1
|
Chaudhary R, Rehman M, Agarwal V, Kumar A, Kaushik AS, Srivastava S, Srivastava S, Verma R, Rajinikanth PS, Mishra V. Terra incognita of glial cell dynamics in the etiology of leukodystrophies: Broadening disease and therapeutic perspectives. Life Sci 2024; 354:122953. [PMID: 39122110 DOI: 10.1016/j.lfs.2024.122953] [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: 02/05/2024] [Revised: 07/09/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Neuroglial cells, also known as glia, are primarily characterized as auxiliary cells within the central nervous system (CNS). The recent findings have shed light on their significance in numerous physiological processes and their involvement in various neurological disorders. Leukodystrophies encompass an array of rare and hereditary neurodegenerative conditions that were initially characterized by the deficiency, aberration, or degradation of myelin sheath within CNS. The primary cellular populations that experience significant alterations are astrocytes, oligodendrocytes and microglia. These glial cells are either structurally or metabolically impaired due to inherent cellular dysfunction. Alternatively, they may fall victim to the accumulation of harmful by-products resulting from metabolic disturbances. In either situation, the possible replacement of glial cells through the utilization of implanted tissue or stem cell-derived human neural or glial progenitor cells hold great promise as a therapeutic strategy for both the restoration of structural integrity through remyelination and the amelioration of metabolic deficiencies. Various emerging treatment strategies like stem cell therapy, ex-vivo gene therapy, infusion of adeno-associated virus vectors, emerging RNA-based therapies as well as long-term therapies have demonstrated success in pre-clinical studies and show promise for rapid clinical translation. Here, we addressed various leukodystrophies in a comprehensive and detailed manner as well as provide prospective therapeutic interventions that are being considered for clinical trials. Further, we aim to emphasize the crucial role of different glial cells in the pathogenesis of leukodystrophies. By doing so, we hope to advance our understanding of the disease, elucidate underlying mechanisms, and facilitate the development of potential treatment interventions.
Collapse
Affiliation(s)
- Rishabh Chaudhary
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Mujeeba Rehman
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vipul Agarwal
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Anand Kumar
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Arjun Singh Kaushik
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Siddhi Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Sukriti Srivastava
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Rajkumar Verma
- University of Connecticut School of Medicine, 200 Academic Way, Farmington, CT 06032, USA
| | - P S Rajinikanth
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India
| | - Vikas Mishra
- Department of Pharmaceutical Sciences, Babasaheb Bhimrao Ambedkar University, Vidya Vihar, Raebareli Road, Lucknow 226025, U.P., India.
| |
Collapse
|
2
|
Bido S, Nannoni M, Muggeo S, Gambarè D, Ruffini G, Bellini E, Passeri L, Iaia S, Luoni M, Provinciali M, Giannelli SG, Giannese F, Lazarevic D, Gregori S, Broccoli V. Microglia-specific IL-10 gene delivery inhibits neuroinflammation and neurodegeneration in a mouse model of Parkinson's disease. Sci Transl Med 2024; 16:eadm8563. [PMID: 39167665 DOI: 10.1126/scitranslmed.adm8563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 04/23/2024] [Accepted: 07/31/2024] [Indexed: 08/23/2024]
Abstract
Neuroinflammation plays a key role in exacerbating dopaminergic neuron (DAN) loss in Parkinson's disease (PD). However, it remains unresolved how to effectively normalize this immune response given the complex interplay between the innate and adaptive immune responses occurring within a scarcely accessible organ like the brain. In this study, we uncovered a consistent correlation between neuroinflammation, brain parenchymal lymphocytes, and DAN loss among several commonly used mouse models of PD generated by a variety of pathological triggers. We validated a viral therapeutic approach for the microglia-specific expression of interleukin 10 (IL-10) to selectively mitigate the excessive inflammatory response. We found that this approach induced a local nigral IL-10 release that alleviated DAN loss in mice overexpressing the human SNCA gene in the substantia nigra. Single-cell transcriptomics revealed that IL-10 induced the emergence of a molecularly distinct microglial cell state, enriched in markers of cell activation with enhanced expression of prophagocytic pathways. IL-10 promoted microglial phagocytotic and clearance activities in vitro and reduced αSYN aggregate burden in the nigral area in mice overexpressing SNCA. Furthermore, IL-10 stimulated the differentiation of CD4+ T lymphocytes into active T regulatory cells and promoted inhibitory characteristics in CD8+ T cells. In summary, our results show that local and microglia-specific IL-10 transduction elicited strong immunomodulation in the nigral tissue with enhanced suppression of lymphocyte toxicity that was associated with DAN survival. These results offer insights into the therapeutic benefits of IL-10 and showcase a promising gene delivery approach that could minimize undesired side effects.
