1
|
Renner A, Stahringer A, Ruppel KE, Fricke S, Koehl U, Schmiedel D. Development of KoRV-pseudotyped lentiviral vectors for efficient gene transfer into freshly isolated immune cells. Gene Ther 2024; 31:378-390. [PMID: 38684788 PMCID: PMC11257948 DOI: 10.1038/s41434-024-00454-0] [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: 10/11/2023] [Revised: 04/15/2024] [Accepted: 04/18/2024] [Indexed: 05/02/2024]
Abstract
Allogeneic cell therapies, such as those involving macrophages or Natural Killer (NK) cells, are of increasing interest for cancer immunotherapy. However, the current techniques for genetically modifying these cell types using lenti- or gamma-retroviral vectors present challenges, such as required cell pre-activation and inefficiency in transduction, which hinder the assessment of preclinical efficacy and clinical translation. In our study, we describe a novel lentiviral pseudotype based on the Koala Retrovirus (KoRV) envelope protein, which we identified based on homology to existing pseudotypes used in cell therapy. Unlike other pseudotyped viral vectors, this KoRV-based envelope demonstrates remarkable efficiency in transducing freshly isolated primary human NK cells directly from blood, as well as freshly obtained monocytes, which were differentiated to M1 macrophages as well as B cells from multiple donors, achieving up to 80% reporter gene expression within three days post-transduction. Importantly, KoRV-based transduction does not compromise the expression of crucial immune cell receptors, nor does it impair immune cell functionality, including NK cell viability, proliferation, cytotoxicity as well as phagocytosis of differentiated macrophages. Preserving immune cell functionality is pivotal for the success of cell-based therapeutics in treating various malignancies. By achieving high transduction rates of freshly isolated immune cells before expansion, our approach enables a streamlined and cost-effective automated production of off-the-shelf cell therapeutics, requiring fewer viral particles and less manufacturing steps. This breakthrough holds the potential to significantly reduce the time and resources required for producing e.g. NK cell therapeutics, expediting their availability to patients in need.
Collapse
Affiliation(s)
- Alexander Renner
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Department for Cell and Gene Therapy Development, Leipzig, Germany
| | - Anika Stahringer
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Department for Cell and Gene Therapy Development, Leipzig, Germany
| | - Katharina Eva Ruppel
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Department for Cell and Gene Therapy Development, Leipzig, Germany
| | - Stephan Fricke
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Department for Cell and Gene Therapy Development, Leipzig, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, CIMD, Leipzig, Deutschland
| | - Ulrike Koehl
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Department for Cell and Gene Therapy Development, Leipzig, Germany
- Fraunhofer Cluster of Excellence for Immune-Mediated Diseases, CIMD, Leipzig, Deutschland
- Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany
| | - Dominik Schmiedel
- Fraunhofer Institute for Cell Therapy and Immunology (IZI), Department for Cell and Gene Therapy Development, Leipzig, Germany.
- Institute for Clinical Immunology, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
2
|
Kaito Y, Hirano M, Futami M, Nojima M, Tamura H, Tojo A, Imai Y. CD155 and CD112 as possible therapeutic targets of FLT3 inhibitors for acute myeloid leukemia. Oncol Lett 2022; 23:51. [PMID: 34992684 PMCID: PMC8721849 DOI: 10.3892/ol.2021.13169] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 11/17/2021] [Indexed: 12/26/2022] Open
Abstract
Acute myeloid leukemia (AML) relapse is considered to be related to escape from antitumor immunity. Changes in the expression of immune checkpoints, including B7 homolog (H)1 and B7-H2, have been reported to contribute to AML progression. Binding of T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) among other immune checkpoints on natural killer (NK) and T cells to CD155/CD112 in tumors is supposed to be inhibitory; however, the mechanism by which changes in CD155 and CD112 expression affect tumor immunity remains unclear. When the increased expression of CD155 and CD112 activates Raf-MEK-ERK pathway and Raf-MEK-ERK pathway is one of the targets of FMS-like tyrosine kinase 3 (FLT3) inhibition. The present study investigated the alterations in CD155 and CD112 expression under FLT3 inhibition (quizartinib and gilteritinib) and studied its effect on NK and T cell cytotoxicity. CD155 and CD112 expression was analyzed using flow cytometry and reverse transcription-quantitative PCR in AML cell lines with or without FLT3 mutation using FLT3 inhibitors. CD155 and CD112 expression was specifically downregulated by FLT3 inhibition in FLT3-mutated cell lines. Direct cytotoxicity and antibody-dependent cellular cytotoxicity against these cells by NK cells were enhanced. However, the cytotoxicity of γδ T cells with low TIGIT expression compared with NK cells was not enhanced in direct cytotoxicity assay using luciferase luminescence. The analysis of clinical trials from The Cancer Genome Atlas (TCGA) revealed that high CD155 and CD112 expression is associated with poor overall survival. The enhanced cytotoxicity of NK cells against CD155- and CD112-downregulated cells following FLT3 inhibition indicated CD155 and CD112 as possible targets of immunotherapy for AML using FLT3 inhibitors.
Collapse
Affiliation(s)
- Yuta Kaito
- Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Mitsuhito Hirano
- Department of Molecular Therapy, Advanced Clinical Research Center, The University of Tokyo, Tokyo 108-8639, Japan
| | - Muneyoshi Futami
- Department of Molecular Therapy, Advanced Clinical Research Center, The University of Tokyo, Tokyo 108-8639, Japan
| | - Masanori Nojima
- Department of Translational Research, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Hideto Tamura
- Department of Hematology, Saitama Medical Center, Dokkyo Medical University, Saitama 343-8555, Japan
| | - Arinobu Tojo
- Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan.,Department of Molecular Therapy, Advanced Clinical Research Center, The University of Tokyo, Tokyo 108-8639, Japan
| | - Yoichi Imai
- Department of Hematology/Oncology, Research Hospital, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| |
Collapse
|
3
|
Heide M, Haffner C, Murayama A, Kurotaki Y, Shinohara H, Okano H, Sasaki E, Huttner WB. Human-specific ARHGAP11B increases size and folding of primate neocortex in the fetal marmoset. Science 2020; 369:546-550. [PMID: 32554627 DOI: 10.1126/science.abb2401] [Citation(s) in RCA: 118] [Impact Index Per Article: 29.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Accepted: 06/01/2020] [Indexed: 12/16/2022]
Abstract
The neocortex has expanded during mammalian evolution. Overexpression studies in developing mouse and ferret neocortex have implicated the human-specific gene ARHGAP11B in neocortical expansion, but the relevance for primate evolution has been unclear. Here, we provide functional evidence that ARHGAP11B causes expansion of the primate neocortex. ARHGAP11B expressed in fetal neocortex of the common marmoset under control of the gene's own (human) promoter increased the numbers of basal radial glia progenitors in the marmoset outer subventricular zone, increased the numbers of upper-layer neurons, enlarged the neocortex, and induced its folding. Thus, the human-specific ARHGAP11B drives changes in development in the nonhuman primate marmoset that reflect the changes in evolution that characterize human neocortical development.
Collapse
Affiliation(s)
- Michael Heide
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.
| | - Christiane Haffner
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany
| | - Ayako Murayama
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan.,Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan
| | - Yoko Kurotaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Haruka Shinohara
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo 160-8582, Japan.,Laboratory for Marmoset Neural Architecture, RIKEN Center for Brain Science, Wako City, Saitama 351-0198, Japan
| | - Erika Sasaki
- Department of Marmoset Biology and Medicine, Central Institute for Experimental Animals, Kawasaki 210-0821, Japan
| | - Wieland B Huttner
- Max Planck Institute of Molecular Cell Biology and Genetics, 01307 Dresden, Germany.
| |
Collapse
|
4
|
Murata K, Tsukuda S, Suizu F, Kimura A, Sugiyama M, Watashi K, Noguchi M, Mizokami M. Immunomodulatory Mechanism of Acyclic Nucleoside Phosphates in Treatment of Hepatitis B Virus Infection. Hepatology 2020; 71:1533-1545. [PMID: 31529730 DOI: 10.1002/hep.30956] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2018] [Accepted: 09/06/2019] [Indexed: 12/12/2022]
Abstract
BACKGROUND AND AIMS Current treatment with nucleos(t)ide analogs (NUCs) safely controls the replication of hepatitis B virus (HBV) and improves prognosis in patients with HBV. However, the inability to completely clear HBV is problematic, and novel therapies are desired. It has been believed that all NUCs have similar functions to inhibit HBV reverse transcriptase. However, our recent findings that only acyclic nucleoside phosphonates (ANPs; adefovir dipivoxil and tenofovir disoproxil fumarate) had an additional effect of inducing interferon (IFN)-λ3 in the gastrointestinal tract suggests that ANPs are not only distinct from nucleoside analogs (lamivudine and entecavir) in their structures but also in their functions. Because enteric lipopolysaccharide (LPS) can cross the intestine and affect peripheral blood mononuclear cells (PBMCs), we hypothesized that orally administered ANPs could have further additional effects to modulate LPS-mediated cytokine profile in PBMCs. APPROACH AND RESULTS This study showed that pretreatment of PBMCs, from either healthy volunteers or patients with HBV, with ANPs inhibited LPS-mediated interleukin (IL)-10 production, which reciprocally induced IL-12p70 and tumor necrosis factor-α production in a dose-dependent manner. Furthermore, the combination of IFN-α and ANPs synergistically enhanced LPS-mediated IL-12p70 production in PBMCs. Mechanistic analyses revealed that cellular metabolites of ANPs directly bound the Akt protein, inhibiting its translocation to the plasma membrane, thereby impairing Akt phosphorylation. Therefore, pretreatment of PBMCs with ANPs impairs LPS-mediated IL-10 production. CONCLUSIONS Among NUCs, only ANPs have an additional pharmacological effect modulating LPS-mediated cytokine production, which is expected to produce favorable immune responses toward HBV elimination. This additional immunomodulation by ANPs in PBMCs, as well as IFN-λ3 induction in the gastrointestinal tract, provides insights into HBV treatment.
Collapse
Affiliation(s)
- Kazumoto Murata
- Department of Gastroenterology, Graduate School of Medical Sciences, International University of Health and Welfare, Nasushiobara, Japan.,Genome Medical Sciences Project, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Senko Tsukuda
- RIKEN Center for Integrative Medical Sciences (IMS), Wako, Japan.,Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Futoshi Suizu
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Akihiro Kimura
- Department of Immunology and Pathology, Research Center for Hepatitis and Immunology, Research Institute, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Masaya Sugiyama
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Masayuki Noguchi
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Masashi Mizokami
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Ichikawa, Japan
| |
Collapse
|
5
|
Hirata N, Suizu F, Matsuda-Lennikov M, Tanaka T, Edamura T, Ishigaki S, Donia T, Lithanatudom P, Obuse C, Iwanaga T, Noguchi M. Functional characterization of lysosomal interaction of Akt with VRK2. Oncogene 2018; 37:5367-5386. [PMID: 29872222 PMCID: PMC6172193 DOI: 10.1038/s41388-018-0330-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2017] [Revised: 03/31/2018] [Accepted: 04/25/2018] [Indexed: 01/07/2023]
Abstract
Serine-threonine kinase Akt (also known as PKB, protein kinase B), a core intracellular mediator of cell survival, is involved in various human cancers and has been suggested to play an important role in the regulation of autophagy in mammalian cells. Nonetheless, the physiological function of Akt in the lysosomes is currently unknown. We have reported previously that PtdIns(3)P-dependent lysosomal accumulation of the Akt-Phafin2 complex is a critical step for autophagy induction. Here, to characterize the molecular function of activated Akt in the lysosomes in the process of autophagy, we searched for the molecules that interact with the Akt complex at the lysosomes after induction of autophagy. By time-of-flight-mass spectrometry (TOF/MS) analysis, kinases of the VRK family, a unique serine-threonine family of kinases in the human kinome, were identified. VRK2 interacts with Akt1 and Akt2, but not with Akt3; the C terminus of Akt and the N terminus of VRK2 facilitate the interaction of Akt and VRK2 in mammalian cells. The kinase-dead form of VRK2A (KD VRK2A) failed to interact with Akt in coimmunoprecipitation assays. Bimolecular fluorescence complementation (BiFC) experiments showed that, in the lysosomes, Akt interacted with VRK2A but not with VRK2B or KD VRK2A. Immunofluorescent assays revealed that VRK2 and phosphorylated Akt accumulated in the lysosomes after autophagy induction. WT VRK2A, but not KD VRK2A or VRK2B, facilitated accumulation of phosphorylated Akt in the lysosomes. Downregulation of VRK2 abrogated the lysosomal accumulation of phosphorylated Akt and impaired nuclear localization of TFEB; these events coincided to inhibition of autophagy induction. The VRK2-Akt complex is required for control of lysosomal size, acidification, bacterial degradation, and for viral replication. Moreover, lysosomal VRK2-Akt controls cellular proliferation and mitochondrial outer-membrane stabilization. Given the roles of autophagy in the pathogenesis of human cancer, the current study provides a novel insight into the oncogenic activity of VRK2-Akt complexes in the lysosomes via modulation of autophagy.
