1
|
Mattar CN, Chew WL, Lai PS. Embryo and fetal gene editing: Technical challenges and progress toward clinical applications. Mol Ther Methods Clin Dev 2024; 32:101229. [PMID: 38533521 PMCID: PMC10963250 DOI: 10.1016/j.omtm.2024.101229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Gene modification therapies (GMTs) are slowly but steadily making progress toward clinical application. As the majority of rare diseases have an identified genetic cause, and as rare diseases collectively affect 5% of the global population, it is increasingly important to devise gene correction strategies to address the root causes of the most devastating of these diseases and to provide access to these novel therapies to the most affected populations. The main barriers to providing greater access to GMTs continue to be the prohibitive cost of developing these novel drugs at clinically relevant doses, subtherapeutic effects, and toxicity related to the specific agents or high doses required. In vivo strategy and treating younger patients at an earlier course of their disease could lower these barriers. Although currently regarded as niche specialties, prenatal and preconception GMTs offer a robust solution to some of these barriers. Indeed, treating either the fetus or embryo benefits from economy of scale, targeting pre-pathological tissues in the fetus prior to full pathogenesis, or increasing the likelihood of complete tissue targeting by correcting pluripotent embryonic cells. Here, we review advances in embryo and fetal GMTs and discuss requirements for clinical application.
Collapse
Affiliation(s)
- Citra N.Z. Mattar
- Experimental Fetal Medicine Group, Department of Obstetrics and Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, Singapore 119228
- Department of Obstetrics and Gynaecology, National University Health System, Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, Singapore 119228
| | - Wei Leong Chew
- Genome Institute of Singapore, Agency for Science, Technology and Research (A∗STAR), Singapore, 60 Biopolis St, Singapore, Singapore 138672
| | - Poh San Lai
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, 1E Kent Ridge Road, NUHS Tower Block, Level 12, Singapore, Singapore 119228
| |
Collapse
|
2
|
Sung YL, Wang TW, Lin TT, Lin SF. Optogenetics in cardiology: methodology and future applications. INTERNATIONAL JOURNAL OF ARRHYTHMIA 2022. [DOI: 10.1186/s42444-022-00060-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
AbstractOptogenetics is an emerging biological approach with the unique capability of specific targeting due to the precise light control with high spatial and temporal resolution. It uses selected light wavelengths to control and modulate the biological functions of cells, tissues, and organ levels. Optogenetics has been instrumental in different biomedical applications, including neuroscience, diabetes, and mitochondria, based on distinctive optical biomedical effects with light modulation. Nowadays, optogenetics in cardiology is rapidly evolving for the understanding and treatment of cardiovascular diseases. Several in vitro and in vivo research for cardiac optogenetics demonstrated visible progress. The optogenetics technique can be applied to address critical cardiovascular problems such as heart failure and arrhythmia. To this end, this paper reviews cardiac electrophysiology and the technical progress about experimental and clinical cardiac optogenetics and provides the background and evolution of cardiac optogenetics. We reviewed the literature to demonstrate the servo type, transfection efficiency, signal recording, and heart disease targets in optogenetic applications. Such literature review would hopefully expedite the progress of optogenetics in cardiology and further expect to translate into the clinical terminal in the future.
Collapse
|
3
|
Liu Y, Zhang H, Chen R, Wu Y, Yang X, Liu X, Zeng S, Guo W. UnaG as a reporter in adeno-associated virus-mediated gene transfer for biomedical imaging. JOURNAL OF BIOPHOTONICS 2020; 13:e202000182. [PMID: 32894647 DOI: 10.1002/jbio.202000182] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/11/2020] [Revised: 08/21/2020] [Accepted: 09/02/2020] [Indexed: 06/11/2023]
Abstract
Adeno-associated virus (AAV) is one of the most common gene transfer vectors, but it has a limited capacity. A smaller fluorescent protein is urgently needed since it is more suitable to act as a reporter in AAV. In this study, a bilirubin-dependent reporter smaller than EGFP, termed UnaG, was found to have the ability to label the neurons of a mouse brain as clearly as EGFP without the addition of exogenous bilirubin. We also found that UnaG's pH tolerance is better than that of EGFP; however, its fluorescence recovery after protonated quenching is not as good as that of EGFP. In addition, UnaG preserved its fluorescence better than EGFP in SeeDB clearing. Taken together, this study demonstrates that UnaG can act as a small intrinsically fluorescent reporter in the mouse brain without an additional ligand, thus providing an alternative over EGFP for AAV-mediated neuron labeling in mammals.
Collapse
Affiliation(s)
- Yurong Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Huimin Zhang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Ruixi Chen
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Wu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Xiong Yang
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Xiuli Liu
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Shaoqun Zeng
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Wenyan Guo
- Britton Chance Center for Biomedical Photonics, Wuhan National Laboratory for Optoelectronics-Huazhong University of Science and Technology, Wuhan, China
- MOE Key Laboratory for Biomedical Photonics, Collaborative Innovation Center for Biomedical Engineering, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
4
|
Abstract
Baculoviruses are arthropod-specific, enveloped viruses with circular, supercoiled double-stranded deoxyribonucleic acid genomes. While many viruses are studied to seek solutions for their adverse impact on human, veterinary, and plant health, the study of baculoviruses was stimulated initially by their potential utility to control insect pests. Later, the utility of baculovirus as gene expression vectors was evidenced leading to numerous applications. Several strategies are employed to obtain recombinant viruses that express large quantities of heterologous proteins. A major step forward was the development of bacmid technology (the construction of bacterial artificial chromosomes containing the genome of the baculovirus) which allows the manipulation of the baculovirus genome in bacteria. With this technology, foreign genes can be introduced into the bacmid by homologous and site-directed recombination or by transposition. Baculoviruses have been used to explore fundamental questions in molecular biology such as the nature of programmed cell-death. Moreover, the ability of baculoviruses to transduce mammalian cells led to the consideration of their use as gene-therapy and vaccine vectors. Strategies for genetic engineering of baculoviruses have been developed to meet the requirements of new application areas. Display of foreign proteins on the surface of virions or in nucleocapsid structures, the assembly of expressed proteins to form virus-like particles or protein complexes have been explored and validated as vaccines. The aim of this chapter is to update the areas of application of the baculoviruses in protein expression, alternative vaccine designs and gene therapy of infectious diseases and genetic disorders. Finally, we review the baculovirus-derived products on the market and in the pipeline for biomedical and veterinary use.
Collapse
|
5
|
Ryan CT, Ghanta RK. Commentary: Doubling down on adeno-associated viruses for cardiac gene therapy. J Thorac Cardiovasc Surg 2019; 159:1823-1824. [PMID: 31839232 DOI: 10.1016/j.jtcvs.2019.10.111] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Revised: 10/18/2019] [Accepted: 10/21/2019] [Indexed: 10/25/2022]
Affiliation(s)
- Christopher T Ryan
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex
| | - Ravi K Ghanta
- Division of Cardiothoracic Surgery, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, Tex.
| |
Collapse
|
6
|
Maes ME, Colombo G, Schulz R, Siegert S. Targeting microglia with lentivirus and AAV: Recent advances and remaining challenges. Neurosci Lett 2019; 707:134310. [PMID: 31158432 PMCID: PMC6734419 DOI: 10.1016/j.neulet.2019.134310] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2018] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
Abstract
Microglia have emerged as a critical component of neurodegenerative diseases. Genetic manipulation of microglia can elucidate their functional impact in disease. In neuroscience, recombinant viruses such as lentiviruses and adeno-associated viruses (AAVs) have been successfully used to target various cell types in the brain, although effective transduction of microglia is rare. In this review, we provide a short background of lentiviruses and AAVs, and strategies for designing recombinant viral vectors. Then, we will summarize recent literature on successful microglial transductions in vitro and in vivo, and discuss the current challenges. Finally, we provide guidelines for reporting the efficiency and specificity of viral targeting in microglia, which will enable the microglial research community to assess and improve methodologies for future studies.
Collapse
Affiliation(s)
- Margaret E Maes
- Institute of Science and Technology (IST) Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Gloria Colombo
- Institute of Science and Technology (IST) Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Rouven Schulz
- Institute of Science and Technology (IST) Austria, Am Campus 1, 3400 Klosterneuburg, Austria
| | - Sandra Siegert
- Institute of Science and Technology (IST) Austria, Am Campus 1, 3400 Klosterneuburg, Austria.
| |
Collapse
|
7
|
Abstract
Novel affinity agents with high specificity are needed to make progress in disease diagnosis and therapy. Over the last several years, peptides have been considered to have fundamental benefits over other affinity agents, such as antibodies, due to their fast blood clearance, low immunogenicity, rapid tissue penetration, and reproducible chemical synthesis. These features make peptides ideal affinity agents for applications in disease diagnostics and therapeutics for a wide variety of afflictions. Virus-derived peptide techniques provide a rapid, robust, and high-throughput way to identify organism-targeting peptides with high affinity and selectivity. Here, we will review viral peptide display techniques, how these techniques have been utilized to select new organism-targeting peptides, and their numerous biomedical applications with an emphasis on targeted imaging, diagnosis, and therapeutic techniques. In the future, these virus-derived peptides may be used as common diagnosis and therapeutics tools in local clinics.
