1
|
Słyk Ż, Stachowiak N, Małecki M. Recombinant Adeno-Associated Virus Vectors for Gene Therapy of the Central Nervous System: Delivery Routes and Clinical Aspects. Biomedicines 2024; 12:1523. [PMID: 39062095 PMCID: PMC11274884 DOI: 10.3390/biomedicines12071523] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
The Central Nervous System (CNS) is vulnerable to a range of diseases, including neurodegenerative and oncological conditions, which present significant treatment challenges. The blood-brain barrier (BBB) restricts molecule penetration, complicating the achievement of therapeutic concentrations in the CNS following systemic administration. Gene therapy using recombinant adeno-associated virus (rAAV) vectors emerges as a promising strategy for treating CNS diseases, demonstrated by the registration of six gene therapy products in the past six years and 87 ongoing clinical trials. This review explores the implementation of rAAV vectors in CNS disease treatment, emphasizing AAV biology and vector engineering. Various administration methods-such as intravenous, intrathecal, and intraparenchymal routes-and experimental approaches like intranasal and intramuscular administration are evaluated, discussing their advantages and limitations in different CNS contexts. Additionally, the review underscores the importance of optimizing therapeutic efficacy through the pharmacokinetics (PK) and pharmacodynamics (PD) of rAAV vectors. A comprehensive analysis of clinical trials reveals successes and challenges, including barriers to commercialization. This review provides insights into therapeutic strategies using rAAV vectors in neurological diseases and identifies areas requiring further research, particularly in optimizing rAAV PK/PD.
Collapse
Affiliation(s)
- Żaneta Słyk
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Natalia Stachowiak
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
2
|
Słyk Ż, Wrzesień R, Barszcz S, Gawrychowski K, Małecki M. Adeno-associated virus vector hydrogel formulations for brain cancer gene therapy applications. Biomed Pharmacother 2024; 170:116061. [PMID: 38154269 DOI: 10.1016/j.biopha.2023.116061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023] Open
Abstract
Gelatin-based formulations are utilized in neurosurgical procedures, with Medisponge® serving as an illustration of a secure and biocompatible hemostatic formulation. Noteworthy are combined hemostatic products that integrate pharmacological agents with gelatin. Gelatin matrices, which host biologically active substances, provide a platform for a variety of molecules. Biopolymers function as carriers for chemicals and genes, a facet particularly pertinent in brain cancer therapy, as gene therapy complement conventional approaches. The registration of Zolgensma underscores the efficacy of rAAV vectors in therapeutic gene delivery to the CNS. rAAVs, renowned for their safety, stability, and neuron-targeting capabilities, predominate in CNS gene therapy studies. The effectiveness of rAAV vector therapy varies based on the serotype and administration route. Local gene therapy employing hydrogel (e.g., post-tumor resection) enables the circumvention of the blood-brain barrier and restricts formulation diffusion. This study formulates gelatin rAAV gene formulations and evaluates vector transduction potential. Transduction efficiency was assessed using ex vivo mouse brains and in vitro cancer cell lines. In vitro, the transduction of rAAV vectors in gelatin matrices was quantified through qPCR, measuring the itr and Gfp expression. rAAVDJ and rAAV2 demonstrated superior transduction in ex vivo and in vitro models. Among the cell lines tested (Hs683, B16-F10, NIH:OVCAR-3), gelatin matrix F1 exhibited selective transduction, particularly with Hs683 human glioma cells, surpassing the performance Medisponge®. This research highlights the exploration of local brain cancer therapy, emphasizing the potential of gelatin as an rAAV vector carrier for gene therapy. The functional transduction activity of gelatin rAAV formulations is demonstrated.
Collapse
Affiliation(s)
- Żaneta Słyk
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland; Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland.
| | - Robert Wrzesień
- Central Laboratory of Experimental Animals, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Sławomir Barszcz
- Department of Neurosurgery, Children's Clinical Hospital, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Gawrychowski
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland; Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
3
|
Qi Z, Han S, Wang S, Gu X, Deng J, Huang C, Yin X. Visual three-dimensional spatial distribution of motor neurons innervating superficial limb muscles in mice. Front Cell Neurosci 2022; 16:904172. [PMID: 35936500 PMCID: PMC9354668 DOI: 10.3389/fncel.2022.904172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Accepted: 06/29/2022] [Indexed: 11/13/2022] Open
Abstract
The coordination of motor function in the spinal cord depends on selective connections between distinct classes of motor neurons and their target muscles. However, knowledge regarding the anatomical connections between the superficial limb skeletal muscles and the motor neurons that innervate them is limited. In this study, with a combination of the multiple retrograde tracing method with 3DISCO clearing, we explored the spatial distribution of different motor neuron pools targeting specific superficial muscles of the forelimbs or hindlimbs in mouse spinal cords, which were dominated by the radial, median, ulnar, or sciatic nerve. This study reveals the precise interrelationship among different motor neuron pools innervating limb muscles under the same space and time. The data will help to further understand the neural loop and muscular motor coordination.
Collapse
Affiliation(s)
- Zhidan Qi
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Shuai Han
- Department of Orthopaedics, Peking University Third Hospital, Beijing, China
| | - Shen Wang
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Xinyi Gu
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Jin Deng
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Chen Huang
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
| | - Xiaofeng Yin
- Department of Orthopedics and Trauma, Peking University People’s Hospital, Beijing, China
- *Correspondence: Xiaofeng Yin,
| |
Collapse
|
4
|
Xu J, Xuan A, Liu Z, Li Y, Zhu J, Yao Y, Yu T, Zhu D. An Approach to Maximize Retrograde Transport Based on the Spatial Distribution of Motor Endplates in Mouse Hindlimb Muscles. Front Cell Neurosci 2021; 15:707982. [PMID: 34456685 PMCID: PMC8385196 DOI: 10.3389/fncel.2021.707982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2021] [Accepted: 07/16/2021] [Indexed: 11/13/2022] Open
Abstract
Knowledge regarding the relationship between muscles and the corresponding motor neurons would allow therapeutic genes to transport into specific spinal cord segments. Retrograde tracing technique by targeting the motor endplate (MEP), a highly specialized structure that offers direct access to the spinal motor neurons, has been used to elucidate the connectivity between skeletal muscles and the innervating motor neuron pools. However, current injection strategies mainly based on blind injection or the local MEP region might lead to an underestimation of the motor neuron number due to the uneven distribution of MEP in skeletal muscles. In this work, we proposed a novel intramuscular injection strategy based on the 3D distribution of the MEPs in skeletal muscles, applied the 3D intramuscular injection to the gastrocnemius and tibialis anterior for retrograde tracing of the corresponding motor neurons, and compared this with the existing injection strategy. The intramuscular diffusion of the tracer demonstrated that 3D injection could maximize the retrograde transport by ensuring a greater uptake of the tracer by the MEP region. In combination with optical clearing and imaging, we performed 3D mapping and quantification of the labeled motor neurons and confirmed that 3D injection could label more motor neurons than the current injection method. It is expected that 3D intramuscular injection strategy will help elucidate the connective relationship between muscles and motor neurons faithfully and becomes a promising tool in the development of gene therapy strategies for motor neuron diseases.
Collapse
Affiliation(s)
- Jianyi Xu
- 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, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Ang Xuan
- 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, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Zhang 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, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Yusha Li
- 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, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Jingtan Zhu
- 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, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Yingtao Yao
- 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, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Tingting Yu
- 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, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| | - Dan Zhu
- 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, School of Engineering Sciences, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
5
|
Wright JH, Huang LY, Weaver S, Archila LD, McAfee MS, Hirayama AV, Chapuis AG, Bleakley M, Rongvaux A, Turtle CJ, Chanthaphavong RS, Campbell JS, Pierce RH. Detection of engineered T cells in FFPE tissue by multiplex in situ hybridization and immunohistochemistry. J Immunol Methods 2021; 492:112955. [PMID: 33383062 PMCID: PMC7979489 DOI: 10.1016/j.jim.2020.112955] [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: 07/29/2020] [Revised: 11/24/2020] [Accepted: 12/25/2020] [Indexed: 10/22/2022]
Abstract
Identifying engineered T cells in situ is important to understand the location, persistence, and phenotype of these cells in patients after adoptive T cell therapy. While engineered cells are routinely characterized in fresh tissue or blood from patients by flow cytometry, it is difficult to distinguish them from endogenous cells in formalin-fixed, paraffin-embedded (FFPE) tissue biopsies. To overcome this limitation, we have developed a method for characterizing engineered T cells in fixed tissue using in situ hybridization (ISH) to the woodchuck hepatitis post-transcriptional regulatory element (WPRE) common in many lentiviral vectors used to transduce chimeric antigen receptor T (CAR-T) and T cell receptor T (TCR-T) cells, coupled with alternative permeabilization conditions that allows subsequent multiplex immunohistochemical (mIHC) staining within the same image. This new method provides the ability to mark the cells by ISH, and simultaneously stain for cell-associated proteins to immunophenotype CAR/TCR modified T cells within tumors, as well as assess potential roles of these cells in on-target/off-tumor toxicity in other tissue.