Collapse
Affiliation(s)
- Noriyuki Hirata
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Futoshi Suizu
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Mami Matsuda-Lennikov
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Tsutomu Tanaka
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Tatsuma Edamura
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Satoko Ishigaki
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
| | - Thoria Donia
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Chemistry Department, Faculty of Science, Tanta University, Tanta, Egypt
| | - Pathrapol Lithanatudom
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan
- Department of Biology, Faculty of Science, Center of Excellence in Bioresources for Agriculture, Industry and Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Chikashi Obuse
- Division of Molecular Life Science, Graduate School of Life Science, Hokkaido University, Sapporo, Japan
| | - Toshihiko Iwanaga
- Laboratory of Histology and Cytology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Masayuki Noguchi
- Division of Cancer Biology, Institute for Genetic Medicine, Hokkaido University, Sapporo, Japan.
| |
Collapse
|
6
|
Holstein M, Mesa-Nuñez C, Miskey C, Almarza E, Poletti V, Schmeer M, Grueso E, Ordóñez Flores JC, Kobelt D, Walther W, Aneja MK, Geiger J, Bonig HB, Izsvák Z, Schleef M, Rudolph C, Mavilio F, Bueren JA, Guenechea G, Ivics Z. Efficient Non-viral Gene Delivery into Human Hematopoietic Stem Cells by Minicircle Sleeping Beauty Transposon Vectors. Mol Ther 2018; 26:1137-1153. [PMID: 29503198 PMCID: PMC6079369 DOI: 10.1016/j.ymthe.2018.01.012] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 01/08/2018] [Accepted: 01/12/2018] [Indexed: 12/26/2022] Open
Abstract
The Sleeping Beauty (SB) transposon system is a non-viral gene delivery platform that combines simplicity, inexpensive manufacture, and favorable safety features in the context of human applications. However, efficient correction of hematopoietic stem and progenitor cells (HSPCs) with non-viral vector systems, including SB, demands further refinement of gene delivery techniques. We set out to improve SB gene transfer into hard-to-transfect human CD34+ cells by vectorizing the SB system components in the form of minicircles that are devoid of plasmid backbone sequences and are, therefore, significantly reduced in size. As compared to conventional plasmids, delivery of the SB transposon system as minicircle DNA is ∼20 times more efficient, and it is associated with up to a 50% reduction in cellular toxicity in human CD34+ cells. Moreover, providing the SB transposase in the form of synthetic mRNA enabled us to further increase the efficacy and biosafety of stable gene delivery into hematopoietic progenitors ex vivo. Genome-wide insertion site profiling revealed a close-to-random distribution of SB transposon integrants, which is characteristically different from gammaretroviral and lentiviral integrations in HSPCs. Transplantation of gene-marked CD34+ cells in immunodeficient mice resulted in long-term engraftment and hematopoietic reconstitution, which was most efficient when the SB transposase was supplied as mRNA and nucleofected cells were maintained for 4–8 days in culture before transplantation. Collectively, implementation of minicircle and mRNA technologies allowed us to further refine the SB transposon system in the context of HSPC gene delivery to ultimately meet clinical demands of an efficient and safe non-viral gene therapy protocol.
Collapse
Affiliation(s)
- Marta Holstein
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Cristina Mesa-Nuñez
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIEMAT/CIBERER), Madrid, Spain; Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM) Madrid, Spain
| | - Csaba Miskey
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Elena Almarza
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIEMAT/CIBERER), Madrid, Spain; Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM) Madrid, Spain
| | | | | | - Esther Grueso
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Juan Carlos Ordóñez Flores
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany
| | - Dennis Kobelt
- Translational Oncology, Experimental and Clinical Research Center, Charité University Medicine, Berlin, Germany
| | - Wolfgang Walther
- Translational Oncology, Experimental and Clinical Research Center, Charité University Medicine, Berlin, Germany
| | | | | | - Halvard B Bonig
- Department of Transfusion Medicine and Immunohematology, Johann-Wolfgang-Goethe Universität, Frankfurt, Germany
| | - Zsuzsanna Izsvák
- Mobile DNA, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | | | - Carsten Rudolph
- ethris GmbH, Planegg, Germany; Department of Pediatrics, Ludwig Maximilian University, Munich, Germany
| | - Fulvio Mavilio
- Genethon, Evry, France; Department of Life Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Juan A Bueren
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIEMAT/CIBERER), Madrid, Spain; Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM) Madrid, Spain
| | - Guillermo Guenechea
- Division of Hematopoietic Innovative Therapies, Centro de Investigaciones Energéticas Medioambientales y Tecnológicas and Centro de Investigación Biomédica en Red de Enfermedades Raras (CIEMAT/CIBERER), Madrid, Spain; Advanced Therapies Unit, Instituto de Investigación Sanitaria Fundación Jiménez Díaz (IIS-FJD, UAM) Madrid, Spain
| | - Zoltán Ivics
- Transposition and Genome Engineering, Division of Medical Biotechnology, Paul Ehrlich Institute, Langen, Germany.
| |
Collapse
|
7
|
Futami M, Sato K, Miyazaki K, Suzuki K, Nakamura T, Tojo A. Efficacy and Safety of Doubly-Regulated Vaccinia Virus in a Mouse Xenograft Model of Multiple Myeloma. MOLECULAR THERAPY-ONCOLYTICS 2017; 6:57-68. [PMID: 28808676 PMCID: PMC5545772 DOI: 10.1016/j.omto.2017.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 07/19/2017] [Indexed: 02/06/2023]
Abstract
Multiple myeloma is a malignancy of plasma cells of the bone marrow. Although the prognosis is variable, no curative therapy has been defined. Vaccinia virus infects cancer cells and kills such cells in a variety of ways. These include direct infection, triggering of immunomediated cell death, and vascular collapse. The potential of the vaccinia virus as an anti-tumor therapy has attracted the attention of oncologists. Interestingly, our preliminary experiments revealed that myeloma cells were particularly susceptible to vaccinia virus. To exploit this susceptibility and to render vaccinia more myeloma specific, we generated thymidine-kinase-deleted microRNA (miRNA)-regulated vaccinia viruses in which the essential viral gene B5R was regulated by miRNAs of normal human cells. Of the miRNAs examined, let-7a was found to be the most reliable in terms of regulating viral transmission. Exposure to unregulated vaccinia virus killed myeloma-transplanted severe combined immunodeficiency (SCID) mice; the animals succumbed to viral toxicity. In contrast, the thymidine-kinase-deleted let-7a-regulated virus remained localized within myeloma cells, triggering tumor regression and improving overall survival. In conclusion, a thymidine-kinase-deleted let-7a-regulated vaccinia virus was safe and effective for mice, warranting clinical trials in humans.
Collapse
Affiliation(s)
- Muneyoshi Futami
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Corresponding author: Muneyoshi Futami, Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan.
| | - Kota Sato
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo 150-8935, Japan
| | - Kanji Miyazaki
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo 150-8935, Japan
| | - Kenshi Suzuki
- Department of Hematology, Japanese Red Cross Medical Center, 4-1-22 Hiroo, Shibuya-ku, Tokyo 150-8935, Japan
| | - Takafumi Nakamura
- Division of Integrative Bioscience, Department of Biomedical Science, Institute of Regenerative Medicine and Biofunction, Graduate School of Medical Science, Tottori University, 86 Nishi-cho, Yonago, Tottori 683-8503, Japan
| | - Arinobu Tojo
- Division of Molecular Therapy, Advanced Clinical Research Center, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| |
Collapse
|
8
|
Shirakawa K, Wang L, Man N, Maksimoska J, Sorum AW, Lim HW, Lee IS, Shimazu T, Newman JC, Schröder S, Ott M, Marmorstein R, Meier J, Nimer S, Verdin E. Salicylate, diflunisal and their metabolites inhibit CBP/p300 and exhibit anticancer activity. eLife 2016; 5. [PMID: 27244239 PMCID: PMC4931907 DOI: 10.7554/elife.11156] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 05/26/2016] [Indexed: 12/19/2022] Open
Abstract
Salicylate and acetylsalicylic acid are potent and widely used anti-inflammatory drugs. They are thought to exert their therapeutic effects through multiple mechanisms, including the inhibition of cyclo-oxygenases, modulation of NF-κB activity, and direct activation of AMPK. However, the full spectrum of their activities is incompletely understood. Here we show that salicylate specifically inhibits CBP and p300 lysine acetyltransferase activity in vitro by direct competition with acetyl-Coenzyme A at the catalytic site. We used a chemical structure-similarity search to identify another anti-inflammatory drug, diflunisal, that inhibits p300 more potently than salicylate. At concentrations attainable in human plasma after oral administration, both salicylate and diflunisal blocked the acetylation of lysine residues on histone and non-histone proteins in cells. Finally, we found that diflunisal suppressed the growth of p300-dependent leukemia cell lines expressing AML1-ETO fusion protein in vitro and in vivo. These results highlight a novel epigenetic regulatory mechanism of action for salicylate and derivative drugs. DOI:http://dx.doi.org/10.7554/eLife.11156.001 People have been using a chemical called salicylate, which was once extracted from willow tree bark, as medicine for pain, fever and inflammation since ancient Greece. Aspirin is derived from salicylate but is a more potent drug. Aspirin exerts its anti-inflammatory effect by shutting down the activity of proteins that would otherwise boost inflammation. Aspirin achieves this by releasing a chemical marker, called an acetyl group, to be added to these proteins via a process known as protein acetylation. However, salicylate cannot trigger protein acetylation and so it was not clear how it reduces inflammation. An anti-diabetes drug that is converted into salicylate in the body reduces inflammation by inhibiting a protein called NF-κB. In 2001, a group of researchers reported that NF-κB becomes active when an enzyme called p300 adds an acetyl group to it. This raised the question: does salicylate reduce inflammation by blocking, instead of triggering, protein acetylation. Now, Shirakawa et al. – who include a researcher involved in the 2001 study – show that salicylate does indeed block the activity of the p300 enzyme. Shirakawa et al. then searched a database looking for drugs that have salicylate as part of their molecular structure. The search led to a drug called diflunisal, which was even more effective at blocking p300 in laboratory tests. Some cancers, including a blood cancer, rely on p300 to grow; diflunisal was shown to stop this kind of cancer cell from growing, both in the laboratory and in mice. Together, the experiments suggest that salicylate and drugs that share some of its structure might represent useful treatments for certain cancers, as well as other diseases that involve the p300 enzyme. DOI:http://dx.doi.org/10.7554/eLife.11156.002
Collapse
Affiliation(s)
- Kotaro Shirakawa
- Gladstone Institutes, University of California, San Francisco, United States.,Department of Medicine, University of California, San Francisco, United States.,Department of Hematology and Oncology, Kyoto University, Kyoto, Japan.,Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Lan Wang
- University of Miami, Gables, United States.,Sylvester Comprehensive Cancer Center, Miami, United States
| | - Na Man
- University of Miami, Gables, United States.,Sylvester Comprehensive Cancer Center, Miami, United States
| | - Jasna Maksimoska
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States.,Department of Biochemistry and Biophysics, Abramson Family Cancer Research Institute, Philadelphia, United States
| | - Alexander W Sorum
- Chemical Biology Laboratory, National Cancer Institute, Frederick, United States
| | - Hyung W Lim
- Gladstone Institutes, University of California, San Francisco, United States.,Department of Medicine, University of California, San Francisco, United States
| | - Intelly S Lee
- Gladstone Institutes, University of California, San Francisco, United States.,Department of Medicine, University of California, San Francisco, United States
| | - Tadahiro Shimazu
- Gladstone Institutes, University of California, San Francisco, United States.,Department of Medicine, University of California, San Francisco, United States
| | - John C Newman
- Gladstone Institutes, University of California, San Francisco, United States.,Department of Medicine, University of California, San Francisco, United States
| | - Sebastian Schröder
- Gladstone Institutes, University of California, San Francisco, United States.,Department of Medicine, University of California, San Francisco, United States
| | - Melanie Ott
- Gladstone Institutes, University of California, San Francisco, United States.,Department of Medicine, University of California, San Francisco, United States
| | - Ronen Marmorstein
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, United States.,Department of Biochemistry and Biophysics, Abramson Family Cancer Research Institute, Philadelphia, United States
| | - Jordan Meier
- Chemical Biology Laboratory, National Cancer Institute, Frederick, United States
| | - Stephen Nimer
- University of Miami, Gables, United States.,Sylvester Comprehensive Cancer Center, Miami, United States
| | - Eric Verdin
- Gladstone Institutes, University of California, San Francisco, United States.,Department of Medicine, University of California, San Francisco, United States
| |
Collapse
|
9
|
A novel long non-coding RNA in the rheumatoid arthritis risk locus TRAF1-C5 influences C5 mRNA levels. Genes Immun 2015; 17:85-92. [PMID: 26673966 DOI: 10.1038/gene.2015.54] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2015] [Revised: 11/06/2015] [Accepted: 11/11/2015] [Indexed: 01/05/2023]
Abstract
Long non-coding RNAs (lncRNAs) can regulate the transcript levels of genes in the same genomic region. These locally acting lncRNAs have been found deregulated in human disease and some have been shown to harbour quantitative trait loci (eQTLs) in autoimmune diseases. However, lncRNAs linked to the transcription of candidate risk genes in loci associated to rheumatoid arthritis (RA) have not yet been identified. The TRAF1 and C5 risk locus shows evidence of multiple eQTLs and transcription of intergenic non-coding sequences. Here, we identified a non-coding transcript (C5T1lncRNA) starting in the 3' untranslated region (UTR) of C5. RA-relevant cell types express C5T1lncRNA and RNA levels are further enhanced by specific immune stimuli. C5T1lncRNA is expressed predominantly in the nucleus and its expression correlates positively with C5 mRNA in various tissues (P=0.001) and in peripheral blood mononuclear cells (P=0.02) indicating transcriptional co-regulation. Knockdown results in a concurrent decrease in C5 mRNA levels but not of other neighbouring genes. Overall, our data show the identification of a novel lncRNA C5T1lncRNA that is fully located in the associated region and influences transcript levels of C5, a gene previously linked to RA pathogenesis.
