1
|
Shyam M, Bm O, Srirangan P, N N, Sabina EP. Targeted miRNA Delivery in Epilepsy: Mechanisms, Advances, and Therapeutic Potential. Mol Biol Rep 2025; 52:368. [PMID: 40192852 DOI: 10.1007/s11033-025-10436-z] [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: 11/22/2024] [Accepted: 03/11/2025] [Indexed: 04/23/2025]
Abstract
Epilepsy, a neurological disorder characterized by recurrent seizures, presents significant therapeutic challenges, with roughly 30% of individuals demonstrating resistance to antiepileptic drugs. Drug-resistant epilepsy diminishes patients' quality of life and underscores the critical need for innovative therapeutic approaches. MicroRNAs, small non-coding RNA molecules, have emerged as key regulators in the pathogenesis of epilepsy, influencing neuronal excitability, synaptic plasticity, and neuroinflammatory processes. By targeting multiple genes and pathways involved in epileptogenesis, miRNAs offer promising opportunities for precision medicine. This review explores the dual roles of specific miRNAs in epilepsy, acting as both promoters and inhibitors of pathogenic pathways, and highlights recent advancements in miRNA-based therapeutic delivery systems. State-of-the-art approaches, including lipid nanoparticles, viral vectors, and exosome-based systems, are being developed to address challenges such as blood-brain barrier penetration, targeted delivery, and minimizing systemic side effects. These advancements lay the groundwork for more effective and personalized treatment strategies.
Collapse
Affiliation(s)
- Mukul Shyam
- Department of Biotechnology, School of Biosciences and Technology, VIT University, Tamil Nadu, Vellore, 632014, India
| | - Oveyaa Bm
- Department of Biotechnology, School of Biosciences and Technology, VIT University, Tamil Nadu, Vellore, 632014, India
| | - Prathap Srirangan
- Department of Biotechnology, School of Biosciences and Technology, VIT University, Tamil Nadu, Vellore, 632014, India
| | - Nivedita N
- Department of Biotechnology, School of Biosciences and Technology, VIT University, Tamil Nadu, Vellore, 632014, India
| | - Evan Prince Sabina
- Department of Biotechnology, School of Biosciences and Technology, VIT University, Tamil Nadu, Vellore, 632014, India.
| |
Collapse
|
2
|
Park J, Rhoo KY, Kim Y, Kim YS, Paik SR. Cell-Division-Independent Rapid Expression of DNA Delivered with α-Synuclein-Gold Nanoparticle Conjugates. ACS APPLIED MATERIALS & INTERFACES 2025; 17:14846-14858. [PMID: 40014054 DOI: 10.1021/acsami.4c17967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
Gene delivery is a primary technology employed in diverse areas of biomedical science, from gene therapy to gene editing, cancer treatment, and stem cell research. Here, we introduce a gene delivery system utilizing an intrinsically disordered protein of α-synuclein (αS) demonstrated to interact with lipid membranes by transforming its original random structure to an α-helix. Since the helix bundle formation is a signature of cell-penetrating peptides for membrane translocation, a multitude of αS(Y136C)s replacing tyrosine at the C-terminus with cysteine were covalently attached onto gold nanoparticles (AuNPs) in a specific orientation with the helix-forming basic N-termini exposed outward. The resulting αS(Y136C)-AuNP conjugates were found to exhibit a rapid gene expression without causing cytotoxicity when the gene of the enhanced green fluorescent protein (EGFP) was delivered with the conjugates into the cells. Based on inhibition studies toward endocytosis and mitosis, the αS(Y136C)-AuNP/DNA complex was demonstrated to take both endosomal and non-endosomal intracellular transport pathways. The DNA translocation into the nucleus was independent of cell division. This nondisruptive and rapid DNA transfection by αS(Y136C)-AuNPs allowed a successful delivery of granzyme A gene leading to cellular pyroptosis. Modifications of αS(Y136C)-AuNP/DNA complex, such as antibody immobilization and replacement of DNA with biological suprastructures including RNA, protein, and nonbiological fusion materials, would allow the intracellular delivery system to be applied in diverse areas of future biotechnology.
Collapse
Affiliation(s)
- Jeongha Park
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Kun Yil Rhoo
- Interdisciplinary Program of Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Yunsoo Kim
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Young-Sik Kim
- Department of Pathology, Ansan Hospital, Korea University College of Medicine, Ansan 15355, Republic of Korea
| | - Seung R Paik
- School of Chemical and Biological Engineering, Institute of Engineering Research, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
- Interdisciplinary Program of Bioengineering, College of Engineering, Seoul National University, Seoul 08826, Republic of Korea
| |
Collapse
|
3
|
Mani S, Jindal D, Singh M. Gene Therapy, A Potential Therapeutic Tool for Neurological and Neuropsychiatric Disorders: Applications, Challenges and Future Perspective. Curr Gene Ther 2023; 23:20-40. [PMID: 35345999 DOI: 10.2174/1566523222666220328142427] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/18/2022] [Accepted: 02/02/2022] [Indexed: 02/08/2023]
Abstract
Neurological and neuropsychiatric disorders are the main risks for the health care system, exhibiting a huge socioeconomic load. The available range of pharmacotherapeutics mostly provides palliative consequences and fails to treat such conditions. The molecular etiology of various neurological and neuropsychiatric disorders is mostly associated with a change in genetic background, which can be inherited/triggered by other environmental factors. To address such conditions, gene therapy is considered a potential approach claiming a permanent cure of the disease primarily by deletion, silencing, or edition of faulty genes and by insertion of healthier genes. In gene therapy, vectors (viral/nonvial) play an important role in delivering the desired gene to a specific region of the brain. Targeted gene therapy has unraveled opportunities for the treatment of many neurological and neuropsychiatric disorders. For improved gene delivery, the current techniques mainly focus on designing a precise viral vector, plasmid transfection, nanotechnology, microRNA, and in vivo clustered regulatory interspaced short palindromic repeats (CRISPR)-based therapy. These latest techniques have great benefits in treating predominant neurological and neurodevelopmental disorders, including Parkinson's disease, Alzheimer's disease, and autism spectrum disorder, as well as rarer diseases. Nevertheless, all these delivery methods have their limitations, including immunogenic reactions, off-target effects, and a deficiency of effective biomarkers to appreciate the effectiveness of therapy. In this review, we present a summary of the current methods in targeted gene delivery, followed by the limitations and future direction of gene therapy for the cure of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Shalini Mani
- Department of Biotechnology, Centre for Emerging Diseases, Jaypee Institute of Information Technology, Noida, U.P., India
| | - Divya Jindal
- Department of Biotechnology, Centre for Emerging Diseases, Jaypee Institute of Information Technology, Noida, U.P., India
| | - Manisha Singh
- Department of Biotechnology, Centre for Emerging Diseases, Jaypee Institute of Information Technology, Noida, U.P., India
| |
Collapse
|
4
|
Podetz-Pedersen KM, Laoharawee K, Singh S, Nguyen TT, Smith MC, Temme A, Kozarsky K, McIvor RS, Belur LR. Neurologic Recovery in MPS I and MPS II Mice by AAV9-Mediated Gene Transfer to the CNS After the Development of Cognitive Dysfunction. Hum Gene Ther 2023; 34:8-18. [PMID: 36541357 PMCID: PMC10024071 DOI: 10.1089/hum.2022.162] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 10/25/2022] [Indexed: 12/24/2022] Open
Abstract
The mucopolysaccharidoses (MPS) are a group of recessively inherited conditions caused by deficiency of lysosomal enzymes essential to the catabolism of glycosaminoglycans (GAG). MPS I is caused by deficiency of the lysosomal enzyme alpha-L-iduronidase (IDUA), while MPS II is caused by a lack of iduronate-2-sulfatase (IDS). Lack of these enzymes leads to early mortality and morbidity, often including neurological deficits. Enzyme replacement therapy has markedly improved the quality of life for MPS I and MPS II affected individuals but is not effective in addressing neurologic manifestations. For MPS I, hematopoietic stem cell transplant has shown effectiveness in mitigating the progression of neurologic disease when carried out in early in life, but neurologic function is not restored in patients transplanted later in life. For both MPS I and II, gene therapy has been shown to prevent neurologic deficits in affected mice when administered early, but the effectiveness of treatment after the onset of neurologic disease manifestations has not been characterized. To test if neurocognitive function can be recovered in older animals, human IDUA or IDS-encoding AAV9 vector was administered by intracerebroventricular injection into MPS I and MPS II mice, respectively, after the development of neurologic deficit. Vector sequences were distributed throughout the brains of treated animals, associated with high levels of enzyme activity and normalized GAG storage. Two months after vector infusion, treated mice exhibited spatial navigation and learning skills that were normalized, that is, indistinguishable from those of normal unaffected mice, and significantly improved compared to untreated, affected animals. We conclude that cognitive function was restored by AAV9-mediated, central nervous system (CNS)-directed gene transfer in the murine models of MPS I and MPS II, suggesting that gene transfer may result in neurodevelopment improvements in severe MPS I and MPS II when carried out after the onset of cognitive decline.