Collapse
Affiliation(s)
- Jocelyn H Wright
- Immunopathology Lab, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America.
| | - Li-Ya Huang
- Experimental Histopathology, Fred Hutchinson Cancer Research Center, United States of America
| | - Stephanie Weaver
- Experimental Histopathology, Fred Hutchinson Cancer Research Center, United States of America
| | - L Diego Archila
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America
| | - Megan S McAfee
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America
| | - Alexandre V Hirayama
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America
| | - Aude G Chapuis
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America; Department of Medicine, University of Washington, United States of America
| | - Marie Bleakley
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America; Department of Pediatrics, University of Washington School of Medicine, United States of America; Seattle Cancer Care Alliance, University of Washington, United States of America; Seattle Children's Hospital, University of Washington, United States of America
| | - Anthony Rongvaux
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America; Department of Immunology, University of Washington School of Medicine, United States of America
| | - Cameron J Turtle
- Program in Immunology, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America; Department of Medicine, University of Washington, United States of America; Seattle Cancer Care Alliance, University of Washington, United States of America
| | - R Savanh Chanthaphavong
- Experimental Histopathology, Fred Hutchinson Cancer Research Center, United States of America
| | - Jean S Campbell
- Immunopathology Lab, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America; Department of Laboratory Medicine and Pathology, University of Washington, United States of America
| | - Robert H Pierce
- Immunopathology Lab, Clinical Research Division, Fred Hutchinson Cancer Research Center, United States of America
| |
Collapse
|
6
|
Jan A, Richner M, Vægter CB, Nyengaard JR, Jensen PH. Gene Transfer in Rodent Nervous Tissue Following Hindlimb Intramuscular Delivery of Recombinant Adeno-Associated Virus Serotypes AAV2/6, AAV2/8, and AAV2/9. Neurosci Insights 2019; 14:1179069519889022. [PMID: 32363345 PMCID: PMC7176396 DOI: 10.1177/1179069519889022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/15/2019] [Indexed: 01/15/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors have emerged as the safe vehicles of choice for long-term gene transfer in mammalian nervous system. Recombinant adeno-associated virus–mediated localized gene transfer in adult nervous system following direct inoculation, that is, intracerebral or intrathecal, is well documented. However, recombinant adeno-associated virus delivery in defined neuronal populations in adult animals using less-invasive methods as well as avoiding ectopic gene expression following systemic inoculation remain challenging. Harnessing the capability of some recombinant adeno-associated virus serotypes for retrograde transduction may potentially address such limitations (Note: The term retrograde transduction in this manuscript refers to the uptake of injected recombinant adeno-associated virus particles at nerve terminals, retrograde transport, and subsequent transduction of nerve cell soma). In some studies, recombinant adeno-associated virus serotypes 2/6, 2/8, and 2/9 have been shown to exhibit transduction of connected neuroanatomical tracts in adult animals following lower limb intramuscular recombinant adeno-associated virus delivery in a pattern suggestive of retrograde transduction. However, an extensive side-by-side comparison of these serotypes following intramuscular delivery regarding tissue viral load, and the effect of promoter on transgene expression, has not been performed. Hence, we delivered recombinant adeno-associated virus serotypes 2/6, 2/8, or 2/9 encoding enhanced green fluorescent protein (eGFP), under the control of either cytomegalovirus (CMV) or human synapsin (hSyn) promoter, via a single unilateral hindlimb intramuscular injection in the bicep femoris of adult C57BL/6J mice. Four weeks post injection, we quantified viral load and transgene (enhanced green fluorescent protein) expression in muscle and related nervous tissues. Our data show that the select recombinant adeno-associated virus serotypes transduce sciatic nerve and groups of neurons in the dorsal root ganglia on the injected side, indicating that the intramuscular recombinant adeno-associated virus delivery is useful for achieving gene transfer in local neuroanatomical tracts. We also observed sparse recombinant adeno-associated virus viral delivery or eGFP transduction in lumbar spinal cord and a noticeable lack thereof in brain. Therefore, further improvements in recombinant adeno-associated virus design are warranted to achieve efficient widespread retrograde transduction following intramuscular and possibly other peripheral routes of delivery.
Collapse
Affiliation(s)
- Asad Jan
- Aarhus Institute of Advanced Studies and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mette Richner
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Christian B Vægter
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, and Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Poul H Jensen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
7
|
Chen Z, Fan G, Li A, Yuan J, Xu T. rAAV2-Retro Enables Extensive and High-Efficient Transduction of Lower Motor Neurons following Intramuscular Injection. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 17:21-33. [PMID: 31890738 PMCID: PMC6926343 DOI: 10.1016/j.omtm.2019.11.006] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 11/09/2019] [Indexed: 12/27/2022]
Abstract
The motor system controls muscle movement through lower motor neurons in the spinal cord and brainstem. Lower motor neurons are efferent neurons in the central nervous system (CNS) characterized by axonal projections that reach specific targets in the periphery. Lower motor neuron lesions result in the denervation and dysfunction of peripheral skeletal muscle. Great progress has been made to develop therapeutic strategies to transduce lower motor neurons with genes. However, the widespread distribution of lower motor neurons makes their specific, extensive, and efficient transduction a challenge. In this study, we demonstrated that, compared to the other tested recombinant adeno-associated virus (rAAV) serotypes, rAAV2-retro mediated the most efficient retrograde transduction of lower motor neurons in the spinal cord following intramuscular injection in neonatal mice. A single injection of rAAV2-retro in a single muscle enabled the efficient and extensive transduction of lower motor neurons in the spinal cord and brainstem rather than transducing only the lower motor neurons connected to the injected muscle. rAAV2-retro achieved the extensive transduction of lower motor neurons by the cerebrospinal fluid pathway. Our work suggests that gene delivery via the intramuscular injection of rAAV2-retro represents a promising tool in the development of gene therapy strategies for motor neuron diseases.
Collapse
Affiliation(s)
- Zhilong 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
| | - Guoqing Fan
- 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
| | - Anan Li
- 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
| | - Jing Yuan
- 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
| | - Tonghui Xu
- 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.,Institute of Life Sciences, Nanchang University, Nanchang, China
| |
Collapse
|
8
|
Hudry E, Vandenberghe LH. Therapeutic AAV Gene Transfer to the Nervous System: A Clinical Reality. Neuron 2019; 101:839-862. [PMID: 30844402 PMCID: PMC11804970 DOI: 10.1016/j.neuron.2019.02.017] [Citation(s) in RCA: 235] [Impact Index Per Article: 39.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 02/07/2023]
Abstract
Gene transfer has long been a compelling yet elusive therapeutic modality. First mainly considered for rare inherited disorders, gene therapy may open treatment opportunities for more challenging and complex diseases such as Alzheimer's or Parkinson's disease. Today, examples of striking clinical proof of concept, the first gene therapy drugs coming onto the market, and the emergence of powerful new molecular tools have broadened the number of avenues to target neurological disorders but have also highlighted safety concerns and technology gaps. The vector of choice for many nervous system targets currently is the adeno-associated viral (AAV) vector due to its desirable safety profile and strong neuronal tropism. In aggregate, the clinical success, the preclinical potential, and the technological innovation have made therapeutic AAV drug development a reality, particularly for nervous system disorders. Here, we discuss the rationale, opportunities, limitations, and progress in clinical AAV gene therapy.
Collapse
Affiliation(s)
- Eloise Hudry
- Department of Neurology, The Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, USA.
| | - Luk H Vandenberghe
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Harvard Program in Therapeutic Science, Harvard University, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
9
|
Anderson HE, Weir RFF. On the development of optical peripheral nerve interfaces. Neural Regen Res 2019; 14:425-436. [PMID: 30539808 PMCID: PMC6334609 DOI: 10.4103/1673-5374.245461] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2018] [Accepted: 09/19/2018] [Indexed: 11/04/2022] Open
Abstract
Limb loss and spinal cord injury are two debilitating conditions that continue to grow in prevalence. Prosthetic limbs and limb reanimation present two ways of providing affected individuals with means to interact in the world. These techniques are both dependent on a robust interface with the peripheral nerve. Current methods for interfacing with the peripheral nerve tend to suffer from low specificity, high latency and insufficient robustness for a chronic implant. An optical peripheral nerve interface may solve some of these problems by decreasing invasiveness and providing single axon specificity. In order to implement such an interface three elements are required: (1) a transducer capable of translating light into a neural stimulus or translating neural activity into changes in fluorescence, (2) a means for delivering said transducer and (3) a microscope for providing the stimulus light and detecting the fluorescence change. There are continued improvements in both genetically encoded calcium and voltage indicators as well as new optogenetic actuators for stimulation. Similarly, improvements in specificity of viral vectors continue to improve expression in the axons of the peripheral nerve. Our work has recently shown that it is possible to virally transduce axons of the peripheral nerve for recording from small fibers. The improvements of these components make an optical peripheral nerve interface a rapidly approaching alternative to current methods.