Collapse
|
10
|
Araya N, Sato T, Ando H, Tomaru U, Yoshida M, Coler-Reilly A, Yagishita N, Yamauchi J, Hasegawa A, Kannagi M, Hasegawa Y, Takahashi K, Kunitomo Y, Tanaka Y, Nakajima T, Nishioka K, Utsunomiya A, Jacobson S, Yamano Y. HTLV-1 induces a Th1-like state in CD4+CCR4+ T cells. J Clin Invest 2014; 124:3431-42. [PMID: 24960164 PMCID: PMC4109535 DOI: 10.1172/jci75250] [Citation(s) in RCA: 84] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 05/08/2014] [Indexed: 12/14/2022] Open
Abstract
Human T-lymphotropic virus type 1 (HTLV-1) is linked to multiple diseases, including the neuroinflammatory disease HTLV-1-associated myelopathy/tropical spastic paraparesis (HAM/TSP) and adult T cell leukemia/lymphoma. Evidence suggests that HTLV-1, via the viral protein Tax, exploits CD4+ T cell plasticity and induces transcriptional changes in infected T cells that cause suppressive CD4+CD25+CCR4+ Tregs to lose expression of the transcription factor FOXP3 and produce IFN-γ, thus promoting inflammation. We hypothesized that transformation of HTLV-1-infected CCR4+ T cells into Th1-like cells plays a key role in the pathogenesis of HAM/TSP. Here, using patient cells and cell lines, we demonstrated that Tax, in cooperation with specificity protein 1 (Sp1), boosts expression of the Th1 master regulator T box transcription factor (T-bet) and consequently promotes production of IFN-γ. Evaluation of CSF and spinal cord lesions of HAM/TSP patients revealed the presence of abundant CD4+CCR4+ T cells that coexpressed the Th1 marker CXCR3 and produced T-bet and IFN-γ. Finally, treatment of isolated PBMCs and CNS cells from HAM/TSP patients with an antibody that targets CCR4+ T cells and induces cytotoxicity in these cells reduced both viral load and IFN-γ production, which suggests that targeting CCR4+ T cells may be a viable treatment option for HAM/TSP.
Collapse
MESH Headings
- Adult
- Aged
- Antibodies, Monoclonal/therapeutic use
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/virology
- Cell Line
- Cytotoxicity, Immunologic
- Female
- Gene Products, tax/immunology
- Human T-lymphotropic virus 1/immunology
- Human T-lymphotropic virus 1/pathogenicity
- Humans
- Immunotherapy
- Interferon-gamma/biosynthesis
- Interferon-gamma/genetics
- Male
- Middle Aged
- Paraparesis, Tropical Spastic/genetics
- Paraparesis, Tropical Spastic/immunology
- Paraparesis, Tropical Spastic/virology
- Receptors, CCR4/antagonists & inhibitors
- Receptors, CCR4/immunology
- Receptors, CCR4/metabolism
- Sp1 Transcription Factor/immunology
- T-Box Domain Proteins/genetics
- T-Box Domain Proteins/immunology
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/virology
- Th1 Cells/immunology
- Th1 Cells/virology
- Viral Load/immunology
Collapse
Affiliation(s)
- Natsumi Araya
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Tomoo Sato
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Hitoshi Ando
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Utano Tomaru
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Mari Yoshida
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Ariella Coler-Reilly
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Naoko Yagishita
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Junji Yamauchi
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Atsuhiko Hasegawa
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Mari Kannagi
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Yasuhiro Hasegawa
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Katsunori Takahashi
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Yasuo Kunitomo
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Yuetsu Tanaka
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Toshihiro Nakajima
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Kusuki Nishioka
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Atae Utsunomiya
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Steven Jacobson
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| | - Yoshihisa Yamano
- Department of Rare Diseases Research, Institute of Medical Science, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Pathology, Hokkaido University Graduate School of Medicine, Hokkaido, Japan. Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan. Department of Immunotherapeutics, Tokyo Medical and Dental University, Graduate School, Tokyo, Japan. Department of Neurology, St. Marianna University School of Medicine, Kanagawa, Japan. Department of Immunology, Graduate School of Medicine, University of the Ryukyus, Okinawa, Japan. Institute of Medical Science and Center for Clinical Research, Tokyo Medical University, Tokyo, Japan. Department of Hematology, Imamura Bun-in Hospital, Kagoshima, Japan. Viral Immunology Section, Neuroimmunology Branch, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
11
|
Bai Y, Lu Z, Lin Y, Sun B, Wang S, Wang G. Restoration of INK4a/ARF gene inhibits cell growth and cooperates with imatinib mesylate in Philadelphia chromosome-positive leukemias. Oncol Res 2014; 21:23-31. [PMID: 24330849 DOI: 10.3727/096504013x13786659070271] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
VSV-G-pseudotyped lentiviral vectors expressing p16(INK4a) or p14(ARF) were used to infect at high-efficiency Philadelphia chromosome (Ph)-positive leukemia cell lines lacking endogenous transcripts. Restoration of p16(INK4a) accumulated cells in the G0/G1 phase of cell cycle and restoration of p14(ARF) induced their apoptosis, followed by significant growth inhibition. Transduction of primary blast cells from chronic myeloid leukemia in blast crisis (CML-BC) and Ph-positive acute lymphoblastic leukemia (ALL) with p16(INK4a) or p14(ARF) virus also resulted in cell growth inhibition and/or apoptosis with a patient-to-patient variation, whereas clonal growth and differentiation of cord blood progenitor cells were not affected by enforced expression of INK4a/ARF. Furthermore, upon viral transduction at low multiplicity of infection, INK4a/ARF potentiated the effect of imatinib mesylate on Ph-positive leukemia cell lines in an additive but not synergistic manner. These results suggest that INK4a/ARF protein-mimetic agents may be promising options for Ph-positive leukemias in combination with imatinib mesylate.
Collapse
Affiliation(s)
- Yuansong Bai
- Department of Hematology/Oncology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | | | | | | | | | | |
Collapse
|
12
|
Gong Y, Qian Y, Yang F, Wang H, Yu Y. Lentiviral-mediated expression of SATB2 promotes osteogenic differentiation of bone marrow stromal cells in vitro and in vivo. Eur J Oral Sci 2014; 122:190-7. [PMID: 24666017 DOI: 10.1111/eos.12122] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/23/2014] [Indexed: 12/21/2022]
Affiliation(s)
- Yiming Gong
- Department of Stomatology; Zhongshan Hospital, Fudan University; Shanghai China
| | - Yanyan Qian
- Department of Biochemistry and Molecular Biology; Shanghai Medical College; Fudan University; Shanghai China
| | - Fei Yang
- Department of Stomatology; Zhongshan Hospital, Fudan University; Shanghai China
| | - Huijun Wang
- Department of Biochemistry and Molecular Biology; Shanghai Medical College; Fudan University; Shanghai China
| | - Youcheng Yu
- Department of Stomatology; Zhongshan Hospital, Fudan University; Shanghai China
| |
Collapse
|
13
|
Shin JW, Suzuki T, Ninomiya N, Kishima M, Hasegawa Y, Kubosaki A, Yabukami H, Hayashizaki Y, Suzuki H. Establishment of single-cell screening system for the rapid identification of transcriptional modulators involved in direct cell reprogramming. Nucleic Acids Res 2012; 40:e165. [PMID: 22879381 PMCID: PMC3505982 DOI: 10.1093/nar/gks732] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Combinatorial interactions of transcription modulators are critical to regulate cell-specific expression and to drive direct cell reprogramming (e.g. trans-differentiation). However, the identification of key transcription modulators from myriad of candidate genes is laborious and time consuming. To rapidly identify key regulatory factors involved in direct cell reprogramming, we established a multiplex single-cell screening system using a fibroblast-to-monocyte transition model. The system implements a single-cell 'shotgun-transduction' strategy followed by nested-single-cell-polymerase chain reaction (Nesc-PCR) gene expression analysis. To demonstrate this, we simultaneously transduced 18 monocyte-enriched transcription modulators in fibroblasts followed by selection of single cells expressing monocyte-specific CD14 and HLA-DR cell-surface markers from a heterogeneous population. Highly multiplex Nesc-PCR expression analysis revealed a variety of gene combinations with a significant enrichment of SPI1 (86/86) and a novel transcriptional modulator, HCLS1 (76/86), in the CD14(+)/HLA-DR(+) single cells. We could further demonstrate the synergistic role of HCLS1 in regulating monocyte-specific gene expressions and phagocytosis in dermal fibroblasts in the presence of SPI1. This study establishes a platform for a multiplex single-cell screening of combinatorial transcription modulators to drive any direct cell reprogramming.