Collapse
Affiliation(s)
- Simone Bido
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Melania Nannoni
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Sharon Muggeo
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Diana Gambarè
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Giorgia Ruffini
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Edoardo Bellini
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Laura Passeri
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Silvia Iaia
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mirko Luoni
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- CNR Institute of Neuroscience, 20129 Milan, Italy
| | - Martino Provinciali
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Serena Gea Giannelli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Francesca Giannese
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Dejan Lazarevic
- Center for Omics Sciences, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Silvia Gregori
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, IRCCS San Raffaele Scientific Institute, 20132 Milan, Italy
- CNR Institute of Neuroscience, 20129 Milan, Italy
| |
Collapse
|
3
|
Ingusci S, Hall BL, Goins WF, Cohen JB, Glorioso JC. Viral vectors for gene delivery to the central nervous system. HANDBOOK OF CLINICAL NEUROLOGY 2024; 205:59-81. [PMID: 39341663 DOI: 10.1016/b978-0-323-90120-8.00001-0] [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: 10/01/2024]
Abstract
Brain diseases with a known or suspected genetic basis represent an important frontier for advanced therapeutics. The central nervous system (CNS) is an intricate network in which diverse cell types with multiple functions communicate via complex signaling pathways, making therapeutic intervention in brain-related diseases challenging. Nevertheless, as more information on the molecular genetics of brain-related diseases becomes available, genetic intervention using gene therapeutic strategies should become more feasible. There remain, however, several significant hurdles to overcome that relate to (i) the development of appropriate gene vectors and (ii) methods to achieve local or broad vector delivery. Clearly, gene delivery tools must be engineered for distribution to the correct cell type in a specific brain region and to accomplish therapeutic transgene expression at an appropriate level and duration. They also must avoid all toxicity, including the induction of inflammatory responses. Over the last 40 years, various types of viral vectors have been developed as tools to introduce therapeutic genes into the brain, primarily targeting neurons. This review describes the most prominent vector systems currently approaching clinical application for CNS disorders and highlights both remaining challenges as well as improvements in vector designs that achieve greater safety, defined tropism, and therapeutic gene expression.
Collapse
Affiliation(s)
- Selene Ingusci
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Bonnie L Hall
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - William F Goins
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Justus B Cohen
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States
| | - Joseph C Glorioso
- Department of Microbiology and Molecular Genetics, University of Pittsburgh, Pittsburgh, PA, United States.
| |
Collapse
|
4
|
Moore TL, Pannuzzo G, Costabile G, Palange AL, Spanò R, Ferreira M, Graziano ACE, Decuzzi P, Cardile V. Nanomedicines to treat rare neurological disorders: The case of Krabbe disease. Adv Drug Deliv Rev 2023; 203:115132. [PMID: 37918668 DOI: 10.1016/j.addr.2023.115132] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 10/26/2023] [Accepted: 10/29/2023] [Indexed: 11/04/2023]
Abstract
The brain remains one of the most challenging therapeutic targets due to the low and selective permeability of the blood-brain barrier and complex architecture of the brain tissue. Nanomedicines, despite their relatively large size compared to small molecules and nucleic acids, are being heavily investigated as vehicles to delivery therapeutics into the brain. Here we elaborate on how nanomedicines may be used to treat rare neurodevelopmental disorders, using Krabbe disease (globoid cell leukodystrophy) to frame the discussion. As a monogenetic disorder and lysosomal storage disease affecting the nervous system, the lessons learned from examining nanoparticle delivery to the brain in the context of Krabbe disease can have a broader impact on the treatment of various other neurodevelopmental and neurodegenerative disorders. In this review, we introduce the epidemiology and genetic basis of Krabbe disease, discuss current in vitro and in vivo models of the disease, as well as current therapeutic approaches either approved or at different stage of clinical developments. We then elaborate on challenges in particle delivery to the brain, with a specific emphasis on methods to transport nanomedicines across the blood-brain barrier. We highlight nanoparticles for delivering therapeutics for the treatment of lysosomal storage diseases, classified by the therapeutic payload, including gene therapy, enzyme replacement therapy, and small molecule delivery. Finally, we provide some useful hints on the design of nanomedicines for the treatment of rare neurological disorders.