Collapse
Affiliation(s)
- Hans E. Anderson
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA
| | - Richard F. ff. Weir
- Department of Bioengineering, University of Colorado Denver | Anschutz Medical Campus, Aurora, CO, USA
| |
Collapse
|
10
|
Zheng C, Wang S, Bai Y, Luo T, Wang J, Dai C, Guo B, Luo S, Wang D, Yang Y, Wang Y. Lentiviral Vectors and Adeno-Associated Virus Vectors: Useful Tools for Gene Transfer in Pain Research. Anat Rec (Hoboken) 2018; 301:825-836. [PMID: 29149775 PMCID: PMC6585677 DOI: 10.1002/ar.23723] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2017] [Accepted: 05/19/2017] [Indexed: 11/09/2022]
Abstract
Pain, especially chronic pain, has always been a heated point in both basic and clinical researches since it puts heavy burdens on both individuals and the whole society. A better understanding of the role of biological molecules and various ionic channels involved in pain can shed light on the mechanism under pain and advocate the development of pain management. Using viral vectors to transfer specific genes at targeted sites is a promising method for both research and clinical applications. Lentiviral vectors and adeno-associated virus (AAV) vectors which allow stable and long-term expression of transgene in non-dividing cells are widely applied in pain research. In this review, we thoroughly outline the structure, category, advantages and disadvantages and the delivery methods of lentiviral and AAV vectors. The methods through which lentiviral and AAV vectors are delivered to targeted sites are closely related with the sites, level and period of transgene expression. Focus is placed on the various delivery methods applied to deliver vectors to spinal cord and dorsal root ganglion both of which play important roles in primary nociception. Our goal is to provide insight into the features of these two viral vectors and which administration approach can be chosen for different pain researches. Anat Rec, 301:825-836, 2018. © 2017 The Authors. The Anatomical Record published by Wiley Periodicals, Inc. on behalf of American Association of Anatomists.
Collapse
Affiliation(s)
- Chen‐Xi Zheng
- Department of Anatomy, Histology and EmbryologyK.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi'an 710032China
| | - Sheng‐Ming Wang
- Department of Anatomy, Histology and EmbryologyK.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi'an 710032China
| | - Yun‐Hu Bai
- Department of Hepatobiliary Surgery, Xi‐Jing HospitalThe Fourth Military Medical UniversityXi'an 710032China
| | - Ting‐Ting Luo
- Department of Neurobiology and Collaborative Innovation Center for Brain Science, School of Basic MedicineThe Fourth Military Medical UniversityXi'an 710032China
| | - Jia‐Qi Wang
- Department of Anatomy, Histology and EmbryologyK.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi'an 710032China
| | - Chun‐Qiu Dai
- Department of Anatomy, Histology and EmbryologyK.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi'an 710032China
| | - Bao‐Lin Guo
- Department of Anatomy, Histology and EmbryologyK.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi'an 710032China
| | - Shi‐Cheng Luo
- Department of Anatomy, Histology and EmbryologyK.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi'an 710032China
| | - Dong‐Hui Wang
- Department of Anatomy, Histology and EmbryologyK.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi'an 710032China
| | - Yan‐Ling Yang
- Department of Hepatobiliary Surgery, Xi‐Jing HospitalThe Fourth Military Medical UniversityXi'an 710032China
| | - Ya‐Yun Wang
- Department of Anatomy, Histology and EmbryologyK.K. Leung Brain Research Centre, The Fourth Military Medical UniversityXi'an 710032China
| |
Collapse
|
11
|
Imaging of electrical activity in small diameter fibers of the murine peripheral nerve with virally-delivered GCaMP6f. Sci Rep 2018; 8:3219. [PMID: 29459701 PMCID: PMC5818512 DOI: 10.1038/s41598-018-21528-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 02/06/2018] [Indexed: 12/24/2022] Open
Abstract
Current neural interfaces are hampered by lack of specificity and selectivity for neural interrogation. A method that might improve these interfaces is an optical peripheral nerve interface which communicates with individual axons via optogenetic reporters. To determine the feasibility of such an interface, we delivered the genetically encoded calcium indicator GCaMP6f to the mouse peripheral nerve by intramuscular injection of adenoassociated viral vector (AAV1) under the control of the CAG (chicken beta actin- cytomegalovirus hybrid promoter). Small diameter axons in the common peroneal nerve were transduced and demonstrated electrically inducible calcium transients ex vivo. Responses to single electrical stimuli were resolvable, and increasing the number of stimuli resulted in a monotonic increase in maximum fluorescence and a prolongation of calcium transient kinetics. This work demonstrates the viability of using a virally-delivered, genetically-encoded calcium indicator to read-out from peripheral nerve axons.
Collapse
|
12
|
Hardcastle N, Boulis NM, Federici T. AAV gene delivery to the spinal cord: serotypes, methods, candidate diseases, and clinical trials. Expert Opin Biol Ther 2017; 18:293-307. [PMID: 29249183 DOI: 10.1080/14712598.2018.1416089] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
INTRODUCTION Adeno-associated viral (AAV) vector-mediated gene delivery to the spinal cord has finally entered the pathway towards regulatory approval. Phase 1 clinical trials using AAV gene therapy for pediatric disorders - spinal muscular atrophy (SMA) and giant axonal neuropathy (GAN) - are now underway. AREAS COVERED This review addresses the latest progress in the field of AAV gene delivery to the spinal cord, particularly focusing on the most prominent AAV serotypes and delivery methodologies to the spinal cord. Candidate diseases and scaling up experiments in large animals are also discussed. EXPERT OPINION Intravenous (IV) and intrathecal (IT) deliveries seem to undoubtedly be the preferred routes of administration for diffuse spinal cord delivery of therapeutic AAV vectors that can cross the blood-brain barrier (BBB) and correct inherited genetic disorders. Conversely, intraparenchymal delivery is still an undervalued but very viable approach for segmental therapy in afflictions such as ALS or Pompe Disease as a means to prevent respiratory dysfunction.
Collapse
Affiliation(s)
- Nathan Hardcastle
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| | - Nicholas M Boulis
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| | - Thais Federici
- a Department of Neurosurgery , Emory University , Atlanta , GA , USA
| |
Collapse
|
13
|
Abstract
Transferring genetic molecules into the peripheral sensory nervous system to manipulate nociceptive pathophysiology is a powerful approach for experimental modulation of sensory signaling and potentially for translation into therapy for chronic pain. This can be efficiently achieved by the use of recombinant adeno-associated virus (rAAV) in conjunction with nociceptor-specific regulatory transgene cassettes. Among different routes of delivery, direct injection into the dorsal root ganglia (DRGs) offers the most efficient AAV-mediated gene transfer selectively into the peripheral sensory nervous system. Here, we briefly discuss the advantages and applications of intraganglionic microinjection, and then provide a detailed approach for DRG injection, including a list of the necessary materials and description of a method for performing DRG microinjection experiments. We also discuss our experience with several adeno-associated virus (AAV) options for in vivo transgene expression in DRG neurons.
Collapse
|
14
|
Gene therapy and respiratory neuroplasticity. Exp Neurol 2016; 287:261-267. [PMID: 27697480 DOI: 10.1016/j.expneurol.2016.09.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2016] [Revised: 09/19/2016] [Accepted: 09/27/2016] [Indexed: 01/01/2023]
Abstract
Breathing is a life-sustaining behavior that in mammals is accomplished by activation of dedicated muscles responsible for inspiratory and expiratory forces acting on the lung and chest wall. Motor control is exerted by specialized pools of motoneurons in the medulla and spinal cord innervated by projections from multiple centers primarily in the brainstem that act in concert to generate both the rhythm and pattern of ventilation. Perturbations that prevent the accomplishment of the full range of motor behaviors by respiratory muscles commonly result in significant morbidity and increased mortality. Recent developments in gene therapy and novel targeting strategies have contributed to deeper understanding of the organization of respiratory motor systems. Gene therapy has received widespread attention and substantial progress has been made in recent years with the advent of improved tools for vector design. Genes can be delivered via a variety of plasmids, synthetic or viral vectors and cell therapies. In recent years, adeno-associated viruses (AAV) have become one of the most commonly used vector systems, primarily because of the extensive characterization conducted to date and the versatility in targeting strategies. Recent studies highlight the power of using AAV to selectively and effectively transduce respiratory motoneurons and muscle fibers with promising therapeutic effects. This brief review summarizes current evidence for the use of gene therapy in respiratory disorders with a primary focus on interventions that address motor control and neuroplasticity, including regeneration, in the respiratory system.