Collapse
Affiliation(s)
- Mingying Yang
- Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou 310058, China
| | - Kegan Sunderland
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Chuanbin Mao
- Department of Chemistry & Biochemistry, Stephenson Life Science Research Center, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
- School of Materials Science and Engineering, Zhejiang University, Hangzhou, Zhejiang 310027, China
| |
Collapse
|
8
|
Xiao PJ, Mitchell AM, Huang L, Li C, Samulski RJ. Disruption of Microtubules Post-Virus Entry Enhances Adeno-Associated Virus Vector Transduction. Hum Gene Ther 2016; 27:309-24. [PMID: 26942476 DOI: 10.1089/hum.2016.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Perinuclear retention of viral particles is a poorly understood phenomenon observed during many virus infections. In this study, we investigated whether perinuclear accumulation acts as a barrier to limit recombinant adeno-associated virus (rAAV) transduction. After nocodazole treatment to disrupt microtubules at microtubule-organization center (MT-MTOC) after virus entry, we observed higher rAAV transduction. To elucidate the role of MT-MTOC in rAAV infection and study its underlying mechanisms, we demonstrated that rAAV's perinuclear localization was retained by MT-MTOC with fluorescent analysis, and enhanced rAAV transduction from MT-MTOC disruption was dependent on the rAAV capsid's nuclear import signals. Interestingly, after knocking down RhoA or inhibiting its downstream effectors (ROCK and Actin), MT-MTOC disruption failed to increase rAAV transduction or nuclear entry. These data suggest that enhancement of rAAV transduction is the result of increased trafficking to the nucleus via the RhoA-ROCK-Actin pathway. Ten-fold higher rAAV transduction was also observed by disrupting MT-MTOC in brain, liver, and tumor in vivo. In summary, this study indicates that virus perinuclear accumulation at MT-MTOC is a barrier-limiting parameter for effective rAAV transduction and defines a novel defense mechanism by which host cells restrain viral invasion.
Collapse
Affiliation(s)
- Ping-Jie Xiao
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,2 Cell and Developmental Biology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - Angela M Mitchell
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,3 Department of Microbiology and Immunology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - Lu Huang
- 4 Department of Statistics, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - Chengwen Li
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| | - R Jude Samulski
- 1 Gene Therapy Center, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina.,5 Department of Pharmacology, University of North Carolina at Chapel Hill , Chapel Hill, North Carolina
| |
Collapse
|
9
|
Expression mediated by three partial sequences of the human tyrosine hydroxylase promoter in vivo. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2016; 3:16062. [PMID: 27689101 PMCID: PMC5031092 DOI: 10.1038/mtm.2016.62] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2016] [Revised: 06/13/2016] [Accepted: 07/21/2016] [Indexed: 12/25/2022]
Abstract
The use of viral vectors to transfect postmitotic neurons has provided an important research tool, and it offers promise for treatment of neurologic disease. The utility of vectors is enhanced by the use of selective promoters that permit control of the cellular site of expression. One potential clinical application is in the neurorestorative treatment of Parkinson’s disease by the induction of new axon growth. However, many of the genes with an ability to restore axons have oncogenic potential. Therefore, clinical safety would be enhanced by restriction of expression to neurons affected by the disease, particularly dopamine neurons. To achieve this goal we have evaluated in vivo three partial sequences of the promoter for human tyrosine hydroxylase, the rate limiting enzyme in catecholamine synthesis. All sequences induced expression in dopamine neurons. None of them induced expression in glia or in nondopaminergic neurons in striatum or cortex. We conclude that these sequences have potential use for targeting dopamine neurons in research and clinical applications.
Collapse
|
10
|
Winters AA, Bou-Ghannam S, Thorp H, Hawayek JA, Atkinson DL, Bartlett CE, Silva FJ, Hsu EW, Moreno AP, Grainger DA, Patel AN. Evaluation of Multiple Biological Therapies for Ischemic Cardiac Disease. Cell Transplant 2016; 25:1591-1607. [DOI: 10.3727/096368916x691501] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Affiliation(s)
| | - Sophia Bou-Ghannam
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
| | - Hallie Thorp
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
| | - Jose A. Hawayek
- University of Utah School of Medicine, Salt Lake City, UT, USA
| | | | | | | | - Edward W. Hsu
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
| | - Alonso P. Moreno
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
- Nora Eccles Cardiovascular and Training Research Institute, Salt Lake City, UT, USA
| | - David A. Grainger
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
| | - Amit N. Patel
- University of Utah School of Medicine, Salt Lake City, UT, USA
- Department of Bioengineering, University of Utah, Salt Lake City, UT, USA
| |
Collapse
|
11
|
Arden E, Metzger JM. Inexpensive, serotype-independent protocol for native and bioengineered recombinant adeno-associated virus purification. J Biol Methods 2016; 3. [PMID: 27294171 PMCID: PMC4902285 DOI: 10.14440/jbm.2016.102] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Recombinant adeno-associated virus (AAV) is a valuable and often used gene therapy vector. With increased demand for highly purified virus comes the need for a standardized purification procedure that is applicable across many serotypes and includes bioengineered viruses. Currently cesium chloride banding or affinity chromatography are the predominate forms of purification. These approaches expose the final purified virus to toxic contaminants or are highly capsid dependent and may require significant optimization to isolate purified AAV. These methods may also limit crude viral lysate processing volume resulting in a significant loss of viral titer. To circumvent these issues, we have developed an AAV purification protocol independent of toxic compounds, supernatant volume and capsid moiety. This purification method standardizes virus purification across native serotype and bioengineered mosaic capsids.
Collapse
Affiliation(s)
- Erik Arden
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Joseph M Metzger
- Department of Integrative Biology & Physiology, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| |
Collapse
|
12
|
Dogbevia GK, Roβmanith M, Sprengel R, Hasan MT. Flexible, AAV-equipped Genetic Modules for Inducible Control of Gene Expression in Mammalian Brain. MOLECULAR THERAPY. NUCLEIC ACIDS 2016; 5:e309. [PMID: 27070301 PMCID: PMC5014524 DOI: 10.1038/mtna.2016.23] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2015] [Accepted: 03/01/2016] [Indexed: 12/17/2022]
Abstract
Controlling gene expression in mammalian brain is of utmost importance to causally link the role of gene function to cell circuit dynamics under normal conditions and disease states. We have developed recombinant adeno-associated viruses equipped with tetracycline-controlled genetic switches for inducible and reversible control of gene expression in a cell type specific and brain subregion selective manner. Here, we characterize a two-virus approach to efficiently and reliably switch gene expression on and off, repetitively, both in vitro and in vivo. Our recombinant adeno-associated virus (rAAV)-Tet approach is highly flexible and it has great potential for application in basic and biomedical neuroscience research and gene therapy.
Collapse
Affiliation(s)
- Godwin K Dogbevia
- Department of Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany.,Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck, Lübeck, Germany
| | - Martin Roβmanith
- Department of Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany
| | - Rolf Sprengel
- Max Planck Research Group at the Institute for Anatomy and Cell Biology, Heidelberg University, Heidelberg, Germany
| | - Mazahir T Hasan
- Department of Neurobiology, Max Planck Institute for Medical Research, Heidelberg, Germany.,Charité-Universitätsmedizin, NeuroCure Cluster of Excellence, Berlin, Germany
| |
Collapse
|
13
|
Mateu MG. Assembly, Engineering and Applications of Virus-Based Protein Nanoparticles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 940:83-120. [PMID: 27677510 DOI: 10.1007/978-3-319-39196-0_5] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Viruses and their protein capsids can be regarded as biologically evolved nanomachines able to perform multiple, complex biological functions through coordinated mechano-chemical actions during the infectious cycle. The advent of nanoscience and nanotechnology has opened up, in the last 10 years or so, a vast number of novel possibilities to exploit engineered viral capsids as protein-based nanoparticles for multiple biomedical, biotechnological or nanotechnological applications. This chapter attempts to provide a broad, updated overview on the self-assembly and engineering of virus capsids, and on applications of virus-based nanoparticles. Different sections provide outlines on: (i) the structure, functions and properties of virus capsids; (ii) general approaches for obtaining assembled virus particles; (iii) basic principles and events related to virus capsid self-assembly; (iv) genetic and chemical strategies for engineering virus particles; (v) some applications of engineered virus particles being developed; and (vi) some examples on the engineering of virus particles to modify their physical properties, in order to improve their suitability for different uses.
