1
|
Pérez-Luz S, Gimenez-Cassina A, Fernández-Frías I, Wade-Martins R, Díaz-Nido J. Delivery of the 135 kb human frataxin genomic DNA locus gives rise to different frataxin isoforms. Genomics 2015; 106:76-82. [PMID: 26027909 DOI: 10.1016/j.ygeno.2015.05.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Revised: 05/21/2015] [Accepted: 05/23/2015] [Indexed: 11/25/2022]
Abstract
Friedreich's ataxia (FRDA) is the most common form of hereditary ataxia caused by recessive mutations in the FXN gene. Recent results have indicated the presence of different frataxin isoforms due to alternative gene expression mechanisms. Our previous studies demonstrated the advantages of using high-capacity herpes simplex virus type 1 (HSV-1) amplicon vectors containing the entire FXN genomic locus (iBAC-FXN) as a gene-delivery vehicle capable of ensuring physiologically-regulated and long-term persistence. Here we describe how expression from the 135 kb human FXN genomic locus produces the three frataxin isoforms both in cultured neuronal cells and also in vivo. Moreover, we also observed the correct expression of these frataxin isoforms in patient-derived cells after delivery of the iBAC-FXN. These results lend further support to the potential use of HSV-1 vectors containing entire genomic loci whose expression is mediated by complex transcriptional and posttranscriptional mechanisms for gene therapy applications.
Collapse
Affiliation(s)
- S Pérez-Luz
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda, Spain
| | | | - I Fernández-Frías
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda, Spain
| | | | - J Díaz-Nido
- Departamento Biología Molecular and Centro de Biología Molecular "Severo Ochoa" (UAM-CSIC), Universidad Autónoma de Madrid, 28049 Madrid, Spain; CIBER de Enfermedades Raras (CIBERER), Spain; Instituto de Investigación Sanitaria Puerta de Hierro-Majadahonda, Spain.
| |
Collapse
|
2
|
Nishiyama J, Hayashi Y, Nomura T, Miura E, Kakegawa W, Yuzaki M. Selective and regulated gene expression in murine Purkinje cells by in utero electroporation. Eur J Neurosci 2012; 36:2867-76. [PMID: 22775058 DOI: 10.1111/j.1460-9568.2012.08203.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cerebellar Purkinje cells, which convey the only output from the cerebellar cortex, play an essential role in cerebellar functions, such as motor coordination and motor learning. To understand how Purkinje cells develop and function in the mature cerebellum, an efficient method for molecularly perturbing them is needed. Here we demonstrate that Purkinje cell progenitors at embryonic day (E)11.5 could be efficiently and preferentially transfected by spatially directed in utero electroporation (IUE) with an optimized arrangement of electrodes. Electrophysiological analyses indicated that the electroporated Purkinje cells maintained normal membrane properties, synaptic responses and synaptic plasticity at postnatal days 25-28. By combining the L7 promoter and inducible Cre/loxP system with IUE, transgenes were expressed even more specifically in Purkinje cells and in a temporally controlled manner. We also show that three different fluorescent proteins could be simultaneously expressed, and that Bassoon, a large synaptic protein, could be expressed in the electroporated Purkinje cells. Moreover, phenotypes of staggerer mutant mice, which have a deletion in the gene encoding retinoid-related orphan receptor α (RORα1), were recapitulated by electroporating a dominant-negative form of RORα1 into Purkinje cells at E11.5. Together, these results indicate that this new IUE protocol, which allows the selective, effective and temporally regulated expression of multiple foreign genes transfected into Purkinje cell progenitors in vivo, without changing the cells' physiological characteristics, is a powerful tool for elucidating the molecular mechanisms underlying early Purkinje cell developmental events, such as dendritogenesis and migration, and synaptic plasticity in mature Purkinje cells.
