1
|
Andreyanov M, Heinrich R, Berlin S. Design of Ultrapotent Genetically Encoded Inhibitors of Kv4.2 for Gating Neural Plasticity. J Neurosci 2024; 44:e2295222023. [PMID: 38154956 PMCID: PMC10869153 DOI: 10.1523/jneurosci.2295-22.2023] [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: 12/15/2022] [Revised: 11/05/2023] [Accepted: 11/22/2023] [Indexed: 12/30/2023] Open
Abstract
The Kv4.2 potassium channel plays established roles in neuronal excitability, while also being implicated in plasticity. Current means to study the roles of Kv4.2 are limited, motivating us to design a genetically encoded membrane tethered Heteropodatoxin-2 (MetaPoda). We find that MetaPoda is an ultrapotent and selective gating-modifier of Kv4.2. We narrow its site of contact with the channel to two adjacent residues within the voltage sensitive domain (VSD) and, with docking simulations, suggest that the toxin binds the VSD from within the membrane. We also show that MetaPoda does not require an external linker of the channel for its activity. In neurons (obtained from female and male rat neonates), MetaPoda specifically, and potently, inhibits all Kv4 currents, leaving all other A-type currents unaffected. Inhibition of Kv4 in hippocampal neurons does not promote excessive excitability, as is expected from a simple potassium channel blocker. We do find that MetaPoda's prolonged expression (1 week) increases expression levels of the immediate early gene cFos and prevents potentiation. These findings argue for a major role of Kv4.2 in facilitating plasticity of hippocampal neurons. Lastly, we show that our engineering strategy is suitable for the swift engineering of another potent Kv4.2-selective membrane-tethered toxin, Phrixotoxin-1, denoted MetaPhix. Together, we provide two uniquely potent genetic tools to study Kv4.2 in neuronal excitability and plasticity.
Collapse
Affiliation(s)
- Michael Andreyanov
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa 3525433, Israel
| | - Ronit Heinrich
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa 3525433, Israel
| | - Shai Berlin
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa 3525433, Israel
| |
Collapse
|
2
|
Castro MG, Candolfi M, Wilson TJ, Calinescu A, Paran C, Kamran N, Koschmann C, Moreno-Ayala MA, Assi H, Lowenstein PR. Adenoviral vector-mediated gene therapy for gliomas: coming of age. Expert Opin Biol Ther 2014; 14:1241-57. [PMID: 24773178 DOI: 10.1517/14712598.2014.915307] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
INTRODUCTION Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults and it carries a dismal prognosis. Adenoviral vector (Ad)-mediated gene transfer is being developed as a promising therapeutic strategy for GBM. Preclinical studies have demonstrated safety and efficacy of adenovirus administration into the brain and tumor mass in rodents and into the non-human primates' brain. Importantly, Ads have been safely administered within the tumor resection cavity in humans. AREAS COVERED This review gives background on GBM and Ads; we describe gene therapy strategies for GBM and discuss the value of combination approaches. Finally, we discuss the results of the human clinical trials for GBM that have used Ads. EXPERT OPINION The transduction characteristics of Ads, and their safety profile, added to their capacity to achieve high levels of transgene expression have made them powerful vectors for the treatment of GBM. Recent gene therapy successes in the treatment of retinal diseases and systemic brain metabolic diseases encourage the development of gene therapy for malignant glioma. Exciting clinical trials are currently recruiting patients; although, it is the large randomized Phase III controlled clinical trials that will provide the final decision on the success of gene therapy for the treatment of GBM.
Collapse
Affiliation(s)
- Maria G Castro
- University of Michigan Medical School, Department of Neurosurgery , 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689 , USA +734 764 0850 ; +734 764 7051 ;
| | | | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Castro MG, Candolfi M, Wilson TJ, Calinescu A, Paran C, Kamran N, Koschmann C, Moreno-Ayala MA, Assi H, Lowenstein PR. Adenoviral vector-mediated gene therapy for gliomas: coming of age. Expert Opin Biol Ther 2014. [PMID: 24773178 DOI: 10.1517/14712598.2014.91530] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults and it carries a dismal prognosis. Adenoviral vector (Ad)-mediated gene transfer is being developed as a promising therapeutic strategy for GBM. Preclinical studies have demonstrated safety and efficacy of adenovirus administration into the brain and tumor mass in rodents and into the non-human primates' brain. Importantly, Ads have been safely administered within the tumor resection cavity in humans. AREAS COVERED This review gives background on GBM and Ads; we describe gene therapy strategies for GBM and discuss the value of combination approaches. Finally, we discuss the results of the human clinical trials for GBM that have used Ads. EXPERT OPINION The transduction characteristics of Ads, and their safety profile, added to their capacity to achieve high levels of transgene expression have made them powerful vectors for the treatment of GBM. Recent gene therapy successes in the treatment of retinal diseases and systemic brain metabolic diseases encourage the development of gene therapy for malignant glioma. Exciting clinical trials are currently recruiting patients; although, it is the large randomized Phase III controlled clinical trials that will provide the final decision on the success of gene therapy for the treatment of GBM.