Collapse
Affiliation(s)
- Kelly M. Podetz-Pedersen
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Kanut Laoharawee
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Sajya Singh
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Tam T. Nguyen
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Miles C. Smith
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Alexa Temme
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - R. Scott McIvor
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lalitha R. Belur
- Department of Genetics, Cell Biology and Development, Center for Genome Engineering, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
5
|
Eftekharpour E, Shcholok T. Cre-recombinase systems for induction of neuron-specific knockout models: a guide for biomedical researchers. Neural Regen Res 2023; 18:273-279. [PMID: 35900402 PMCID: PMC9396489 DOI: 10.4103/1673-5374.346541] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Gene deletion has been a valuable tool for unraveling the mysteries of molecular biology. Early approaches included gene trapping and gene targetting to disrupt or delete a gene randomly or at a specific location, respectively. Using these technologies in mouse embryos led to the generation of mouse knockout models and many scientific discoveries. The efficacy and specificity of these approaches have significantly increased with the advent of new technology such as clustered regularly interspaced short palindromic repeats for targetted gene deletion. However, several limitations including unwanted off-target gene deletion have hindered their widespread use in the field. Cre-recombinase technology has provided additional capacity for cell-specific gene deletion. In this review, we provide a summary of currently available literature on the application of this system for targetted deletion of neuronal genes. This article has been constructed to provide some background information for the new trainees on the mechanism and to provide necessary information for the design, and application of the Cre-recombinase system through reviewing the most frequent promoters that are currently available for genetic manipulation of neurons. We additionally will provide a summary of the latest technological developments that can be used for targeting neurons. This may also serve as a general guide for the selection of appropriate models for biomedical research.
Collapse
|
6
|
Chang KJ, Wu HY, Yarmishyn AA, Li CY, Hsiao YJ, Chi YC, Lo TC, Dai HJ, Yang YC, Liu DH, Hwang DK, Chen SJ, Hsu CC, Kao CL. Genetics behind Cerebral Disease with Ocular Comorbidity: Finding Parallels between the Brain and Eye Molecular Pathology. Int J Mol Sci 2022; 23:9707. [PMID: 36077104 PMCID: PMC9456058 DOI: 10.3390/ijms23179707] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 08/18/2022] [Accepted: 08/22/2022] [Indexed: 11/30/2022] Open
Abstract
Cerebral visual impairments (CVIs) is an umbrella term that categorizes miscellaneous visual defects with parallel genetic brain disorders. While the manifestations of CVIs are diverse and ambiguous, molecular diagnostics stand out as a powerful approach for understanding pathomechanisms in CVIs. Nevertheless, the characterization of CVI disease cohorts has been fragmented and lacks integration. By revisiting the genome-wide and phenome-wide association studies (GWAS and PheWAS), we clustered a handful of renowned CVIs into five ontology groups, namely ciliopathies (Joubert syndrome, Bardet-Biedl syndrome, Alstrom syndrome), demyelination diseases (multiple sclerosis, Alexander disease, Pelizaeus-Merzbacher disease), transcriptional deregulation diseases (Mowat-Wilson disease, Pitt-Hopkins disease, Rett syndrome, Cockayne syndrome, X-linked alpha-thalassaemia mental retardation), compromised peroxisome disorders (Zellweger spectrum disorder, Refsum disease), and channelopathies (neuromyelitis optica spectrum disorder), and reviewed several mutation hotspots currently found to be associated with the CVIs. Moreover, we discussed the common manifestations in the brain and the eye, and collated animal study findings to discuss plausible gene editing strategies for future CVI correction.
Collapse
Affiliation(s)
- Kao-Jung Chang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
| | - Hsin-Yu Wu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | | | - Cheng-Yi Li
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yu-Jer Hsiao
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yi-Chun Chi
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Tzu-Chen Lo
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - He-Jhen Dai
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Yi-Chiang Yang
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Ding-Hao Liu
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - De-Kuang Hwang
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Shih-Jen Chen
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan
| | - Chih-Chien Hsu
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Ophthalmology, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung 80708, Taiwan
| | - Chung-Lan Kao
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Department of Physical Medicine and Rehabilitation, Taipei Veterans General Hospital, Taipei 11217, Taiwan
- Department of Physical Medicine and Rehabilitation, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan
- Center for Intelligent Drug Systems and Smart Bio-Devices (IDS2B), National Yang Ming Chiao Tung University, Hsinchu 300093, Taiwan
| |
Collapse
|
7
|
Gallagher T. Interview with Beverly Davidson, PhD. Hum Gene Ther 2022; 33:347-350. [PMID: 35442068 DOI: 10.1089/hum.2022.29205.int] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
8
|
Abd El-Hamid BN, Khalil IA, Harashima H. Viral Gene Delivery. THE ADME ENCYCLOPEDIA 2022:1183-1192. [DOI: 10.1007/978-3-030-84860-6_117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/01/2023]
|
9
|
Graceffa V. Clinical Development of Cell Therapies to Halt Lysosomal Storage Diseases: Results and Lessons Learned. Curr Gene Ther 2021; 22:191-213. [PMID: 34323185 DOI: 10.2174/1566523221666210728141924] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/31/2021] [Accepted: 06/13/2021] [Indexed: 11/22/2022]
Abstract
Although cross-correction was discovered more than 50 years ago, and held the promise of drastically improving disease management, still no cure exists for lysosomal storage diseases (LSDs). Cell therapies hold the potential to halt disease progression: either a subset of autologous cells can be ex vivo/ in vivo transfected with the functional gene or allogenic wild type stem cells can be transplanted. However, majority of cell-based attempts have been ineffective, due to the difficulties in reversing neuronal symptomatology, in finding appropriate gene transfection approaches, in inducing immune tolerance, reducing the risk of graft versus host disease (GVHD) when allogenic cells are used and that of immune response when engineered viruses are administered, coupled with a limited secretion and uptake of some enzymes. In the last decade, due to advances in our understanding of lysosomal biology and mechanisms of cross-correction, coupled with progresses in gene therapy, ongoing pre-clinical and clinical investigations have remarkably increased. Even gene editing approaches are currently under clinical experimentation. This review proposes to critically discuss and compare trends and advances in cell-based and gene therapy for LSDs. Systemic gene delivery and transplantation of allogenic stem cells will be initially discussed, whereas proposed brain targeting methods will be then critically outlined.
Collapse
Affiliation(s)
- Valeria Graceffa
- Cellular Health and Toxicology Research Group (CHAT), Institute of Technology Sligo, Ash Ln, Bellanode, Sligo, Ireland
| |
Collapse
|
10
|
Abd El-Hamid BN, Khalil IA, Harashima H. Viral Gene Delivery. THE ADME ENCYCLOPEDIA 2021:1-10. [DOI: 10.1007/978-3-030-51519-5_117-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Accepted: 02/01/2021] [Indexed: 09/01/2023]
|
11
|
Plekhanova O, Parfyonova Y, Beloglazova I, Berk BC, Tkachuk V. Oligonucleotide Microarrays Identified Potential Regulatory Genes Related to Early Outward Arterial Remodeling Induced by Tissue Plasminogen Activator. Front Physiol 2019; 10:493. [PMID: 31114508 PMCID: PMC6502959 DOI: 10.3389/fphys.2019.00493] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 04/08/2019] [Indexed: 01/18/2023] Open
Abstract
Constrictive vascular remodeling limiting blood flow, as well as compensatory outward remodeling, has been observed in many cardiovascular diseases; however, the underlying mechanisms regulating the remodeling response of the vessels remain unclear. Plasminogen activators (PA) are involved in many of the processes of vascular remodeling. We have shown previously that increased levels of tissue-type PA (tPA) contributes to outward vascular remodeling. To elucidate the mechanisms involved in the induction of outward remodeling we characterized changes in the expression profiles of 8799 genes in injured rat carotid arteries 1 and 4 days after recombinant tPA treatment compared to vehicle. Periadventitial tPA significantly increased lumen size and vessel area, encompassed by the external elastic lamina, at both one and 4 days after treatment. Among 41 differentially expressed known genes 1 day after tPA application, five genes were involved in gene transcription, five genes were related to the regulation of vascular tone [for example, thromboxane A2 receptor (D32080) or non-selective-type endothelin receptor (S65355)], and eight genes were identified as participating in vascular innervation [for example, calpain (D14478) or neural cell adhesion molecule L1 (X59149)]. Four days after injury in tPA-treated arteries, four genes, regulating vascular tone, were differentially expressed. Thus, tPA promotes outward arterial remodeling after injury, at least in part, by regulating expression of genes in the vessel wall related to function of the nervous system and vascular tone.