Collapse
Affiliation(s)
- Jun Nishiyama
- Department of Physiology, School of Medicine, Keio University, 35 Shinanomachi, Shinjuku-ku, Tokyo 160-8582, Japan
| | | | | | | | | | | |
Collapse
|
3
|
Gimenez-Cassina A, Wade-Martins R, Gomez-Sebastian S, Corona JC, Lim F, Diaz-Nido J. Infectious delivery and long-term persistence of transgene expression in the brain by a 135-kb iBAC-FXN genomic DNA expression vector. Gene Ther 2011; 18:1015-9. [PMID: 21490681 DOI: 10.1038/gt.2011.45] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2010] [Revised: 02/15/2011] [Accepted: 02/20/2011] [Indexed: 01/21/2023]
Abstract
Novel gene-based therapies for disease will depend in many cases on long-term persistent transgene expression. To develop gene therapy strategies for Friedreich's ataxia (FRDA), we have examined the persistence of transgene expression in the brain in vivo provided by the entire 135 kb FXN genomic DNA locus delivered as an infectious bacterial artificial chromosome (iBAC) herpes simplex virus type 1 (HSV-1)-based vector injected in the adult mouse cerebellum. We constructed genomic DNA-reporter fusion vectors carrying a complete 135 kb FXN genomic locus with an insertion of the Escherichia coli lacZ gene at the ATG start codon (iBAC-FXN-lacZ). SHSY5Y human neuroblastoma cells transduced by iBAC-FXN-lacZ showed high efficiency of vector delivery and LacZ expression. Direct intracranial injection of iBAC-FXN-lacZ into the adult mouse cerebellum resulted in a large number of easily detectable transduced cells, with LacZ expression driven by the FXN genomic locus, which persisted for at least 75 days. Green fluorescent protein expression driven from the same vector but by the strong HSV-1 IE4/5 promoter was transient. Our data demonstrate for the first time sustained transgene expression in vivo by infectious delivery of a genomic DNA locus >100 kb in size. Such an approach may be suitable for gene rescue strategies in neurological disease, such as FRDA.
Collapse
Affiliation(s)
- A Gimenez-Cassina
- Department of Molecular Neurobiology, Centro de Biología Molecular Severo Ochoa, Cantoblanco, Spain
| | | | | | | | | | | |
Collapse
|
4
|
Louboutin JP, Reyes BAS, Van Bockstaele EJ, Strayer DS. Gene transfer to the cerebellum. THE CEREBELLUM 2011; 9:587-97. [PMID: 20700772 DOI: 10.1007/s12311-010-0202-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
There are several diseases for which gene transfer therapy to the cerebellum might be practicable. In these studies, we used recombinant Tag-deleted SV40-derived vectors (rSV40s) to study gene delivery targeting the cerebellum. These vectors transduce neurons and microglia very effectively in vitro and in vivo, and so we tested them to evaluate gene transfer to the cerebellum in vivo. Using a rSV40 vector carrying human immunodeficiency virus (HIV)-Nef with a C-terminal FLAG epitope, we characterized the distribution, duration, and cell types transduced. Rats received test and control vectors by stereotaxic injection into the cerebellum. Transgene expression was assessed 1, 2, and 4 weeks later by immunostaining of serial brain sections. FLAG epitope-expressing cells were seen, at all times after vector administration, principally detected in the Purkinje cells of the cerebellum, identified as immunopositive for calbindin. Occasional microglial cells were tranduced; transgene expression was not detected in astrocytes or oligodendrocytes. No inflammatory or other reaction was detected at any time. Thus, SV40-derived vectors can deliver effective, safe, and durable transgene expression to the cerebellum.
Collapse
Affiliation(s)
- Jean-Pierre Louboutin
- Department of Pathology, Jefferson Medical College, Thomas Jefferson University, 1020 Locust Street, Room 255, Philadelphia, PA 19107, USA.