Collapse
|
15
|
Goganau I, Blesch A. Gene Therapy for Spinal Cord Injury. Transl Neurosci 2016. [DOI: 10.1007/978-1-4899-7654-3_8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
16
|
Abstract
Spinal cord injury is a complex pathology often resulting in functional impairment and paralysis. Gene therapy has emerged as a possible solution to the problems of limited neural tissue regeneration through the administration of factors promoting axonal growth, while also offering long-term local delivery of therapeutic molecules at the injury site. Of note, gene therapy is our response to the requirements of neural and glial cells following spinal cord injury, providing, in a time-dependent manner, growth substances for axonal regeneration and eliminating axonal growth inhibitors. Herein, we explore different gene therapy strategies, including targeting gene expression to modulate the presence of neurotrophic growth or survival factors and increase neural tissue plasticity. Special attention is given to describing advances in viral and non-viral gene delivery systems, as well as the available routes of gene delivery. Finally, we discuss the future of combinatorial gene therapies and give consideration to the implementation of gene therapy in humans.
Collapse
|
17
|
de Winter F, Hoyng S, Tannemaat M, Eggers R, Mason M, Malessy M, Verhaagen J. Gene therapy approaches to enhance regeneration of the injured peripheral nerve. Eur J Pharmacol 2013; 719:145-152. [DOI: 10.1016/j.ejphar.2013.04.057] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Revised: 03/28/2013] [Accepted: 04/03/2013] [Indexed: 01/26/2023]
|
18
|
Intraspinal AAV Injections Immediately Rostral to a Thoracic Spinal Cord Injury Site Efficiently Transduces Neurons in Spinal Cord and Brain. MOLECULAR THERAPY-NUCLEIC ACIDS 2013; 2:e108. [PMID: 23881451 PMCID: PMC3731889 DOI: 10.1038/mtna.2013.34] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/21/2013] [Accepted: 05/03/2013] [Indexed: 12/18/2022]
Abstract
In the vast majority of studies utilizing adeno-associated virus (AAV) in central nervous system applications, including those published with spinal cord injury (SCI) models, AAV has been administered at the level of the cell body of neurons targeted for genetic modification, resulting in transduction of neurons in the vicinity of the injection site. However, as SCI interrupts many axon tracts, it may be more beneficial to transduce a diverse pool of supraspinal neurons. We determined if descending axons severed by SCI are capable of retrogradely transporting AAV to remotely transduce a variety of brain regions. Different AAV serotypes encoding the reporter green fluorescent protein (GFP) were injected into gray and white matter immediately rostral to a spinal transection site. This resulted in the transduction of thousands of neurons within the spinal cord and in multiple regions within the brainstem that project to spinal cord. In addition, we established that different serotypes had disparate regional specificity and that AAV5 transduced the most brain and spinal cord neurons. This is the first demonstration that retrograde transport of AAV by axons severed by SCI is an effective means to transduce a collection of supraspinal neurons. Thus, we identify a novel, minimally invasive means to transduce a variety of neuronal populations within both the spinal cord and the brain following SCI. This paradigm to broadly distribute viral vectors has the potential to be an important component of a combinatorial strategy to promote functional axonal regeneration.
Collapse
|
19
|
Cellular and molecular approaches to motor neuron therapy in amyotrophic lateral sclerosis and spinal muscular atrophy. Neurosci Lett 2012; 527:78-84. [PMID: 22579818 DOI: 10.1016/j.neulet.2012.04.079] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2012] [Accepted: 04/29/2012] [Indexed: 12/12/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA) are progressive fatal neurodegenerative diseases. They differ in their disease development but have in common a loss of motor neuron as they progress. Research is ongoing to further understand the origin of these diseases but this common thread of motor neuron loss has provided a target for the development of therapies for both ALS and SMA. It is the linked fields of gene and cell therapy that are providing some of the most interesting therapeutic possibilities.
Collapse
|
20
|
Franz S, Weidner N, Blesch A. Gene therapy approaches to enhancing plasticity and regeneration after spinal cord injury. Exp Neurol 2012; 235:62-9. [PMID: 21281633 PMCID: PMC3116048 DOI: 10.1016/j.expneurol.2011.01.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2010] [Revised: 01/17/2011] [Accepted: 01/24/2011] [Indexed: 01/09/2023]
Abstract
During the past decades, new insights into mechanisms that limit plasticity and functional recovery after spinal cord injury have spurred the development of novel approaches to enhance axonal regeneration and rearrangement of spared circuitry. Gene therapy may provide one means to address mechanisms that underlie the insufficient regenerative response of injured neurons and can also be used to identify factors important for axonal growth. Several genetic approaches aimed to modulate the environment of injured axons, for example by localized expression of growth factors, to enhance axonal sprouting and regeneration and to guide regenerating axons towards their target have been described. In addition, genetic modification of injured neurons via intraparenchymal injection, or via retrograde transport of viral vectors has been used to manipulate the intrinsic growth capacity of injured neurons. In this review we will summarize some of the progress and limitations of cell transplantation and gene therapy to enhance axonal bridging and regeneration across a lesion site, and to maximize the function, collateral sprouting and connectivity of spared axonal systems.
Collapse
Affiliation(s)
- Steffen Franz
- Spinal Cord Injury Center, Heidelberg University Hospital, Germany
| | - Norbert Weidner
- Spinal Cord Injury Center, Heidelberg University Hospital, Germany
| | - Armin Blesch
- Spinal Cord Injury Center, Heidelberg University Hospital, Germany
- Department of Neurosciences, University of California, San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
21
|
Yang P. Lentiviral vector mediates exogenous gene expression in adult rat DRG following peripheral nerve remote delivery. J Mol Neurosci 2012; 47:173-9. [PMID: 22318316 DOI: 10.1007/s12031-012-9710-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2011] [Accepted: 01/16/2012] [Indexed: 02/05/2023]
Abstract
The primary sensory neurons with cell bodies in the dorsal root ganglion (DRG) have been extensively used as models in neurobiology and provide a useful model to study the mechanism of neural regeneration. Therefore, efficient and stable gene delivery to these postmitotic cells has significant therapeutic potential. Various studies involving the viral vector systems capable of neuronal transduction have been extensively evaluated in the cultured DRG neurons by adeno-associated virus. In the present study, we investigated the transduction performance of the lentiviral vector that mediates the catalytic subunit of protein kinase A (PKAc) and green fluorescent protein (GFP) expression in the DRG by sciatic nerve retrograde transport and tested whether PKAc expression in the DRG could inhibit the activation of RhoA after spinal cord injury. Five days after sciatic nerve remote delivery of lentiviral vector (LV)/PKAc-internal ribosome entry site (IRES)-GFP or LV/GFP, the L4-L6 DRGs were dissected for primary culture or immunostaining to observe the exogenous gene expression, or transecting the dorsal part of lumbar enlargement was performed, and 16 h later, the function of the exogenous gene was tested by RhoA pull-down analysis. The results showed that the lentiviral vector could mediate exogenous gene PKAc expression in the DRG and then inhibit spinal cord injury-induced RhoA activation by remote delivery of LV/PKAc-IRES-GFP through the sciatic nerve.
Collapse
Affiliation(s)
- Ping Yang
- Department of Neurobiology, Chongqing Key Laboratory of Neurobiology, Third Military Medical University, Chongqing 400038, People's Republic of China.
| |
Collapse
|
22
|
Rubin AD, Hogikyan ND, Oh A, Feldman EL. Potential for promoting recurrent laryngeal nerve regeneration by remote delivery of viral gene therapy. Laryngoscope 2012; 122:349-55. [PMID: 22241608 DOI: 10.1002/lary.22436] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2011] [Revised: 10/06/2011] [Accepted: 10/13/2011] [Indexed: 12/22/2022]
Abstract
OBJECTIVES/HYPOTHESIS The aims of this study were to demonstrate the ability to enhance nerve regeneration by remote delivery of a viral vector to the crushed recurrent laryngeal nerve (RLN), to demonstrate the usefulness of a crushed RLN model to test the efficacy of viral gene therapy, and to discuss future potential applications of this approach. STUDY DESIGN Animal study. METHODS Adult Sprague-Dawley rats were assigned to two groups. In the experimental group, an adeno-associated viral (AAV) vector carrying a zinc-finger transcription factor, which stimulates endogenous insulinlike growth factor I production (AAV2-TO-6876vp16), was injected into the crushed RLN. In the control group, an AAV vector carrying the gene for green fluorescent protein was injected into the crushed RLN. Unilateral RLN paralysis was confirmed endoscopically. At 1 week, laryngeal endoscopies were repeated and recorded. Larynges were cryosectioned in 15-μm sections and processed for acetylcholine histochemistry (motor endplates) followed by neurofilament immunoperoxidase (nerve fibers). Percentage nerve-endplate contact (PEC) was determined and compared. Vocal fold motion was evaluated by blinded reviewers using a visual analogue scale (VAS). RESULTS The difference between PEC on the crushed and uncrushed sides was statistically less in the experimental group (0.54 ± 0.18 vs. 0.30 ± 0.26, P = .0006). The VAS score at 1 week was significantly better in the experimental group (P = .002). CONCLUSIONS AAV2-TO-6876vp16 demonstrated a neurotrophic effect when injected into the crushed RLN. The RLN offers a conduit for viral gene therapy to the brainstem that could be useful for the treatment of RLN injury or bulbar motor neuron disease.