Collapse
Affiliation(s)
- Thomas Lee Moore
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy.
| | - Giovanna Pannuzzo
- Department of Biomedical and Biotechnological Sciences, Università di Catania, Catania 95123, CT, Italy
| | - Gabriella Costabile
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy; Department of Pharmacy, Università degli Studi di Napoli Federico II, Naples 80131, NA, Italy
| | - Anna Lisa Palange
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy
| | - Raffaele Spanò
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy
| | - Miguel Ferreira
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy
| | - Adriana Carol Eleonora Graziano
- Department of Biomedical and Biotechnological Sciences, Università di Catania, Catania 95123, CT, Italy; Facolta di Medicina e Chirurgia, Università degli Studi di Enna "Kore", Enna 94100, EN, Italy
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Istituto Italiano di Tecnologia, Genoa 16163, GE, Italy
| | - Venera Cardile
- Department of Biomedical and Biotechnological Sciences, Università di Catania, Catania 95123, CT, Italy.
| |
Collapse
|
5
|
Nakamura S, Morohoshi K, Inada E, Sato Y, Watanabe S, Saitoh I, Sato M. Recent Advances in In Vivo Somatic Cell Gene Modification in Newborn Pups. Int J Mol Sci 2023; 24:15301. [PMID: 37894981 PMCID: PMC10607593 DOI: 10.3390/ijms242015301] [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: 08/31/2023] [Revised: 10/12/2023] [Accepted: 10/16/2023] [Indexed: 10/29/2023] Open
Abstract
Germline manipulation at the zygote stage using the CRISPR/Cas9 system has been extensively employed for creating genetically modified animals and maintaining established lines. However, this approach requires a long and laborious task. Recently, many researchers have attempted to overcome these limitations by generating somatic mutations in the adult stage through tail vein injection or local administration of CRISPR reagents, as a new strategy called "in vivo somatic cell genome editing". This approach does not require manipulation of early embryos or strain maintenance, and it can test the results of genome editing in a short period. The newborn is an ideal stage to perform in vivo somatic cell genome editing because it is immune-privileged, easily accessible, and only a small amount of CRISPR reagents is required to achieve somatic cell genome editing throughout the entire body, owing to its small size. In this review, we summarize in vivo genome engineering strategies that have been successfully demonstrated in newborns. We also report successful in vivo genome editing through the neonatal introduction of genome editing reagents into various sites in newborns (as exemplified by intravenous injection via the facial vein), which will be helpful for creating models for genetic diseases or treating many genetic diseases.
Collapse
Affiliation(s)
- Shingo Nakamura
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Tokorozawa 359-8513, Japan;
| | - Kazunori Morohoshi
- Division of Biomedical Engineering, National Defense Medical College Research Institute, Tokorozawa 359-8513, Japan;
| | - Emi Inada
- Department of Pediatric Dentistry, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima 890-8544, Japan;
| | - Yoko Sato
- Graduate School of Public Health, Shizuoka Graduate University of Public Health, Aoi-ku, Shizuoka 420-0881, Japan;
| | - Satoshi Watanabe
- Institute of Livestock and Grassland Science, NARO, Tsukuba 305-0901, Japan;
| | - Issei Saitoh
- Department of Pediatric Dentistry, Asahi University School of Dentistry, Mizuho 501-0296, Japan;
| | - Masahiro Sato
- Department of Genome Medicine, National Center for Child Health and Development, Setagaya-ku, Tokyo 157-8535, Japan;
| |
Collapse
|
6
|
Intracerebral lentiviral ABCD1 gene therapy in an early disease onset ALD mouse model. Gene Ther 2023; 30:18-30. [PMID: 35790794 DOI: 10.1038/s41434-022-00355-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 05/06/2022] [Accepted: 05/30/2022] [Indexed: 02/17/2023]
Abstract
X-linked adrenoleukodystrophy (ALD) is a genetic disorder of the ABCD1 gene. We aimed to treat ALD via direct intracerebral injection of lentiviral ABCD1 (LV.ABCD1). Lentiviral vectors (LVs) were injected into the brain of wild type mice to access toxicities and biodistribution. Confocal microscopy illustrated supraphysiological ABCD1 expression surrounding the injection sites, and LVs were also detected in the opposite site of the unilaterally injected brain. In multi-site bilateral injections (4, 6, 8, and 9 sites), LV.ABCD1 transduced most brain regions including the cerebellum. Investigation of neuronal loss, astrogliosis and microglia activation did not detect abnormality. For efficacy evaluation, a novel ALD knockout (KO) mouse model was established by deleting exons 3 to 9 of the ABCD1 gene based on CRISPR/Cas9 gene editing. The KO mice showed behavioral deficit in open-field test (OFT) and reduced locomotor activities in rotarod test at 6 and 7 months of age, respectively. We treated 3-month-old KO mice with bilateral LV.ABCD1 injections into the external capsule and thalamus. ABCD1 expression was detected 15 days later, and the impaired motor ability was gradually alleviated. Our studies established an early onset ALD model and illustrated neurological improvement after LV.ABCD1 intracerebral injection without immunopathological toxicity.