| | | | | | | |
Collapse
|
5
|
Pérez-Luz S, Díaz-Nido J. Prospects for the use of artificial chromosomes and minichromosome-like episomes in gene therapy. J Biomed Biotechnol 2010; 2010:642804. [PMID: 20862363 PMCID: PMC2938438 DOI: 10.1155/2010/642804] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Revised: 06/02/2010] [Accepted: 07/05/2010] [Indexed: 01/19/2023] Open
Abstract
Artificial chromosomes and minichromosome-like episomes are large DNA molecules capable of containing whole genomic loci, and be maintained as nonintegrating, replicating molecules in proliferating human somatic cells. Authentic human artificial chromosomes are very difficult to engineer because of the difficulties associated with centromere structure, so they are not widely used for gene-therapy applications. However, OriP/EBNA1-based episomes, which they lack true centromeres, can be maintained stably in dividing cells as they bind to mitotic chromosomes and segregate into daughter cells. These episomes are more easily engineered than true human artificial chromosomes and can carry entire genes along with all their regulatory sequences. Thus, these constructs may facilitate the long-term persistence and physiological regulation of the expression of therapeutic genes, which is crucial for some gene therapy applications. In particular, they are promising vectors for gene therapy in inherited diseases that are caused by recessive mutations, for example haemophilia A and Friedreich's ataxia. Interestingly, the episome carrying the frataxin gene (deficient in Friedreich's ataxia) has been demonstrated to rescue the susceptibility to oxidative stress which is typical of fibroblasts from Friedreich's ataxia patients. This provides evidence of their potential to treat genetic diseases linked to recessive mutations through gene therapy.
Collapse
Affiliation(s)
- Sara Pérez-Luz
- Departamento de Biología Molecular, Universidad Autónoma de Madrid, 28049 Madrid, Spain.
| | | |
Collapse
|
6
|
Abstract
During postnatal cerebellar development, Purkinje cells form the most elaborate dendritic trees among neurons in the brain, which have been of great interest to many investigators. This article overviews various examples of cellular and molecular mechanisms of formation of Purkinje cell dendrites as well as the methodological aspects of investigating those mechanisms.
Collapse
Affiliation(s)
- Masahiko Tanaka
- Department of Cellular Biophysics, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan.
| |
Collapse
|
7
|
Hirai H. Progress in transduction of cerebellar Purkinje cells in vivo using viral vectors. THE CEREBELLUM 2009; 7:273-8. [PMID: 18418690 DOI: 10.1007/s12311-008-0012-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Expression of a foreign gene in cerebellar Purkinje cells in vivo is a powerful method for exploring the pathophysiology of the cerebellum. Although using developmental engineering many gene-modified mice have been generated, this approach is time-consuming and requires a lot of effort for crossing different lines of mice, genotyping and maintenance of animals. If a gene of interest can be transferred to and efficiently expressed in Purkinje cells of developing and mature animals, it saves much time, effort and money. Recent advances in viral vectors have markedly contributed to selective and efficient gene transfer to Purkinje cells in vivo. There are two approaches for selective gene expression in Purkinje cells: one is to take advantage of the viral tropism for Purkinje cells, which includes the tropism of adeno-associated virus and the vesicular stomatitis virus glycoprotein (VSV-G)-pseudotyped lentivirus. Another method, which might be used in combination with the first one, is utilization of a Purkinje-cell-specific promoter. Focusing mainly on these points, recent progress in viral-vector-mediated transduction of Purkinje cells in vivo is reviewed.
Collapse
Affiliation(s)
- Hirokazu Hirai
- Department of Neurophysiology, Gunma University Graduate School of Medicine, Maebashi, Gunma, 371-8511, Japan.
| |
Collapse
|
8
|
Lufino MMP, Edser PAH, Wade-Martins R. Advances in high-capacity extrachromosomal vector technology: episomal maintenance, vector delivery, and transgene expression. Mol Ther 2008; 16:1525-38. [PMID: 18628754 DOI: 10.1038/mt.2008.156] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Recent developments in extrachromosomal vector technology have offered new ways of designing safer, physiologically regulated vectors for gene therapy. Extrachromosomal, or episomal, persistence in the nucleus of transduced cells offers a safer alternative to integrating vectors which have become the subject of safety concerns following serious adverse events in recent clinical trials. Extrachromosomal vectors do not cause physical disruption in the host genome, making these vectors safe and suitable tools for several gene therapy targets, including stem cells. Moreover, the high insert capacity of extrachromosomal vectors allows expression of a therapeutic transgene from the context of its genomic DNA sequence, providing an elegant way to express normal splice variants and achieve physiologically regulated levels of expression. Here, we describe past and recent advances in the development of several different extrachromosomal systems, discuss their retention mechanisms, and evaluate their use as expression vectors to deliver and express genomic DNA loci. We also discuss a variety of delivery systems, viral and nonviral, which have been used to deliver episomal vectors to target cells in vitro and in vivo. Finally, we explore the potential for the delivery and expression of extrachromosomal transgenes in stem cells. The long-term persistence of extrachromosomal vectors combined with the potential for stem cell proliferation and differentiation into a wide range of cell types offers an exciting prospect for therapeutic interventions.