Collapse
Affiliation(s)
- Mauricio G Mateu
- Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Universidad Autónoma de Madrid, Campus de Cantoblanco, 28049, Madrid, Spain. .,Department of Molecular Biology, Universidad Autónoma de Madrid, Campus de Cantoblanco, 28049, Madrid, Spain.
| |
Collapse
|
14
|
Zhang C, Yao T, Zheng Y, Li Z, Zhang Q, Zhang L, Zhou D. Development of next generation adeno-associated viral vectors capable of selective tropism and efficient gene delivery. Biomaterials 2015; 80:134-145. [PMID: 26708090 DOI: 10.1016/j.biomaterials.2015.11.066] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/11/2015] [Accepted: 11/29/2015] [Indexed: 12/12/2022]
Abstract
Virus-based nanoparticles have shown promise as vehicles for delivering therapeutic genes. However, the rational design of viral vectors that enable selective tropism towards particular types of cells and tissues remains challenging. Here, we explored structural-functional relationships of the adeno-associated virus 2 (AAV2) vector by expanding its genetic code during production. As a proof-of-principle, an azide moiety was strategically displayed on the vector capsid as a bioorthogonal chemical reporter. Upon bioorthogonal conjugation of AAV2 with fluorophores and cyclic arginyl-glycyl-aspartic acid ligands at certain modifiable sites, we characterized in vitro and in vivo AAV2 movement and enhanced tropism selectivity towards integrin-expressing tumor cells. Targeting AAV2 vectors resulted in selective killing of U87 glioblastoma cells and derived xenografts via the herpes simplex virus suicide gene thymidine kinase, with the potency of ganciclovir being increased by 25-fold. Our results demonstrated successful rational modification of AAV2 as a targeting delivery vehicle, establishing a facile platform for precision engineering of virus-based nanoparticles in basic research and therapeutic applications.
Collapse
Affiliation(s)
- Chuanling Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Tianzhuo Yao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Yongxiang Zheng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Zhongjun Li
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Qiang Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Lihe Zhang
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China
| | - Demin Zhou
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, 100191, China.
| |
Collapse
|
15
|
Rincon MY, VandenDriessche T, Chuah MK. Gene therapy for cardiovascular disease: advances in vector development, targeting, and delivery for clinical translation. Cardiovasc Res 2015; 108:4-20. [PMID: 26239654 PMCID: PMC4571836 DOI: 10.1093/cvr/cvv205] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 07/22/2015] [Indexed: 01/06/2023] Open
Abstract
Gene therapy is a promising modality for the treatment of inherited and acquired cardiovascular diseases. The identification of the molecular pathways involved in the pathophysiology of heart failure and other associated cardiac diseases led to encouraging preclinical gene therapy studies in small and large animal models. However, the initial clinical results yielded only modest or no improvement in clinical endpoints. The presence of neutralizing antibodies and cellular immune responses directed against the viral vector and/or the gene-modified cells, the insufficient gene expression levels, and the limited gene transduction efficiencies accounted for the overall limited clinical improvements. Nevertheless, further improvements of the gene delivery technology and a better understanding of the underlying biology fostered renewed interest in gene therapy for heart failure. In particular, improved vectors based on emerging cardiotropic serotypes of the adeno-associated viral vector (AAV) are particularly well suited to coax expression of therapeutic genes in the heart. This led to new clinical trials based on the delivery of the sarcoplasmic reticulum Ca2+-ATPase protein (SERCA2a). Though the first clinical results were encouraging, a recent Phase IIb trial did not confirm the beneficial clinical outcomes that were initially reported. New approaches based on S100A1 and adenylate cyclase 6 are also being considered for clinical applications. Emerging paradigms based on the use of miRNA regulation or CRISPR/Cas9-based genome engineering open new therapeutic perspectives for treating cardiovascular diseases by gene therapy. Nevertheless, the continuous improvement of cardiac gene delivery is needed to allow the use of safer and more effective vector doses, ultimately bringing gene therapy for heart failure one step closer to reality.
Collapse
Affiliation(s)
- Melvin Y Rincon
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Centro de Investigaciones, Fundacion Cardiovascular de Colombia, Floridablanca, Colombia
| | - Thierry VandenDriessche
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| | - Marinee K Chuah
- Department of Gene Therapy and Regenerative Medicine, Free University of Brussels (VUB), Building D, room D306, Laarbeeklaan 103, Brussels, Belgium Center for Molecular and Vascular Biology, Department of Cardiovascular Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|
16
|
Fan Q, Hu W, Ohta AT. Light-induced microbubble poration of localized cells. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2015; 2013:4482-5. [PMID: 24110729 DOI: 10.1109/embc.2013.6610542] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Molecular delivery into localized NIH/3T3 cells was achieved with microbubbles produced by laser pulses focused on an optically absorbent substrate. The laser-induced bubble expansion and contraction resulted in cell poration. The microbubbles are localized at the laser focal point, so molecular delivery can be directed at specific localized cells. This was demonstrated with the delivery of 3-kDa FITC-Dextran. Single-cell molecular delivery was achieved, even in the presence of nearby cells. The efficiency of the cell poration was up to 95%, with a corresponding cell viability of 98%.
Collapse
|
17
|
Benzler J, Ganjam GK, Pretz D, Oelkrug R, Koch CE, Legler K, Stöhr S, Culmsee C, Williams LM, Tups A. Central inhibition of IKKβ/NF-κB signaling attenuates high-fat diet-induced obesity and glucose intolerance. Diabetes 2015; 64:2015-27. [PMID: 25626735 DOI: 10.2337/db14-0093] [Citation(s) in RCA: 97] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2014] [Accepted: 01/20/2015] [Indexed: 11/13/2022]
Abstract
Metabolic inflammation in the central nervous system might be causative for the development of overnutrition-induced metabolic syndrome and related disorders, such as obesity, leptin and insulin resistance, and type 2 diabetes. Here we investigated whether nutritive and genetic inhibition of the central IκB kinase β (IKKβ)/nuclear factor-κB (NF-κB) pathway in diet-induced obese (DIO) and leptin-deficient mice improves these metabolic impairments. A known prominent inhibitor of IKKβ/NF-κB signaling is the dietary flavonoid butein. We initially determined that oral, intraperitoneal, and intracerebroventricular administration of this flavonoid improved glucose tolerance and hypothalamic insulin signaling. The dose-dependent glucose-lowering capacity was profound regardless of whether obesity was caused by leptin deficiency or high-fat diet (HFD). To confirm the apparent central role of IKKβ/NF-κB signaling in the control of glucose and energy homeostasis, we genetically inhibited this pathway in neurons of the arcuate nucleus, one key center for control of energy homeostasis, via specific adeno-associated virus serotype 2-mediated overexpression of IκBα, which inhibits NF-κB nuclear translocation. This treatment attenuated HFD-induced body weight gain, body fat mass accumulation, increased energy expenditure, and reduced arcuate suppressor of cytokine signaling 3 expression, indicative for enhanced leptin signaling. These results reinforce a specific role of central proinflammatory IKKβ/NF-κB signaling in the development and potential treatment of DIO-induced comorbidities.
Collapse
Affiliation(s)
- Jonas Benzler
- Department of Animal Physiology, Faculty of Biology, Philipps-University Marburg, Marburg, Germany
| | - Goutham K Ganjam
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Dominik Pretz
- Department of Animal Physiology, Faculty of Biology, Philipps-University Marburg, Marburg, Germany
| | - Rebecca Oelkrug
- Department of Animal Physiology, Faculty of Biology, Philipps-University Marburg, Marburg, Germany
| | - Christiane E Koch
- Department of Animal Physiology, Faculty of Biology, Philipps-University Marburg, Marburg, Germany
| | - Karen Legler
- Department of Animal Physiology, Faculty of Biology, Philipps-University Marburg, Marburg, Germany
| | - Sigrid Stöhr
- Department of Animal Physiology, Faculty of Biology, Philipps-University Marburg, Marburg, Germany
| | - Carsten Culmsee
- Institute for Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Philipps-University Marburg, Marburg, Germany
| | - Lynda M Williams
- Metabolic Health Group, Rowett Institute of Nutrition and Health, University of Aberdeen, Aberdeen, U.K
| | - Alexander Tups
- Department of Animal Physiology, Faculty of Biology, Philipps-University Marburg, Marburg, Germany Centre for Neuroendocrinology, Department of Physiology, School of Medical Sciences, University of Otago, Dunedin, New Zealand
| |
Collapse
|
18
|
Dogbevia GK, Marticorena-Alvarez R, Bausen M, Sprengel R, Hasan MT. Inducible and combinatorial gene manipulation in mouse brain. Front Cell Neurosci 2015; 9:142. [PMID: 25954155 PMCID: PMC4404871 DOI: 10.3389/fncel.2015.00142] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2015] [Accepted: 03/25/2015] [Indexed: 12/30/2022] Open
Abstract
We have deployed recombinant adeno-associated viruses equipped with tetracycline-controlled genetic switches to manipulate gene expression in mouse brain. Here, we show a combinatorial genetic approach for inducible, cell type-specific gene expression and Cre/loxP mediated gene recombination in different brain regions. Our chemical-genetic approach will help to investigate 'when', 'where', and 'how' gene(s) control neuronal circuit dynamics, and organize, for example, sensory signal processing, learning and memory, and behavior.