Collapse
Affiliation(s)
- Adam D Rubin
- Lakeshore Professional Voice Center, St. Clair Shores, Michigan, University of Michigan, Ann Arbor, Michigan 48081, USA.
| | | | | | | |
Collapse
|
23
|
Huang WC, Kuo HS, Tsai MJ, Ma H, Chiu CW, Huang MC, Yang LH, Chang PT, Lin YL, Kuo WC, Lee MJ, Liu JC, Cheng H. Adeno-associated virus-mediated human acidic fibroblast growth factor expression promotes functional recovery of spinal cord-contused rats. J Gene Med 2011; 13:283-9. [PMID: 21557400 DOI: 10.1002/jgm.1568] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Following spinal cord injury, the delivery of neurotrophic factors to the injured spinal cord has been shown to promote axonal regeneration and functional recovery. In previous studies, we showed that acidic fibroblast growth factor (aFGF) is a potent neurotrophic factor that promotes the regeneration of axotomized spinal cord or dorsal root ganglion neurones. METHODS We constructed a recombinant adeno-associated virus (AAV) vector to express human aFGF and evaluated aFGF expression and function in AAV-aFGF-infected PC12 cells. We analyzed AAV-green fluorescent protein (GFP) tropism and AAV-mediated aFGF expression in contused spinal cords. Animals received behavioural testing to evaluate the functional recovery. RESULTS Overexpression of aFGF was shown in AAV-aFGF-infected PC12 cells in a dose-dependent manner. Concurrently, neurite extension and cell number were significantly increased in AAV-aFGF infected cells. AAV-mediated GFP expression persisted for at least 5 weeks in contused spinal cords, and the most prominently transduced cells were neurones. Contusive injury reduced endogenous aFGF expression in spinal cords. Overexpression of aFGF was demonstrated in AAV-aFGF transduced spinal cords compared to AAV-GFP transduced spinal cords at 3 and 14 days post-injury. Evaluation of motor function revealed that the improvement of AAV-aFGF-treated rats was prominent. Both AAV-aFGF- and recombinant human aFGF-treated rats revealed significantly better recovery at 5 weeks post-injury, compared to vehicle- and AAV-GFP-treated rats. CONCLUSIONS These data suggest that supplement of aFGF improve the functional recovery of spinal cord-contused rats and that AAV-aFGF-mediated gene transfer could be a clinically feasible therapeutic approach for patients after nervous system injuries.
Collapse
Affiliation(s)
- Wen-Cheng Huang
- Centre for Neural Regeneration, Department of Neurosurgery, Neurological Institute, Taipei Veterans General Hospital, Taipei, Taiwan
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Federici T, Taub JS, Baum GR, Gray SJ, Grieger JC, Matthews KA, Handy CR, Passini MA, Samulski RJ, Boulis NM. Robust spinal motor neuron transduction following intrathecal delivery of AAV9 in pigs. Gene Ther 2011; 19:852-9. [DOI: 10.1038/gt.2011.130] [Citation(s) in RCA: 122] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
25
|
Stepanichev MY. Current approaches and future directions of gene therapy in Alzheimer’s disease. NEUROCHEM J+ 2011. [DOI: 10.1134/s181971241103010x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
|
26
|
Handy CR, Krudy C, Boulis N. Gene therapy: a potential approach for cancer pain. PAIN RESEARCH AND TREATMENT 2011; 2011:987597. [PMID: 22110939 PMCID: PMC3196247 DOI: 10.1155/2011/987597] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/01/2010] [Revised: 12/14/2010] [Accepted: 01/21/2011] [Indexed: 12/21/2022]
Abstract
Chronic pain is experienced by as many as 90% of cancer patients at some point during the disease. This pain can be directly cancer related or arise from a sensory neuropathy related to chemotherapy. Major pharmacological agents used to treat cancer pain often lack anatomical specificity and can have off-target effects that create new sources of suffering. These concerns establish a need for improved cancer pain management. Gene therapy is emerging as an exciting prospect. This paper discusses the potential for viral vector-based treatment of cancer pain. It describes studies involving vector delivery of transgenes to laboratory pain models to modulate the nociceptive cascade. It also discusses clinical investigations aimed at regulating pain in cancer patients. Considering the prevalence of pain among cancer patients and the growing potential of gene therapy, these studies could set the stage for a new class of medicines that selectively disrupt nociceptive signaling with limited off-target effects.
Collapse
Affiliation(s)
- Chalonda R. Handy
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Rm 6339, Atlanta, GA 30322, USA
| | - Christina Krudy
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Rm 6339, Atlanta, GA 30322, USA
| | - Nicholas Boulis
- Department of Neurosurgery, Emory University, 101 Woodruff Circle, Rm 6339, Atlanta, GA 30322, USA
| |
Collapse
|
27
|
Zhang Y, Zheng Y, Zhang YP, Shields LBE, Hu X, Yu P, Burke DA, Wang H, Jun C, Byers J, Whittemore SR, Shields CB. Enhanced adenoviral gene delivery to motor and dorsal root ganglion neurons following injection into demyelinated peripheral nerves. J Neurosci Res 2010; 88:2374-84. [PMID: 20623527 DOI: 10.1002/jnr.22394] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Injection of viral vectors into peripheral nerves may transfer specific genes into their dorsal root ganglion (DRG) neurons and motoneurons. However, myelin sheaths of peripheral axons block the entry of viral particles into nerves. We studied whether mild, transient peripheral nerve demyelination prior to intraneural viral vector injection would enhance gene transfer to target DRG neurons and motoneurons. The right sciatic nerve of C57BL/6 mice was focally demyelinated with 1% lysolecithin, and the left sciatic nerve was similarly injected with saline (control). Five days after demyelination, 0.5 microl of Ad5-GFP was injected into both sciatic nerves at the site of previous injection. The effectiveness of gene transfer was evaluated by counting GFP(+) neurons in the DRGs and ventral horns. After peripheral nerve demyelination, there was a fivefold increase in the number of infected DRG neurons and almost a 15-fold increase in the number of infected motoneurons compared with the control, nondemyelinated side. Focal demyelination reduced the myelin sheath barrier, allowing greater virus-axon contact. Increased CXADR expression on the demyelinated axons facilitated axoplasmic viral entry. No animals sustained any prolonged neurological deficits. Increased gene delivery into DRG neurons and motoneurons may provide effective treatment for amyotrophic lateral sclerosis, pain, and spinal cord injury.
Collapse
Affiliation(s)
- Yongjie Zhang
- Kentucky Spinal Cord Injury Research Center, Louisville, Kentucky, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Tsai MJ, Pan HA, Liou DY, Weng CF, Hoffer BJ, Cheng H. Adenoviral gene transfer of bone morphogenetic protein-7 enhances functional recovery after sciatic nerve injury in rats. Gene Ther 2010; 17:1214-24. [PMID: 20520648 DOI: 10.1038/gt.2010.72] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Bone morphogenetic proteins (BMPs), members of the transforming growth factor-β subfamily, function as instructive signals for neuronal lineage commitment and promote neuronal differentiation. However, the mechanism of BMP7 action in vivo after peripheral nerve injury is poorly understood. This study examines the efficacy of gene transfer of adenoviral (Ad) BMP7 on peripheral neuropathy. Transgene expression was found in both Ad-infected sciatic nerves and their respective remote neurons, indicating Ad transduction by a retrograde transport. After AdBMP7 infection to nerves, the sciatic nerves were crushed or transected. Hind limb functional behavior, including rotarod test and sciatic functional index, were conducted in rats weekly after nerve injury. Interestingly, enhanced BMP7 expression significantly improved hind limb functional recovery in AdBMP7-transduced rats when compared with AdGFP-transduced nerve-crushed or transected rats. Furthermore, AdBMP7 transduction reduced injury-induced macrophage activation, nerve demyelination and axonal degeneration. By contrast, AdBMP7 infection did not affect the hyperalgesia paw-withdrawal latency after nerve injury. We further examined the effect of AdBMP7 infection on sciatic nerve explant and Schwann cell cultures. Enhanced cell proliferation was significantly increased by AdBMP7 transduction in both cultures. Taken together, BMP7 overexpression by Ad gene transfer was beneficial in both nerves and Schwann cells on functional recovery after sciatic nerve injury in rats.