Collapse
Affiliation(s)
- Michele M P Lufino
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | | | | |
Collapse
|
9
|
Cuchet D, Potel C, Thomas J, Epstein AL. HSV-1 amplicon vectors: a promising and versatile tool for gene delivery. Expert Opin Biol Ther 2007; 7:975-95. [PMID: 17665988 DOI: 10.1517/14712598.7.7.975] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Amplicons are defective and non-integrative vectors derived from herpes simplex virus type 1. They carry no virus genes in the vector genome and are, therefore, not toxic to the infected cells or pathogenic for the transduced organisms, making these vectors safe. In addition, the large transgenic capacity of amplicons, which allow delivery of < or = 150 Kbp of foreign DNA, make these vectors one of the most powerful, interesting and versatile gene delivery platforms. Here, the authors present recent technological developments that have significantly improved and extended the use of amplicons, both in cultured cells and in living organisms. In addition, this review illustrates the many possible applications that are presently being developed with amplicons and discuss the many difficulties still pending to be solved in order to achieve stable and physiologically regulated transgenic expression.
Collapse
|
10
|
Lim F, Palomo GM, Mauritz C, Giménez-Cassina A, Illana B, Wandosell F, Díaz-Nido J. Functional recovery in a Friedreich's ataxia mouse model by frataxin gene transfer using an HSV-1 amplicon vector. Mol Ther 2007; 15:1072-8. [PMID: 17375064 DOI: 10.1038/sj.mt.6300143] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
There is currently no effective treatment for Friedreich's ataxia (FA), the most common of the hereditary ataxias. The disease is caused by mutations in FRDA that drastically reduce expression levels of the mitochondrial protein frataxin. In FA animal models, a key difficulty is obtaining the precise levels of frataxin expression in the appropriate tissues to provoke pathology without early lethality. To develop strategies to circumvent these problems, conditional frataxin transgenic mice have been generated. We now show that frataxin expression can be eliminated in neurons from these loxP[frda] mice by infection with CRE-expressing herpes simplex virus type 1 (HSV-1) amplicon vectors. We have also achieved in vivo delivery by stereotaxic injection of these CRE-expressing vectors into the brainstem of loxP[frda] mice to generate a localized gene knockout model. These mice develop a behavioral deficit in the rotarod assay detectable after 4 weeks, and when re-injected with HSV-1 amplicon vectors expressing human frataxin complementary DNA (cDNA) exhibit behavioral recovery as early as 4 weeks after the second injection. To the best of our knowledge, this is the first proof of principle of recovery of neurological function by a therapeutic agent aimed at correcting frataxin deficiency.
Collapse
Affiliation(s)
- Filip Lim
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Universidad Autónoma de, Madrid, Madrid, Spain.