Collapse
Affiliation(s)
- Jay W Shin
- Omics Science Center, RIKEN Yokohama, 1-7-22 Suehiro-cho Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Tsai HJ, Kobayashi S, Izawa K, Ishida T, Watanabe T, Umezawa K, Lin SF, Tojo A. Bioimaging analysis of nuclear factor-κB activity in Philadelphia chromosome-positive acute lymphoblastic leukemia cells reveals its synergistic upregulation by tumor necrosis factor-α-stimulated changes to the microenvironment. Cancer Sci 2011; 102:2014-21. [PMID: 21777350 PMCID: PMC11158770 DOI: 10.1111/j.1349-7006.2011.02039.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
To gain an insight into the microenvironmental regulation of nuclear factor (NF)-κB activity in the progression of leukemia, we established a bioluminescent imaging model of Philadelphia chromosome-positive acute lymphoblastic leukemia (Ph+ALL) cells transduced with a NF-κB/luciferase (Luc) reporter and cocultured with murine stromal cells and cytokines. Stromal cells alone did not augment Luc activity, taken as an index of NF-κB, but Luc activity was synergistically upregulated by the combination of stromal cells and tumor necrosis factor (TNF)-α. Dehydroxymethylepoxyquinomicin (DHMEQ), a specific inhibitor of NF-κB DNA binding, rapidly induced the apoptosis of Ph+ALL cells, indicating that NF-κB is necessary for the growth and survival of these cells. However, the DHMEQ-induced suppression of NF-κB activity and the apoptosis of leukemia cells were attenuated by the presence of stromal cells and TNF-α. In NOD-SCID mice transplanted with NF-κB/Luc reporter-containing Ph+ALL cell lines and monitored periodically during the progression of the leukemia, murine TNF-α was significantly expressed in lesions in which the leukemia cells emitted a significant NF-κB signal. These results support the notion that TNF-α also triggers microenvironmental upregulation of NF-κB activity in vivo. Collectively, the results indicated that TNF-α-stimulated microenvironment may contribute to the survival and progression of Ph+ALL cells through the synergistic upregulation of NF-κB activity.
Collapse
Affiliation(s)
- Hui-Jen Tsai
- Division of Molecular Therapy, Advanced Clinical Research Center, Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | |
Collapse
|
15
|
The interaction between AID and CIB1 is nonessential for antibody gene diversification by gene conversion or class switch recombination. PLoS One 2010; 5:e11660. [PMID: 20652029 PMCID: PMC2907397 DOI: 10.1371/journal.pone.0011660] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2010] [Accepted: 06/10/2010] [Indexed: 11/19/2022] Open
Abstract
Activation-induced deaminase (AID) initiates somatic hypermutation, gene conversion and class switch recombination by deaminating variable and switch region DNA cytidines to uridines. AID is predominantly cytoplasmic and must enter the nuclear compartment to initiate these distinct antibody gene diversification reactions. Nuclear AID is relatively short-lived, as it is efficiently exported by a CRM1-dependent mechanism and it is susceptible to proteasome-dependent degradation. To help shed light on mechanisms of post-translational regulation, a yeast-based screen was performed to identify AID-interacting proteins. The calcium and integrin binding protein CIB1 was identified by sequencing and the interaction was confirmed by immunoprecipitation experiments. The AID/CIB1 resisted DNase and RNase treatment, and it is therefore unlikely to be mediated by nucleic acid. The requirement for CIB1 in AID-mediated antibody gene diversification reactions was assessed in CIB1-deficient DT40 cells and in knockout mice, but immunoglobulin gene conversion and class switch recombination appeared normal. The DT40 system was also used to show that CIB1 over-expression has no effect on gene conversion and that AID-EGFP subcellular localization is normal. These combined data demonstrate that CIB1 is not required for AID to mediate antibody gene diversification processes. It remains possible that CIB1 has an alternative, a redundant or a subtle non-limiting regulatory role in AID biology.
Collapse
|
16
|
Salerni BL, Bates DJ, Albershardt TC, Lowrey CH, Eastman A. Vinblastine induces acute, cell cycle phase-independent apoptosis in some leukemias and lymphomas and can induce acute apoptosis in others when Mcl-1 is suppressed. Mol Cancer Ther 2010; 9:791-802. [PMID: 20371726 DOI: 10.1158/1535-7163.mct-10-0028] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Chemotherapeutic agents modify intracellular signaling that culminates in the inhibition of Bcl-2 family members and initiates apoptosis. Inhibition of the extracellular signal-regulated kinase by PD98059 dramatically accelerates vinblastine-mediated apoptosis in ML-1 leukemia with cells dying in 4 hours from all phases of the cell cycle. Inhibition of protein synthesis by cycloheximide also markedly accelerated vinblastine-induced apoptosis, showing that the proteins required for this acute apoptosis are constitutively expressed. Vinblastine induced the rapid induction of Mcl-1 that was inhibited by PD98059 and cycloheximide. No change in Bcl-2 or Bcl-X was observed. We hypothesize that ML-1 cells use Mcl-1 for protection from the rapid vinblastine-induced apoptosis. This was confirmed by targeting Mcl-1 with short hairpin RNA. We also investigated the response of 13 other leukemia and lymphoma cell lines and cells from seven chronic lymphocytic leukemia patients. Four cell lines and all chronic lymphocytic leukemia cells were killed in 6 hours by vinblastine alone. Two additional cell lines were sensitized to vinblastine by PD98059, which suppressed Mcl-1. This acute apoptosis either alone or in combination with PD98059 required vinblastine-mediated activation of c-Jun-NH(2)-terminal kinase. PD98059 did not suppress Mcl-1 in other cell lines whereas sorafenib did, but this did not sensitize the cells to vinblastine, suggesting that the acute apoptosis varies depending on which Bcl-2 protein mediates protection. Most of the cell lines were sensitized to vinblastine by cycloheximide, suggesting that inhibition of a short-lived protein in addition to Mcl-1 can acutely sensitize cells. These results suggest several clinical strategies that might provide an effective therapy for selected patients. Mol Cancer Ther; 9(4); 791-802. (c)2010 AACR.
Collapse
Affiliation(s)
- Bethany L Salerni
- Norris Cotton Cancer Center, Rubin Building Level 6, Dartmouth Medical School, Lebanon, NH 03756, USA
| | | | | | | | | |
Collapse
|
17
|
Hall SL, Chen ST, Gysin R, Gridley DS, Mohan S, Lau KHW. Stem cell antigen-1+ cell-based bone morphogenetic protein-4 gene transfer strategy in mice failed to promote endosteal bone formation. J Gene Med 2009; 11:877-88. [PMID: 19629966 DOI: 10.1002/jgm.1369] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
BACKGROUND This study assessed whether a Sca-1+ cell-based ex vivo gene transfer strategy, which has been shown to promote robust endosteal bone formation with a modified fibroblast growth factor-2 (FGF2) gene, can be extended to use with bone morphogenetic protein (BMP)2/4 hybrid gene. METHODS Sublethally irradiated recipient mice were transplanted with lentiviral (LV)-BMP2/4-transduced Sca-1+ cells. Bone parameters were monitored by pQCT and microCT. Gene expression was assessed by the real-time reverse transcriptase-polymerase chain reaction. RESULTS Recipient mice of LV-BMP2/4-transduced Sca-1+ cells yielded high engraftment and increased BMP4 mRNA levels in marrow cells; but exhibited only insignificant increases in serum and bone alkaline phosphatase activity compared to control mice. pQCT and microCT analyses of femurs showed that, with the exception of small changes in trabecular bone mineral density and cortical bone mineral content in LV-BMP2/4 mice, there were no differences in measured bone parameters between mice of the LV-BMP2/4 group and controls. The lack of large endosteal bone formation effects with the BMP4 strategy could not be attributed to ineffective engraftment or expansion of BMP4-expressing Sca-1+ cells, an inability of the transduced cells to secrete active BMP4 proteins, or to use of the LV-based vector. CONCLUSIONS Sca-1+ cell-based BMP4 ex vivo strategy did not promote robust endosteal bone formation, raising the possibility of intrinsic differences between FGF2- and BMP4-based strategies in their ability to promote endosteal bone formation. It emphasizes the importance of choosing an appropriate bone growth factor gene for delivery by this Sca-1+ cell-based ex vivo systemic gene transfer strategy to promote bone formation.
Collapse
Affiliation(s)
- Susan L Hall
- Musculoskeletal Disease Center (151), Jerry L. Pettis Memorial VA Medical Center, 11201 Benton Street, Loma Linda, California 92357, USA.
| | | | | | | | | | | |
Collapse
|
18
|
Murohashi M, Hinohara K, Kuroda M, Isagawa T, Tsuji S, Kobayashi S, Umezawa K, Tojo A, Aburatani H, Gotoh N. Gene set enrichment analysis provides insight into novel signalling pathways in breast cancer stem cells. Br J Cancer 2009; 102:206-12. [PMID: 19997106 PMCID: PMC2813736 DOI: 10.1038/sj.bjc.6605468] [Citation(s) in RCA: 70] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Background: Tumour-initiating cells (TICs) or cancer stem cells can exist as a small population in malignant tissues. The signalling pathways activated in TICs that contribute to tumourigenesis are not fully understood. Methods: Several breast cancer cell lines were sorted with CD24 and CD44, known markers for enrichment of breast cancer TICs. Tumourigenesis was analysed using sorted cells and total RNA was subjected to gene expression profiling and gene set enrichment analysis (GSEA). Results: We showed that several breast cancer cell lines have a small population of CD24−/low/CD44+ cells in which TICs may be enriched, and confirmed the properties of TICs in a xenograft model. GSEA revealed that CD24−/low/CD44+ cell populations are enriched for genes involved in transforming growth factor-β, tumour necrosis factor, and interferon response pathways. Moreover, we found the presence of nuclear factor-κB (NF-κB) activity in CD24−/low/CD44+ cells, which was previously unrecognised. In addition, NF-κB inhibitor dehydroxymethylepoxyquinomicin (DHMEQ) prevented tumourigenesis of CD24−/low/CD44+ cells in vivo. Conclusion: Our findings suggest that signalling pathways identified using GSEA help to identify molecular targets and biomarkers for TIC-like cells.
Collapse
Affiliation(s)
- M Murohashi
- Division of Systems Biomedical Technology, Institute of Medical Science, University of Tokyo, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Generation of transgenic non-human primates with germline transmission. Nature 2009; 459:523-7. [PMID: 19478777 DOI: 10.1038/nature08090] [Citation(s) in RCA: 524] [Impact Index Per Article: 34.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2008] [Accepted: 04/30/2009] [Indexed: 11/08/2022]
Abstract
The common marmoset (Callithrix jacchus) is increasingly attractive for use as a non-human primate animal model in biomedical research. It has a relatively high reproduction rate for a primate, making it potentially suitable for transgenic modification. Although several attempts have been made to produce non-human transgenic primates, transgene expression in the somatic tissues of live infants has not been demonstrated by objective analyses such as polymerase chain reaction with reverse transcription or western blots. Here we show that the injection of a self-inactivating lentiviral vector in sucrose solution into marmoset embryos results in transgenic common marmosets that expressed the transgene in several organs. Notably, we achieved germline transmission of the transgene, and the transgenic offspring developed normally. The successful creation of transgenic marmosets provides a new animal model for human disease that has the great advantage of a close genetic relationship with humans. This model will be valuable to many fields of biomedical research.