Collapse
Affiliation(s)
- Godwin K Dogbevia
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research Heidelberg, Germany ; Institute of Experimental and Clinical Pharmacology and Toxicology, University of Lübeck Lübeck, Germany
| | | | - Melanie Bausen
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research Heidelberg, Germany
| | - Rolf Sprengel
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research Heidelberg, Germany
| | - Mazahir T Hasan
- Department of Molecular Neurobiology, Max Planck Institute for Medical Research Heidelberg, Germany ; NeuroCure Cluster of Excellence, Charité-Universitätsmedizin Berlin, Germany
| |
Collapse
|
19
|
Krook-Magnuson E, Soltesz I. Beyond the hammer and the scalpel: selective circuit control for the epilepsies. Nat Neurosci 2015; 18:331-8. [PMID: 25710834 PMCID: PMC4340083 DOI: 10.1038/nn.3943] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Accepted: 01/03/2015] [Indexed: 12/12/2022]
Abstract
Current treatment options for epilepsy are inadequate, as too many patients suffer from uncontrolled seizures and from negative side effects of treatment. In addition to these clinical challenges, our scientific understanding of epilepsy is incomplete. Optogenetic and designer receptor technologies provide unprecedented and much needed specificity, allowing for spatial, temporal and cell type-selective modulation of neuronal circuits. Using such tools, it is now possible to begin to address some of the fundamental unanswered questions in epilepsy, to dissect epileptic neuronal circuits and to develop new intervention strategies. Such specificity of intervention also has the potential for direct therapeutic benefits, allowing healthy tissue and network functions to continue unaffected. In this Perspective, we discuss promising uses of these technologies for the study of seizures and epilepsy, as well as potential use of these strategies for clinical therapies.
Collapse
Affiliation(s)
- Esther Krook-Magnuson
- Department of Anatomy & Neurobiology, University of California, Irvine, 192 Irvine Hall, Irvine, California 92697
| | - Ivan Soltesz
- Department of Anatomy & Neurobiology, University of California, Irvine, 192 Irvine Hall, Irvine, California 92697
| |
Collapse
|
20
|
Fan Q, Hu W, Ohta AT. Efficient single-cell poration by microsecond laser pulses. LAB ON A CHIP 2015; 15:581-8. [PMID: 25421758 PMCID: PMC4304703 DOI: 10.1039/c4lc00943f] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Payloads including FITC-Dextran dye and plasmids were delivered into NIH/3T3 fibroblasts using microbubbles produced by microsecond laser pulses to induce pores in the cell membranes. Two different operational modes were used to achieve molecular delivery. Smaller molecules, such as the FITC-Dextran dye, were delivered via a scanning-laser mode. The poration efficiency and the cell viability were both 95.1 ± 3.0%. Relatively larger GFP plasmids can be delivered efficiently via a fixed-laser mode, which is a more vigorous method that can create larger transient pores in the cell membrane. The transfection efficiency of 5.7 kb GFP plasmid DNA can reach to 86.7 ± 3.3%. Using this cell poration system, targeted single cells can be porated with high resolution, and cells can be porated in arbitrary patterns.
Collapse
Affiliation(s)
- Qihui Fan
- Department of Mechanical Engineering, University of Hawaii at Manoa, 2540 Dole Street, Holmes Hall 302, Honolulu, USA., Fax: +1-808-956-3427; Tel: 808-956-3427
| | - Wenqi Hu
- Department of Electrical Engineering, University of Hawaii at Manoa, 2540 Dole Street, Holmes Hall 483, Honolulu, USA
| | - Aaron T. Ohta
- Department of Electrical Engineering, University of Hawaii at Manoa, 2540 Dole Street, Holmes Hall 483, Honolulu, USA
| |
Collapse
|
21
|
Maguire CA, Ramirez SH, Merkel SF, Sena-Esteves M, Breakefield XO. Gene therapy for the nervous system: challenges and new strategies. Neurotherapeutics 2014; 11:817-39. [PMID: 25159276 PMCID: PMC4391389 DOI: 10.1007/s13311-014-0299-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Current clinical treatments for central nervous system (CNS) diseases, such as Parkinson's disease and glioblastoma do not halt disease progression and have significant treatment morbidities. Gene therapy has the potential to "permanently" correct disease by bringing in a normal gene to correct a mutant gene deficiency, knocking down mRNA of mutant alleles, and inducing cell-death in cancer cells using transgenes encoding apoptosis-inducing proteins. Promising results in clinical trials of eye disease (Leber's congenital aumorosis) and Parkinson's disease have shown that gene-based neurotherapeutics have great potential. The recent development of genome editing technology, such as zinc finger nucleases, TALENS, and CRISPR, has made the ultimate goal of gene correction a step closer. This review summarizes the challenges faced by gene-based neurotherapeutics and the current and recent strategies designed to overcome these barriers. We have chosen the following challenges to focus on in this review: (1) delivery vehicles (both virus and nonviral), (2) use of promoters for vector-mediated gene expression in CNS, and (3) delivery across the blood-brain barrier. The final section (4) focuses on promising pre-clinical/clinical studies of neurotherapeutics.
Collapse
Affiliation(s)
- Casey A Maguire
- Department of Neurology, Massachusetts General Hospital, and Neuroscience Program, Harvard Medical School, Molecular Neurogenetics Unit, 13th Street, Building 149, Charlestown, MA, 02129, USA,
| | | | | | | | | |
Collapse
|
22
|
Belzung C, Turiault M, Griebel G. Optogenetics to study the circuits of fear- and depression-like behaviors: A critical analysis. Pharmacol Biochem Behav 2014; 122:144-57. [DOI: 10.1016/j.pbb.2014.04.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/06/2013] [Revised: 03/31/2014] [Accepted: 04/03/2014] [Indexed: 02/05/2023]
|
23
|
Kaestner L, Scholz A, Tian Q, Ruppenthal S, Tabellion W, Wiesen K, Katus HA, Müller OJ, Kotlikoff MI, Lipp P. Genetically encoded Ca2+ indicators in cardiac myocytes. Circ Res 2014; 114:1623-39. [PMID: 24812351 DOI: 10.1161/circresaha.114.303475] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Genetically encoded Ca(2+) indicators constitute a powerful set of tools to investigate functional aspects of Ca(2+) signaling in isolated cardiomyocytes, cardiac tissue, and whole hearts. Here, we provide an overview of the concepts, experiences, state of the art, and ongoing developments in the use of genetically encoded Ca(2+) indicators for cardiac cells and heart tissue. This review is supplemented with in vivo viral gene transfer experiments and comparisons of available genetically encoded Ca(2+) indicators with each other and with the small molecule dye Fura-2. In the context of cardiac myocytes, we provide guidelines for selecting a genetically encoded Ca(2+) indicator. For future developments, we discuss improvements of a broad range of properties, including photophysical properties such as spectral spread and biocompatibility, as well as cellular and in vivo applications.
Collapse
Affiliation(s)
- Lars Kaestner
- From the Institute for Molecular Cell Biology and Research Center for Molecular Imaging and Screening, School of Medicine, Saarland University, Homburg-Saar, Germany (L.K., A.S., Q.T., S.R., W.T., K.W., P.L.); Department of Internal Medicine III, University of Heidelberg, Heidelberg, Germany (H.A.K., O.J.M.); DZHK (German Centre for Cardiovascular Research), Partner Site Heidelberg/Mannheim, Germany (H.A.K., O.J.M.); and Biomedical Sciences Department, College of Veterinary Medicine, Cornell University, Ithaca, NY (M.I.K.)
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Evans-Galea MV, Pébay A, Dottori M, Corben LA, Ong SH, Lockhart PJ, Delatycki MB. Cell and gene therapy for Friedreich ataxia: progress to date. Hum Gene Ther 2014; 25:684-93. [PMID: 24749505 DOI: 10.1089/hum.2013.180] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Neurodegenerative disorders such as Friedreich ataxia (FRDA) present significant challenges in developing effective therapeutic intervention. Current treatments aim to manage symptoms and thus improve quality of life, but none can cure, nor are proven to slow, the neurodegeneration inherent to this disease. The primary clinical features of FRDA include progressive ataxia and shortened life span, with complications of cardiomyopathy being the major cause of death. FRDA is most commonly caused by an expanded GAA trinucleotide repeat in the first intron of FXN that leads to reduced levels of frataxin, a mitochondrial protein important for iron metabolism. The GAA expansion in FRDA does not alter the coding sequence of FXN. It results in reduced production of structurally normal frataxin, and hence any increase in protein level is expected to be therapeutically beneficial. Recently, there has been increased interest in developing novel therapeutic applications like cell and/or gene therapies, and these cutting-edge applications could provide effective treatment options for FRDA. Importantly, since individuals with FRDA produce frataxin at low levels, increased expression should not elicit an immune response. Here we review the advances to date and highlight the future potential for cell and gene therapy to treat this debilitating disease.