Collapse
Affiliation(s)
- Maria G Castro
- University of Michigan Medical School, Department of Neurosurgery , 4570 MSRB II, 1150 West Medical Center Drive, Ann Arbor, MI 48109-5689 , USA +734 764 0850 ; +734 764 7051 ;
| | | | | | | | | | | | | | | | | | | |
Collapse
|
4
|
Abstract
PURPOSE OF REVIEW During the past few years novel gene-based approaches emerged attempting to treat chronic pain experimentally in animal models. This review will discuss some of the most recent developments in this area with special emphasis on vector-mediated targeted transfer of DNA at the spinal level. RECENT FINDINGS Local overexpression of precursors of opioid peptides, mainly at the spinal level, induces antihyperalgesic effects in various animal models of persistent pain. Different techniques enabling the in vivo transfer of these precursors have been described. Virus-derived vectors appear as potent systems, providing targeted and sustained overproduction of opioid peptides. Interestingly, overexpression of proenkephalin A in primary sensory neurones induced antinociceptive effects in persistent pain of inflammatory, neuropathic and cancerous origins. Targeted overproduction of many other proteins may be relevant to the relief of ongoing pain. For instance, local overproduction of brain derived neurotrophic factor in the spinal cord has been reported to treat neuropathic pain induced by chronic constrictive injury of the sciatic nerve. SUMMARY Gene-based techniques may contribute to the search for a better management of chronic pain. In this respect, tempting data were obtained in animal models of persistent pain using viral vector-mediated overproduction of opioid peptides and neurotrophins. Gene-based protocols targeting some molecules involved in pain induction and perpetuation also raise the interesting possibility of blocking the development of chronic pain, rather than relieving it. Apart from the 'gene therapy' of chronic pain, the clinical application of which still remains to be established, these techniques might help in evaluating the potential interest of some recently identified molecules involved in pain transduction mechanisms or sensory nerve sensitization. They might finally lead to the development of new classical pharmacological tools.
Collapse
Affiliation(s)
- Michel Pohl
- Pitié-Salpêtrière Faculty of Medicine, INSERM E 0331, Paris, France.
| | | |
Collapse
|
5
|
Wilson RB, Kunchithapautham K, Rohrer B. Paradoxical role of BDNF: BDNF+/- retinas are protected against light damage-mediated stress. Invest Ophthalmol Vis Sci 2007; 48:2877-86. [PMID: 17525224 PMCID: PMC1964504 DOI: 10.1167/iovs.06-1079] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
PURPOSE Photoreceptors can be prevented from undergoing apoptosis in response to constant light by the application of exogenous neuroprotective agents, including brain-derived neurotrophic factor (BDNF). BDNF, however, cannot exert its effect directly on photoreceptors because they do not express receptors for BDNF. It has been proposed that BDNF released from Müller cells provides a feed-forward loop, increasing ciliary neurotrophic factor (CNTF) and basic fibroblast growth factor (bFGF) production in Müller cells, which may enhance photoreceptor survival. The authors hypothesized that retinas with reduced BDNF levels in which the BDNF-mediated release of neuroprotective signals is dampened are more susceptible to light-induced photoreceptor degeneration. METHODS Young adult BDNF+/+ and BDNF+/- littermates (B6.129-BDNF(tm1-LT)) were analyzed. Retinal neurotrophin and growth factor mRNA levels were determined by quantitative RT-PCR, photoreceptor function was assessed through electroretinography, and survival was documented in morphologic sections and in TUNEL assays. Oxidative stress was assayed by measuring glutathione peroxidase activity. RESULTS At baseline, BDNF+/- animals had significantly increased levels of glial-derived neurotrophic factor (GDNF) mRNA compared with their wild-type littermates. After light damage GDNF, CNTF, and BDNF mRNA levels dropped 14- to 16-fold in the BDNF+/+ mice but remained almost unchanged compared with baseline levels in the BDNF+/- mice. Preservation of neurotrophin levels in BDNF+/- mice correlated with photoreceptor cell survival, preservation of function, and reduced oxidative stress. CONCLUSIONS Contrary to the hypothesis, reducing BDNF levels resulted in photoreceptor protection against light damage. Survival was paralleled by a reduction in oxidative stress and the preservation of neurotrophin levels, suggesting that chronic reduction of BDNF in the retina provides a level of preconditioning against stress.