Collapse
Affiliation(s)
- Olga Plekhanova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,National Medical Research Center of Cardiology, Moscow, Russia
| | - Yelena Parfyonova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,National Medical Research Center of Cardiology, Moscow, Russia
| | - Irina Beloglazova
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,National Medical Research Center of Cardiology, Moscow, Russia
| | - Bradford C Berk
- Aab Cardiovascular Research Institute, University of Rochester Medical Center, Rochester, NY, United States
| | - Vsevolod Tkachuk
- Faculty of Medicine, Lomonosov Moscow State University, Moscow, Russia.,National Medical Research Center of Cardiology, Moscow, Russia
| |
Collapse
|
12
|
Choudhury SR, Hudry E, Maguire CA, Sena-Esteves M, Breakefield XO, Grandi P. Viral vectors for therapy of neurologic diseases. Neuropharmacology 2017; 120:63-80. [PMID: 26905292 PMCID: PMC5929167 DOI: 10.1016/j.neuropharm.2016.02.013] [Citation(s) in RCA: 123] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2015] [Revised: 02/07/2016] [Accepted: 02/15/2016] [Indexed: 12/21/2022]
Abstract
Neurological disorders - disorders of the brain, spine and associated nerves - are a leading contributor to global disease burden with a shockingly large associated economic cost. Various treatment approaches - pharmaceutical medication, device-based therapy, physiotherapy, surgical intervention, among others - have been explored to alleviate the resulting extent of human suffering. In recent years, gene therapy using viral vectors - encoding a therapeutic gene or inhibitory RNA into a "gutted" viral capsid and supplying it to the nervous system - has emerged as a clinically viable option for therapy of brain disorders. In this Review, we provide an overview of the current state and advances in the field of viral vector-mediated gene therapy for neurological disorders. Vector tools and delivery methods have evolved considerably over recent years, with the goal of providing greater and safer genetic access to the central nervous system. Better etiological understanding of brain disorders has concurrently led to identification of improved therapeutic targets. We focus on the vector technology, as well as preclinical and clinical progress made thus far for brain cancer and various neurodegenerative and neurometabolic disorders, and point out the challenges and limitations that accompany this new medical modality. Finally, we explore the directions that neurological gene therapy is likely to evolve towards in the future. This article is part of the Special Issue entitled "Beyond small molecules for neurological disorders".
Collapse
Affiliation(s)
- Sourav R Choudhury
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Eloise Hudry
- Alzheimer's Disease Research Unit, Harvard Medical School & Massachusetts General Hospital, Charlestown, MA 02129, USA.
| | - Casey A Maguire
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA 01605, USA; Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| | - Xandra O Breakefield
- Department of Neurology and Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA 02114, USA.
| | - Paola Grandi
- Department of Neurological Surgery, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15219, USA.
| |
Collapse
|
13
|
Antyborzec I, O'Leary VB, Dolly JO, Ovsepian SV. Low-Affinity Neurotrophin Receptor p75 Promotes the Transduction of Targeted Lentiviral Vectors to Cholinergic Neurons of Rat Basal Forebrain. Neurotherapeutics 2016; 13:859-870. [PMID: 27220617 PMCID: PMC5081123 DOI: 10.1007/s13311-016-0445-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Basal forebrain cholinergic neurons (BFCNs) are one of the most affected neuronal types in Alzheimer's disease (AD), with their extensive loss documented at late stages of the pathology. While discriminatory provision of neuroprotective agents and trophic factors to these cells is thought to be of substantial therapeutic potential, the intricate topography and structure of the forebrain cholinergic system imposes a major challenge. To overcome this, we took advantage of the physiological enrichment of BFCNs with a low-affinity p75 neurotrophin receptor (p75NTR) for their targeting by lentiviral vectors within the intact brain of adult rat. Herein, a method is described that affords selective and effective transduction of BFCNs with a green fluorescence protein (GFP) reporter, which combines streptavidin-biotin technology with anti-p75NTR antibody-coated lentiviral vectors. Specific GFP expression in cholinergic neurons was attained in the medial septum and nuclei of the diagonal band Broca after a single intraventricular administration of such targeted vectors. Bioelectrical activity of GFP-labeled neurons was proven to be unchanged. Thus, proof of principle is obtained for the utility of the low-affinity p75NTR for targeted transduction of vectors to BFCNs in vivo.
Collapse
Affiliation(s)
- Inga Antyborzec
- International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland
| | - Valerie B O'Leary
- International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland
- Institute of Radiation Biology, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg, Germany
| | - James O Dolly
- International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland
| | - Saak V Ovsepian
- International Centre for Neurotherapeutics, Dublin City University, Dublin, Ireland.
- Institute for Biological and Medical Imaging, Helmholtz Zentrum Munich, German Research Center for Environmental Health, Neuherberg, Germany.
- Munich School of Bioengineering, Technical University Munich, Munich, Germany.
| |
Collapse
|
14
|
Parente MK, Rozen R, Seeholzer SH, Wolfe JH. Integrated analysis of proteome and transcriptome changes in the mucopolysaccharidosis type VII mouse hippocampus. Mol Genet Metab 2016; 118:41-54. [PMID: 27053151 PMCID: PMC4832927 DOI: 10.1016/j.ymgme.2016.03.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Accepted: 03/05/2016] [Indexed: 12/15/2022]
Abstract
Mucopolysaccharidosis type VII (MPS VII) is a lysosomal storage disease caused by the deficiency of β-glucuronidase. In this study, we compared the changes relative to normal littermates in the proteome and transcriptome of the hippocampus in the C57Bl/6 mouse model of MPS VII, which has well-documented histopathological and neurodegenerative changes. A completely different set of significant changes between normal and MPS VII littermates were found in each assay. Nevertheless, the functional annotation terms generated by the two methods showed agreement in many of the processes, which also corresponded to known pathology associated with the disease. Additionally, assay-specific changes were found, which in the proteomic analysis included mitochondria, energy generation, and cytoskeletal differences in the mutant, while the transcriptome differences included immune, vesicular, and extracellular matrix changes. In addition, the transcriptomic changes in the mutant hippocampus were concordant with those in a MPS VII mouse caused by the same mutation but on a different background inbred strain.
Collapse
Affiliation(s)
- Michael K Parente
- Research Institute of the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ramona Rozen
- Research Institute of the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Steven H Seeholzer
- Research Institute of the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - John H Wolfe
- Research Institute of the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; W. F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
15
|
King B, Setford ML, Hassiotis S, Trim PJ, Duplock S, Tucker JN, Hattersley K, Snel MF, Hopwood JJ, Hemsley KM. Low-dose, continual enzyme delivery ameliorates some aspects of established brain disease in a mouse model of a childhood-onset neurodegenerative disorder. Exp Neurol 2016; 278:11-21. [DOI: 10.1016/j.expneurol.2015.11.013] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 11/19/2015] [Accepted: 11/23/2015] [Indexed: 12/12/2022]
|
16
|
Biffi A. Gene therapy for lysosomal storage disorders: a good start. Hum Mol Genet 2015; 25:R65-75. [PMID: 26604151 DOI: 10.1093/hmg/ddv457] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022] Open
Abstract
Lysosomal storage disorders (LSDs) are a heterogeneous group of inherited diseases with a collective frequency of ∼1 in 7000 births, resulting from the deficiency in one or more enzymes or transporters that normally reside within the lysosomes. Pathology results from the progressive accumulation of uncleaved lipids, glycoproteins and/or glycosaminoglycans in the lysosomes and secondary damages that affect the brain, viscera, bones and connective tissues. Most treatment modalities developed for LSD, including gene therapy (GT), are based on the lysosome-specific cross-correction mechanism, by which close proximity of normal cells leads to the correction of the biochemical consequences of enzymatic deficiency within the neighboring cells. Here, GT efforts addressing these disorders are reviewed with an up-to-date discussion of their impact on the LSD disease phenotype in animal models and patients.
Collapse
Affiliation(s)
- Alessandra Biffi
- Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
17
|
Gadalla KKE, Ross PD, Hector RD, Bahey NG, Bailey MES, Cobb SR. Gene therapy for Rett syndrome: prospects and challenges. FUTURE NEUROLOGY 2015. [DOI: 10.2217/fnl.15.29] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Rett syndrome (RTT) is a neurological disorder that affects females and is caused by loss-of-function mutations in the X-linked gene MECP2. Deletion of Mecp2 in mice results in a constellation of neurological features that resemble those seen in RTT patients. Experiments in mice have demonstrated that restoration of MeCP2, even at adult stages, reverses several aspects of the RTT-like pathology suggesting that the disorder may be inherently treatable. This has provided an impetus to explore several therapeutic approaches targeting RTT at the level of the gene, including gene therapy, activation of MECP2 on the inactive X chromosome and read-through and repair of RTT-causing mutations. Here, we review these different strategies and the challenges of gene-based approaches in RTT.