Collapse
|
20
|
Lee HJ, Lee YS, Kim HS, Kim YK, Kim JH, Jeon SH, Lee HW, Kim S, Miyoshi H, Chung HM, Kim DK. Retronectin enhances lentivirus-mediated gene delivery into hematopoietic progenitor cells. Biologicals 2009; 37:203-9. [PMID: 19264508 DOI: 10.1016/j.biologicals.2009.01.008] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2008] [Revised: 01/18/2009] [Accepted: 01/19/2009] [Indexed: 11/26/2022] Open
Abstract
Genetic modification of hematopoietic stem cells holds great promise in the treatment of hematopoietic disorders. However, clinical application of gene delivery has been limited, in part, by low gene transfer efficiency. To overcome this problem, we investigated the effect of retronectin (RN) on lentiviral-mediated gene delivery into hematopoietic progenitor cells (HPCs) derived from bone marrow both in vitro and in vivo. RN has been shown to enhance transduction by promoting colocalization of lentivirus and target cells. We found that RN enhanced lentiviral transfer of the VENUS transgene into cultured c-Kit(+) Lin(-) HPCs. As a complementary approach, in vivo gene delivery was performed by subjecting mice to intra-bone marrow injection of lentivirus or a mixture of RN and lentivirus. We found that co-injection with RN increased the number of VENUS-expressing c-Kit(+) Lin(-) HPCs in bone marrow by 2-fold. Further analysis of VENUS expression in colony-forming cells from the bone marrow of these animals revealed that RN increased gene delivery among these cells by 4-fold. In conclusion, RN is effective in enhancing lentivirus-mediated gene delivery into HPCs.
Collapse
Affiliation(s)
- Hyun-Joo Lee
- Graduate School of Life Science and Biotechnology, Pochon CHA University, CHA Stem Cell Institute, 605 Yeoksam 1-dong, Kangnam-gu, Seoul 135-081, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
The effects of lentiviral gene therapy with bone morphogenetic protein-2-producing bone marrow cells on spinal fusion in rats. ACTA ACUST UNITED AC 2008; 21:372-9. [PMID: 18600149 DOI: 10.1097/bsd.0b013e31814cf51d] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
STUDY DESIGN Rat spinal fusion model. OBJECTIVE This study aimed to assess the ability of rat bone marrow cells (RBMCs) transfected with bone morphogenetic protein (BMP)-2-containing lentivirus to induce a posterolateral spinal fusion in a rat model. SUMMARY OF BACKGROUND DATA Spinal arthrodesis is a commonly performed spinal procedure and autograft remains the standard for achieving spinal fusion. However, its procurement is associated with significant morbidity, and the rate of pseudoarthrosis has been reported to be 5% to 43%. Nonunion frequently leads to an unsatisfactory resolution of clinical symptoms and usually results in high medical costs and morbidity as well as the need for additional surgeries. These problems have led surgeons to search for alternative solutions to stimulate bone formation. Recombinant BMPs have also been used successfully in clinical trials. However, large doses of BMPs were required to induce adequate bone repair. The development of a regional gene therapy may be a more efficient method to deliver proteins to a specific anatomic site. Furthermore, adeno-BMP-2-producing rat bone marrow-derived cells have been used successfully to induce posterior spinal fusion. Recently, lentiviral vectors on the basis of human immunodeficiency virus have been developed for gene therapy. Lentiviruses are capable of insertion into the host genome, ensuring a prolonged gene expression. However, safety issues are a major concern when adopting these vectors for clinical use. METHODS In vitro study, we used RBMCs transfected with lentivirus vectors encoding BMP-2 (Lenti-BMP-2), RBMCs transfected with lentivirus vectors encoding the green fluorescent protein (GFP) (Lenti-GFP), and untransfected RBMCs; the latter 2 were used as controls. Alkaline phosphatase (ALP) staining and ALP activity were compared between the groups to assess the ability of the Lenti-BMP-2-transfected RBMCs to stimulate osteoblastic differentiation. In the rat posterolateral spine fusion model, the experimental study comprised 4 groups. Group 1 comprised 6 animals that were implanted with a collagen sponge containing 5 million RBMCs transfected with Lenti-BMP-2. Group 2 comprised 3 animals that were implanted with a collagen sponge containing 5 million RBMCs transfected with Lenti-GFP. Group 3 comprised 6 animals that were implanted with a collagen sponge containing 5 million untransfected RBMCs. Group 4 comprised 3 animals that were implanted with a collagen sponge alone. The rats were assessed by radiographs obtained at 4, 6, and 8 weeks. After death, their spines were explanted and assessed by manual palpation, high-resolution microcomputerized tomography, and histologic analysis. RESULTS The ALP staining was significantly greater in the Lenti-BMP-2-transfected RBMCs than in the untransfected RBMCs and the Lenti-GFP-transfected RBMCs. The ALP activity was 3-fold greater in the Lenti-BMP-2-transfected RBMCs than in the untransfected RBMCs and the Lenti-GFP-transfected RBMCs. In the rat spine fusion model, radiographic evaluation, high-resolution microcomputerized tomography, and manual palpation revealed spinal fusion in all the rats in Group 1 at 8 weeks. Groups 2, 3, and 4 comprised the control group. None of the rats in the control group (0 of 12) developed fusion at L4-L5. CONCLUSIONS The present study demonstrated that BMP-2-producing RBMCs, created through lentiviral gene transfer, induced sufficient spinal fusion. The use of lentiviral vectors that contain the cDNA for BMP-2 will be a novel and promising approach for a spinal fusion strategy.
Collapse
|
22
|
Kurita R, Oikawa T, Okada M, Yokoo T, Kurihara Y, Honda Y, Kageyama R, Suehiro Y, Okazaki T, Iga M, Miyoshi H, Tani K. Construction of a high-performance human fetal liver-derived lentiviral cDNA library. Mol Cell Biochem 2008; 319:181-7. [DOI: 10.1007/s11010-008-9891-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2008] [Accepted: 07/24/2008] [Indexed: 12/16/2022]
|
23
|
Gu W, Putral L, McMillan N. siRNA and shRNA as anticancer agents in a cervical cancer model. Methods Mol Biol 2008; 442:159-72. [PMID: 18369785 DOI: 10.1007/978-1-59745-191-8_12] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2023]
Abstract
We describe the protocols of using siRNAs, or shRNAs delivered by a lentiviral vector, as a means to silence cancer-causing genes. We use cervical cancer as a model to demonstrate the inhibition of the human papillomavirus (HPV) oncogenes E6 and E7 in cervical cancer cells by RNAi and inhibition of the cell growth in vitro and tumor growth in mouse models. The protocols include methods on siRNA and shRNA design, production of lentiviral-vectored shRNA, transfection or transduction of cervical cancer cells with siRNA or shRNA, and detection of the inhibitory effects of siRNA or shRNA both in vitro and in vitro.
Collapse
Affiliation(s)
- Wenyi Gu
- Cancer Biology Program, Centre for Immunology and Cancer Research, Princess Alexandra Hospital, University of Queensland, Brisbane, Australia
| | | | | |
Collapse
|
24
|
Abstract
STUDY DESIGN Rat spinal fusion model. OBJECTIVE This study aimed to compare the efficacy of lentiviral gene therapy, and adenoviral gene therapy in inducing spinal fusion in an immune competent rat spinal fusion model. SUMMARY OF BACKGROUND DATA Recombinant bone morphogenetic proteins (BMPs) have also been used for spinal fusion successfully in clinical trials. However, large doses of BMPs are required to induce adequate bone repair. Hence, regional gene therapy may be a more efficient method to deliver proteins to a specific anatomic site. Recently, lentiviral vectors based on human immunodeficiency virus have been developed for gene therapy. However, lentiviral gene therapy for spinal fusion has not been compared with adenoviral gene therapy. METHODS Lewis rats were divided into 7 groups. group I, II, III, and IV rats were implanted with a collagen sponge containing rat bone marrow cells (RBMCs) transfected with Lenti-BMP-2, Adeno-BMP-2, Lenti-GFP, Adeno-LacZ, respectively. Group V, VI, and VII rats were implanted with a collagen sponge containing recombinant BMP-2, a collagen sponge containing untransfected RBMCs, and a collagen sponge alone, respectively. The rats were assessed at 4, 6, and 8 weeks after implantation. After sacrificing the rats, their spines were explanted and assessed by manual palpation, high-resolution microcomputed tomography, and histologic analysis. RESULTS Spinal fusion was observed in all animals in group I, II, and V rats at 8 weeks. None of the rats in groups III, IV, VI, and VII showed spinal fusion. The volumes of the new bone in the area between the L4 and L5 transverse processes were greater in group I rats than in group II, and V rats with a significant difference. CONCLUSION BMP-2-producing RBMCs developed using lentiviral gene transfer induced more abundant bone within the fusion mass than the RBMCs transduced via adenoviral gene transfer and recombinant protein therapy.
Collapse
|
25
|
Virk MS, Conduah A, Park SH, Liu N, Sugiyama O, Cuomo A, Kang C, Lieberman JR. Influence of short-term adenoviral vector and prolonged lentiviral vector mediated bone morphogenetic protein-2 expression on the quality of bone repair in a rat femoral defect model. Bone 2008; 42:921-31. [PMID: 18295562 DOI: 10.1016/j.bone.2007.12.216] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/05/2007] [Revised: 12/10/2007] [Accepted: 12/12/2007] [Indexed: 01/26/2023]
Abstract
The objective of this study was to compare the efficacy of adenoviral and lentiviral regional gene therapy in a rat critical sized femoral defect model. The healing rates and quality of bone repair of femoral defects treated with syngeneic rat bone marrow cells (RBMCs) transduced with either lentiviral vector (Group I) or adenoviral vector (Group II) expressing bone morphogenetic protein-2 (BMP-2) gene were assessed. RBMCs transduced with the adenoviral vectors produced more than 3 times greater (p<0.001) BMP-2 when compared to RBMCs transduced with lentiviral vectors in an in vitro evaluation. Serial bioluminescent imaging demonstrated short duration luciferase expression (less than 3 weeks) in defects treated with RBMCs co-transduced with two adenoviral vectors (Group IV; adenovirus expressing BMP-2 and luciferase [Ad-BMP-2+Ad-Luc]). In contrast, the luciferase signal was present for 8 weeks in defects treated with RBMCs co-transduced with two lentiviral vectors (Group III; lentivirus expressing BMP-2 and luciferase gene [LV-BMP-2+LV-Luc]). There were no significant differences with respect to the radiological healing rates (p=0.12) in defects treated with lentiviral versus adenoviral mediated BMP-2 gene transfer. Biomechanical testing of healed Group I femoral specimens demonstrated significantly higher energy to failure (p<0.05) when compared to Group II defects. Micro CT analysis revealed higher bone volume/tissue volume fraction (p=0.04) in Group I defects when compared to Group II defects. In conclusion, prolonged BMP-2 expression associated with lentiviral mediated gene transfer demonstrated a trend towards superior quality of bone repair when compared to adenoviral mediated transfer of BMP-2. These results suggest that the bone repair associated with regional gene therapy is influenced not just by the amount of protein expression but also by duration of protein production. This observation needs validation in a more biologically challenging environment where differences in healing rates and quality of bone repair are more likely to be significantly different.