| | | | | | | | | | | | | |
Collapse
|
11
|
Gomez-Sebastian S, Gimenez-Cassina A, Diaz-Nido J, Lim F, Wade-Martins R. Infectious delivery and expression of a 135 kb human FRDA genomic DNA locus complements Friedreich's ataxia deficiency in human cells. Mol Ther 2007; 15:248-54. [PMID: 17235301 DOI: 10.1038/sj.mt.6300021] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2006] [Accepted: 09/06/2006] [Indexed: 01/18/2023] Open
Abstract
Friedreich's ataxia (FA) is the most common recessive ataxia, affecting 1-2 in 50,000 Caucasians, and there is currently no effective cure or treatment. FA results from a deficiency of the mitochondrial protein frataxin brought about by a repeat expansion in intron 1 of the FRDA gene. The main areas affected are the central nervous system (particularly the spinocerebellar system) and cardiac tissue. Therapies aimed at alleviating the neurological degeneration have proved unsuccessful to date. Here, we describe the construction and delivery of high capacity herpes simplex virus type 1 (HSV-1) amplicon vectors expressing the entire 80 kb FRDA genomic locus, driven by the endogenous FRDA promoter and including all introns and flanking regulatory sequences within a 135 kb genomic DNA insert. FA patient primary fibroblasts deficient in frataxin protein and exhibiting sensitivity to oxidative stress were transduced at high efficiency by FRDA genomic locus vectors. Following vector transduction, expression of FRDA protein by immunofluorescence was shown. Finally, functional complementation studies demonstrated restoration of the wild-type cellular phenotype in response to oxidative stress in transduced FA patient cells. These results suggest the potential of the infectious bacterial artificial chromosome-FRDA vectors for gene therapy of FA.
Collapse
|
12
|
Gimenez-Cassina A, Lim F, Diaz-Nido J. Gene transfer into Purkinje cells using herpesviral amplicon vectors in cerebellar cultures. Neurochem Int 2006; 50:181-8. [PMID: 16989924 DOI: 10.1016/j.neuint.2006.08.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2006] [Revised: 07/26/2006] [Accepted: 08/01/2006] [Indexed: 11/30/2022]
Abstract
Purkinje cells play a crucial role in sensory motor coordination since they are the only output projection neurons in the cerebellar cortex and are affected in most spinocerebellar ataxias. They stand out in the central nervous system due to their large size and their profusely branched dendritic arbor. However, molecular and cellular studies on Purkinje cells are often hampered by the difficulty of maintaining these cells in culture. Here we report an easy, robust and reproducible method to obtain Purkinje-enriched mixed cerebellar cell cultures from day 16 mouse embryos using papain digestion and a semi-defined culture medium, being the composition of the culture approximately 20% Purkinje cells, 70% non-Purkinje neurons and 10% glial cells. We demonstrate that efficient gene transfer into Purkinje cells (as well as into other cerebellar populations) is possible using herpes simplex virus-1 (HSV-1)-derived vectors. Indeed, up to 50% of the Purkinje cells can be transduced and gene expression may persist for at least 14 days. As a result, this procedure permits functional gene expression studies to be carried out on cultured Purkinje neurons. To demonstrate this, we show that the expression of a dominant-negative form of glycogen synthase kinase-3 protects Purkinje neurons against cell death triggered by a chemical inhibitor of phosphatidylinositol-3 kinase. In summary, we have established reproducible and reliable cerebellar cell cultures enriched for Purkinje cells which enables gene transfer studies to be carried out using herpesviral vectors.
Collapse
Affiliation(s)
- Alfredo Gimenez-Cassina
- Departamento de Biologia Molecular, Centro de Biología Molecular Severo Ochoa, Universidad Autónoma de Madrid, Campus de Cantoblanco, 28049 Madrid, Spain
| | | | | |
Collapse
|
13
|
Torashima T, Yamada N, Itoh M, Yamamoto A, Hirai H. Exposure of lentiviral vectors to subneutral pH shifts the tropism from Purkinje cell to Bergmann glia. Eur J Neurosci 2006; 24:371-80. [PMID: 16836635 DOI: 10.1111/j.1460-9568.2006.04927.x] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Cerebellar Purkinje cells play an important role in cerebellar function; lesions of Purkinje cells result in the disruption of motor coordination and motor learning. Although selective gene delivery to Purkinje cells would be a powerful technique for the study of pathophysiology in the cerebellum, a method for such a delivery has not yet been established. Here we employed human immunodeficiency virus-derived lentiviral vectors pseudotyped with vesicular stomatitis virus glycoprotein to transduce Purkinje cells and examined factors that critically affect the viral tropism for Purkinje cells. Viral vectors encoding GFP were generated using different protocols, and were then injected into the mouse cerebellum. At 7 days and 2 months post-transduction, the relative proportions of transduced Purkinje cells were determined. Lentiviral vectors harvested from a medium of pH 7.2 preferentially transduced Purkinje cells (about half of the transduced cells). In contrast, when the viral vector was harvested from medium of <or= pH 7.0, only 12-26% of transduced cells were identified as Purkinje cells and 68-77% as Bergmann glia. A similar decrease in the efficiency of transduction for Purkinje cells, depending on the pH of the medium at the viral harvest, was observed in dissociated cell cultures. These results indicate that lentivector tropism for Purkinje cells is extremely sensitive to pH: a subtle decrease in the pH of the medium at the harvest shifts viral tropism strikingly towards Bergmann glia.