Collapse
Affiliation(s)
- Marguerite V Evans-Galea
- 1 Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute , Parkville Victoria 3052, Australia
| | | | | | | | | | | | | |
Collapse
|
25
|
Mah CS, Soustek MS, Todd AG, McCall A, Smith BK, Corti M, Falk DJ, Byrne BJ. Adeno-associated virus-mediated gene therapy for metabolic myopathy. Hum Gene Ther 2014; 24:928-36. [PMID: 24164240 DOI: 10.1089/hum.2013.2514] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Metabolic myopathies are a diverse group of rare diseases in which impaired breakdown of stored energy leads to profound muscle dysfunction ranging from exercise intolerance to severe muscle wasting. Metabolic myopathies are largely caused by functional deficiency of a single gene and are generally subcategorized into three major types of metabolic disease: mitochondrial, lipid, or glycogen. Treatment varies greatly depending on the biochemical nature of the disease, and unfortunately no definitive treatments exist for metabolic myopathy. Since this group of diseases is inherited, gene therapy is being explored as an approach to personalized medical treatment. Adeno-associated virus-based vectors in particular have shown to be promising in the treatment of several forms of metabolic myopathy. This review will discuss the most recent advances in gene therapy efforts for the treatment of metabolic myopathies.
Collapse
Affiliation(s)
- Cathryn S Mah
- 1 Powell Gene Therapy Center, Department of Pediatrics, College of Medicine, University of Florida , Gainesville, FL 32610
| | | | | | | | | | | | | | | |
Collapse
|
26
|
Fan Q, Hu W, Ohta AT. Laser-induced microbubble poration of localized single cells. LAB ON A CHIP 2014; 14:1572-8. [PMID: 24632785 PMCID: PMC4004443 DOI: 10.1039/c3lc51394g] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Laser-induced microbubbles were used to porate the cell membranes of localized single NIH/3T3 fibroblasts. Microsecond laser pulses were focused on an optically absorbent substrate, creating a vapour microbubble that oscillated in size at the laser focal point in a fluidic chamber. The shear stress accompanying the bubble size oscillation was able to porate nearby cells. Cell poration was demonstrated with the delivery of FITC-dextran dye with various molecular weights. Under optimal poration conditions, the cell poration efficiency was up to 95.2 ± 4.8%, while maintaining 97.6 ± 2.4% cell viability. The poration system is able to target a single cell without disturbing surrounding cells.
Collapse
Affiliation(s)
- Qihui Fan
- Department of Mechanical Engineering, University of Hawaii at Manoa, 2540 Dole Street, Holmes Hall 302, Honolulu, USA. Fax: +1-808-956-3427; Tel: 808-956-3427
| | - Wenqi Hu
- Department of Electrical Engineering, University of Hawaii at Manoa, 2540 Dole Street, Holmes Hall 483, Honolulu, USA
| | - Aaron T. Ohta
- Department of Electrical Engineering, University of Hawaii at Manoa, 2540 Dole Street, Holmes Hall 483, Honolulu, USA
| |
Collapse
|
27
|
Gene Therapy Used in Cancer Treatment. Biomedicines 2014; 2:149-162. [PMID: 28548065 PMCID: PMC5423469 DOI: 10.3390/biomedicines2020149] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2014] [Revised: 03/12/2014] [Accepted: 03/18/2014] [Indexed: 12/13/2022] Open
Abstract
Cancer has been, from the beginning, a target of intense research for gene therapy approaches. Currently, more than 60% of all on-going clinical gene therapy trials worldwide are targeting cancer. Indeed, there is a clear unmet medical need for novel therapies. This is further urged by the fact that current conventional cancer therapies are frequently troubled by their toxicities. Different gene therapy strategies have been employed for cancer, such as pro-drug activating suicide gene therapy, anti-angiogenic gene therapy, oncolytic virotherapy, gene therapy-based immune modulation, correction/compensation of gene defects, genetic manipulation of apoptotic and tumor invasion pathways, antisense, and RNAi strategies. Cancer types, which have been targeted with gene therapy, include brain, lung, breast, pancreatic, liver, colorectal, prostate, bladder, head and neck, skin, ovarian, and renal cancer. Currently, two cancer gene therapy products have received market approval, both of which are in China. In addition, the stimulation of the host’s immune system, using gene therapeutic approaches, has gained vast interest. The intention of this review is to point out the most commonly viral and non-viral vectors and methods used in cancer gene therapy, as well as highlight some key results achieved in clinical trials.
Collapse
|
28
|
Ali HM, Urbinati G, Raouane M, Massaad-Massade L. Significance and applications of nanoparticles in siRNA delivery for cancer therapy. Expert Rev Clin Pharmacol 2014; 5:403-12. [PMID: 22943120 DOI: 10.1586/ecp.12.33] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Hafiz Muhammad Ali
- UMR CNRS 8203, Institut Gustave Roussy, 114 rue Edouard Vaillant, 94805 Villejuif, France
| | | | | | | |
Collapse
|
29
|
Ma H, Zhang Y, Wang H, Han C, Lei R, Zhang L, Yang Z, Rao L, Qing H, Xiang J, Deng Y. Effect and mechanism of Mitomycin C combined with recombinant adeno-associated virus type II against glioma. Int J Mol Sci 2013; 15:1-14. [PMID: 24451124 PMCID: PMC3907794 DOI: 10.3390/ijms15010001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Revised: 12/09/2013] [Accepted: 12/09/2013] [Indexed: 11/25/2022] Open
Abstract
The effect of chemotherapy drug Mitomycin C (MMC) in combination with recombinant adeno-associated virus II (rAAV2) in cancer therapy was investigated, and the mechanism of MMC affecting rAAV2’s bioactivity was also studied. The combination effect was evaluated by the level of GFP and TNF expression in a human glioma cell line, and the mechanism of MMC effects on rAAV mediated gene expression was investigated by AAV transduction related signal molecules. C57 and BALB/c nude mice were injected with rAAV-EGFP or rAAV-TNF alone, or mixed with MMC, to evaluate the effect of MMC on AAV-mediated gene expression and tumor suppression. MMC was shown to improve the infection activity of rAAV2 both in vitro and in vivo. Enhancement was found to be independent of initial rAAV2 receptor binding stage or subsequent second-strand synthesis of target DNA, but was related to cell cycle retardation followed by blocked genome degradation. In vivo injection of MMC combined with rAAV2 into the tumors of the animals resulted in significant suppression of tumor growth. It was thus demonstrated for the first time that MMC could enhance the expression level of the target gene mediated by rAAV2. The combination of rAAV2 and MMC may be a promising strategy in cancer therapy.
Collapse
Affiliation(s)
- Hong Ma
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Yunjia Zhang
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Hailong Wang
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Chuanhui Han
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Runhong Lei
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Lei Zhang
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Zuye Yang
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Ling Rao
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Jim Xiang
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| | - Yulin Deng
- School of Life Science, Beijing Institute of Technology, Haidian District, Beijing 100081, China.
| |
Collapse
|
30
|
The role of apoptosis in immune hyporesponsiveness following AAV8 liver gene transfer. Mol Ther 2013; 21:2227-35. [PMID: 24126962 DOI: 10.1038/mt.2013.94] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 03/28/2013] [Indexed: 12/13/2022] Open
Abstract
Gene therapy provides a significant opportunity to treat a variety of inherited and acquired diseases. However, adverse immune responses toward the adeno-associated virus (AAV) antigens may limit its success. The mechanisms responsible for immunity or tolerance toward AAV-encoded transgene products remain poorly defined. Studies in mice demonstrate that AAV2/8 gene transfer to liver is associated with immunological hyporesponsiveness toward both AAV vector and antigenic transgene product. To evaluate the role of activation-induced cell death (AICD) and cytokine withdrawal (intrinsic cell death) in the deletion of mature T lymphocytes, we compared immunological responses in hepatic AAV2/8 transfer in murine recipients lacking the Fas receptor, and recipients overexpressing Bcl-xL, to WT murine counterparts. Prolonged transgene expression was dependent on both Fas signaling and Bcl-xL-regulated apoptosis in T cells. Abrogation of intrinsic cell death enhanced Th1 responses, whereas AICD functioned to limit neutralizing antibody production toward AAV2/8. In addition, immune hyporesponsiveness and stable transgene expression was dependent on upregulation of FasL expression on transduced hepatocytes and a corresponding apoptosis of infiltrating Fas (+) cells. These data provide evidence that both AICD and apoptosis due to cytokine withdrawal of lymphocytes are essential for immune hyporesponsiveness toward hepatic AAV2/8-encoded transgene product in the setting of liver gene transfer.