Collapse
Affiliation(s)
- Kamal KE Gadalla
- University of Glasgow, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, Glasgow, UK
- Pharmacology Department, Faculty of Medicine, Tanta University, Egypt
| | - Paul D Ross
- University of Glasgow, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, Glasgow, UK
| | - Ralph D Hector
- University of Glasgow, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, Glasgow, UK
| | - Noha G Bahey
- University of Glasgow, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, Glasgow, UK
- Histology Department, Faculty of Medicine, Tanta University, Egypt
| | - Mark ES Bailey
- School of Life Sciences, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Stuart R Cobb
- University of Glasgow, Institute of Neuroscience and Psychology, College of Medical, Veterinary and Life Sciences, Glasgow, UK
| |
Collapse
|
18
|
Recent studies of ovine neuronal ceroid lipofuscinoses from BARN, the Batten Animal Research Network. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2279-86. [PMID: 26073432 DOI: 10.1016/j.bbadis.2015.06.013] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2015] [Revised: 05/26/2015] [Accepted: 06/08/2015] [Indexed: 12/19/2022]
Abstract
Studies on naturally occurring New Zealand and Australian ovine models of the neuronal ceroid-lipofuscinoses (Batten disease, NCLs) have greatly aided our understanding of these diseases. Close collaborations between the New Zealand groups at Lincoln University and the University of Otago, Dunedin, and a group at the University of Sydney, Australia, led to the formation of BARN, the Batten Animal Research Network. This review focusses on presentations at the 14th International Conference on Neuronal Ceroid Lipofuscinoses (Batten Disease), recent relevant background work, and previews of work in preparation for publication. Themes include CLN5 and CLN6 neuronal cell culture studies, studies on tissues from affected and control animals and whole animal in vivo studies. Topics include the effect of a CLN6 mutation on endoplasmic reticulum proteins, lysosomal function and the interactions of CLN6 with other lysosomal activities and trafficking, scoping gene-based therapies, a molecular dissection of neuroinflammation, identification of differentially expressed genes in brain tissue, an attempted therapy with an anti-inflammatory drug in vivo and work towards gene therapy in ovine models of the NCLs. This article is part of a Special Issue entitled: "Current Research on the Neuronal Ceroid Lipofuscinoses (Batten Disease)".
Collapse
|
19
|
Beck M. Enzyme replacement and gene therapy for mucopolysaccharidoses: current progress and future directions. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1021777] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
20
|
Hassiotis S, Beard H, Luck A, Trim PJ, King B, Snel MF, Hopwood JJ, Hemsley KM. Disease stage determines the efficacy of treatment of a paediatric neurodegenerative disease. Eur J Neurosci 2015; 39:2139-50. [PMID: 25068161 DOI: 10.1111/ejn.12557] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Lysosomal storage disorders are a large group of inherited metabolic conditions resulting from the deficiency of proteins involved in lysosomal catabolism, with resulting accumulation of substrates inside the cell. Two-thirds of these disorders are associated with a neurodegenerative phenotype and, although few therapeutic options are available to patients at present, clinical trials of several treatments including lysosomal enzyme replacement are underway. Although animal studies indicate the efficacy of presymptomatic treatment, it is largely unknown whether symptomatic disease-related pathology and functional deficits are reversible. To begin to address this, we used a naturally-occurring mouse model with Sanfilippo syndrome (mucopolysaccharidosis type IIIA) to examine the effectiveness of intracisternal cerebrospinal fluid enzyme replacement in early, mid- and symptomatic disease stage mice. We observed a disease-stage-dependent treatment effect, with the most significant reductions in primary and secondary substrate accumulation, astrogliosis and protein aggregate accumulation seen in mucopolysaccharidosis type IIIA mice treated very early in the disease course. Affected mice treated at a symptomatic age exhibited little change in these neuropathological markers in the time-frame of the study. Microgliosis was refractory to treatment regardless of the age at which treatment was instigated. Although longer-term studies are warranted, these findings indicate the importance of early intervention in this condition.
Collapse
|
21
|
Liu W, Griffin G, Clarke T, Parente MK, Valentino RJ, Wolfe JH, Fraser NW. Bilateral single-site intracerebral injection of a nonpathogenic herpes simplex virus-1 vector decreases anxiogenic behavior in MPS VII mice. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:14059. [PMID: 26052529 PMCID: PMC4448997 DOI: 10.1038/mtm.2014.59] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2014] [Revised: 11/11/2014] [Accepted: 11/11/2014] [Indexed: 01/17/2023]
Abstract
Genetic diseases of the brain usually have pathologic lesions distributed throughout, thus requiring global correction. Herpes simplex virus-1 (HSV-1) vectors may be especially useful for gene delivery in these disorders since they can spread trans-synaptically along neuronal pathways to distal sites from a localized injection. We have previously shown that a nonpathogenic HSV-1 (strain 1716), which is deleted in the ICP34.5 gene, and expressing the lysosomal enzyme β-glucuronidase (GUSB) from the latency-associated transcript (LAT) promoter, spreads within the brains of GUSB-deficient mucopolysaccharidosis VII mice to reverse the pathognomonic storage lesions throughout the diseased brain. In this study, we tested the ability of the 1716 LAT-GUSB vector to improve behavioral deficits. The treatment significantly decreased anxiogenic behaviors associated with the mutation, as indicated by open-field behavior and decreased neophobia in a novel object-recognition task. The treated mice also exhibited an improvement in cognitive function associated with the cerebral cortex in a familiar object test. The results indicate the functional therapeutic potential of the 1716 LAT-GUSB vector.
Collapse
Affiliation(s)
- Wenpei Liu
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania, USA
| | - Gerald Griffin
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania, USA
| | - Trena Clarke
- Stokes Institute, Children's Hospital of Philadelphia , Philadelphia, Pennsylvania, USA
| | - Michael K Parente
- Stokes Institute, Children's Hospital of Philadelphia , Philadelphia, Pennsylvania, USA
| | - Rita J Valentino
- Stokes Institute, Children's Hospital of Philadelphia , Philadelphia, Pennsylvania, USA ; Department of Anesthesiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania, USA
| | - John H Wolfe
- Stokes Institute, Children's Hospital of Philadelphia , Philadelphia, Pennsylvania, USA ; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania, USA ; W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania , Philadelphia, Pennsylvania, USA
| | - Nigel W Fraser
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania , Philadelphia, Pennsylvania, USA
| |
Collapse
|
22
|
McIntyre C, Derrick-Roberts ALK, Byers S, Anson DS. Correction of murine mucopolysaccharidosis type IIIA central nervous system pathology by intracerebroventricular lentiviral-mediated gene delivery. J Gene Med 2014; 16:374-87. [DOI: 10.1002/jgm.2816] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2014] [Revised: 11/07/2014] [Accepted: 11/14/2014] [Indexed: 11/06/2022] Open
Affiliation(s)
- Chantelle McIntyre
- School of Paediatrics and Reproductive Health; University of Adelaide; South Australia Australia
| | - Ainslie L. K. Derrick-Roberts
- School of Paediatrics and Reproductive Health; University of Adelaide; South Australia Australia
- Genetics and Molecular Pathology, SA Pathology; North Adelaide South Australia Australia
| | - Sharon Byers
- School of Paediatrics and Reproductive Health; University of Adelaide; South Australia Australia
- Genetics and Molecular Pathology, SA Pathology; North Adelaide South Australia Australia
- School of Molecular and Biomedical Science; University of Adelaide; South Australia Australia
| | - Donald S. Anson
- School of Paediatrics and Reproductive Health; University of Adelaide; South Australia Australia
- Genetics and Molecular Pathology, SA Pathology; North Adelaide South Australia Australia
| |
Collapse
|
23
|
Cachón-González MB, Wang SZ, Ziegler R, Cheng SH, Cox TM. Reversibility of neuropathology in Tay-Sachs-related diseases. Hum Mol Genet 2014; 23:730-48. [PMID: 24057669 PMCID: PMC3888261 DOI: 10.1093/hmg/ddt459] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Accepted: 09/16/2013] [Indexed: 01/22/2023] Open
Abstract
The GM2 gangliosidoses are progressive neurodegenerative disorders due to defects in the lysosomal β-N-acetylhexosaminidase system. Accumulation of β-hexosaminidases A and B substrates is presumed to cause this fatal condition. An authentic mouse model of Sandhoff disease (SD) with pathological characteristics resembling those noted in infantile GM2 gangliosidosis has been described. We have shown that expression of β-hexosaminidase by intracranial delivery of recombinant adeno-associated viral vectors to young adult SD mice can prevent many features of the disease and extends lifespan. To investigate the nature of the neurological injury in GM2 gangliosidosis and the extent of its reversibility, we have examined the evolution of disease in the SD mouse; we have moreover explored the effects of gene transfer delivered at key times during the course of the illness. Here we report greatly increased survival only when the therapeutic genes are expressed either before the disease is apparent or during its early manifestations. However, irrespective of when treatment was administered, widespread and abundant expression of β-hexosaminidase with consequent clearance of glycoconjugates, α-synuclein and ubiquitinated proteins, and abrogation of inflammatory responses and neuronal loss was observed. We also show that defects in myelination occur in early life and cannot be easily resolved when treatment is given to the adult brain. These results indicate that there is a limited temporal opportunity in which function and survival can be improved-but regardless of resolution of the cardinal pathological features of GM2 gangliosidosis, a point is reached when functional deterioration and death cannot be prevented.