Collapse
Affiliation(s)
- M-J Tsai
- Neural Regeneration Laboratory, Neurological Institute, Taipei Veterans General Hospital, No. 322 Shih-pai Road Sec. 2, Taipei, Taiwan
| | | | | | | | | | | |
Collapse
|
29
|
Federici T, Boulis NM. Invited review: festschrift edition of neurosurgery peripheral nervous system as a conduit for delivering therapies for diabetic neuropathy, amyotrophic lateral sclerosis, and nerve regeneration. Neurosurgery 2010; 65:A87-92. [PMID: 19927084 DOI: 10.1227/01.neu.0000335653.52938.f2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
In this review, we describe how therapies that promote axonal regeneration and neuronal protection can complement surgery for a successful functional restoration in peripheral nerve disorders. We discuss the advantages of peripheral drug delivery and the role of the neurosurgeon in the precise delivery of molecular therapies to surgically inaccessible structures. Strategies for enhancing uptake and retrograde transport of therapeutics, including gene therapy, are emphasized as conduits for delivery of therapeutics. Finally, candidate therapeutic proteins and genes are discussed in the context of application to degenerative disorders of the nervous system, including nerve injury, peripheral neuropathy, and amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Thais Federici
- Department of Neurosurgery, Emory University, Atlanta, Georgia 30322, USA
| | | |
Collapse
|
30
|
Snyder BR, Boulis NM, Federici T. Viral vector-mediated gene transfer for CNS disease. Expert Opin Biol Ther 2010; 10:381-94. [DOI: 10.1517/14712590903514074] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
31
|
Towne C, Pertin M, Beggah AT, Aebischer P, Decosterd I. Recombinant adeno-associated virus serotype 6 (rAAV2/6)-mediated gene transfer to nociceptive neurons through different routes of delivery. Mol Pain 2009; 5:52. [PMID: 19737386 PMCID: PMC2747840 DOI: 10.1186/1744-8069-5-52] [Citation(s) in RCA: 120] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2009] [Accepted: 09/08/2009] [Indexed: 11/27/2022] Open
Abstract
Background Gene transfer to nociceptive neurons of the dorsal root ganglia (DRG) is a promising approach to dissect mechanisms of pain in rodents and is a potential therapeutic strategy for the treatment of persistent pain disorders such as neuropathic pain. A number of studies have demonstrated transduction of DRG neurons using herpes simplex virus, adenovirus and more recently, adeno-associated virus (AAV). Recombinant AAV are currently the gene transfer vehicles of choice for the nervous system and have several advantages over other vectors, including stable and safe gene expression. We have explored the capacity of recombinant AAV serotype 6 (rAAV2/6) to deliver genes to DRG neurons and characterized the transduction of nociceptors through five different routes of administration in mice. Results Direct injection of rAAV2/6 expressing green fluorescent protein (eGFP) into the sciatic nerve resulted in transduction of up to 30% eGFP-positive cells of L4 DRG neurons in a dose dependant manner. More than 90% of transduced cells were small and medium sized neurons (< 700 μm2), predominantly colocalized with markers of nociceptive neurons, and had eGFP-positive central terminal fibers in the superficial lamina of the spinal cord dorsal horn. The efficiency and profile of transduction was independent of mouse genetic background. Intrathecal administration of rAAV2/6 gave the highest level of transduction (≈ 60%) and had a similar size profile and colocalization with nociceptive neurons. Intrathecal administration also transduced DRG neurons at cervical and thoracic levels and resulted in comparable levels of transduction in a mouse model for neuropathic pain. Subcutaneous and intramuscular delivery resulted in low levels of transduction in the L4 DRG. Likewise, delivery via tail vein injection resulted in relatively few eGFP-positive cells within the DRG, however, this transduction was observed at all vertebral levels and corresponded to large non-nociceptive cell types. Conclusion We have found that rAAV2/6 is an efficient vector to deliver transgenes to nociceptive neurons in mice. Furthermore, the characterization of the transduction profile may facilitate gene transfer studies to dissect mechanisms behind neuropathic pain.
Collapse
Affiliation(s)
- Chris Towne
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, EPFL, Lausanne, Switzerland.
| | | | | | | | | |
Collapse
|
32
|
Kells AP, Hadaczek P, Yin D, Bringas J, Varenika V, Forsayeth J, Bankiewicz KS. Efficient gene therapy-based method for the delivery of therapeutics to primate cortex. Proc Natl Acad Sci U S A 2009; 106:2407-11. [PMID: 19193857 PMCID: PMC2650169 DOI: 10.1073/pnas.0810682106] [Citation(s) in RCA: 109] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2008] [Indexed: 11/18/2022] Open
Abstract
Transduction of the primate cortex with adeno-associated virus (AAV)-based gene therapy vectors has been challenging, because of the large size of the cortex. We report that a single infusion of AAV2 vector into thalamus results in widespread expression of transgene in the cortex through transduction of widely dispersed thalamocortical projections. This finding has important implications for the treatment of certain genetic and neurodegenerative diseases.
Collapse
Affiliation(s)
- Adrian P. Kells
- Department of Neurological Surgery, University of California, 1855 Folsom Street, San Francisco, CA 94103
| | - Piotr Hadaczek
- Department of Neurological Surgery, University of California, 1855 Folsom Street, San Francisco, CA 94103
| | - Dali Yin
- Department of Neurological Surgery, University of California, 1855 Folsom Street, San Francisco, CA 94103
| | - John Bringas
- Department of Neurological Surgery, University of California, 1855 Folsom Street, San Francisco, CA 94103
| | - Vanja Varenika
- Department of Neurological Surgery, University of California, 1855 Folsom Street, San Francisco, CA 94103
| | - John Forsayeth
- Department of Neurological Surgery, University of California, 1855 Folsom Street, San Francisco, CA 94103
| | - Krystof S. Bankiewicz
- Department of Neurological Surgery, University of California, 1855 Folsom Street, San Francisco, CA 94103
| |
Collapse
|
33
|
Aspalter M, Vyas A, Feiner J, Griffin J, Brushart T, Redett R. Modification of Schwann cell gene expression by electroporation in vivo. J Neurosci Methods 2009; 176:96-103. [PMID: 18834904 PMCID: PMC2640232 DOI: 10.1016/j.jneumeth.2008.08.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2008] [Revised: 08/21/2008] [Accepted: 08/24/2008] [Indexed: 11/29/2022]
Abstract
Clinical outcomes of nerve grafting are often inferior to those of end-to-end nerve repair. This may be due, in part, to the routine use of cutaneous nerve to support motor axon regeneration. In previous work, we have demonstrated that Schwann cells express distinct sensory and motor phenotypes, and that these promote regeneration in a modality-specific fashion. Intra-operative modification of graft Schwann cell phenotype might therefore improve clinical outcomes. This paper demonstrates the feasibility of electroporating genes into intact nerve to modify Schwann cell gene expression. Initial trials established 70 V, 5 ms as optimum electroporation parameters. Intact, denervated, and reinnervated rat tibial nerves were electroporated with the YFP gene and evaluated serially by counting S-100 positive cells that expressed YFP. In intact nerve, a mean of 28% of Schwann cells expressed the gene at 3 days, falling to 20% at 7 days with little expression at later times. There were no significant differences among the three groups at each time period. Electronmicroscopic evaluation of treated, intact nerve revealed only occasional demyelination and axon degeneration. Intra-operative electroporation of nerve graft is thus a practical means of altering Schwann cell gene expression without the risks inherent in viral transfection.
Collapse
Affiliation(s)
- Manuela Aspalter
- Department of Orthopaedic Surgery, Johns Hopkins University, 601 North Caroline Street, Baltimore, MD 21287, United States
| | | | | | | | | | | |
Collapse
|
34
|
Federici T, Kutner R, Zhang XY, Kuroda H, Tordo N, Boulis NM, Reiser J. Comparative analysis of HIV-1-based lentiviral vectors bearing lyssavirus glycoproteins for neuronal gene transfer. GENETIC VACCINES AND THERAPY 2009; 7:1. [PMID: 19144125 PMCID: PMC2639530 DOI: 10.1186/1479-0556-7-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/08/2008] [Accepted: 01/13/2009] [Indexed: 01/16/2023]
Abstract
Background The delivery of therapeutic genes to the central nervous system (CNS) using viral vectors represents an appealing strategy for the treatment of nerve injury and disorders of the CNS. Important factors determining CNS targeting include tropism of the viral vectors and retrograde transport of the vector particles. Retrograde transport of equine anemia virus (EIAV)-based lentiviral vectors pseudotyped with the glycoprotein derived from the Rabies virus RabERA strain from peripheral muscle to spinal motor neurons (MNs) was previously reported. Despite therapeutic effects achieved in mouse models of amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), the efficiency of this approach needs to be improved for clinical translation. To date there has not been a quantitative assessment of pseudotyped HIV-1-based lentiviral vectors to transduce MNs. Here, we describe quantitative tests to analyze the retrograde transport capacity of HIV-1 vectors pseudotyped with the G glycoprotein derived from Rabies and Rabies-related viruses (Lyssaviruses). Methods With a view toward optimizing the retrograde transport properties of HIV-1-based lentiviral vectors, we compared the glycoproteins from different enveloped viruses belonging to the Rhabdoviridae family, genus Lyssavirus, and evaluated their ability to transduce specific cell populations and promote retrograde axonal transport. We first tested the transduction performance of these pseudotypes in vitro in SH-SY5Y neuroblastoma cells, NSC-34 neuroblastoma-spinal cord hybrid cells, and primary mixed spinal cord and pure astrocyte cultures. We then analyzed the uptake and retrograde transport of these pseudotyped vectors in vitro, using Campenot chambers. Finally, intraneural injections were performed to evaluate the in vivo retrograde axonal transport of these pseudotypes. Results Both the in vitro and in vivo studies demonstrated that lentiviral vectors pseudotyped with the glycoprotein derived from the Rabies virus PV strain possessed the best performance and neuronal tropism among the vectors tested. Conclusion Our results indicate that HIV-1-based lentiviral vectors pseudotyped with the Rabies PV glycoprotein might provide important vehicles for CNS targeting by peripheral injection in the treatment of motor neuron diseases (MND), pain, and neuropathy.