Collapse
Affiliation(s)
- Takashi Torashima
- Innovative Brain Science Project, Advanced Science Research Center, Kanazawa University, Kanazawa 920-8640, Japan
| | | | | | | | | |
Collapse
|
14
|
Cortés ML, Oehmig A, Perry KF, Sanford JD, Breakefield XO. Expression of human ATM cDNA in Atm-deficient mouse brain mediated by HSV-1 amplicon vector. Neuroscience 2006; 141:1247-56. [PMID: 16809004 DOI: 10.1016/j.neuroscience.2006.05.055] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2006] [Revised: 05/21/2006] [Accepted: 05/23/2006] [Indexed: 11/17/2022]
Abstract
Ataxia-telangiectasia (A-T) is an autosomal recessive disorder characterized by neurodegeneration, immunodeficiency, cancer predisposition, genome instability, and radiation sensitivity. Herpes simplex virus type 1 (HSV-1) amplicon vectors provide a means to deliver large genes to the nervous system efficiently and safely. We have generated an amplicon vector, carrying human FLAG-tagged A-T mutated (ATM), as well as an enhanced green fluorescent protein (EGFP) marker gene. Due to the lack of effective and reliable antibodies for ATM and FLAG appropriate for immunohistochemistry in mouse tissue sections, expression of the human FLAG-tagged ATM was confirmed in the mouse cerebellum at the RNA level by reverse transcription followed by quantitative PCR, and by radioactive in situ hybridization. In addition, we were able to immunoprecipitate the full-length human ATM protein from the cerebella of Atm -/- mice post-infection. This vector has been injected into the cerebella of Atm -/- mice with gene delivery to thousands of cells, including Purkinje cells, based on the EGFP marker gene. The expression of human FLAG-tagged ATM has been demonstrated in the cerebella of Atm-/- mice at the transcription and translational level three days post-infection. To our knowledge, this is the first report of vector-mediated delivery of the human ATM cDNA to an Atm -/- mouse. These vectors provide the groundwork to develop gene therapy approaches for A-T patients.
Collapse
Affiliation(s)
- M L Cortés
- Molecular Neurogenetics Unit, Department of Neurology, Massachusetts General Hospital-East, Harvard Medical School, 13th Street, Building 149, 6th Floor, Charlestown, MA 02129, USA.
| | | | | | | | | |
Collapse
|
15
|
Croci C, Fasano S, Superchi D, Perani L, Martellosio A, Brambilla R, Consalez G, Bongarzone ER. Cerebellar neurons and glial cells are transducible by lentiviral vectors without decrease of cerebellar functions. Dev Neurosci 2006; 28:216-21. [PMID: 16679768 DOI: 10.1159/000091919] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2005] [Accepted: 09/02/2005] [Indexed: 11/19/2022] Open
Abstract
Due to the profuse connections of the cerebellum to the rest of the central nervous system, cerebellar dysfunction impacts tremendously on movement coordination, maintenance of equilibrium, muscle tone and motor memory. Efficient gene transfer of therapeutic genes to this central nervous system structure would constitute a relevant step ahead the design of treatments to ameliorate cerebellar dysfunction. Lentiviral vectors (LVs) have been used as efficient vehicles to integrate transgenes into dividing and non-dividing cells, such as postmitotic adult neurons, with minimal toxicity and immune response. This study aimed to use LVs carrying green fluorescent protein (GFP) cDNA for transduction of cerebellar cells in vivo without compromising neurological cerebellar functions. Our results indicate that LVs, injected in the lobulus simplex, transduced different cerebellar neurons including stellate, Purkinje cells, granular neurons and glial cells such as astrocytes, oligodendrocytes, and that this gene transfer approach was not accompanied by cerebellar deficits.