Collapse
Affiliation(s)
- Mandeep Singh Virk
- New England Musculoskeletal Institute, Department of Orthopaedic Surgery, University of Connecticut Health Center, Medical Arts and Research Building N4046, 263 Farmington Avenue, Farmington, CT 06030-5456, USA
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Futami M, Hatano T, Soda Y, Kobayashi S, Miyagishi M, Tojo A. RNAi-mediated silencing of p190Bcr-Abl inactivates Stat5 and cooperates with imatinib mesylate and 17-allylamino-17-demetoxygeldanamycin in selective killing of p190Bcr-Abl-expressing leukemia cells. Leukemia 2008; 22:1131-8. [DOI: 10.1038/leu.2008.60] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
27
|
Abstract
The cAMP-responsive element binding protein (CREB) is a 43-kDa nuclear transcription factor that regulates cell growth, memory, and glucose homeostasis. We showed previously that CREB is amplified in myeloid leukemia blasts and expressed at higher levels in leukemia stem cells from patients with myeloid leukemia. CREB transgenic mice develop myeloproliferative disease after 1 year, but not leukemia, suggesting that CREB contributes to but is not sufficient for leukemogenesis. Here, we show that CREB is most highly expressed in lineage negative hematopoietic stem cells (HSCs). To understand the role of CREB in hematopoietic progenitors and leukemia cells, we examined the effects of RNA interference (RNAi) to knock down CREB expression in vitro and in vivo. Transduction of primary HSCs or myeloid leukemia cells with lentiviral CREB shRNAs resulted in decreased proliferation of stem cells, cell- cycle abnormalities, and inhibition of CREB transcription. Mice that received transplants of bone marrow transduced with CREB shRNA had decreased committed progenitors compared with control mice. Mice injected with Ba/F3 cells expressing either Bcr-Abl wild-type or T315I mutation with CREB shRNA had delayed leukemic infiltration by bioluminescence imaging and prolonged median survival. Our results suggest that CREB is critical for normal myelopoiesis and leukemia cell proliferation.
Collapse
|
28
|
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
|
29
|
Ito Y, Demachi-Okamura A, Ohta R, Akatsuka Y, Nishida K, Tsujimura K, Morishima Y, Takahashi T, Kuzushima K. Full-length EBNA1 mRNA-transduced dendritic cells stimulate cytotoxic T lymphocytes recognizing a novel HLA-Cw*0303- and -Cw*0304-restricted epitope on EBNA1-expressing cells. J Gen Virol 2007; 88:770-780. [PMID: 17325349 DOI: 10.1099/vir.0.82519-0] [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: 12/20/2022] Open
Abstract
Epstein–Barr virus (EBV)-encoded nuclear antigen 1 (EBNA1) is an attractive target for immunotherapy against EBV-associated malignancies because it is expressed in all EBV-positive cells. Although CD8+ cytotoxic T-lymphocyte (CTL) epitope presentation is largely prevented by its glycine–alanine-repeat domain (GAr), the use of mRNA-transduced dendritic cells (DCs) would offer the advantage of priming EBNA1-specific CTLs. After stimulation with GAr-containing EBNA1-transduced monocyte-derived DCs, two EBNA1-specific CTL clones, B5 and C6, were isolated successfully from a healthy donor. These CTLs recognize peptides in the context of HLA-B*3501 and HLA-Cw*0303, respectively. A novel epitope, FVYGGSKTSL, was then identified, presented by both HLA-Cw*0303 and -Cw*0304, which are expressed by >35 % of Japanese, >20 % of Northern Han Chinese and >25 % of Caucasians. The mixed lymphocyte–peptide culture method revealed that FVYGGSKTSL-specific CTL-precursor frequencies in HLA-Cw*0303- or -Cw*0304-positive donors were between 1×10−5 and 1×10−4 CD8+ T cells. Moreover, both CTL clones inhibited growth of HLA-matched EBV-transformed B lymphocytes in vitro, and B5 CTLs produced a gamma interferon response to EBNA1-expressing gastric carcinoma cells in the context of HLA-Cw*0303. These data demonstrate that EBNA1 mRNA-transduced DCs may be useful tools for inducing EBNA1-specific CTLs that might be of clinical interest for CTL therapy of EBV-associated malignancies.
Collapse
Affiliation(s)
- Yoshinori Ito
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | | | - Rieko Ohta
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yoshiki Akatsuka
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Keiko Nishida
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kunio Tsujimura
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Yasuo Morishima
- Department of Cell Therapy, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Toshitada Takahashi
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Kiyotaka Kuzushima
- Division of Immunology, Aichi Cancer Center Research Institute, Nagoya, Japan
| |
Collapse
|
30
|
Saito Y. Making a stealth organ by RNA inhibition: will it fly? Transfusion 2007; 47:4-5. [PMID: 17207222 DOI: 10.1111/j.1537-2995.2007.01086.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
31
|
Zhang X, Soda Y, Takahashi K, Bai Y, Mitsuru A, Igura K, Satoh H, Yamaguchi S, Tani K, Tojo A, Takahashi TA. Successful immortalization of mesenchymal progenitor cells derived from human placenta and the differentiation abilities of immortalized cells. Biochem Biophys Res Commun 2006; 351:853-9. [PMID: 17094946 DOI: 10.1016/j.bbrc.2006.10.125] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2006] [Accepted: 10/23/2006] [Indexed: 12/28/2022]
Abstract
We reported previously that mesenchymal progenitor cells derived from chorionic villi of the human placenta could differentiate into osteoblasts, adipocytes, and chondrocytes under proper induction conditions and that these cells should be useful for allogeneic regenerative medicine, including cartilage tissue engineering. However, similar to human mesenchymal stem cells (hMSCs), though these placental cells can be isolated easily, they are difficult to study in detail because of their limited life span in vitro. To overcome this problem, we attempted to prolong the life span of human placenta-derived mesenchymal cells (hPDMCs) by modifying hTERT and Bmi-1, and investigated whether these modified hPDMCs retained their differentiation capability and multipotency. Our results indicated that the combination of hTERT and Bmi-1 was highly efficient in prolonging the life span of hPDMCs with differentiation capability to osteogenic, adipogenic, and chondrogenic cells in vitro. Clonal cell lines with directional differentiation ability were established from the immortalized parental hPDMC/hTERT+Bmi-1. Interestingly, hPDMC/Bmi-1 showed extended proliferation after long-term growth arrest and telomerase was activated in the immortal hPDMC/Bmi-1 cells. However, the differentiation potential was lost in these cells. This study reports a method to extend the life span of hPDMCs with hTERT and Bmi-1 that should become a useful tool for the study of mesenchymal stem cells.
Collapse
Affiliation(s)
- Xiaohong Zhang
- Division of Cell Processing, The Institute of Medical Science, The University of Tokyo, 4-6-1 Shirokanedai, Minato-ku, Tokyo 108-8639, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Tsurutani N, Yasuda J, Yamamoto N, Choi BI, Kadoki M, Iwakura Y. Nuclear import of the preintegration complex is blocked upon infection by human immunodeficiency virus type 1 in mouse cells. J Virol 2006; 81:677-88. [PMID: 17079325 PMCID: PMC1797461 DOI: 10.1128/jvi.00870-06] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Mouse cells do not support human immunodeficiency virus type 1 (HIV-1) replication because of host range barriers at steps including virus entry, transcription, RNA splicing, polyprotein processing, assembly, and release. The exact mechanisms for the suppression, however, are not completely understood. To elucidate further the barriers against HIV-1 replication in mouse cells, we analyzed the replication of the virus in lymphocytes from human CD4/CXCR4 transgenic mice. Although primary splenocytes and thymocytes allowed the entry and reverse transcription of HIV-1, the integration efficiency of the viral DNA was greatly reduced in these cells relative to human peripheral blood mononuclear cells, suggesting an additional block(s) before or at the point of host chromosome integration of the viral DNA. Preintegration processes were further analyzed using HIV-1 pseudotyped viruses. The reverse transcription step of HIV-1 pseudotyped with the envelope of murine leukemia virus or vesicular stomatitis virus glycoprotein was efficiently supported in both human and mouse cells, but nuclear import of the preintegration complex (PIC) of HIV-1 was blocked in mouse cells. We found that green fluorescent protein (GFP)-labeled HIV-1 integrase, which is known to be important in the nuclear localization of the PIC, could not be imported into the nucleus of mouse cells, in contrast to human cells. On the other hand, GFP-Vpr localized exclusively to the nuclei of both mouse and human cells. These observations suggest that, due to the dysfunction of integrase, the nuclear localization of PIC is suppressed in mouse cells.
Collapse
Affiliation(s)
- Naomi Tsurutani
- Center for Experimental Medicine, Institute of Medical Science, University of Tokyo, 4-6-1 Shirokanedai, Tokyo 108-8639, Japan
| | | | | | | | | | | |
Collapse
|
33
|
Micucci F, Zingoni A, Piccoli M, Frati L, Santoni A, Galandrini R. High-efficient lentiviral vector-mediated gene transfer into primary human NK cells. Exp Hematol 2006; 34:1344-52. [PMID: 16982327 DOI: 10.1016/j.exphem.2006.06.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2006] [Revised: 05/08/2006] [Accepted: 06/01/2006] [Indexed: 10/24/2022]
Abstract
OBJECTIVE The long-term transfection of genes into primary natural killer (NK) cells without disrupting normal cellular functions has been proven to be difficult with currently available gene-transfer methods. In this study, we establish a lentiviral vector-based technique for improved gene transfer into human NK cells in vitro and we report on high-efficient transduction of freshly isolated as well as cultured primary NK cells. METHODS Freshly isolated or primary cultured human NK cells, as well as the human NK cell line YTS, were transduced with replication-incompetent human immunodeficiency virus (HIV)-based lentiviral vector bearing a GFP reporter gene or a gene of interest under the control of the elongation factor 1alpha (EF1alpha) promoter. Transduction efficiencies were monitored by flow cytometry. RESULTS A long-term transgene expression was detected in up to 98% of YTS NK cells, whereas in freshly isolated or primary cultured NK cells exposed to interleukin (IL)-2 plus IL-12 upon infection, efficiency was in the range of 50% to 90%. Moreover, in freshly isolated quiescent NK cells a transfection efficiency of 18% to 20% was achieved without stimulation. Notably, no major phenotypic and functional modifications were observed in transduced cells with respect to control cells: the expression levels of activating receptors, CD69-antigen induction as well as cytotoxic function were unaffected. CONCLUSION Results of our study demonstrate that NK cells can be efficiently transduced by lentiviral vectors.
Collapse
MESH Headings
- Antigens, CD/biosynthesis
- Antigens, Differentiation, T-Lymphocyte/biosynthesis
- Cells, Cultured
- Genes, Reporter
- Genetic Vectors
- Humans
- Interleukin-12/pharmacology
- Killer Cells, Natural/cytology
- Killer Cells, Natural/metabolism
- Lectins, C-Type
- Lentivirus
- Lymphocyte Activation/drug effects
- Lymphocyte Activation/genetics
- Peptide Elongation Factor 1/genetics
- Promoter Regions, Genetic/genetics
- Time Factors
- Transduction, Genetic/methods
Collapse
Affiliation(s)
- Federica Micucci
- Department of Experimental Medicine and Pathology, Istituto Pasteur-Fondazione Cenci-Bolognetti, University "La Sapienza," Rome, Italy
| | | | | | | | | | | |
Collapse
|
34
|
Koch P, Siemen H, Biegler A, Itskovitz-Eldor J, Brüstle O. Transduction of human embryonic stem cells by ecotropic retroviral vectors. Nucleic Acids Res 2006; 34:e120. [PMID: 16998181 PMCID: PMC1636442 DOI: 10.1093/nar/gkl674] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The steadily increasing availability of human embryonic stem (hES) cell lines has created strong interest in applying available tools for gene transfer in murine cells to human systems. Here we present a method for the transduction of hES cells with ecotropic retroviral vectors. hES cells were transiently transfected with a construct carrying the murine retrovirus receptor mCAT1. Subsequently, the cells were exposed to replication-deficient Moloney murine leukemia virus (MoMuLV) derivatives or pseudotyped lentiviral vectors. With oncoretroviral vectors, this procedure yields overall transduction efficiencies of up to 20% and permits selection of permanently transduced clones with high frequency. Selected clones maintained expression of pluripotency-associated markers and exhibited multi-germ layer differentiation both in vitro and in vivo. HES cell-derived somatic cells including neural progeny maintained high levels of transgene expression. Lentiviral vectors pseudotyped with the MoMuLV envelope could be introduced in the same manner with efficiencies of up to 33%. Transgene expression of lentivirally transduced hES cells remained permanent after differentiation even without selection pressure. Bypassing the regulatory issues associated with the use of amphotropic retroviral systems and exploiting the large pool of existing murine vectors, this method provides a safe and versatile tool for gene transfer and lineage analysis in hES cells and their progeny.