Collapse
Affiliation(s)
| | - Susan Z. Wang
- Department of Medicine, University of Cambridge, Cambridge, UK and
| | | | | | - Timothy M. Cox
- Department of Medicine, University of Cambridge, Cambridge, UK and
| |
Collapse
|
24
|
Naz H, Islam A, Waheed A, Sly WS, Ahmad F, Hassan MI. Humanβ-Glucuronidase: Structure, Function, and Application in Enzyme Replacement Therapy. Rejuvenation Res 2013; 16:352-63. [DOI: 10.1089/rej.2013.1407] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Affiliation(s)
- Huma Naz
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Asimul Islam
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Abdul Waheed
- The Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis, University School of Medicine, St. Louis, Missouri
| | - William S. Sly
- The Edward A. Doisy Department of Biochemistry and Molecular Biology, St. Louis, University School of Medicine, St. Louis, Missouri
| | - Faizan Ahmad
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| | - Md. Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, India
| |
Collapse
|
25
|
Biomaterial-Based Vectors for Targeted Delivery of Nucleic Acids to the Nervous System. DRUG DELIVERY SYSTEMS: ADVANCED TECHNOLOGIES POTENTIALLY APPLICABLE IN PERSONALISED TREATMENT 2013. [DOI: 10.1007/978-94-007-6010-3_7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
|
26
|
Tomanin R, Zanetti A, Zaccariotto E, D'Avanzo F, Bellettato CM, Scarpa M. Gene therapy approaches for lysosomal storage disorders, a good model for the treatment of mendelian diseases. Acta Paediatr 2012; 101:692-701. [PMID: 22428546 DOI: 10.1111/j.1651-2227.2012.02674.x] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
UNLABELLED This review describes the different gene therapy technologies applied to approach lysosomal storage disorders, monogenic conditions, with known genetic and biochemical defects, for many of which animal models are available. Both viral and nonviral procedures are described, underlying the specific needs that the treatment of genetic disorders requires. CONCLUSIONS Lysosomal storage disorders represent a good model of study of gene therapeutic procedures that are, or could be, relevant to the treatment of several other mendelian diseases.
Collapse
Affiliation(s)
- Rosella Tomanin
- Gene Therapy Laboratory, Department of Pediatrics, University of Padova, Italy
| | | | | | | | | | | |
Collapse
|
27
|
Abstract
Feline immunodeficiency virus (FIV)-based lentiviral vectors are useful for introducing integrated transgenes into nondividing human cells. This article describes the production and use of advanced generation FIV vectors. Key properties are discussed in comparison to other lentiviral vectors. Additional topics include the practical implications of species-specific retroviral restriction factors and the production of nonintegrating FIV vectors.
Collapse
|
28
|
Abstract
Mutations in the X-linked gene MECP2 (methyl CpG-binding protein 2) are the primary cause of the neurodevelopmental disorder RTT (Rett syndrome), and are also implicated in other neurological conditions. The expression product of this gene, MeCP2, is a widely expressed nuclear protein, especially abundant in mature neurons of the CNS (central nervous system). The major recognized consequences of MECP2 mutation occur in the CNS, but there is growing awareness of peripheral effects contributing to the full RTT phenotype. MeCP2 is classically considered to act as a DNA methylation-dependent transcriptional repressor, but may have additional roles in regulating gene expression and chromatin structure. Knocking out Mecp2 function in mice recapitulates many of the overt neurological features seen in RTT patients, and the characteristic postnatally delayed onset of symptoms is accompanied by aberrant neuronal morphology and deficits in synaptic physiology. Evidence that reactivation of endogenous Mecp2 in mutant mice, even at adult stages, can reverse aspects of RTT-like pathology and result in apparently functionally mature neurons has provided renewed hope for patients, but has also provoked discussion about traditional boundaries between neurodevelopmental disorders and those involving dysfunction at later stages. In the present paper we review the neurobiology of MeCP2 and consider the various genetic (including gene therapy), pharmacological and environmental interventions that have been, and could be, developed to attempt phenotypic rescue in RTT. Such approaches are already providing valuable insights into the potential tractability of RTT and related conditions, and are useful pointers for the development of future therapeutic strategies.
Collapse
|
29
|
Abstract
INTRODUCTION Lysosomal storage disorders (LSDs) encompass more than 50 distinct diseases, caused by defects in various aspects of lysosomal function. Neurodegeneration and/or dysmyelination are the hallmark of roughly 70% of LSDs. Gene therapy represents a promising approach for the treatment of CNS manifestations in LSDs, as it has the potential to provide a permanent source of the deficient enzyme, either by direct injection of vectors or by transplantation of gene-corrected cells. In this latter approach, the biology of neural stem/progenitor cells and hematopoietic cells might be exploited. AREAS COVERED Based on an extensive literature search up until March 2011, the author reviews and discusses the progress, the crucial aspects and the major challenges towards the development of novel gene therapy strategies aimed to target the CNS, with particular attention to direct intracerebral gene delivery and transplantation of neural stem/progenitor cells. EXPERT OPINION The implementation of viral vector delivery systems with specific tropism, regulated transgene expression, low immunogenicity and low genotoxic risk and the improvement in isolation and manipulation of relevant cell types to be transplanted, are fundamental challenges to the field. Also, combinatorial strategies might be required to achieve full correction in LSDs with neurological involvement.
Collapse
Affiliation(s)
- Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (HSR-TIGET), San Raffaele Scientific Institute, Via Olgettina 58, 20132, Milano, Italy.
| |
Collapse
|
30
|
Mingozzi F, High KA. Therapeutic in vivo gene transfer for genetic disease using AAV: progress and challenges. Nat Rev Genet 2011; 12:341-55. [PMID: 21499295 DOI: 10.1038/nrg2988] [Citation(s) in RCA: 693] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
In vivo gene replacement for the treatment of inherited disease is one of the most compelling concepts in modern medicine. Adeno-associated virus (AAV) vectors have been extensively used for this purpose and have shown therapeutic efficacy in a range of animal models. Successful translation to the clinic was initially slow, but long-term expression of donated genes at therapeutic levels has now been achieved in patients with inherited retinal disorders and haemophilia B. Recent exciting results have raised hopes for the treatment of many other diseases. As we discuss here, the prospects and challenges for AAV gene therapy are to a large extent dependent on the target tissue and the specific disease.
Collapse
Affiliation(s)
- Federico Mingozzi
- Center for Cellular and Molecular Therapeutics, The Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, 5th Floor CTRB, Philadelphia, Pennsylvania 19104, USA
| | | |
Collapse
|
31
|
Pan D. Cell- and gene-based therapeutic approaches for neurological deficits in mucopolysaccharidoses. Curr Pharm Biotechnol 2011; 12:884-96. [PMID: 21235445 PMCID: PMC4040261 DOI: 10.2174/138920111795542679] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2010] [Accepted: 07/15/2010] [Indexed: 11/22/2022]
Abstract
Mucopolysaccharidoses (MPS) are a group of lysosomal storage diseases that are resulted from abnormal accumulation of glycosaminoglycans. Among the progressive multi-organ abnormalities often associated with MPS diseases, the deterioration of central nervous system (CNS) is the most challenging manifestations to be tackled, due to the impermeability of the blood-brain-barrier (BBB). Evolved with recent development in stem cell biotechnology and gene therapy, several novel experimental approaches have been investigated in animal models. In this review, we will address different approaches attempting to bypass the BBB for neuropathic MPS treatment using cell- and gene-based therapies. Several neurological findings in CNS pathophysiology emerged with therapeutic investigation will also be discussed.
Collapse
Affiliation(s)
- Dao Pan
- Molecular and Gene Therapy Program, Division of Experimental Hematology, Cincinnati Children's Hospital Medical Center, OH 45229, USA.
| |
Collapse
|
32
|
Linterman KS, Palmer DN, Kay GW, Barry LA, Mitchell NL, McFarlane RG, Black MA, Sands MS, Hughes SM. Lentiviral-mediated gene transfer to the sheep brain: implications for gene therapy in Batten disease. Hum Gene Ther 2011; 22:1011-20. [PMID: 21595499 DOI: 10.1089/hum.2011.026] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The neuronal ceroid lipofuscinoses (NCLs; Batten disease) are inherited neurodegenerative lysosomal storage diseases with common clinical features of blindness and seizures culminating in premature death. Gene-therapy strategies for these diseases depend on whether the missing activity is a secreted lysosomal protein taken up by neighboring cells, or an intramembrane protein that requires careful targeting. Therapies are best developed in animal models with large complex human-like brains. Lentiviral-mediated gene delivery to neural cell cultures from normal sheep and sheep affected with an NCL resulted in green fluorescent protein (GFP) expression in neurons and neuroblasts, more efficiently than in astrocytes. Similar transgene expression was obtained from two constitutive promoters, the viral MND promoter and the human EF1α promoter. In vivo studies showed stable and persistent GFP expression throughout the cell bodies, axons, and dendrites from intracortical injections and indicated ependymal and subependymal transduction. The sheep showed no ill effects from the injections. These data support continuing gene-therapy trials in the sheep models of Batten disease.