Collapse
|
31
|
Tellez J, Van Vliet K, Tseng YS, Finn JD, Tschernia N, Almeida-Porada G, Arruda VR, Agbandje-McKenna M, Porada CD. Characterization of naturally-occurring humoral immunity to AAV in sheep. PLoS One 2013; 8:e75142. [PMID: 24086458 PMCID: PMC3782463 DOI: 10.1371/journal.pone.0075142] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Accepted: 08/09/2013] [Indexed: 11/18/2022] Open
Abstract
AAV vectors have shown great promise for clinical gene therapy (GT), but pre-existing human immunity against the AAV capsid often limits transduction. Thus, testing promising AAV-based GT approaches in an animal model with similar pre-existing immunity could better predict clinical outcome. Sheep have long been used for basic biological and preclinical studies. Moreover, we have re-established a line of sheep with severe hemophilia A (HA). Given the impetus to use AAV-based GT to treat hemophilia, we characterized the pre-existing ovine humoral immunity to AAV. ELISA revealed naturally-occurring antibodies to AAV1, AAV2, AAV5, AAV6, AAV8, and AAV9. For AAV2, AAV8, and AAV9 these inhibit transduction in a luciferase-based neutralization assay. Epitope mapping identified peptides that were common to the capsids of all AAV serotypes tested (AAV2, AAV5, AAV8 and AAV9), with each animal harboring antibodies to unique and common capsid epitopes. Mapping using X-ray crystallographic AAV capsid structures demonstrated that these antibodies recognized both surface epitopes and epitopes located within regions of the capsid that are internal or buried in the capsid structure. These results suggest that sheep harbor endogenous AAV, which induces immunity to both intact capsid and to capsid epitopes presented following proteolysis during the course of infection. In conclusion, their clinically relevant physiology and the presence of naturally-occurring antibodies to multiple AAV serotypes collectively make sheep a unique model in which to study GT for HA, and other diseases, and develop strategies to circumvent the clinically important barrier of pre-existing AAV immunity.
Collapse
Affiliation(s)
- Joseph Tellez
- Department of Animal Biotechnology, University of Nevada, Reno, Nevada, United States of America
| | - Kim Van Vliet
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States of America
| | - Yu-Shan Tseng
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States of America
| | - Jonathan D. Finn
- University of Pennsylvania School of Medicine, the Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Nick Tschernia
- Department of Animal Biotechnology, University of Nevada, Reno, Nevada, United States of America
| | - Graça Almeida-Porada
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina, United States of America
| | - Valder R. Arruda
- University of Pennsylvania School of Medicine, the Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, United States of America
| | - Mavis Agbandje-McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, United States of America
| | - Christopher D. Porada
- Wake Forest Institute for Regenerative Medicine, Winston-Salem, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
32
|
Smedemark-Margulies N, Trapani JG. Tools, methods, and applications for optophysiology in neuroscience. Front Mol Neurosci 2013; 6:18. [PMID: 23882179 PMCID: PMC3713398 DOI: 10.3389/fnmol.2013.00018] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2013] [Accepted: 06/27/2013] [Indexed: 11/13/2022] Open
Abstract
The advent of optogenetics and genetically encoded photosensors has provided neuroscience researchers with a wealth of new tools and methods for examining and manipulating neuronal function in vivo. There exists now a wide range of experimentally validated protein tools capable of modifying cellular function, including light-gated ion channels, recombinant light-gated G protein-coupled receptors, and even neurotransmitter receptors modified with tethered photo-switchable ligands. A large number of genetically encoded protein sensors have also been developed to optically track cellular activity in real time, including membrane-voltage-sensitive fluorophores and fluorescent calcium and pH indicators. The development of techniques for controlled expression of these proteins has also increased their utility by allowing the study of specific populations of cells. Additionally, recent advances in optics technology have enabled both activation and observation of target proteins with high spatiotemporal fidelity. In combination, these methods have great potential in the study of neural circuits and networks, behavior, animal models of disease, as well as in high-throughput ex vivo studies. This review collects some of these new tools and methods and surveys several current and future applications of the evolving field of optophysiology.
Collapse
|
33
|
Affiliation(s)
- Scott Banta
- Department of Chemical Engineering, Columbia University, New York, NY 10027;
| | - Kevin Dooley
- Department of Chemical Engineering, Columbia University, New York, NY 10027;
| | - Oren Shur
- Department of Chemical Engineering, Columbia University, New York, NY 10027;
- Current affiliation: Boston Consulting Group, New York, NY 10022
| |
Collapse
|
34
|
Abstract
Heart failure is increasing in incidence throughout the world, especially in industrialized countries. Although the current therapeutic modalities have been successful in stabilizing the course of heart failure, morbidity and mortality remain quite high and there remains a great need for innovative breakthroughs that will offer new treatment strategies for patients with advanced forms of the disease. The past few years have witnessed a greater understanding of the molecular underpinnings of the failing heart, paving the way for novel strategies in modulating the cellular environment. As such, gene therapy has recently emerged as a powerful tool offering the promise of a new paradigm for alleviating heart failure. Current gene therapy research for heart failure is focused on exploring potential cellular targets and preclinical and clinical studies are ongoing toward the realization of this goal. Efforts also include the development of sophisticated viral vectors and vector delivery methods for efficient transduction of cardiomyocytes.
Collapse
Affiliation(s)
- Razmig Garo Kratlian
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | |
Collapse
|
35
|
Abstract
Advances in understanding of the molecular basis of myocardial dysfunction, together with the development of increasingly efficient gene transfer technology, has placed heart failure within reach of gene-based therapy. Multiple components of cardiac contractility, including the Beta-adrenergic system, the calcium channel cycling pathway, and cytokine mediated cell proliferation, have been identified as appropriate targets for gene therapy. The development of efficient and safe vectors such as adeno-associated viruses and polymer nanoparticles has provided an opportunity for clinical application for gene therapy. The recent successful and safe completion of a phase 2 trial targeting the sarcoplasmic reticulum calcium ATPase pump (SERCA2a) has the potential to open a new era for gene therapy in the treatment of heart failure.
Collapse
Affiliation(s)
- Charbel Naim
- Cardiovascular Research Center, Mount Sinai School of Medicine, New York, NY 10029, USA
| | | | | |
Collapse
|
36
|
Kim JY, Ash RT, Ceballos-Diaz C, Levites Y, Golde TE, Smirnakis SM, Jankowsky JL. Viral transduction of the neonatal brain delivers controllable genetic mosaicism for visualising and manipulating neuronal circuits in vivo. Eur J Neurosci 2013; 37:1203-20. [PMID: 23347239 DOI: 10.1111/ejn.12126] [Citation(s) in RCA: 104] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2012] [Revised: 12/09/2012] [Accepted: 12/12/2012] [Indexed: 12/19/2022]
Abstract
The neonatal intraventricular injection of adeno-associated virus has been shown to transduce neurons widely throughout the brain, but its full potential for experimental neuroscience has not been adequately explored. We report a detailed analysis of the method's versatility with an emphasis on experimental applications where tools for genetic manipulation are currently lacking. Viral injection into the neonatal mouse brain is fast, easy, and accesses regions of the brain including the cerebellum and brainstem that have been difficult to target with other techniques such as electroporation. We show that viral transduction produces an inherently mosaic expression pattern that can be exploited by varying the titer to transduce isolated neurons or densely-packed populations. We demonstrate that the expression of virally-encoded proteins is active much sooner than previously believed, allowing genetic perturbation during critical periods of neuronal plasticity, but is also long-lasting and stable, allowing chronic studies of aging. We harness these features to visualise and manipulate neurons in the hindbrain that have been recalcitrant to approaches commonly applied in the cortex. We show that viral labeling aids the analysis of postnatal dendritic maturation in cerebellar Purkinje neurons by allowing individual cells to be readily distinguished, and then demonstrate that the same sparse labeling allows live in vivo imaging of mature Purkinje neurons at a resolution sufficient for complete analytical reconstruction. Given the rising availability of viral constructs, packaging services, and genetically modified animals, these techniques should facilitate a wide range of experiments into brain development, function, and degeneration.