Collapse
Affiliation(s)
| | | | | | | | - Oliver Brüstle
- To whom correspondence should be addressed at Institute of Reconstructive Neurobiology, Life and Brain Center, University of Bonn, Sigmund-Freud-Strasse 25, D-53105 Bonn, Germany. Tel: +49 228 6885 500; Fax: +49 228 6885 501;
| |
Collapse
|
35
|
Kurita R, Sasaki E, Yokoo T, Hiroyama T, Takasugi K, Imoto H, Izawa K, Dong Y, Hashiguchi T, Soda Y, Maeda T, Suehiro Y, Tanioka Y, Nakazaki Y, Tani K. Tal1/Scl Gene Transduction Using a Lentiviral Vector Stimulates Highly Efficient Hematopoietic Cell Differentiation from Common Marmoset (Callithrix jacchus) Embryonic Stem Cells. Stem Cells 2006; 24:2014-22. [PMID: 16728561 DOI: 10.1634/stemcells.2005-0499] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The development of embryonic stem cell (ESC) therapies requires the establishment of efficient methods to differentiate ESCs into specific cell lineages. Here, we report the in vitro differentiation of common marmoset (CM) (Callithrix jacchus) ESCs into hematopoietic cells after exogenous gene transfer using vesicular stomatitis virus-glycoprotein-pseudotyped lentiviral vectors. We transduced hematopoietic genes, including tal1/scl, gata1, gata2, hoxB4, and lhx2, into CM ESCs. By immunochemical and morphological analyses, we demonstrated that overexpression of tal1/scl, but not the remaining genes, dramatically increased hematopoiesis of CM ESCs, resulting in multiple blood-cell lineages. Furthermore, flow cytometric analysis demonstrated that CD34, a hematopoietic stem/progenitor cell marker, was highly expressed in tal1/scl-overexpressing embryoid body cells. Similar results were obtained from three independent CM ESC lines. These results suggest that transduction of exogenous tal1/scl cDNA into ESCs is a promising method to induce the efficient differentiation of CM ESCs into hematopoietic stem/progenitor cells.
Collapse
Affiliation(s)
- Ryo Kurita
- Department of Molecular Genetics, Division of Molecular and Clinical Genetics, Medical Institute of Bioregulation, Kyushu University, 3-1-1, Maidashi, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Feeley BT, Conduah AH, Sugiyama O, Krenek L, Chen ISY, Lieberman JR. In vivo molecular imaging of adenoviral versus lentiviral gene therapy in two bone formation models. J Orthop Res 2006; 24:1709-21. [PMID: 16788987 DOI: 10.1002/jor.20229] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Regional gene therapy techniques are promising methods to enhance bone formation in large bone defects that would be difficult to treat with allograft or autograft bone stock. In this study, we compared in vivo temporal expression patterns of adenoviral- and lentiviral-mediated gene therapy in two bone formation models. Primary rat bone marrow cells (RBMC) were transduced with lentiviral or adenoviral vectors containing luciferase (Luc) or BMP-2 cDNA, or cotransduced with vectors containing Luc and bone morphogenetic protein 2 (BMP-2). In vitro protein production was determined with luciferase assay or ELISA (for BMP-2 production) weekly for 12 weeks. Two bone formation models were used -- a hind limb muscle pouch or radial defect -- in SCID mice. A cooled charged-coupled device (CCD) camera was used to image in vivo luciferase expression weekly for 12 weeks. In vitro, adenoviral expression of BMP-2 and luciferase was detected by ELISA or luciferase assay, respectively, for 4 weeks. Lentiviral expression of BMP-2 and luciferase was sustained in culture for 3 months. Using the CCD camera, we found that adenoviral vectors expressed luciferase expression for up to 21 days, but lentiviral vectors expressed target gene expression for 3 months in vivo in both bone formation models. There was no detectable difference in the amount of bone formed between the adenoviral and lentiviral groups. Lentiviral-mediated delivery of BMP-2 can induce long term in vitro and in vivo gene expression, which may be beneficial when developing tissue engineering strategies to heal large bone defects or defects with a compromised biologic environment.
Collapse
Affiliation(s)
- Brian T Feeley
- Department of Orthopaedic Surgery, David Geffen School of Medicine, UCLA AIDS Institute, Los Angeles, California, USA
| | | | | | | | | | | |
Collapse
|
37
|
Baum C, Schambach A, Bohne J, Galla M. Retrovirus Vectors: Toward the Plentivirus? Mol Ther 2006; 13:1050-63. [PMID: 16632409 DOI: 10.1016/j.ymthe.2006.03.007] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2005] [Revised: 03/16/2006] [Accepted: 03/16/2006] [Indexed: 01/19/2023] Open
Abstract
Recombinant retroviral vectors based upon simple gammaretroviruses, complex lentiviruses, or potentially nonpathogenic spumaviruses represent relatively well characterized tools that are widely used for stable gene transfer. Different members of the Retroviridae family have developed distinct and potentially useful features related to their life cycle. These natural differences can be exploited for specialized applications in gene therapy and could conceivably be combined to create future retroviral hybrid vectors, ideally incorporating the following features: an efficient, noncytopathic packaging system with low likelihood of recombination; serum resistance; an ability to pseudotype with cell-specific envelopes; high-fidelity reverse transcription before cell entry; unrestricted cytoplasmic transport and nuclear import; an insulated expression cassette; specific chromosomal targeting; and physiologic or regulated levels of transgene expression. We envisage that, compared to contemporary vectors, a hybrid vector combining these properties would have increased therapeutic efficacy and an enhanced biosafety profile. Many of the above goals will require the inclusion of nonretroviral components into vector particles or transgenes.
Collapse
Affiliation(s)
- Christopher Baum
- Department of Experimental Hematology, Hannover Medical School, D-30625 Hannover, Germany.
| | | | | | | |
Collapse
|
38
|
Zeng ZJ, Li ZB, Luo SQ, Hu WX. Retrovirus-mediated tk gene therapy of implanted human breast cancer in nude mice under the regulation of Tet-On. Cancer Gene Ther 2006; 13:290-7. [PMID: 16110312 DOI: 10.1038/sj.cgt.7700889] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Tight regulation of the therapeutic gene expression is critical in gene therapy. In this report, a doxycycline (Dox)-regulated retrovirus-mediated gene expression system was used to study the effects of suicide gene therapy on human breast cancer cell line MCF-7 and the nude mice model of implanted human breast cancer. To render the expression of suicide gene under control, we used two pseudoviruses simultaneously, RevTRE/HSVtk and RevTet-On, to infect MCF-7 cells or xenografts of nude mice. When infected by the pseudoviruses and followed by Dox and Ganciclovir (GCV) treatment, MCF-7 cells were arrested at S phase and the growth was suppressed. We then evaluated the antitumor efficiency of this system in vivo through studying the mice bearing human breast cancer xenografts. Compared with control groups, the HSVtk mRNA level increased significantly in tumor tissues, mass of the tumors shrank remarkably, and tumor necrosis features occurred after treatment with Dox and GCV. These data suggest that suicide gene therapy using the Dox-induced Tet-On-controlled HSVtk gene expression system is a feasible method to treat human breast cancer.
Collapse
Affiliation(s)
- Z-J Zeng
- Molecular Biology Research Center, Xiangya Medical College, Central South University, Changsha, Hunan, PR China
| | | | | | | |
Collapse
|
39
|
Tahvanainen J, Pykäläinen M, Kallonen T, Lähteenmäki H, Rasool O, Lahesmaa R. Enrichment of nucleofected primary human CD4+ T cells: a novel and efficient method for studying gene function and role in human primary T helper cell differentiation. J Immunol Methods 2006; 310:30-9. [PMID: 16516225 DOI: 10.1016/j.jim.2005.11.024] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2005] [Revised: 11/29/2005] [Accepted: 11/30/2005] [Indexed: 02/04/2023]
Abstract
Identification of key factors mediating the differentiation of naïve CD4(+) T helper cells into Th1 and Th2 subsets is important for understanding the molecular mechanisms of the development of autoimmune diseases as well as asthma and allergy. Functional importance of a given gene in the initiation of human T helper cell differentiation has been hard to study due to the difficulty in transfecting primary resting human T lymphocytes. In this study we have successfully transfected human primary CD4(+) T helper cells using Amaxa's Nucleofection technology. To overcome the background caused by untransfected cells, we have developed a system for enriching nucleofected unstimulated human primary T helper cells that express the gene of interest. This is achieved by introducing a plasmid construct containing a bicistronic unit coding for a truncated mouse MHC class l H-2K(k) cell surface marker followed by selection of H-2K(k) positive cells using antibody coated beads. We demonstrate that the nucleofected and enriched H-2K(k) positive T helper cells differentiate into Th1 and Th2 cells as well as the non-transfected control cells. We also show that by using this novel method, introduction of an shRNA targeting Stat6, a key molecule driving the Th2 cell development, results in impaired Th2 cell differentiation, as expected. The method described here, enables fast and feasible preparation of highly pure transfected primary CD4(+) T cell cultures ideal for studying the influence of overexpression or knockdown of a given gene on T helper cell differentiation and other primary human T cell functions.
Collapse
Affiliation(s)
- Johanna Tahvanainen
- Turku Centre for Biotechnology, Turku University and Abo Akademi University, Finland.
| | | | | | | | | | | |
Collapse
|
40
|
Dekker RJ, Boon RA, Rondaij MG, Kragt A, Volger OL, Elderkamp YW, Meijers JCM, Voorberg J, Pannekoek H, Horrevoets AJG. KLF2 provokes a gene expression pattern that establishes functional quiescent differentiation of the endothelium. Blood 2006; 107:4354-63. [PMID: 16455954 DOI: 10.1182/blood-2005-08-3465] [Citation(s) in RCA: 265] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The flow-responsive transcription factor KLF2 is acquiring a leading role in the regulation of endothelial cell gene expression. A genome-wide microarray expression profiling is described employing lentivirus-mediated, 7-day overexpression of human KLF2 at levels observed under prolonged flow. KLF2 is not involved in lineage typing, as 42 endothelial-specific markers were unaffected. Rather, KLF2 generates a gene transcription profile (> 1000 genes) affecting key functional pathways such as cell migration, vasomotor function, inflammation, and hemostasis and induces a morphology change typical for shear exposure including stress fiber formation. Protein levels for thrombomodulin, endothelial nitric oxide synthase, and plasminogen activator inhibitor type-1 are altered to atheroprotective levels, even in the presence of the inflammatory cytokine TNF-alpha. KLF2 attenuates cell migration by affecting multiple genes including VEGFR2 and the potent antimigratory SEMA3F. The distribution of Weibel-Palade bodies in cultured cell populations is normalized at the single-cell level without interfering with their regulated, RalA-dependent release. In contrast, thrombin-induced release of Weibel-Palade bodies is significantly attenuated, consistent with the proposed role of VWF release at low-shear stress regions of the vasculature in atherosclerosis. These results establish that KLF2 acts as a central transcriptional switch point between the quiescent and activated states of the adult endothelial cell.