Collapse
Affiliation(s)
- Kathryn S Linterman
- Department of Biochemistry, School of Medical Sciences, University of Otago, Dunedin 9054, New Zealand
| | | | | | | | | | | | | | | | | |
Collapse
|
33
|
ElAli A, Hermann DM. ATP-binding cassette transporters and their roles in protecting the brain. Neuroscientist 2011; 17:423-36. [PMID: 21518814 DOI: 10.1177/1073858410391270] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The blood-brain barrier is a network of endothelial cells that are tightly attached with each other via specialized cell-cell contacts. This passive diffusion barrier is complemented by ATP-binding cassette (ABC) transporters, which are localized on the surface of the endothelial cells. ABC transporters play important roles in the maintenance of blood-brain barrier integrity, as they carry a wide range of organic molecules, cell metabolites, and nutrients both out of the brain and into the brain. Recent studies have unraveled important roles of ABC transporters in the preservation of tissue homeostasis, pointing out the fact that ABC transporters protect both brain parenchymal cells and microvascular cells from injury. As such, ABC transporters have been involved in the pathogenesis of age-related neurodegenerative diseases, such as Parkinson and Alzheimer diseases, recently. This has led to the idea that neurodegenerative processes might be targeted by restoration of transport processes across the blood-brain barrier.
Collapse
Affiliation(s)
- Ayman ElAli
- Department of Neurology, University Hospital Essen, Essen, Germany
| | | |
Collapse
|
34
|
Lattanzi A, Neri M, Maderna C, di Girolamo I, Martino S, Orlacchio A, Amendola M, Naldini L, Gritti A. Widespread enzymatic correction of CNS tissues by a single intracerebral injection of therapeutic lentiviral vector in leukodystrophy mouse models. Hum Mol Genet 2010; 19:2208-27. [PMID: 20203170 DOI: 10.1093/hmg/ddq099] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Leukodystrophies are rare diseases caused by defects in the genes coding for lysosomal enzymes that degrade several glycosphingolipids. Gene therapy for leukodystrophies requires efficient distribution of the missing enzymes in CNS tissues to prevent demyelination and neurodegeneration. In this work, we targeted the external capsule (EC), a white matter region enriched in neuronal projections, with the aim of obtaining maximal protein distribution from a single injection site. We used bidirectional (bd) lentiviral vectors (LV) (bdLV) to ensure coordinate expression of a therapeutic gene (beta-galactocerebrosidase, GALC; arylsulfatase A, ARSA) and of a reporter gene, thus monitoring simultaneously transgene distribution and enzyme reconstitution. A single EC injection of bdLV.GALC in early symptomatic twitcher mice (a murine model of globoid cell leukodystrophy) resulted in rapid and robust expression of a functional GALC protein in the telencephalon, cerebellum, brainstem and spinal cord. This led to global rescue of enzymatic activity, significant reduction of tissue storage and decrease of activated astroglia and microglia. Widespread protein distribution and complete metabolic correction were also observed after EC injection of bdLV.ARSA in a mouse model of metachromatic leukodystrophy. Our data indicated axonal transport, distribution through cerebrospinal fluid flow and cross-correction as the mechanisms contributing to widespread bioavailability of GALC and ARSA proteins in CNS tissues. LV-mediated gene delivery of lysosomal enzymes by targeting highly interconnected CNS regions is a potentially effective strategy that, combined with a treatment able to target the PNS and peripheral organs, may provide significant therapeutic benefit to patients affected by leukodystrophies.
Collapse
Affiliation(s)
- Annalisa Lattanzi
- San Raffaele Scientific Institute, Telethon Institute for Gene Therapy (HSR-TIGET), Via Olgettina 58, 20132 Milano, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Gateway to the diseased brain. Nat Med 2009; 15:1123-4. [PMID: 19812565 DOI: 10.1038/nm1009-1123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
36
|
Lin LH, Langasek JE, Talman LS, Taktakishvili OM, Talman WT. Feline immunodeficiency virus as a gene transfer vector in the rat nucleus tractus solitarii. Cell Mol Neurobiol 2009; 30:339-46. [PMID: 19777342 DOI: 10.1007/s10571-009-9456-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2009] [Accepted: 09/09/2009] [Indexed: 01/25/2023]
Abstract
Gene transfer has been used to examine the role of putative neurotransmitters in the nucleus tractus solitarii (NTS). Most such studies used adenovirus vector-mediated gene transfer although adenovirus vector transfects both neuronal and non-neuronal cells. Successful transfection in the NTS has also been reported with lentivirus as the vector. Feline immunodeficiency virus (FIV), a lentivirus, may preferentially transfect neurons and could be a powerful tool to delineate physiological effects produced by altered synthesis of transmitters in neurons. However, it has not been studied in NTS. Therefore, we sought to determine whether FIV transfects rat NTS cells and to define the type of cell transfected. We found that injection of FIV encoding LacZ gene (FIVLacZ) into the NTS led to transfection of numerous NTS cells. Injection of FIVLacZ did not alter immunoreactivity (IR) for neuronal nitric oxide synthase, which we have shown resides in NTS neurons. A majority (91.7 +/- 3.9%) of transfected cells contained IR for neuronal nuclear antigen, a neuronal marker; 2.1 +/- 3.8% of transfected cells contained IR for glial fibrillary acidic protein, a glial marker. No transfected neurons or fibers were observed in the nodose ganglion, which sends afferents to the NTS. We conclude that FIV almost exclusively transfects neurons in the rat NTS from which it is not retrogradely transported. The cell-type specificity of FIV in the NTS may provide a molecular method to study local physiological functions mediated by potential neurotransmitters in the NTS.
Collapse
Affiliation(s)
- L H Lin
- Department of Neurology, 1191 ML, University of Iowa, Iowa City, IA 52242, USA.
| | | | | | | | | |
Collapse
|
37
|
Macauley SL, Sands MS. Promising CNS-directed enzyme replacement therapy for lysosomal storage diseases. Exp Neurol 2009; 218:5-8. [PMID: 19361502 PMCID: PMC2701189 DOI: 10.1016/j.expneurol.2009.03.040] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2009] [Revised: 03/27/2009] [Accepted: 03/31/2009] [Indexed: 11/19/2022]
Affiliation(s)
- Shannon L. Macauley
- Washington University School of Medicine, Departments of Internal Medicine and Genetics, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | - Mark S. Sands
- Washington University School of Medicine, Departments of Internal Medicine and Genetics, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| |
Collapse
|
38
|
The trigeminal retrograde transfer pathway in the treatment of neurodegeneration. J Neuroimmunol 2009; 209:139-42. [DOI: 10.1016/j.jneuroim.2009.02.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 02/04/2009] [Accepted: 02/05/2009] [Indexed: 11/22/2022]
|
39
|
Abstract
he development of therapeutic interventions for genetic disorders and diseases that affect the central nervous system (CNS) has proven challenging. There has been significant progress in the development of gene therapy strategies in murine models of human disease, but gene therapy outcomes in these models do not always translate to the human setting. Therefore, large animal models are crucial to the development of diagnostics, treatments, and eventual cures for debilitating neurological disorders. This review focuses on the description of large animal models of neurological diseases such as lysosomal storage diseases, Parkinsons disease, Huntingtons disease, and neuroAIDS. The review also describes the contributions of these models to progress in gene therapy research.
Collapse
|
40
|
Kilic E, Spudich A, Kilic U, Rentsch KM, Vig R, Matter CM, Wunderli-Allenspach H, Fritschy JM, Bassetti CL, Hermann DM. ABCC1: a gateway for pharmacological compounds to the ischaemic brain. Brain 2008; 131:2679-89. [PMID: 18796513 DOI: 10.1093/brain/awn222] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
By preventing access of drugs to the CNS, the blood-brain barrier hampers developments in brain pharmacotherapy. Strong efforts are currently being made to identify drugs that accumulate more efficaciously in ischaemic brain tissue. We identified an ATP-binding cassette (ABC) transporter, ABCC1, which is expressed on the abluminal surface of the brain capillary endothelium and mildly downregulated in response to focal cerebral ischaemia, induced by intraluminal middle cerebral artery occlusion. In biodistribution studies we show that ABCC1 promotes the accumulation of known neuroprotective and neurotoxic compounds in the ischaemic and non-ischaemic brain, ABCC1 deactivation reducing tissue concentrations by up to two orders of magnitude. As such, ABCC1's expression and functionality in the brain differs from the liver, spleen and testis, where ABCC1 is strongly expressed on parenchymal cells, resulting -- in case of liver and testis -- in directed transport from the tissue into the blood. After focal cerebral ischaemia, ABCC1 deactivation abolished the efficacy of both neuroprotective and neurotoxic compounds. Our data indicate that ABCC1 acts as gateway for pharmacological compounds to the stroke brain. We suggest that the tailoring of compounds binding to abluminal but not luminal ABC transporters may facilitate stroke pharmacotherapy.