Collapse
|
7
|
Sala D, Ornaghi F, Morena F, Argentati C, Valsecchi M, Alberizzi V, Di Guardo R, Bolino A, Aureli M, Martino S, Gritti A. Therapeutic advantages of combined gene/cell therapy strategies in a murine model of GM2 gangliosidosis. Mol Ther Methods Clin Dev 2022; 25:170-189. [PMID: 35434178 PMCID: PMC8983315 DOI: 10.1016/j.omtm.2022.03.011] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/13/2022] [Indexed: 11/28/2022]
Abstract
Genetic deficiency of β-N-acetylhexosaminidase (Hex) functionality leads to accumulation of GM2 ganglioside in Tay-Sachs disease and Sandhoff disease (SD), which presently lack approved therapies. Current experimental gene therapy (GT) approaches with adeno-associated viral vectors (AAVs) still pose safety and efficacy issues, supporting the search for alternative therapeutic strategies. Here we leveraged the lentiviral vector (LV)-mediated intracerebral (IC) GT platform to deliver Hex genes to the CNS and combined this strategy with bone marrow transplantation (BMT) to provide a timely, pervasive, and long-lasting source of the Hex enzyme in the CNS and periphery of SD mice. Combined therapy outperformed individual treatments in terms of lifespan extension and normalization of the neuroinflammatory/neurodegenerative phenotypes of SD mice. These benefits correlated with a time-dependent increase in Hex activity and a remarkable reduction in GM2 storage in brain tissues that single treatments failed to achieve. Our results highlight the synergic mode of action of LV-mediated IC GT and BMT, clarify the contribution of treatments to the therapeutic outcome, and inform on the realistic threshold of corrective enzymatic activity. These results have important implications for interpretation of ongoing experimental therapies and for design of more effective treatment strategies for GM2 gangliosidosis.
Collapse
Affiliation(s)
- Davide Sala
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Francesca Ornaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Francesco Morena
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Chiara Argentati
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Manuela Valsecchi
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Via Fratelli Cervi 93, 20090 Segrate, MI, Italy
| | - Valeria Alberizzi
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, 20132 Milan, Italy
| | - Roberta Di Guardo
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, 20132 Milan, Italy
| | - Alessandra Bolino
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, 20132 Milan, Italy
| | - Massimo Aureli
- Department of Medical Biotechnology and Translational Medicine, University of Milano, Via Fratelli Cervi 93, 20090 Segrate, MI, Italy
| | - Sabata Martino
- Department of Chemistry, Biology, and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| |
Collapse
|
8
|
Rintz E, Higuchi T, Kobayashi H, Galileo DS, Wegrzyn G, Tomatsu S. Promoter considerations in the design of lentiviral vectors for use in treating lysosomal storage diseases. Mol Ther Methods Clin Dev 2022; 24:71-87. [PMID: 34977274 PMCID: PMC8688940 DOI: 10.1016/j.omtm.2021.11.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
More than 50 lysosomal storage diseases (LSDs) are associated with lysosomal dysfunctions with the frequency of 1:5,000 live births. As a result of missing enzyme activity, the lysosome dysfunction accumulates undegraded or partially degraded molecules, affecting the entire body. Most of them are life-threatening diseases where patients could die within the first or second decade of life. Approximately 20 LSDs have the approved treatments, which do not provide the cure for the disorder. Therefore, the delivery of missing genes through gene therapy is a promising approach for LSDs. Over the years, ex vivo lentiviral-mediated gene therapy for LSDs has been approached using different strategies. Several clinical trials for LSDs are under investigation.Ex vivo lentiviral-mediated gene therapy needs optimization in dose, time of delivery, and promoter-driven expression. Choosing suitable promoters seems to be one of the important factors for the effective expression of the dysfunctional enzyme. This review summarizes the research on therapy for LSDs that has used different lentiviral vectors, emphasizing gene promoters.