Collapse
Affiliation(s)
- C Croci
- San Raffaele Telethon Institute for Gene Therapy, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
16
|
Torashima T, Okoyama S, Nishizaki T, Hirai H. In vivo transduction of murine cerebellar Purkinje cells by HIV-derived lentiviral vectors. Brain Res 2006; 1082:11-22. [PMID: 16516872 DOI: 10.1016/j.brainres.2006.01.104] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2005] [Revised: 01/18/2006] [Accepted: 01/22/2006] [Indexed: 11/28/2022]
Abstract
Cerebellar Purkinje cells are key elements in motor learning and motor coordination, and therefore, it is important to clarify the mechanisms by which Purkinje cells integrate information and control cerebellar function. Gene transfer into neurons, followed by the assessment of the effects on neural function, is an effective approach for examining gene function. However, this method has not been used fully in the study of the cerebellum because adenovirus vectors, the vectors most commonly used for in vivo gene transfer, have very low affinity for Purkinje cells. In this study, we used a human immunodeficiency virus (HIV)-derived lentiviral vector and examined the transduction profile of the vector in the cerebellum. A lentiviral vector carrying the GFP gene was injected into the cerebellar cortex. Seven days after the injection, Purkinje cells were efficiently transduced without significant influence on the cell viability and synaptic functions. GFP was also expressed, though less efficiently, in other cortical interneurons and Bergmann glias. In contrast to reported findings with other viral vectors, no transduced cells were observed outside of the cerebellar cortex. Thus, when HIV-derived lentiviral vectors were injected into the cerebellar cortex, transduction was limited to the cells in the cerebellar cortex, with the highest tropism for Purkinje cells. These results suggest that HIV-derived lentiviral vectors are useful for the study of gene function in Purkinje cells as well as for application as a gene therapy tool for the treatment of diseases that affect Purkinje cells.
Collapse
Affiliation(s)
- Takashi Torashima
- Innovative Brain Science Project, Advanced Science Research Center, Kanazawa University, Kanazawa 920-8640, Japan
| | | | | | | |
Collapse
|
17
|
Gimenez-Cassina A, Lim F, Diaz-Nido J. Differentiation of a human neuroblastoma into neuron-like cells increases their susceptibility to transduction by herpesviral vectors. J Neurosci Res 2006; 84:755-67. [PMID: 16802347 DOI: 10.1002/jnr.20976] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Gene transfer is a powerful tool for functional gene analysis in human cells. In this respect, there is a need to develop experimental models that involve homogeneous cultures of human neuron-like cells susceptible to gene transduction and that are easy to handle. Here we describe an optimized and reproducible procedure to differentiate human SH-SY5Y neuroblastoma cells into a homogeneous population of neuron-like cells. The fully differentiated cells are postmitotic and resemble primary cultured neurons in terms of their cytoskeletal polarity. Notably, differentiated SH-SY5Y cells are far more susceptible to transduction by herpes simplex virus (HSV-1)-based vectors than proliferating SH-SY5Y cells. This increase in transduction efficiency after neuronal differentiation may be due to the up-regulation of cell surface receptors for herpesvirus entry. In summary, we propose that fully differentiated human neuron-like cells obtained from the SH-SY5Y neuroblastoma may constitute an excellent and versatile experimental tool for gene transfer and functional genomic studies with HSV-1 vectors.