Collapse
Affiliation(s)
- Ertugrul Kilic
- Department of Neurology, University Hospital Zurich, Zurich, Switzerland
| | | | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Nasr P, Sullivan PG, Smith GM. Mitochondrial imaging in dorsal root ganglion neurons following the application of inducible adenoviral vector expressing two fluorescent proteins. J Neurosci Methods 2008; 172:185-94. [PMID: 18541307 PMCID: PMC2657596 DOI: 10.1016/j.jneumeth.2008.04.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 04/06/2008] [Accepted: 04/22/2008] [Indexed: 01/31/2023]
Abstract
Mitochondrial morphology and dynamics are known to vary considerably depending on the cell type and organism studied. The objective of this study was to assess the potential application of adenoviral-fluorescent protein constructs for long-term tracking of mitochondria in neurons. An adenoviral vector containing two fluorescent proteins, the enhanced green fluorescent protein (eGFP) targeted to the cytoplasm to highlight the neuronal processes, and the red fluorescent protein (RFP) directed to mitochondria under the control of an inducible promoter, facilitated an efficient and accurate method to study mitochondrial dynamics in long-term studies. Dorsal root ganglion neurons from rat embryos were cultured and infected. The infected neurons exhibited green fluorescence after 24h, while 16 h following induction with doxycycline, red fluorescence protein began to localize within mitochondria. The red fluorescent protein was transported into mitochondria at the cell body followed by distribution within processes. As the neurons aged, the expression of red fluorescent protein was confined to cytoplasmic vacuoles and not mitochondria. Further analysis suggested that the cytoplasmic vacuoles were likely of lysosomal origin. Taken together, the current study presents novel strategies to study the life history of cellular organelles such as mitochondria in long-term studies.
Collapse
Affiliation(s)
- Payman Nasr
- Department of Biological Sciences, Kent State University, Ashtabula, OH 44004, United States.
| | | | | |
Collapse
|
42
|
Abstract
Lentiviral vectors are useful for gene transfer to dividing and nondividing cells. Feline immunodeficiency virus (FIV) vectors transduce most human cell types with good efficiency and may have advantages for clinical gene therapy applications. This article reviews significant progress in the development and refinement of FIV vector systems.
Collapse
Affiliation(s)
- Román A Barraza
- Molecular Medicine Program, Guggenheim 18, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, MN 55905, United States.
| | | |
Collapse
|
43
|
Abstract
UNLABELLED Lysosomal storage diseases (LSDs) are a group of inherited metabolic disorders usually caused by deficient activity of a single lysosomal enzyme. As most lysosomal enzymes are ubiquitously expressed, a deficiency in a single enzyme can affect multiple organ systems, including the central nervous system (CNS). At least 75% of all LSDs have a significant CNS component. Approaches such as bone marrow transplantation (BMT) or enzyme replacement therapy (ERT) can effectively treat the systemic disease associated with LSDs in some patients. However, CNS disease remains a major challenge. Gene therapy represents a promising approach for the treatment of CNS disease as it has the potential to provide a permanent source of the deficient enzyme. Direct intracranial injection of viral gene transfer vectors has resulted in reduced lysosomal storage and functional improvement in certain small (rodent) and large (canine and feline) animal models of LSDs. The addition of protein transduction domains (PTDs) to the recombinant enzymes increased the distribution of enzyme and the extent of correction. Therapeutic levels of lysosomal enzymes can also be delivered to distant sites in the brain by anterograde and retrograde axonal transport. Finally, combining disparate approaches such as BMT and CNS-directed gene therapy can increase treatment efficacy in LSDs with severe CNS disease that are refractory to more conventional approaches. CONCLUSION The development of gene transfer vectors that mediate persistent expression in vivo, the addition of PTDs, a better understanding of lysosomal enzyme trafficking and combining different therapies provide hope that the CNS component of LSDs can be effectively treated.
Collapse
Affiliation(s)
- Mark S Sands
- Department of Internal Medicine and Department of Genetics, Washington University School of Medicine, St. Louis, MO, USA.
| | | |
Collapse
|
44
|
Berges BK, Wolfe JH, Fraser NW. Transduction of brain by herpes simplex virus vectors. Mol Ther 2008; 15:20-9. [PMID: 17164771 DOI: 10.1038/sj.mt.6300018] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
An imposing obstacle to gene therapy is the inability to transduce all of the necessary cells in a target organ. This certainly applies to gene transfer to the brain, especially when one considers the challenges involved in scaling up transduction from animal models to use in the clinic. Non-neurotropic viral gene transfer vectors (e.g., adenovirus, adeno-associated virus, and lentivirus) do not spread very far in the nervous system, and consequently these vectors transduce brain regions mostly near the injection site in adult animals. This indicates that numerous, well-spaced injections would be required to achieve widespread transduction in a large brain with these vectors. In contrast, herpes simplex virus type 1 (HSV-1) is a promising vector for widespread gene transfer to the brain owing to the innate ability of the virus to spread through the nervous system and form latent infections in neurons that last for the lifetime of the infected individual. In this review, we summarize the published literature of the transduction patterns produced by attenuated HSV-1 vectors in small animals as a function of the injection site, and discuss the implications of the distribution for widespread gene transfer to the large animal brain.
Collapse
Affiliation(s)
- Bradford K Berges
- Department of Microbiology, School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
45
|
Roberts ALK, Rees MH, Klebe S, Fletcher JM, Byers S. Improvement in behaviour after substrate deprivation therapy with rhodamine B in a mouse model of MPS IIIA. Mol Genet Metab 2007; 92:115-21. [PMID: 17681480 DOI: 10.1016/j.ymgme.2007.06.016] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2007] [Revised: 06/13/2007] [Accepted: 06/13/2007] [Indexed: 11/17/2022]
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA) is a specific lysosomal storage disorder caused by an enzyme deficiency in sulphamidase, which is required for the degradation of heparan sulphate glycosaminoglycan (gag). This deficiency results in widespread gag storage and leads to severe CNS degeneration and mild somatic pathology. We have developed substrate deprivation as a therapy (SDT) for MPS disorders to reduce the initial production of gag substrate for the deficient enzyme, using the compound rhodamine B as an inhibitor of gag biosynthesis. This should restore the balance between gag level and residual enzyme activity towards normal and improve patient outcome. To determine if SDT improved CNS function, MPS IIIA mice were treated for 6months with weekly, intravenous 1mg/kg rhodamine B and then tested in a 4-arm water cross maze, which measures spatial learning and memory. MPS IIIA untreated mice were unable to perform to the same level as normal littermates, having increased escape latency, increased incorrect entries and decreased correct entries. Rhodamine B treatment improved MPS IIIA performance towards normal with treated mice having decreased escape latency, decreased incorrect entries and increased correct entries when compared to MPS IIIA untreated littermates. This provides the first report of SDT resulting in a beneficial effect on CNS function in an MPS disorder and SDT targeting gag synthesis may be a viable treatment option for children with MPS.
Collapse
Affiliation(s)
- Ainslie L K Roberts
- Department of Genetic Medicine, Children, Youth and Women's Health Service, North Adelaide, Australia
| | | | | | | | | |
Collapse
|
46
|
Abstract
Mucopolysaccharidoses (MPS) are due to deficiencies in activities of lysosomal enzymes that degrade glycosaminoglycans. Some attempts at gene therapy for MPS in animal models have involved intravenous injection of vectors derived from an adeno-associated virus (AAV), adenovirus, retrovirus or a plasmid, which primarily results in expression in liver and secretion of the relevant enzyme into blood. Most vectors can correct disease in liver and spleen, although correction in other organs including the brain requires high enzyme activity in the blood. Alternative approaches are to transduce hematopoietic stem cells, or to inject a vector locally into difficult-to-reach sites such as the brain. Gene therapy holds great promise for providing a long-lasting therapeutic effect for MPS if safety issues can be resolved.
Collapse
Affiliation(s)
- Katherine P Ponder
- Washington University School of Medicine, Department of Internal Medicine, 660 South Euclid Avenue, St. Louis, MO 63110, USA.
| | | |
Collapse
|
47
|
Biffi A, Naldini L. Novel candidate disease for gene therapy: metachromatic leukodystrophy. Expert Opin Biol Ther 2007; 7:1193-205. [PMID: 17696818 DOI: 10.1517/14712598.7.8.1193] [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] [Indexed: 12/17/2022]
Abstract
Metachromatic leukodystrophy (MLD) is a rare, fatal, inherited, autosomal recessive, lysosomal storage disorder, characterized by severe and progressive demyelination affecting the central and peripheral nervous systems. Despite some initial expectations in hematopoietic stem cell transplantation, and despite the ameliorated supportive therapy, MLD remains a life-threatening disease, with an extremely poor quality of life and a severe prognosis for all affected patients. Prospectively, in children affected by MLD, who have no other therapeutic option and an extremely poor prognosis, the potential risks associated with the use of a novel technology, such as gene therapy, might be well balanced by the potential benefit of a positive outcome. Thus, MLD might be considered an optimal candidate disease for testing innovative and potentially efficacious therapeutic approaches. Some of the gene therapy approaches discussed here, such as hematopoietic stem cells gene therapy, are likely to enter clinical testing in the near future.