Collapse
Affiliation(s)
- R. Brooks Wilson
- Neurosciences Division of Research, Medical University of South Carolina, Charleston, South Carolina
| | - Kannan Kunchithapautham
- Neurosciences Division of Research, Medical University of South Carolina, Charleston, South Carolina
| | - Bärbel Rohrer
- Department of Ophthalmology, Medical University of South Carolina, Charleston, South Carolina
- Neurosciences Division of Research, Medical University of South Carolina, Charleston, South Carolina
| |
Collapse
|
6
|
Shevtsova Z, Malik JMI, Michel U, Schöll U, Bähr M, Kügler S. Evaluation of epitope tags for protein detection after in vivo CNS gene transfer. Eur J Neurosci 2006; 23:1961-9. [PMID: 16630044 DOI: 10.1111/j.1460-9568.2006.04725.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Functional characterization of disease-related proteins, their splice variants and dominant negative mutants in the context of complex CNS tissues such as brain and retina is frequently assessed by in vivo gene transfer. For correct interpretation of results it is imperative that the protein under investigation is unambiguously detected in the transduced cell types and can be distinguished from any endogenously expressed physiological variants. Therefore the first systematic evaluation of epitope tags used to trace ectopically expressed proteins in the central nervous system is presented here. Substantial differences in the performances of various epitope tag-antibody combinations with respect to sensitivity, specificity and influence of the epitope tag on the fusion protein are elucidated. Epitope tags already established for protein detection in vitro and to some extent in vivo (c-Myc, HA and FLAG tags) were immunohistochemically detected with high sensitivity. However, detection of these tags revealed problems with background staining and we also document structural and functional influence of the tags on the fusion protein. In order to prevent such unwanted side-effects, epitope tags which have not yet been used for in vivo applications (IRS, EE and AU1 tags) were characterized in brain, retina and cultured neurons. While use of the IRS and EE tags was hindered by low sensitivity or specificity, optimal results were obtained with the AU1 epitope, which may develop into a standard tool for detection of ectopic protein expression in the central nervous system.
Collapse
Affiliation(s)
- Z Shevtsova
- Department of Neurology; University of Göttingen, Medical School, Waldweg 33, 37073 Göttingen, Germany
| | | | | | | | | | | |
Collapse
|
7
|
Ogawa Y, Morikawa N, Ohkubo-Suzuki A, Miyoshi S, Arakawa H, Kita Y, Nishimura S. An epoch-making application of discharge plasma phenomenon to gene-transfer. Biotechnol Bioeng 2006; 92:865-70. [PMID: 16170826 DOI: 10.1002/bit.20659] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
We discovered an epoch-making gene transfer method utilizing discharge plasma. Although an electroporation method is commonly used in present gene transfer experiments, it cannot transfer genes into primary cells sufficiently. The atmospheric pressure discharge plasma employed in this study was originally used for surface treatment of non-biological materials. We hypothesized that it could provide a suitable effect on the surface of target cells and applied it to gene transfer into various types of cells. The plasma technology succeeded in the efficient transfer of green fluorescence protein (GFP) plasmid into post-mitotic neuronal cells obtained from cerebral cortices of rats, into which an electroporation with conventional equipment cannot transfer genes sufficiently, as the cells were attached. After the transfection of rat pheochromocytoma PC12 cells with the GFP gene by plasma treatment, the cells retained their function, that is, nerve growth factor-induced differentiation. Furthermore, gene transfer with the plasma technology was also applicable to other types of cell lines such as HeLa cells and Chinese hamster lung (CHL) cells as adherent cell lines, and Jurkat cells as a suspended cell line, and another type of primary cell, human umbilical vein endothelial cells (HUVEC). In conclusion, the plasma method is an epoch-making gene transfer technology which efficiently transfers genes into primary cells into which electroporation cannot transfer genes. Moreover, the method is able to universally transfer genes into various types of cells as the function of the cells was maintained.