Collapse
Affiliation(s)
- Thais Federici
- Department of Neurosurgery, Emory University, Atlanta, GA, USA.
| | | | | | | | | | | | | |
Collapse
|
35
|
Carlton E, Teng Q, Federici T, Yang J, Riley J, Boulis NM. FUSION OF THE TETANUS TOXIN C FRAGMENT BINDING DOMAIN AND BCL-XL FOR PROTECTION OF PERIPHERAL NERVE NEURONS. Neurosurgery 2008; 63:1175-82; discussion 1182-4. [DOI: 10.1227/01.neu.0000334415.45003.ea] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Erin Carlton
- Department of Cell Biology, Cleveland Clinic, Cleveland, Ohio
| | - Qingshan Teng
- Department of Neurosurgery, Emory University, Atlanta, Georgia
| | - Thais Federici
- Department of Neurosurgery, Emory University, Atlanta, Georgia
| | - Jun Yang
- Department of Neurosurgery, Cleveland Clinic, Cleveland, Ohio
| | | | | |
Collapse
|
36
|
Federici T, Liu JK, Teng Q, Garrity-Moses M, Yang J, Boulis NM. Neuronal affinity of a C7C loop peptide identified through phage display. J Drug Target 2008; 14:263-71. [PMID: 16882546 DOI: 10.1080/10611860600763079] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Phage display is a promising tool for the screening of peptides with high affinity for specific cells. Here we describe a novel peptide with neuronal affinity isolated from a C7C library. We designed a two-tiered biopanning strategy initially selecting for ganglioside binding and subsequently selecting for binding to PC12 cells. At the completion of biopanning, 54.8% of phage clones bore the identical peptide (Tet.C7C.1). Immunofluorescence confirmed selective binding of this clone to differentiated PC12 cells. Tet.C7C.1 was synthesized and fluorescein conjugated. The synthetic peptide binds neuronal cell lines (SH-SY5Y, NSC-34 and PC12 cells) and tissue (DRG and spinal cord). The C7C structure creates a loop that minimizes the impact of peptide insertion on the confirmation of the recipient protein. Small loop peptides have the ideal characteristics for modification of viral vector capsids without undermining genome packaging. The neuronal binding properties of this peptide may be applied in the development of neurotropic viral vectors.
Collapse
Affiliation(s)
- Thais Federici
- Department of Neuroscience and Center for Neurological Restoration, Cleveland Clinic, 9500 Euclid Ave, Cleveland, OH, 44195, USA.
| | | | | | | | | | | |
Collapse
|
37
|
McClelland S, Teng Q, Benson LS, Boulis NM. Motor neuron inhibition-based gene therapy for spasticity. Am J Phys Med Rehabil 2007; 86:412-21. [PMID: 17449986 DOI: 10.1097/phm.0b013e31804a83cf] [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] [Indexed: 11/27/2022]
Abstract
Spasticity is a condition resulting from excess motor neuron excitation, leading to involuntary muscle contraction in response to increased velocity of movement, for which there is currently no cure. Existing symptomatic therapies face a variety of limitations. The extent of relief that can be delivered by ablative techniques such as rhizotomy is limited by the potential for sensory denervation. Pharmacological approaches, including intrathecal baclofen, can be undermined by tolerance. One potential new approach to the treatment of spasticity is the control of neuromuscular overactivity through the delivery of genes capable of inducing synaptic inhibition. A variety of experiments in cell culture and animal models have demonstrated the ability of neural gene transfer to inhibit neuronal activity and suppress transmission. Similarly, enthusiasm for the application of gene therapy to neurodegenerative diseases of motor neurons has led to the development of a variety of strategies for motor neuron gene delivery. In this review, we discuss the limitations of existing spasticity therapies, the feasibility of motor neuron inhibition as a gene-based treatment for spasticity, potential inhibitory transgene candidates, strategies for control of transgene expression, and applicable motor neuron gene targeting strategies. Finally, we discuss future directions and the potential for gene-based motor neuron inhibition in therapeutic clinical trials to serve as an effective treatment modality for spasticity, either in conjunction with or as a replacement for presently available therapies.
Collapse
Affiliation(s)
- Shearwood McClelland
- Department of Neurosurgery, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | | | | | | |
Collapse
|
38
|
Federici T, Liu JK, Teng Q, Yang J, Boulis NM. A Means for Targeting Therapeutics to Peripheral Nervous System Neurons with Axonal Damage. Neurosurgery 2007; 60:911-8; discussion 911-8. [PMID: 17460527 DOI: 10.1227/01.neu.0000255444.44365.b9] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE Delivery of biological therapeutics to motor and dorsal root ganglion neurons remains a major hurdle in the development of treatments for a variety of neurological processes, including peripheral nerve injury, pain, and motor neuron diseases. Because nerve cell bodies are important in initiating and controlling axonal regeneration, targeted delivery is an appealing strategy to deliver therapeutic proteins after peripheral nerve injury. METHODS Tet1 is a 12-aa peptide, isolated through phage display that is selected for tetanus toxin C fragment-like binding properties. In this study, we surveyed its uptake and retrograde transport using compartmented cultures and sciatic nerve injections. We then characterized the time course of this delivery. Finally, to confirm the retrograde transport involvement, a colchicine pretreatment was performed. We also performed competitive binding studies between Tet1 and a recombinant tetanus toxin C fragment using recombinant tetanus toxin C fragment enzyme-linked immunosorbent assay. RESULTS We were able to demonstrate efficient uptake and retrograde axonal transport of the Tet1 peptide in vitro and in vivo. Intraneural colchicine pretreatment partially blocked fluorescence detection in the spinal cord, revealing a retrograde axonal transport mechanism. Finally, a competitive enzyme-linked immunosorbent assay experiment revealed Tet1-specific binding to the recombinant tetanus toxin C fragment axon terminal trisialogangliosides receptor. CONCLUSION These properties of Tet1 can be applied to the development of therapeutic viral vectors and fusion proteins for neuronal targeting and enhanced spinal cord delivery in the treatment of nerve regeneration, neuroprotection, analgesia, and spasticity. Small peptides can be easily fused to larger proteins without significantly modifying their function and can be used to alter the binding and uptake properties of these proteins.
Collapse
|
39
|
Pearse DD, Bunge MB. Designing cell- and gene-based regeneration strategies to repair the injured spinal cord. J Neurotrauma 2006; 23:438-52. [PMID: 16629628 DOI: 10.1089/neu.2006.23.437] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
There is an array of new and promising strategies being developed to improve function after spinal cord injury (SCI). The targeting of a diversity of deleterious processes within the tissue after SCI will necessitate a multi-factorial intervention, such as the combination of cell- and gene-based approaches. To ensure proper development and design of these experiments, many issues need to be addressed. It is the purpose of this review to consider the strategies involved in testing the efficacy of these new combinations to improve axonal regeneration. For cell-based therapy, issues are choosing a SCI model, the time of cell implantation, placement of cells and their subsequent migration, fluid versus solid grafts, use of agents to prevent immune rejection, and tracking of implanted cells. Grafting is also discussed in view of improving function, reducing secondary damage, bridging the injured spinal cord, supporting axonal regrowth, replacing lost neurons, facilitating myelination, and promoting axonal growth from the implant into the cord. The choice of a gene delivery system, gene-based therapies in vivo to provide chemoattractant and guidance cues, altering the intrinsic regenerative capacity of neurons, enhancing endogenous non-neuronal cell functions, and targeting the synthesis of growth inhibitory molecules are also discussed, as well as combining ex vivo gene and cell therapies.
Collapse
Affiliation(s)
- D D Pearse
- The Miami Project to Cure Paralysis, Department of Neurological Surgery, University of Miami Leonard M. Miller School of Medicine, Miami, Florida 33101, USA.
| | | |
Collapse
|
40
|
Garrity-Moses ME, Teng Q, Liu J, Tanase D, Boulis NM. Neuroprotective adeno-associated virus Bcl-xL gene transfer in models of motor neuron disease. Muscle Nerve 2006; 32:734-44. [PMID: 16116646 DOI: 10.1002/mus.20418] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recent work implicates excitotoxicity-induced apoptosis as the mechanism triggering motor neuron death in amyotrophic lateral sclerosis (ALS). Our laboratory has previously utilized glutamate excitotoxicity in vitro to study this process. The present experiment tests whether overexpression of the gene for Bcl-xL can inhibit excitotoxicity in this model system. To track Bcl-xL expression, the gene for green fluorescent protein (GFP) was inserted in-frame, upstream of the Bcl-xL gene. The GFP-Bcl-xL gene was then cloned into an adeno-associated viral (AAV2) vector. GFP expression in both SH-SY5Y and embryonic day 15 (E15) motor neurons (MNs) peaked 48 hours after infection. Bcl-xL expression in SH-SY5Y cells significantly reduced terminal deoxy-UTP nick-end labeling (TUNEL)-positive cells and maintained cell density after glutamate exposure. Similarly, Bcl-xL expression inhibited the development of TUNEL staining in E15 MNs and supported cell density after glutamate exposure. These findings suggest that AAV-mediated expression of genes for antiapoptotic proteins may provide a means for ALS gene therapy.
