AU-rich element-binding protein, to mRNAs elicits mRNA decay. Mol Cell Biol 2006; 26:3695-706. [PMID: 16648466 PMCID: PMC1489004 DOI: 10.1128/mcb.26.10.3695-3706.2006] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Inherently unstable mRNAs contain AU-rich elements (AREs) in their 3' untranslated regions that act as mRNA stability determinants by interacting with ARE-binding proteins (ARE-BPs). We have destabilized two mRNAs by fusing sequence-specific RNA-binding proteins to KSRP, a decay-promoting ARE-BP, in a tethering assay. These results support a model that KSRP recruits mRNA decay machinery/factors to elicit decay. The ability of tethered KSRP to elicit mRNA decay depends on functions of known mRNA decay enzymes. By targeting the Rev response element of human immunodeficiency virus type 1 by using Rev-KSRP fusion protein, we degraded viral mRNA, resulting in a dramatic reduction of viral replication. These results provide a foundation for the development of novel therapeutic strategies to inhibit specific gene expression in patients with acquired or hereditary diseases.
Collapse
Affiliation(s)
- Chu-Fang Chou
- Department of Biochemistry & Molecular Genetics, University of Alabama at Birmingham, Birmingham, Alabama 35294, USA
| | | | | | | | | | | | | |
Collapse
|
32
|
Swan CH, Bühler B, Steinberger P, Tschan MP, Barbas CF, Torbett BE. T-cell protection and enrichment through lentiviral CCR5 intrabody gene delivery. Gene Ther 2006; 13:1480-92. [PMID: 16738691 DOI: 10.1038/sj.gt.3302801] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
CCR5 is the chemokine co-receptor for R5-tropic human immunodeficiency virus type 1 (HIV-1) isolates most often associated with primary infection. We have developed an HIV-1 self-inactivating vector, CAD-R5, containing a CCR5 single-chain antibody (intrabody) gene, which when expressed in T-cell lines and primary CD4+ T cells disrupts CCR5 cell surface expression and provides protection from R5-tropic isolate exposure. Furthermore, CAD-R5 intrabody expression in primary CD4+ T cells supports significant growth and enrichment over time during HIV-1-pulsed dendritic cell-T-cell interactions. These results indicate that CCR5 intrabody-expressing CD4+ T cells are refractory against this highly efficient primary route of infection. CD34+ cells transduced with the CAD-R5 vector gave rise to CD4+ and CD8+ thymocytes in non-obese diabetic (NOD)/ severely combined-immunodeficient (SCID)-human thymus/liver (hu thy/liv) mice, suggesting that CCR5 intrabody expression can be maintained throughout differentiation without obvious cellular effects. CD4+ T cells isolated from NOD/SCID-hu thy/liv mice were resistant to R5-tropic HIV-1 challenge demonstrating the maintenance of protection. Our findings demonstrate delivery of anti-HIV-1 activity through CCR5 intrabodies in primary CD4+ T cells and CD34+ cell-derived T-cell progeny. Thus, gene delivery strategies that provide a selective survival and growth advantage for T effector cells may provide a therapeutic benefit for HIV-1-infected individuals who have failed conventional therapies.
Collapse
Affiliation(s)
- C H Swan
- Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA
| | | | | | | | | | | |
Collapse
|
33
|
Konstantinova P, de Vries W, Haasnoot J, ter Brake O, de Haan P, Berkhout B. Inhibition of human immunodeficiency virus type 1 by RNA interference using long-hairpin RNA. Gene Ther 2006; 13:1403-13. [PMID: 16708080 PMCID: PMC7091653 DOI: 10.1038/sj.gt.3302786] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Inhibition of virus replication by means of RNA interference has been reported for several important human pathogens, including human immunodeficiency virus type 1 (HIV-1). RNA interference against these pathogens has been accomplished by introduction of virus-specific synthetic small interfering RNAs (siRNAs) or DNA constructs encoding short-hairpin RNAs (shRNAs). Their use as therapeutic antiviral against HIV-1 is limited, because of the emergence of viral escape mutants. In order to solve this durability problem, we tested DNA constructs encoding virus-specific long-hairpin RNAs (lhRNAs) for their ability to inhibit HIV-1 production. Expression of lhRNAs in mammalian cells may result in the synthesis of many siRNAs targeting different viral sequences, thus providing more potent inhibition and reducing the chance of viral escape. The lhRNA constructs were compared with in vitro diced double-stranded RNA and a DNA construct encoding an effective nef-specific shRNA for their ability to inhibit HIV-1 production in cells. Our results show that DNA constructs encoding virus-specific lhRNAs are capable of inhibiting HIV-1 production in a sequence-specific manner, without inducing the class I interferon genes.