Collapse
Affiliation(s)
- Yasuhiro Ogawa
- Astellas Pharma, Inc., 10-2, Kanda Tomiyama-cho, Chiyoda-ku, Tokyo 101-0043, Japan.
| | | | | | | | | | | | | |
Collapse
|
8
|
Shamoo AE, Cole J. Ethics of genetic modifications of behavior. Account Res 2005; 11:201-14. [PMID: 15812966 DOI: 10.1080/08989620490891395] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Adil E Shamoo
- University of Maryland School of Medicine, 108 N. Green St., Baltimore, MD 21201, USA.
| | | |
Collapse
|
9
|
Whittlesey KJ, Shea LD. Delivery systems for small molecule drugs, proteins, and DNA: the neuroscience/biomaterial interface. Exp Neurol 2005; 190:1-16. [PMID: 15473976 DOI: 10.1016/j.expneurol.2004.06.020] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2004] [Revised: 04/30/2004] [Accepted: 06/18/2004] [Indexed: 11/18/2022]
Abstract
Manipulation of cellular processes in vivo by the delivery of drugs, proteins or DNA is of paramount importance to neuroscience research. Methods for the presentation of these molecules vary widely, including direct injection (either systemic or stereotactic), osmotic pump-mediated chronic delivery, or even implantation of cells engineered to indefinitely secrete a factor of interest. Biomaterial-based delivery systems represent an alternative to more traditional approaches, with the possibility of increased efficacy. Drug-releasing biomaterials, either as injectable microspheres or as three-dimensional implants, can deliver a molecule of interest (including small molecule drugs, biologically active proteins, or DNA) over a more prolonged period of time than by standard bolus injection, avoiding the need for repeated administration. Furthermore, sustained-release systems can maintain therapeutic concentrations at a target site, thus reducing the chance for toxicity. This review summarizes applications of polymer-based delivery of small molecule drugs, proteins, and DNA specifically relevant to neuroscience research. We detail the fabrication procedures for the polymeric systems and their utility in various experimental models. The biomaterial field offers unique experimental tools with downstream clinical application for the study and treatment of neurologic disease.
Collapse
Affiliation(s)
- Kevin J Whittlesey
- Interdepartmental Biological Sciences Program, Northwestern University, 2145 Sheridan Road, Evanston, IL 60208, USA.
| | | |
Collapse
|
10
|
Jones TL, Sweitzer SM, Wilson SP, Yeomans DC. Afferent fiber-selective shift in opiate potency following targeted opioid receptor knockdown. Pain 2004; 106:365-371. [PMID: 14659519 DOI: 10.1016/j.pain.2003.08.006] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
Spinal application of opiates is the cornerstone of potent analgesia. In the present study, opiate analgesia was investigated after cutaneous application of a recombinant herpes simplex virus type-1 (HSV-1) encoding micro-opioid receptor (microOR) cDNA in reverse orientation with respect to the human cytomegalovirus early enhancer-promoter. Hind paw application of this recombinant vector was used in order to attenuate expression of the microOR in primary afferents and determine whether recombinant vector application would differentially affect the antinociceptive effects of the specific microOR agonist, [D-Ala(2),N-MePhe(4),Gly-ol(5)] enkephalin (DAMGO), on behavioral responses mediated by C- and Adelta-thermonociceptors. The recombinant vector encoding the Escherichia coli lacZ gene marker, KHZ, served as a control virus. Dorsal hind paw surfaces of female Swiss-Webster mice were treated with one of these two viruses (1x10(8)pfu, 10 microl) or vehicle (uninfected). Immunohistochemistry and quantitative image analyses revealed decreased microOR expression in the superficial dorsal horns ipsilateral to hind paws treated with AMOR, but not KHZ. To add, behavioral foot withdrawal latencies of AMOR- and KHZ-treated hind paws demonstrated dose-dependent antinociception after intrathecal DAMGO administration. However, cutaneous application of dorsal hind paw surfaces treated with AMOR, but not KHZ, caused a rightward shift in the C-fiber dose-response, thus, indicating a loss of potency of intrathecal DAMGO. Loss or diminution of DAMGO potency during Adelta-fiber-mediated responses was not observed. These immunohistochemistry and behavioral results of novel, recombinant HSV-1 vector microOR 'knock-down' in nociceptor afferent fibers provide additional evidence for presynaptic localization of microORs on central C-, but not Adelta-terminals.