Collapse
Affiliation(s)
- Mary E Garrity-Moses
- Department of Neurological Surgery, Lerner Research Institute, Cleveland Clinic Foundation, NB 2 120, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
Motor neuron diseases (MND), such as amyotrophic lateral sclerosis (ALS) and spinal muscular atrophy (SMA), are progressive neurodegenerative diseases that share the common characteristic of upper and/or lower motor neuron degeneration. Therapeutic strategies for MND are designed to confer neuroprotection, using trophic factors, anti-apoptotic proteins, as well as antioxidants and anti-excitotoxicity agents. Although a large number of therapeutic clinical trials have been attempted, none has been shown satisfactory for MND at this time. A variety of strategies have emerged for motor neuron gene transfer. Application of these approaches has yielded therapeutic results in cell culture and animal models, including the SOD1 models of ALS. In this study we describe the gene-based treatment of MND in general, examining the potential viral vector candidates, gene delivery strategies, and main therapeutic approaches currently attempted. Finally, we discuss future directions and potential strategies for more effective motor neuron gene delivery and clinical translation.
Collapse
Affiliation(s)
- Thais Federici
- Department of Neuroscience, Cleveland Clinic Foundation, NB2-126A, 9500 Euclid Avenue, Cleveland, Ohio 44195, USA
| | | |
Collapse
|
42
|
Vincent AM, Feldman EL, Song DK, Jung V, Schild A, Zhang W, Imperiale MJ, Boulis NM. Adeno-associated viral-mediated insulin-like growth factor delivery protects motor neurons in vitro. Neuromolecular Med 2005; 6:79-85. [PMID: 15970625 DOI: 10.1385/nmm:6:2-3:079] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2004] [Revised: 11/23/2004] [Accepted: 11/30/2004] [Indexed: 11/11/2022]
Abstract
Recent work has demonstrated that adeno-associated viral (AAV) vector-mediated delivery of the insulin-like growth factor (IGF-I) gene through retrograde axonal transport can prolong survival and delay disease onset in the superoxide dismutase mutant mouse model of motor neuron (MN) disease. The present experiment examines IGF-I gene transfer in vitro. Adenoviral and AAV vectors for IGF-I infect neurons triggering expression and secretion of biologically active IGF-I. AAV-mediated IGF-I expression in SH-SY5Y neurons protects both cells expressing the transgene, and bystanders without transgene expression from glutamate-induced apoptosis. Similarly, AAV-mediated IGF-I delivery in primary E15 MN culture provides a titer-dependent neuroprotection from glutamate-induced DNA fragmentation. Both infected and noninfected neurons are equally protected. These observations argue that vector-mediated IGF-I gene transfer induces secretion of active IGF-I that acts through direct effects on spinal cord MNs. This mechanism may explain the therapeutic effects observed in vivo despite relatively low affinity AAV spinal cord uptake.
Collapse
|
43
|
Liu JK, Teng Q, Garrity-Moses M, Federici T, Tanase D, Imperiale MJ, Boulis NM. A novel peptide defined through phage display for therapeutic protein and vector neuronal targeting. Neurobiol Dis 2005; 19:407-18. [PMID: 16023583 DOI: 10.1016/j.nbd.2005.01.022] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2004] [Revised: 12/01/2004] [Accepted: 01/20/2005] [Indexed: 01/04/2023] Open
Abstract
A novel peptide with the binding characteristics of tetanus toxin was identified with phage display, for application in therapeutic protein and vector motor and sensory neuron targeting. A 12mer phage library was biopanned on trisialoganglioside (G(T1b)) and eluted with the tetanus toxin C fragment (rTTC). Phage ELISAs revealed increases in G(T1b) binding for the Tet1 and Tet2 phage clones when compared to peptideless phage (PLP). rTTC displaced both Tet1 and Tet2 phage clones from G(T1b), and both clones reduced rTTC-G(T1b) binding. Comparison of Tet1, Tet2, PLP, and the random phage library binding to PC12 and HEK293 cells revealed enhanced cellular binding by Tet1 and Tet2 phage. Tet1 phage binding was selective for neurons. Immunofluorescence also confirmed selective PC12 binding of Tet1 and Tet2 phage. Fluorescein-conjugated synthetic Tet1, but not Tet2, peptide showed strong binding to cultured PC12, primary motor neurons, and dorsal root ganglion (DRG) cells. Synthetic Tet1 bound DRG and motor neurons but not muscle in tissue sections. The enhanced neuronal binding affinity and specificity of Tet1, a novel 12 amino acid peptide, suggests potential utility for targeting neurotherapeutic proteins and viral vectors in the treatment of motor neuron disease, neuropathy, and pain.
Collapse
Affiliation(s)
- James K Liu
- Department of Neuroscience and Center for Neurological Restoration, NB2-126, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | | | | | | | | | |
Collapse
|
44
|
Teng Q, Garrity-Moses M, Federici T, Tanase D, Liu JK, Mazarakis ND, Azzouz M, Walmsley LE, Carlton E, Boulis NM. Trophic activity of Rabies G protein-pseudotyped equine infectious anemia viral vector mediated IGF-I motor neuron gene transfer in vitro. Neurobiol Dis 2005; 20:694-700. [PMID: 16005636 DOI: 10.1016/j.nbd.2005.05.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2004] [Revised: 04/25/2005] [Accepted: 05/02/2005] [Indexed: 11/20/2022] Open
Abstract
The present study examines gene delivery to cultured motor neurons (MNs) with the Rabies G protein (RabG)-pseudotyped lentiviral equine infectious anemia virus (RabG.EIAV) vector. RabG.EIAV-mediated beta-galactosidase (RabG.EIAV-LacZ) gene expression in cultured MNs plateaus 120 h after infection. The rate and percent of gene expression observed are titer-dependent (P < 0.001). The rat IGF-I cDNA sequence was then cloned into a RabG.EIAV vector (RabG.EIAV-IGF-I) and was shown to induce IGF-I expression in HEK 293 cells. MNs infected with RabG.EIAV-IGF-I demonstrate enhanced survival compared to MNs infected with RabG.EIAV-LacZ virus (P < 0.01). In addition, IGF-I expression in cultured MNs induced profound MN axonal elongation compared to control virus (P < 0.01). The enhanced motor neuron tropism of RabG.EIAV previously demonstrated in vivo, together with the trophic effects of RabG.EIAV-IGF-I MN gene expression may lend this vector to therapeutic application in motor neuron disease.
Collapse
Affiliation(s)
- Qingshan Teng
- Department of Neuroscience and Center for Neurological Restoration, Lerner Research Institute, Cleveland Clinic Foundation, NB2-126, 9500 Euclid Avenue, Cleveland, OH 44195, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Teng Q, Tanase D, Tanase DK, Liu JK, Garrity-Moses ME, Baker KB, Boulis NM. Adenoviral clostridial light chain gene-based synaptic inhibition through neuronal synaptobrevin elimination. Gene Ther 2005; 12:108-19. [PMID: 15496959 DOI: 10.1038/sj.gt.3302400] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Clostridial neurotoxins have assumed increasing importance in clinical application. The toxin's light chain component (LC) inhibits synaptic transmission by digesting vesicle-docking proteins without directly altering neuronal health. To study the properties of LC gene expression in the nervous system, an adenoviral vector containing the LC of tetanus toxin (AdLC) was constructed. LC expressed in differentiated neuronal PC12 cells was shown to induce time- and concentration-dependent digestion of mouse brain synaptobrevin in vitro as compared to control transgene products. LC gene expression in the rat lumbar spinal cord disrupted hindlimb sensorimotor function in comparison to control vectors as measured by the Basso-Beattie-Bresnahan (BBB) scale (P<0.001) and rotarod assay (P<0.003). Evoked electromyography (EMG) showed increased stimulus threshold and decreased response current amplitude in LC gene-transferred rats. At the peak of functional impairment, neither neuronal TUNEL staining nor reduced motor neuron density could be detected. Spontaneous functional recovery was observed to parallel the cessation of LC gene expression. These results suggest that light chain gene delivery within the nervous system may provide a nondestructive means for focused neural inhibition to treat a variety of disorders related to excessive synaptic activity, and prove useful for the study of neural circuitry.
Collapse
Affiliation(s)
- Q Teng
- Department of Neuroscience, Lerner Research Institute, Cleveland, OH, USA
| | | | | | | | | | | | | |
Collapse
|