Collapse
Affiliation(s)
- Ji-Yoen Kim
- Department of Neuroscience, Baylor College of Medicine, BCM295, One Baylor Plaza, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
37
|
Liu B, Traini R, Killinger B, Schneider B, Moszczynska A. Overexpression of parkin in the rat nigrostriatal dopamine system protects against methamphetamine neurotoxicity. Exp Neurol 2013; 247:359-72. [PMID: 23313192 DOI: 10.1016/j.expneurol.2013.01.001] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2012] [Revised: 12/05/2012] [Accepted: 01/01/2013] [Indexed: 11/15/2022]
Abstract
Methamphetamine (METH) is a central nervous system psychostimulant with a high potential for abuse. At high doses, METH causes a selective degeneration of dopaminergic terminals in the striatum, sparing other striatal terminals and cell bodies. We previously detected a deficit in parkin after binge METH in rat striatal synaptosomes. Parkin is an ubiquitin-protein E3 ligase capable of protecting dopamine neurons from diverse cellular insults. Whether the deficit in parkin mediates the toxicity of METH and whether parkin can protect from toxicity of the drug is unknown. The present study investigated whether overexpression of parkin attenuates degeneration of striatal dopaminergic terminals exposed to binge METH. Parkin overexpression in rat nigrostriatal dopamine system was achieved by microinjection of adeno-associated viral transfer vector 2/6 encoding rat parkin (AAV2/6-parkin) into the substantia nigra pars compacta. The microinjections of AAV2/6-parkin dose-dependently increased parkin levels in both the substantia nigra pars compacta and striatum. The levels of dopamine synthesizing enzyme, tyrosine hydroxylase, remained at the control levels; therefore, tyrosine hydroxylase immunoreactivity was used as an index of dopaminergic terminal integrity. In METH-exposed rats, the increase in parkin levels attenuated METH-induced decreases in striatal tyrosine hydroxylase immunoreactivity in a dose-dependent manner, indicating that parkin can protect striatal dopaminergic terminals against METH neurotoxicity.
Collapse
Affiliation(s)
- Bin Liu
- Department of Pharmaceutical Sciences, Eugene Applebaum College of Pharmacy and Health Sciences, Wayne State University, Detroit, MI 48202, USA
| | | | | | | | | |
Collapse
|
38
|
Aurnhammer C, Haase M, Muether N, Hausl M, Rauschhuber C, Huber I, Nitschko H, Busch U, Sing A, Ehrhardt A, Baiker A. Universal real-time PCR for the detection and quantification of adeno-associated virus serotype 2-derived inverted terminal repeat sequences. Hum Gene Ther Methods 2013; 23:18-28. [PMID: 22428977 DOI: 10.1089/hgtb.2011.034] [Citation(s) in RCA: 328] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Viral vectors based on various naturally occurring adeno-associated virus (AAV) serotypes are among the most promising tools in human gene therapy. For the production of recombinant AAV (rAAV) vectors, researchers are focusing predominantly on cross-packaging an artificial AAV genome based on serotype 2 (AAV2) into capsids derived from other serotypes. Within the packaged genome the inverted terminal repeats (ITRs) are the only cis-acting viral elements required for rAAV vector generation and depict the lowest common denominator of all AAV2-derived vector genomes. Up to now, no quantitative PCR (qPCR) for the detection and quantification of AAV2 ITRs could be established because of their extensive secondary hairpin structure formation. Current qPCR-based methods are therefore targeting vector-encoded transgenes or regulatory elements. Herein we establish a molecular biological method that allows accurate and reproducible quantification of AAV2 genomes on the basis of an AAV2 ITR sequence-specific qPCR. Primers and labeled probe are located within the ITR sequence and have been designed to detect both wild-type AAV2 and AAV2-based vectors. This method is suitable for detecting single-stranded DNA derived from AAV2 vector particles and double-stranded DNA derived from vector plasmids. The limit of detection has been determined as 50 ITR sequence copies per reaction, by comparison with a plasmid standard. In conclusion, this method describes the first qPCR system facilitating the detection and quantification of AAV2 ITR sequences. Because this method can be used universally for all AAV2 genome-based vectors, it will significantly simplify rAAV2 vector titrations in the future.
Collapse
|
39
|
Braunwald E. Cardiovascular science: opportunities for translating research into improved care. J Clin Invest 2013; 123:6-10. [PMID: 23281404 DOI: 10.1172/jci67541] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Cardiovascular research is progressing on many fronts, as highlighted in the collection of Reviews in this issue of the JCI. MicroRNAs that regulate cardiac function have been implicated in cardiac disorders, and efforts to develop therapeutic antagomirs are underway. The genetic bases of several cardiac disorders, including cardiomyopathies that cause heart failure and channelopathies that underlie cardiac arrhythmias, have been elucidated. Genetic testing can identify asymptomatic individuals at risk, potentially leading to effective preventative measures. Growing evidence supports the role of chronic inflammation in atherosclerosis, providing new opportunities for therapeutic intervention. For heart failure, recent work suggests that cardiac regeneration using stem/progenitor cells, gene transfer, new drugs that restore normal Ca2+ cycling, and agents that reduce reperfusion injury following myocardial infarction are all viable new approaches to managing disease. Cumulatively, it seems likely that the clinical advances emerging from ongoing research will, in the foreseeable future, reduce the number of deaths in the industrialized world from cardiovascular disease.
Collapse
Affiliation(s)
- Eugene Braunwald
- TIMI Study Group, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts 02115, USA.
| |
Collapse
|
40
|
Lipinski DM, Thake M, MacLaren RE. Clinical applications of retinal gene therapy. Prog Retin Eye Res 2013; 32:22-47. [DOI: 10.1016/j.preteyeres.2012.09.001] [Citation(s) in RCA: 76] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2011] [Revised: 09/04/2012] [Accepted: 09/04/2012] [Indexed: 02/08/2023]
|
41
|
Abstract
Myocardial in vivo gene delivery is a valuable technique to investigate the relevance of a protein of interest on cardiac contractile function, hypertrophy, and energy state in healthy animals as well as in a variety of models of cardiovascular disease. Rodent models are used to screen effects and to investigate molecular mechanisms, while large animal models, more closely reflecting human anatomy, physiology, and function, are inevitable for translational therapeutic approaches. The gene of interest, whose expression is driven by a non-cardioselective or cardioselective promotor is cloned into a viral vector. This vehicle is then delivered using an appropriate administration route to target the heart and to achieve efficient protein expression in myocardium. Here we describe myocardial gene therapy in small and large animal models of postischemic heart failure used to reveal the positive inotrope, antihypertrophic, and pro-energetic action of the small calcium sensor protein S100A1.
Collapse
|
42
|
Cytoplasmic trafficking, endosomal escape, and perinuclear accumulation of adeno-associated virus type 2 particles are facilitated by microtubule network. J Virol 2012; 86:10462-73. [PMID: 22811523 DOI: 10.1128/jvi.00935-12] [Citation(s) in RCA: 110] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Understanding adeno-associated virus (AAV) trafficking is critical to advance our knowledge of AAV biology and exploit novel aspects of vector development. Similar to the case for most DNA viruses, after receptor binding and entry, AAV traverses the cytoplasm and deposits the viral genome in the cell nucleus. In this study, we examined the role of the microtubule (MT) network in productive AAV infection. Using pharmacological reagents (e.g., nocodazole), live-cell imaging, and flow cytometry analysis, we demonstrated that AAV type 2 (AAV2) transduction was reduced by at least 2-fold in the absence of the MT network. Cell surface attachment and viral internalization were not dependent on an intact MT network. In treated cells at 2 h postinfection, quantitative three-dimensional (3D) microscopy determined a reproducible difference in number of intracellular particles associated with the nuclear membrane or the nucleus compared to that for controls (6 to 7% versus 26 to 30%, respectively). Confocal microscopy analysis demonstrated a direct association of virions with MTs, further supporting a critical role in AAV infection. To investigate the underling mechanisms, we employed single-particle tracking (SPT) to monitor the viral movement in real time. Surprisingly, unlike other DNA viruses (e.g., adenovirus [Ad] and herpes simplex virus [HSV]) that display bidirectional motion on MTs, AAV2 displays only unidirectional movement on MTs toward the nuclei, with peak instantaneous velocities at 1.5 to 3.5 μm/s. This rapid and unidirectional motion on MTs lasts for about 5 to 10 s and results in AAV particles migrating more than 10 μm in the cytoplasm reaching the nucleus very efficiently. Furthermore, electron microscopy analysis determined that, unlike Ad and HSV, AAV2 particles were transported on MTs within membranous compartments, and surprisingly, the acidification of AAV2-containing endosomes was delayed by the disruption of MTs. These findings together suggest an as-yet-undescribed model in which after internalization, AAV2 exploits MTs for rapid cytoplasmic trafficking in endosomal compartments unidirectionally toward the perinuclear region, where most acidification events for viral escape take place.
Collapse
|
43
|
Tomanin R, Zanetti A, Zaccariotto E, D'Avanzo F, Bellettato CM, Scarpa M. Gene therapy approaches for lysosomal storage disorders, a good model for the treatment of mendelian diseases. Acta Paediatr 2012; 101:692-701. [PMID: 22428546 DOI: 10.1111/j.1651-2227.2012.02674.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
UNLABELLED This review describes the different gene therapy technologies applied to approach lysosomal storage disorders, monogenic conditions, with known genetic and biochemical defects, for many of which animal models are available. Both viral and nonviral procedures are described, underlying the specific needs that the treatment of genetic disorders requires. CONCLUSIONS Lysosomal storage disorders represent a good model of study of gene therapeutic procedures that are, or could be, relevant to the treatment of several other mendelian diseases.