Collapse
Affiliation(s)
- Rob J Dekker
- Department of Medical Biochemistry, Academic Medical Center, University of Amsterdam, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Satou Y, Yasunaga JI, Yoshida M, Matsuoka M. HTLV-I basic leucine zipper factor gene mRNA supports proliferation of adult T cell leukemia cells. Proc Natl Acad Sci U S A 2006; 103:720-5. [PMID: 16407133 PMCID: PMC1334651 DOI: 10.1073/pnas.0507631103] [Citation(s) in RCA: 453] [Impact Index Per Article: 25.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Human T cell leukemia virus type I (HTLV-I) causes adult T cell leukemia (ATL) in 2-5% of carriers after a long latent period. An HTLV-I encoded protein, Tax, induces proliferation and inhibits apoptosis, resulting in clonal proliferation of infected cells. However, tax gene expression in ATL cells is disrupted by several mechanisms, including genetic changes in the tax gene and DNA methylation/deletion of the 5' long terminal repeat (LTR). Because Tax is the major target of cytotoxic T-lymphocytes in vivo, loss of Tax expression should enable ATL cells to escape the host immune system. The 5' LTR of HTLV-I is frequently hypermethylated or deleted in ATL cells, whereas the 3' LTR remains unmethylated and intact, suggesting the involvement of the 3' LTR in leukemogenesis. Here we show that a gene encoded by the minus strand of the HTLV-I proviral genome, HTLV-I basic leucine zipper factor (HBZ), is transcribed from 3'-LTR in all ATL cells. Suppression of HBZ gene transcription by short interfering RNA inhibits proliferation of ATL cells. In addition, HBZ gene expression promotes proliferation of a human T cell line. Analyses of T cell lines transfected with mutated HBZ genes showed that HBZ promotes T cell proliferation in its RNA form, whereas HBZ protein suppresses Tax-mediated viral transcription through the 5' LTR. Thus, the single HBZ gene has bimodal functions in two different molecular forms. The growth-promoting activity of HBZ RNA likely plays an important role in oncogenesis by HTLV-I.
Collapse
Affiliation(s)
- Yorifumi Satou
- Laboratory of Virus Immunology, Institute for Virus Research, Kyoto University, 53 Shogoin Kawahara-cho, Sakyo-ku, Kyoto 606-8507, Japan
| | | | | | | |
Collapse
|
42
|
Okada S, Ishii K, Yamane J, Iwanami A, Ikegami T, Katoh H, Iwamoto Y, Nakamura M, Miyoshi H, Okano HJ, Contag CH, Toyama Y, Okano H. In vivo imaging of engrafted neural stem cells: its application in evaluating the optimal timing of transplantation for spinal cord injury. FASEB J 2005; 19:1839-41. [PMID: 16141363 DOI: 10.1096/fj.05-4082fje] [Citation(s) in RCA: 191] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Neural stem/progenitor cells (NSPCs) hold promise in neural tissue replacement therapy after spinal cord injury. However, understanding the survival time of grafted NSPCs and determining the extent of migration away from transplantation sites are essential for optimizing treatment regimens. Here, we used in vivo bioluminescence imaging to noninvasively assess the survival and residence time of transplanted NSPCs at the injury sites in living animals, and we used histologic analyses to assess cell integration and morphology. Third-generation lentiviral vectors enabled efficient transduction and stable expression of both luciferase and a variant of green fluorescent protein in primary cultured NSPCs. Signals from these cells were detectable for up to 10 months or more after transplantation into the injured spinal cords of C57BL/6J mice. Histological and functional data supported the imaging data and suggest that the timing of NSPC transplantation may be a key determinant of the fates and function of integrated cells since cell survival and migration depended on the time of transplantation relative to injury. Optimization of cell therapies can be greatly accelerated and refined by imaging, and the methods in the present study can be widely applied to various research fields of regeneration medicine, including transplantation study.
Collapse
Affiliation(s)
- Seiji Okada
- Department of Physiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Sugiyama O, An DS, Kung SPK, Feeley BT, Gamradt S, Liu NQ, Chen ISY, Lieberman JR. Lentivirus-mediated gene transfer induces long-term transgene expression of BMP-2 in vitro and new bone formation in vivo. Mol Ther 2005; 11:390-8. [PMID: 15727935 DOI: 10.1016/j.ymthe.2004.10.019] [Citation(s) in RCA: 95] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2004] [Accepted: 10/28/2004] [Indexed: 12/23/2022] Open
Abstract
We examined the potential of ex vivo gene therapy to enhance bone repair using lentiviral vectors encoding either enhanced green fluorescent protein (EGFP) as a reporter gene or bone morphogenetic protein-2 (BMP-2) downstream of either the cytomegalovirus immediate early (CMV) promoter or the murine leukemia virus long terminal repeat (RhMLV) promoter derived from a murine retrovirus adapted to replicate in a rhesus macaque. In vitro, rat bone marrow stromal cells (BMSCs) transduced with Lenti-CMV-EGFP or Lenti-RhMLV-EGFP demonstrated over 90% transduction efficiency at 1 week and continued to demonstrate stable expression for 8 weeks. ELISA results demonstrated that lentivirus-mediated gene transfer into BMSCs induced stable BMP-2 production in vitro for 8 weeks. Increased EGFP and BMP-2 production was noted with the RhMLV promoter. In addition, we implanted BMSCs transduced with Lenti-RhMLV-BMP-2 into a muscle pouch in the hind limbs of severe combined immune deficient mice. Robust bone formation was noted in animals that received Lenti-RhMLV-BMP-2 cells at 3 weeks. These results demonstrate that lentiviral vectors expressing BMP-2 can induce long-term gene expression in vitro and new bone formation in vivo under the control of the RhMLV promoter. Prolonged gene expression may be advantageous when developing tissue engineering strategies to repair large bone defects.
Collapse
Affiliation(s)
- Osamu Sugiyama
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA 90095, USA
| | | | | | | | | | | | | | | |
Collapse
|
44
|
McMillin DW, Landázuri N, Gangadharan B, Hewes B, Archer DR, Spencer HT, Le Doux JM. Highly efficient transduction of repopulating bone marrow cells using rapidly concentrated polymer-complexed retrovirus. Biochem Biophys Res Commun 2005; 330:768-75. [PMID: 15809063 DOI: 10.1016/j.bbrc.2005.03.035] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2005] [Indexed: 02/07/2023]
Abstract
Using the cationic polymer, Polybrene, and the anionic polymer, chondroitin sulfate C, we concentrated recombinant retrovirus pseudotyped with an ecotropic envelope, which is susceptible to inactivation by high-speed concentration methods. To evaluate gene marking, murine bone marrow was harvested from C3H mice, transduced with polymer-concentrated GFP virus, and transplanted into lethally irradiated recipients. Total gene marking in mice averaged 30-35% at 8 weeks post-transplant and transgene expression remained stable for over 16 weeks. Using the polymer concentration method, a second retroviral vector encoding the drug resistant variant of dihydrofolate reductase (L22Y-DHFR) was concentrated and tested. Approximately 40% of transduced murine bone marrow progenitor cells were protected against trimetrexate concentrations that completely eliminated the growth of non-modified cells. These results show that anionic and cationic polymers can be combined to rapidly concentrate viruses that are normally difficult to concentrate, and the concentrated virus efficiently transduces hematopoietic stem cells.
Collapse
Affiliation(s)
- Douglas W McMillin
- Department of Pediatrics, Division of Hematology/Oncology and Bone Marrow Transplantation, Emory University School of Medicine, Atlanta, GA 30322, USA
| | | | | | | | | | | | | |
Collapse
|
45
|
Shi Q, Wilcox DA, Morateck PA, Fahs SA, Kenny D, Montgomery RR. Targeting platelet GPIbalpha transgene expression to human megakaryocytes and forming a complete complex with endogenous GPIbbeta and GPIX. J Thromb Haemost 2004; 2:1989-97. [PMID: 15550031 DOI: 10.1111/j.1538-7836.2004.00961.x] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Bernard-Soulier Syndrome (BSS) is a severe congenital platelet disorder that results from a deficiency of the platelet membrane glycoprotein (GP) Ib/IX complex that is composed of four subunits (GPIbalpha, GPIbbeta, GPIX, and GPV). Mutations in either GPIbalpha, GPIbbeta, or GPIX can result in BSS with many of the known mutations occurring in GPIbalpha. In this study, we have developed a gene therapy strategy to express hemagglutinin (HA)-tagged GPIbalpha in megakaryocytes and potentially correct a hereditary deficiency. To direct GPIbalpha expression in megakaryocytic lineage cells, we designed a GPIbalpha cassette where human GPIbalpha cDNA was placed under control of the megakaryocytic/platelet-specific alphaIIb promoter and inserted into a lentiviral vector. Human CD34+ peripheral blood cells (PBC) and Dami cells were transduced with alphaIIb-HA-GPIbalpha-WPT virus. Flow cytometry analysis demonstrated that 50.1% of the megakaryocytes derived from CD34+ stem cells and 97.3% of Dami cells were transduced and expressed transgene GPIbalpha protein. Immunoprecipitation with Western blot analysis demonstrated that transgene protein associated with endogenous GPIbbeta and GPIX proteins. To address further the lineage-specific expression of the alphaIIb-HA-GPIbalpha construct, three cell lines, Dami, AtT-20 and HepG2, were transfected with GPIbalpha expression plasmids and analyzed by confocal microscopy. The results demonstrated that among these three cell lines, the tissue-specific alphaIIb promoter was active only in Dami cells. Thus, GPIbalpha can be efficiently and specifically expressed in the megakaryocytic compartment of hematopoietic cells and the transgene product associates with endogenous GPIbbeta and GPIX forming a complete complex. This strategy could potentially be utilized for gene therapy of BSS.
Collapse
Affiliation(s)
- Q Shi
- Department of Pediatrics, Medical College of Wisconsin, MACC Fund Research Center (MFRC), Milwaukee, WI 53226, USA.
| | | | | | | | | | | |
Collapse
|
46
|
Anson DS. The use of retroviral vectors for gene therapy-what are the risks? A review of retroviral pathogenesis and its relevance to retroviral vector-mediated gene delivery. GENETIC VACCINES AND THERAPY 2004; 2:9. [PMID: 15310406 PMCID: PMC515179 DOI: 10.1186/1479-0556-2-9] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/11/2004] [Accepted: 08/13/2004] [Indexed: 01/23/2023]
Abstract
Retroviral vector-mediated gene transfer has been central to the development of gene therapy. Retroviruses have several distinct advantages over other vectors, especially when permanent gene transfer is the preferred outcome. The most important advantage that retroviral vectors offer is their ability to transform their single stranded RNA genome into a double stranded DNA molecule that stably integrates into the target cell genome. This means that retroviral vectors can be used to permanently modify the host cell nuclear genome. Recently, retroviral vector-mediated gene transfer, as well as the broader gene therapy field, has been re-invigorated with the development of a new class of retroviral vectors which are derived from lentiviruses. These have the unique ability amongst retroviruses of being able to infect non-cycling cells. Vectors derived from lentiviruses have provided a quantum leap in technology and seemingly offer the means to achieve significant levels of gene transfer in vivo.The ability of retroviruses to integrate into the host cell chromosome also raises the possibility of insertional mutagenesis and oncogene activation. Both these phenomena are well known in the interactions of certain types of wild-type retroviruses with their hosts. However, until recently they had not been observed in replication defective retroviral vector-mediated gene transfer, either in animal models or in clinical trials. This has meant the potential disadvantages of retroviral mediated gene therapy have, until recently, been seen as largely, if not entirely, hypothetical. The recent clinical trial of gammac mediated gene therapy for X-linked severe combined immunodeficiency (X-SCID) has proven the potential of retroviral mediated gene transfer for the treatment of inherited metabolic disease. However, it has also illustrated the potential dangers involved, with 2 out of 10 patients developing T cell leukemia as a consequence of the treatment. A considered review of retroviral induced pathogenesis suggests these events were qualitatively, if not quantitatively, predictable. In addition, it is clear that the probability of such events can be greatly reduced by relatively simple vector modifications, such as the use of self-inactivating vectors and vectors derived from non-oncogenic retroviruses. However, these approaches remain to be fully developed and validated. This review also suggests that, in all likelihood, there are no other major retroviral pathogenetic mechanisms that are of general relevance to replication defective retroviral vectors. These are important conclusions as they suggest that, by careful design and engineering of retroviral vectors, we can continue to use this gene transfer technology with confidence.
Collapse
Affiliation(s)
- Donald S Anson
- Department of Genetic Medicine, Women's and Children's Hospital, 4th Floor Rogerson Building, 72 King William Road, North Adelaide, South Australia, 5006, Australia.
| |
Collapse
|