Collapse
Affiliation(s)
- Alfredo Gimenez-Cassina
- Departamento de Biologia Molecular, Centro de Biologia Molecular Severo Ochoa, Universidad Autonoma de Madrid, Madrid, Spain
| | | | | |
Collapse
|
18
|
Oehmig A, Fraefel C, Breakefield XO. Update on herpesvirus amplicon vectors. Mol Ther 2005; 10:630-43. [PMID: 15451447 DOI: 10.1016/j.ymthe.2004.06.641] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Accepted: 06/17/2004] [Indexed: 12/29/2022] Open
Affiliation(s)
- Angelika Oehmig
- Department of Neurology, Massachusetts General Hospital, and Program in Neuroscience, Harvard Medical School, Boston, MA 02114, USA
| | | | | |
Collapse
|
19
|
Paul CA, Reid PC, Boegle AK, Karten B, Zhang M, Jiang ZG, Franz D, Lin L, Chang TY, Vance JE, Blanchette-Mackie J, Maue RA. Adenovirus expressing an NPC1-GFP fusion gene corrects neuronal and nonneuronal defects associated with Niemann pick type C disease. J Neurosci Res 2005; 81:706-19. [PMID: 16015597 DOI: 10.1002/jnr.20592] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Niemann Pick type C (NPC) disease is an autosomal recessive disorder characterized by abnormal cholesterol metabolism and accumulation in lysosomal and endosomal compartments. Although peripheral organs are affected, the progressive neurodegeneration in the brain is typically most deleterious, leading to dystonia, ataxia, seizures, and premature death. Although the two genes underlying this disorder in humans and mouse models of the disease have been identified (NPC1 in 95% and NPC2/HE1 in 5% of human cases), their cellular roles have not Been fully defined, and there is currently no effective treatment for this disorder. To help address these issues, we constructed a recombinant adenovirus, Ad(NPC1-GFP), which contains a cDNA encoding a mouse NPC1 protein with a green fluorescent protein (GFP) fused to its C-terminus. Fluorescence microscopy and cholesterol trafficking assays demonstrate that the GFP-tagged NPC1 protein is functional and detectable in cells from different species (hamster, mouse, human) and of different types (ovary-derived cells, fibroblasts, astrocytes, neurons from peripheral and central nervous systems) in vitro. Combined with results from time-lapse microscopy and in vivo brain injections, our findings suggest that this adenovirus offers advantages for expressing NPC1 and analyzing its cellular localization, movement, functional properties, and beneficial effects in vitro and in vivo.
Collapse
Affiliation(s)
- C A Paul
- Department of Biochemistry, Dartmouth Medical School, Hanover, New Hampshire 03755, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Qi J, Shackelford R, Manuszak R, Cheng D, Smith M, Link CJ, Wang S. Functional expression of ATM gene carried by HSV amplicon vector in vitro and in vivo. Gene Ther 2004; 11:25-33. [PMID: 14681694 DOI: 10.1038/sj.gt.3302140] [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: 11/08/2022]
Abstract
Ataxia-telangiectasia (AT) is a human autosomal recessive disease with a pleiotropic phenotype characterized by cerebellar degeneration, immunodeficiency, premature aging, cancer predisposition, and radiation sensitivity. The gene mutated in AT, ATM (for AT-mutated), had been cloned and found to have ionizing radiation and oxidative stress-inducible kinase activity. No treatment can stop the progression of the disease. In this study, the complete open-reading frame of ATM cDNA was cloned into a Herpes simplex virus type-1 (HSV-1) amplicon vector (pTO-ATM), and the transduction of cultured AT cells was demonstrated by immunohistochemistry and Western blot analysis. Functional gene expression was evaluated by cell colony-forming assays following exposure to oxidative stress. The survival of AT cells with ATM gene transduction was about 100% higher compared to nontransduced cells after t-butyl hydroperoxide treatments. Next, the normal ATM gene expression in different regions of the rat brain was studied. Immunohistochemistry staining demonstrated weak endogenous ATM protein expression in neurons of the caudate-putamen, with significantly higher levels of expression detected in neurons in other brain regions. Exogenous ATM gene expression from pTO-ATM after viral transduction in the caudate-putamen of the adult rat was examined. At 3 days after injection of the pTO-ATM viral vector, abundant positive ATM staining of the neurons was found at the injection sites, in comparison to the controls. These data demonstrate that the relatively large ATM cDNA can be transduced and expressed in vitro and in vivo from an HSV amplicon viral vector. These data provide initial evidence that the replacement of the ATM gene into the cells of AT patients might be possible some day.
Collapse
Affiliation(s)
- J Qi
- Human Gene Therapy Research Institute, Stoddard Cancer Research Institute, IA 50309, USA
| | | | | | | | | | | | | |
Collapse
|