Collapse
MESH Headings
- Analgesics, Opioid/pharmacology
- Animals
- Dose-Response Relationship, Drug
- Enkephalin, Ala(2)-MePhe(4)-Gly(5)-/pharmacology
- Female
- Genetic Vectors/genetics
- Genetic Vectors/metabolism
- Herpesvirus 1, Human/genetics
- Herpesvirus 1, Human/metabolism
- Male
- Mice
- Neurons, Afferent/drug effects
- Neurons, Afferent/metabolism
- Receptors, Opioid, mu/agonists
- Receptors, Opioid, mu/deficiency
- Receptors, Opioid, mu/genetics
- Recombinant Proteins/genetics
- Recombinant Proteins/metabolism
Collapse
Affiliation(s)
- Toni L Jones
- Department of Anesthesia, Stanford University School of Medicine, 300 Pasteur Drive, Stanford, CA 94305-5117, USA Department of Pharmacology, Physiology, and Neuroscience, University of South Carolina School of Medicine, Columbia, SC 29208, USA
| | | | | | | |
Collapse
|
11
|
Lowenstein PR, Suwelack D, Hu J, Yuan X, Jimenez-Dalmaroni M, Goverdhana S, Castro MG. Nonneurotropic adenovirus: a vector for gene transfer to the brain and gene therapy of neurological disorders. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2003; 55:3-64. [PMID: 12968530 PMCID: PMC2902245 DOI: 10.1016/s0074-7742(03)01001-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Pedro R Lowenstein
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Department of Medicine, UCLA, Los Angeles, California 90048, USA
| | | | | | | | | | | | | |
Collapse
|
12
|
Ourednik J, Ourednik V, Lynch WP, Schachner M, Snyder EY. Neural stem cells display an inherent mechanism for rescuing dysfunctional neurons. Nat Biotechnol 2002; 20:1103-10. [PMID: 12379867 DOI: 10.1038/nbt750] [Citation(s) in RCA: 382] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2002] [Accepted: 08/09/2002] [Indexed: 01/09/2023]
Abstract
We investigated the hypothesis that neural stem cells (NSCs) possess an intrinsic capacity to "rescue" dysfunctional neurons in the brains of aged mice. The study focused on a neuronal cell type with stereotypical projections that is commonly compromised in the aged brain-the dopaminergic (DA) neuron. Unilateral implantation of murine NSCs into the midbrains of aged mice, in which the presence of stably impaired but nonapoptotic DA neurons was increased by treatment with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP), was associated with bilateral reconstitution of the mesostriatal system. Functional assays paralleled the spatiotemporal recovery of tyrosine hydroxylase (TH) and dopamine transporter (DAT) activity, which, in turn, mirrored the spatiotemporal distribution of donor-derived cells. Although spontaneous conversion of donor NSCs to TH(+) cells contributed to nigral reconstitution in DA-depleted areas, the majority of DA neurons in the mesostriatal system were "rescued" host cells. Undifferentiated donor progenitors spontaneously expressing neuroprotective substances provided a plausible molecular basis for this finding. These observations suggest that host structures may benefit not only from NSC-derived replacement of lost neurons but also from the "chaperone" effect of some NSC-derived progeny.
Collapse
Affiliation(s)
- Jitka Ourednik
- Department of Neurobiology, Swiss Federal Institute of Technology, Hoenggerberg, CH-8093, Switzerland.
| | | | | | | | | |
Collapse
|
13
|
Abstract
To illuminate the function of the thousands of genes that make up the complexity of the nervous system, it is critical to be able to introduce and express DNA in neurons. Over the past two decades, many gene transfer methods have been developed, including viral vectors, liposomes and electroporation. Although the perfect gene transfer technique for every application has not yet been developed, recent technical advances have facilitated the ease of neuronal gene transfer and have increased the accessibility of these techniques to all laboratories. In order to select a transfection method for any particular experiment, the specific advantages and disadvantages of each technique must be considered.
Collapse
Affiliation(s)
- Philip Washbourne
- Center for Neuroscience, University of California, Davis, Davis, California 95616, USA.
| | | |
Collapse
|
14
|
Hsich G, Sena-Esteves M, Breakefield XO. Critical issues in gene therapy for neurologic disease. Hum Gene Ther 2002; 13:579-604. [PMID: 11916483 DOI: 10.1089/10430340252837198] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Gene therapy for the nervous system is a newly emerging field with special issues related to modes of delivery, potential toxicity, and realistic expectations for treatment of this vital and highly complex tissue. This review focuses on the potential for gene delivery to the brain, as well as possible risks and benefits of these procedures. This includes discussion of appropriate vectors, such as adeno-associated virus, lentivirus, gutless adenovirus, and herpes simplex virus hybrid amplicons, and cell vehicles, such as neuroprogenitor cells. Routes of delivery for focal and global diseases are enumerated, including use of migratory cells, facilitation of vascular delivery across the blood-brain barrier, cerebrospinal fluid delivery, and convection injection. Attention is given to examples of diseases falling into different etiologic types: metabolic deficiency states, including Canavan disease and lysosomal storage disorders; and degenerative conditions, including Parkinson's disease and other neurodegenerative conditions.
Collapse
Affiliation(s)
- Gary Hsich
- Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital, and Neuroscience Program, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|