Collapse
Affiliation(s)
- P Konstantinova
- Department of Human Retrovirology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - W de Vries
- Department of Human Retrovirology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Viruvation BV Wassenaarseweg 72, Leiden, The Netherlands
| | - J Haasnoot
- Department of Human Retrovirology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - O ter Brake
- Department of Human Retrovirology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - P de Haan
- Viruvation BV Wassenaarseweg 72, Leiden, The Netherlands
| | - B Berkhout
- Department of Human Retrovirology, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
34
|
Schambach A, Schiedlmeier B, Kühlcke K, Verstegen M, Margison GP, Li Z, Kamino K, Bohne J, Alexandrov A, Hermann FG, von Laer D, Baum C. Towards hematopoietic stem cell-mediated protection against infection with human immunodeficiency virus. Gene Ther 2006; 13:1037-47. [PMID: 16541120 DOI: 10.1038/sj.gt.3302755] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The failure of pharmacological approaches to cure infection with the human immunodeficiency virus (HIV) has renewed the interest in gene-based therapies. Among the various strategies that are currently explored, the blockade of HIV entry into susceptible T cells and macrophages promises to be the most powerful intervention. For long-term protection of both of these lineages, genetic modification of hematopoietic stem cells (HSCs) would be required. Here, we tested whether HSCs and their progeny can be modified to express therapeutic levels of M87o, a gammaretroviral vector encoding an artificial transmembrane molecule that blocks fusion-mediated uptake of HIV. In serial murine bone marrow transplantations, efficient and multilineage expression of M87o was observed for more than 1 year (range 37-75% of mononuclear cells), without signs of toxicity related to the transmembrane molecule. To allow enrichment of M87o-modified HSCs after transplant, we constructed vectors coexpressing the P140K mutant of O(6)-methylguanine-DNA-methyltransferase (MGMT-P140K). This clinically relevant selection marker mediates a survival advantage in HSCs if exposed to combinations of methylguanine-methyltransferase (MGMT) inhibitors and alkylating agents. A bicistronic vector mediated sufficient expression of both M87o and MGMT to confer a selective survival advantage in the presence of HIV and alkylating agents, respectively. These data encourage further investigations in large animal models and clinical trials.
Collapse
Affiliation(s)
- A Schambach
- Department of Hematology, Hemostaseology and Oncology, Hannover Medical School, Hannover, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Oishi J, Kawamura K, Kang JH, Kodama K, Sonoda T, Murata M, Niidome T, Katayama Y. An intracellular kinase signal-responsive gene carrier for disordered cell-specific gene therapy. J Control Release 2005; 110:431-436. [PMID: 16309776 DOI: 10.1016/j.jconrel.2005.10.007] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2005] [Revised: 09/05/2005] [Accepted: 10/11/2005] [Indexed: 11/21/2022]
Abstract
We have previously reported artificial gene-regulation systems responding to cyclic AMP-dependent protein kinase (PKA) using cationic polymer. This cationic polymer (PAK) was a graft-type polymer with an oligopeptide that is a substrate for PKA and could regulate gene-expression in a cell-free system. In the present study, we carried out a detailed characterization of the PAK-DNA complex (AFM observation and DLS measurement) and tried to apply this polymer to living cells. In the unstimulated NIH 3T3 cells, transfection of the PAK-DNA complex showed no expression of the delivered gene. This means that PAK formed a stable complex with DNA in the normal cells to totally suppress gene expression. In contrast, significant expression was seen when the PAK-DNA complex was delivered to forskolin-treated cells. Thus, activated PKA disintegrates the complexes even in living cells, resulting in gene expression. Our results indicate that this type of intracellular signal-responsive polymer will be useful for the cell-specific release of genes.
Collapse
Affiliation(s)
- Jun Oishi
- Graduate School of Systems Life Sciences, Kyushu University, Hakozaki 6-10-1, Higashi-ku, Fukuoka 812-8581, Japan; CREST, Japan Science and Technology Corporation, 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Kenji Kawamura
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Hakozaki 6-10-1, Higashi-ku, Fukuoka 812-8581, Japan; CREST, Japan Science and Technology Corporation, 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Jeong-Hun Kang
- CREST, Japan Science and Technology Corporation, 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Kota Kodama
- CREST, Japan Science and Technology Corporation, 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Tatsuhiko Sonoda
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Hakozaki 6-10-1, Higashi-ku, Fukuoka 812-8581, Japan
| | - Masaharu Murata
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Hakozaki 6-10-1, Higashi-ku, Fukuoka 812-8581, Japan; CREST, Japan Science and Technology Corporation, 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Takuro Niidome
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Hakozaki 6-10-1, Higashi-ku, Fukuoka 812-8581, Japan; CREST, Japan Science and Technology Corporation, 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan
| | - Yoshiki Katayama
- Department of Applied Chemistry, Faculty of Engineering, Kyushu University, Hakozaki 6-10-1, Higashi-ku, Fukuoka 812-8581, Japan; CREST, Japan Science and Technology Corporation, 4-1-8 Honcho, Kawaguchi-shi, Saitama 332-0012, Japan.
| |
Collapse
|