Collapse
Affiliation(s)
- Rosella Tomanin
- Gene Therapy Laboratory, Department of Pediatrics, University of Padova, Italy
| | | | | | | | | | | |
Collapse
|
44
|
Recombinant adeno-associated virus: clinical application and development as a gene-therapy vector. Ther Deliv 2012; 3:835-56. [DOI: 10.4155/tde.12.63] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Gene therapy is gaining momentum as a method of treating human disease. Initially conceived as a strategy to complement defective genes in monogenic disorders, the scope of gene therapy has expanded to encompass a variety of applications. Likewise, the molecular tools for gene delivery have evolved and diversified to meet these various therapeutic needs. Recombinant adeno-associated virus (rAAV) has made significant strides toward clinical application with an excellent safety profile and successes in several clinical trials. This review covers the basic biology of rAAV as a gene therapy vector as well as its advantages compared with other methods of gene delivery. The status of clinical trials utilizing rAAV is also discussed in detail. In conclusion, methods of engineering the vector to overcome challenges identified from these trials are covered, with emphasis on modification of the viral capsid to increase the tissue/cell-specific targeting and transduction efficiency.
Collapse
|
45
|
Hovlid ML, Steinmetz NF, Laufer B, Lau JL, Kuzelka J, Wang Q, Hyypiä T, Nemerow GR, Kessler H, Manchester M, Finn MG. Guiding plant virus particles to integrin-displaying cells. NANOSCALE 2012; 4:3698-705. [PMID: 22585108 PMCID: PMC3567620 DOI: 10.1039/c2nr30571b] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2023]
Abstract
Viral nanoparticles (VNPs) are structurally regular, highly stable, tunable nanomaterials that can be conveniently produced in high yields. Unmodified VNPs from plants and bacteria generally do not show tissue specificity or high selectivity in binding to or entry into mammalian cells. They are, however, malleable by both genetic and chemical means, making them useful scaffolds for the display of large numbers of cell- and tissue-targeting ligands, imaging moieties, and/or therapeutic agents in a well-defined manner. Capitalizing on this attribute, we modified the genetic sequence of the Cowpea mosaic virus (CPMV) coat protein to display an RGD oligopeptide sequence derived from human adenovirus type 2 (HAdV-2). Concurrently, wild-type CPMV was modified via NHS acylation and Cu(I)-catalyzed azide-alkyne cycloaddition (CuAAC) chemistry to attach an integrin-binding cyclic RGD peptide. Both types of particles showed strong and selective affinity for several different cancer cell lines that express RGD-binding integrin receptors.
Collapse
Affiliation(s)
- Marisa L Hovlid
- Department of Chemistry, The Scripps Research Institute, 10550 N. Torrey Pines Rd, La Jolla, CA 92037, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Congestive heart failure accounts for half a million deaths per year in the United States. Despite its place among the leading causes of morbidity, pharmacological and mechanic remedies have only been able to slow the progression of the disease. Today's science has yet to provide a cure, and there are few therapeutic modalities available for patients with advanced heart failure. There is a critical need to explore new therapeutic approaches in heart failure, and gene therapy has emerged as a viable alternative. Recent advances in understanding of the molecular basis of myocardial dysfunction, together with the evolution of increasingly efficient gene transfer technology, have placed heart failure within reach of gene-based therapy. The recent successful and safe completion of a phase 2 trial targeting the sarcoplasmic reticulum calcium ATPase pump (SERCA2a), along with the start of more recent phase 1 trials, opens a new era for gene therapy for the treatment of heart failure.
Collapse
Affiliation(s)
- Lisa Tilemann
- Cardiovascular Research Center, Mount Sinai Medical Center, New York, NY 10029, USA
| | | | | | | |
Collapse
|
47
|
Wirth T. A short perspective on gene therapy: Clinical experience on gene therapy of gliomablastoma multiforme. World J Exp Med 2011; 1:10-6. [PMID: 24520527 PMCID: PMC3905579 DOI: 10.5493/wjem.v1.i1.10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2011] [Revised: 12/12/2011] [Accepted: 12/16/2011] [Indexed: 02/06/2023] Open
Abstract
More than two decades have passed since the first gene therapy clinical trial was conducted. During this time, we have gained much knowledge regarding gene therapy in general, but also learned to understand the fear that persists in society. We have experienced drawbacks and successes. More than 1700 clinical trials have been conducted where gene therapy is used as a means for therapy. In the very first trial, patients with advanced melanoma were treated with tumor infiltrating lymphocytes genetically modified ex-vivo to express tumor necrosis factor. Around the same time the first gene therapy trial was conducted, the ethical aspects of performing gene therapy on humans was intensively discussed. What are the risks involved with gene therapy? Can we control the technology? What is ethically acceptable and what are the indications gene therapy can be used for? Initially, gene therapy was thought to be implemented mainly for the treatment of monogenetic diseases, such as adenosine deaminase deficiency. However, other therapeutic areas have become of interest and currently cancer is the most studied therapeutic area for gene therapy based medicines. In this review I will be giving a short introduction into gene therapy and will direct the discussion to where we should go from here. Furthermore, I will focus on the use of the Herpes simplex virus-thymidine kinase for gene therapy of malignant gliomas and highlight the efficacy of gene therapy for the treatment of malignant gliomas, but other strategies will also be mentioned.
Collapse
Affiliation(s)
- Thomas Wirth
- Thomas Wirth, AI Virtanen Institute, Department of Biotechnology and Molecular Medicine, University of Eastern Finland, Neulaniementie 2, FIN-70211 Kuopio, Finland
| |
Collapse
|
48
|
Aurnhammer C, Haase M, Muether N, Hausl M, Rauschhuber C, Huber I, Nitschko H, Busch U, Sing A, Ehrhardt A, Baiker A. Universal Real-Time PCR for the Detection and Quantification of Adeno-Associated Virus Serotype 2-Derived Inverted Terminal Repeat Sequences. Hum Gene Ther 2011. [DOI: 10.1089/hum.2011.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
49
|
Abstract
INTRODUCTION Glycogen storage disease (GSD) type Ia and Ib are disorders of impaired glucose homeostasis affecting the liver and kidney. GSD-Ib also affects neutrophils. Current dietary therapies cannot prevent long-term complications. In animal studies, recombinant adeno-associated virus (rAAV) vector-mediated gene therapy can correct or minimize multiple aspects of the disorders, offering hope for human gene therapy. AREAS COVERED A summary of recent progress in rAAV-mediated gene therapy for GSD-I; strategies to improve rAAV-mediated gene delivery, transduction efficiency and immune avoidance; and vector refinements that improve expression. EXPERT OPINION rAAV-mediated gene delivery to the liver can restore glucose homeostasis in preclinical models of GSD-I, but some long-term complications of the liver and kidney remain. Gene therapy for GSD-Ib is less advanced than for GSD-Ia and only transient correction of myeloid dysfunction has been achieved. A question remains as to whether a single rAAV vector can meet the expression efficiency and tropism required to treat all aspects of GSD-I, or if a multi-pronged approach is needed. An understanding of the strengths and weaknesses of rAAV vectors in the context of strategies to achieve efficient transduction of the liver, kidney and hematopoietic stem cells is required for treating GSD-I.
Collapse
Affiliation(s)
- Janice Y Chou
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Section on Cellular Differentiation, Program on Developmental Endocrinology and Genetics, Bethesda, MD 20892 1830, USA.
| | | |
Collapse
|
50
|
Abstract
INTRODUCTION Lysosomal storage disorders (LSDs) encompass more than 50 distinct diseases, caused by defects in various aspects of lysosomal function. Neurodegeneration and/or dysmyelination are the hallmark of roughly 70% of LSDs. Gene therapy represents a promising approach for the treatment of CNS manifestations in LSDs, as it has the potential to provide a permanent source of the deficient enzyme, either by direct injection of vectors or by transplantation of gene-corrected cells. In this latter approach, the biology of neural stem/progenitor cells and hematopoietic cells might be exploited. AREAS COVERED Based on an extensive literature search up until March 2011, the author reviews and discusses the progress, the crucial aspects and the major challenges towards the development of novel gene therapy strategies aimed to target the CNS, with particular attention to direct intracerebral gene delivery and transplantation of neural stem/progenitor cells. EXPERT OPINION The implementation of viral vector delivery systems with specific tropism, regulated transgene expression, low immunogenicity and low genotoxic risk and the improvement in isolation and manipulation of relevant cell types to be transplanted, are fundamental challenges to the field. Also, combinatorial strategies might be required to achieve full correction in LSDs with neurological involvement.
Collapse
Affiliation(s)
- Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milano, Italy.
| |
Collapse
|