Collapse
Affiliation(s)
- Alessandra Biffi
- San Raffaele Telethon Institute for Gene Therapy and Vita Salute University, H. San Raffaele Scientific Institute, Milan, Italy. a.biffi @hsr.it
| | | |
Collapse
|
48
|
Liu G, Chen YH, He X, Martins I, Heth JA, Chiorini JA, Davidson BL. Adeno-associated virus type 5 reduces learning deficits and restores glutamate receptor subunit levels in MPS VII mice CNS. Mol Ther 2007; 15:242-7. [PMID: 17235300 DOI: 10.1038/sj.mt.6300016] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
A major challenge in treating lysosomal storage diseases with enzyme therapy is correcting symptoms in the central nervous system (CNS). This study used a murine model of mucopolysaccharidosis type VII (MPS VII) to test whether pathological and functional CNS defects could be corrected by expressing beta-glucuronidase via bilateral intrastriatal injection of adeno-associated virus type 5 (AAV5betagluc) vectors. After injecting AAV5betagluc, different brain regions expressed active beta-glucuronidase, which corrected lysosomal storage defects. Compared to age-matched littermates, adult MPS VII mice were impaired in spatial learning and memory, as measured by the repeated acquisition and performance chamber (RAPC) assay. AAV5betagluc-treated MPS VII mice improved significantly in the RAPC assay, relative to saline-injected littermates. Moreover, our studies reveal that cognitive changes in MPS VII mice correlate with decreased N-methyl-d-aspartate and alpha-amino-3-hydroxy-5-methyl-isoxazole-4-propionic acid receptor expression. Importantly, AAV5betagluc delivery restored glutamate receptor levels. Together, these data demonstrate that AAV5 vectors deliver a therapeutically effective beta-glucuronidase gene to the CNS and further suggest a possible mechanism underlying spatial learning defects in MPS VII mice.
Collapse
Affiliation(s)
- Gumei Liu
- Program in Gene Therapy, Department of Internal Medicine, University of Iowa, Iowa City, Iowa 52242, USA
| | | | | | | | | | | | | |
Collapse
|
49
|
Anson DS, McIntyre C, Thomas B, Koldej R, Ranieri E, Roberts A, Clements PR, Dunning K, Byers S. Lentiviral-mediated gene correction of mucopolysaccharidosis type IIIA. GENETIC VACCINES AND THERAPY 2007; 5:1. [PMID: 17227588 PMCID: PMC1783652 DOI: 10.1186/1479-0556-5-1] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2006] [Accepted: 01/16/2007] [Indexed: 11/10/2022]
Abstract
BACKGROUND Mucopolysaccharidosis type IIIA (MPS IIIA) is the most common of the mucopolysaccharidoses. The disease is caused by a deficiency of the lysosomal enzyme sulphamidase and results in the storage of the glycosaminoglycan (GAG), heparan sulphate. MPS IIIA is characterised by widespread storage and urinary excretion of heparan sulphate, and a progressive and eventually profound neurological course. Gene therapy is one of the few avenues of treatment that hold promise of a sustainable treatment for this disorder. METHODS The murine sulphamidase gene cDNA was cloned into a lentiviral vector and high-titre virus produced. Human MPS IIIA fibroblast cultures were transduced with the sulphamidase vector and analysed using molecular, enzymatic and metabolic assays. High-titre virus was intravenously injected into six 5-week old MPS IIIA mice. Three of these mice were pre-treated with hyperosmotic mannitol. The weight of animals was monitored and GAG content in urine samples was analysed by polyacrylamide gel electrophoresis. RESULTS Transduction of cultured MPS IIIA fibroblasts with the sulphamidase gene corrected both the enzymatic and metabolic defects. Sulphamidase secreted by gene-corrected cells was able to cross correct untransduced MPS IIIA cells. Urinary GAG was found to be greatly reduced in samples from mice receiving the vector compared to untreated MPS IIIA controls. In addition, the weight of treated mice became progressively normalised over the 6-months post-treatment. CONCLUSION Lentiviral vectors appear promising vehicles for the development of gene therapy for MPS IIIA.
Collapse
Affiliation(s)
- Donald S Anson
- Department of Genetic Medicine, Women's and Children's Hospital, Children, Youth and Women's Health Service, 72 King William Road, North Adelaide, SA 5006, Australia
- Department of Paediatrics, University of Adelaide, SA 5005, Australia
- Department of Biotechnology, Flinders University, GPO Box 2100, Adelaide, SA 5001, Australia
- School of Pharmacy & Medical Sciences, University of South Australia, GPO Box 2471, Adelaide, SA 5001, Australia
| | - Chantelle McIntyre
- Department of Genetic Medicine, Women's and Children's Hospital, Children, Youth and Women's Health Service, 72 King William Road, North Adelaide, SA 5006, Australia
- Department of Paediatrics, University of Adelaide, SA 5005, Australia
| | - Belinda Thomas
- Department of Genetic Medicine, Women's and Children's Hospital, Children, Youth and Women's Health Service, 72 King William Road, North Adelaide, SA 5006, Australia
- Department of Respiratory and Sleep Medicine, Monash Medical Centre, VIC 3168, Australia
| | - Rachel Koldej
- Department of Genetic Medicine, Women's and Children's Hospital, Children, Youth and Women's Health Service, 72 King William Road, North Adelaide, SA 5006, Australia
- Department of Paediatrics, University of Adelaide, SA 5005, Australia
| | - Enzo Ranieri
- Department of Genetic Medicine, Women's and Children's Hospital, Children, Youth and Women's Health Service, 72 King William Road, North Adelaide, SA 5006, Australia
- Department of Paediatrics, University of Adelaide, SA 5005, Australia
| | - Ainslie Roberts
- Department of Genetic Medicine, Women's and Children's Hospital, Children, Youth and Women's Health Service, 72 King William Road, North Adelaide, SA 5006, Australia
- Department of Paediatrics, University of Adelaide, SA 5005, Australia
| | - Peter R Clements
- Department of Genetic Medicine, Women's and Children's Hospital, Children, Youth and Women's Health Service, 72 King William Road, North Adelaide, SA 5006, Australia
- Department of Paediatrics, University of Adelaide, SA 5005, Australia
| | - Kylie Dunning
- Department of Genetic Medicine, Women's and Children's Hospital, Children, Youth and Women's Health Service, 72 King William Road, North Adelaide, SA 5006, Australia
- Department of Obstetrics and Gynaecology, University of Adelaide, SA 5005, Australia
| | - Sharon Byers
- Department of Genetic Medicine, Women's and Children's Hospital, Children, Youth and Women's Health Service, 72 King William Road, North Adelaide, SA 5006, Australia
- Department of Paediatrics, University of Adelaide, SA 5005, Australia
| |
Collapse
|
50
|
Beck M. New therapeutic options for lysosomal storage disorders: enzyme replacement, small molecules and gene therapy. Hum Genet 2006; 121:1-22. [PMID: 17089160 DOI: 10.1007/s00439-006-0280-4] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2006] [Accepted: 10/08/2006] [Indexed: 10/24/2022]
Abstract
During the last few years, much progress has been made in the treatment of lysosomal storage disorders. In the past, no specific therapy was available for the affected patients, and management consisted solely of supportive care and treatment of complications. Since enzyme replacement therapy has been successfully introduced for patients with Gaucher disease, this principle of treatment has been taken into consideration for other lysosomal storage disorders as well. Clinical trials could demonstrate the clinical benefit of this therapeutic principle in Fabry disease, mucopolysaccharidoses type I, II and VI and in Pompe disease. However, the usefulness of enzyme replacement therapy is limited due to the fact that a given enzyme preparation does not have beneficial effects on all aspects of a disorder in the same degree. Additionally, clinical studies have shown that many symptoms of a lysosomal storage disorder even after long-term treatment are no more reversible. A further novel therapeutic option for lysosomal storage disorders consists of the application of small molecules that either inhibit a key enzyme which is responsible for substrate synthesis (substrate deprivation) or act as a chaperone to increase the residual activity of the lysosomal enzyme (enzyme enhancing therapy). Various gene therapeutic techniques (in vivo and ex vivo technique) have been developed in order to administer the gene that is defective in a patient to the bloodstream or directly to the brain in order to overcome the blood-brain barrier. This review will give an insight into these newly developed therapeutic strategies and will discuss their advantages and limitations.
Collapse
Affiliation(s)
- Michael Beck
- Children Hospital, University of Mainz, Langenbeckstrasse 1, 55101 Mainz, Germany.
| |
Collapse
|