Collapse
Affiliation(s)
- Estera Rintz
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza, 59, 80-308 Gdansk, Poland
- Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Road, Wilmington, DE 19803, USA
| | - Takashi Higuchi
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University School of Medicine, 3 Chome-25-8 Nishishinbashi, Minato City, Tokyo 105-8461, Japan
| | - Hiroshi Kobayashi
- Division of Gene Therapy, Research Center for Medical Sciences, The Jikei University School of Medicine, 3 Chome-25-8 Nishishinbashi, Minato City, Tokyo 105-8461, Japan
| | - Deni S. Galileo
- Department of Biological Sciences, University of Delaware, 118 Wolf Hall, Newark, DE 19716, USA
| | - Grzegorz Wegrzyn
- Department of Molecular Biology, Faculty of Biology, University of Gdansk, Wita Stwosza, 59, 80-308 Gdansk, Poland
| | - Shunji Tomatsu
- Nemours/Alfred I. duPont Hospital for Children, 1600 Rockland Road, Wilmington, DE 19803, USA
- Department of Biological Sciences, University of Delaware, 118 Wolf Hall, Newark, DE 19716, USA
- Department of Pediatrics, Gifu University, Gifu, Yanagido 501-1193, Japan
- Department of Pediatrics, Thomas Jefferson University, 901 Walnut Street, Philadelphia, PA 19107, USA
| |
Collapse
|
9
|
Cantore A, Fraldi A, Meneghini V, Gritti A. In vivo Gene Therapy to the Liver and Nervous System: Promises and Challenges. Front Med (Lausanne) 2022; 8:774618. [PMID: 35118085 PMCID: PMC8803894 DOI: 10.3389/fmed.2021.774618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Accepted: 12/16/2021] [Indexed: 12/02/2022] Open
Abstract
In vivo genetic engineering has recently shown remarkable potential as a novel effective treatment for an ever-growing number of diseases, as also witnessed by the recent marketing authorization of several in vivo gene therapy products. In vivo genetic engineering comprises both viral vector-mediated gene transfer and the more recently developed genome/epigenome editing strategies, as long as they are directly administered to patients. Here we first review the most advanced in vivo gene therapies that are commercially available or in clinical development. We then highlight the major challenges to be overcome to fully and broadly exploit in vivo gene therapies as novel medicines, discussing some of the approaches that are being taken to address them, with a focus on the nervous system and liver taken as paradigmatic examples.
Collapse
Affiliation(s)
- Alessio Cantore
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
- School of Medicine, Vita-Salute San Raffaele University, Milan, Italy
- *Correspondence: Alessio Cantore
| | - Alessandro Fraldi
- CEINGE Biotecnologie Avanzate, Naples, Italy
- Department of Translational Medicine, University of Naples “Federico II”, Naples, Italy
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
10
|
Nasir G, Chopra R, Elwood F, Ahmed SS. Krabbe Disease: Prospects of Finding a Cure Using AAV Gene Therapy. Front Med (Lausanne) 2021; 8:760236. [PMID: 34869463 PMCID: PMC8633897 DOI: 10.3389/fmed.2021.760236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/15/2021] [Indexed: 11/13/2022] Open
Abstract
Krabbe Disease (KD) is an autosomal metabolic disorder that affects both the central and peripheral nervous systems. It is caused by a functional deficiency of the lysosomal enzyme, galactocerebrosidase (GALC), resulting in an accumulation of the toxic metabolite, psychosine. Psychosine accumulation affects many different cellular pathways, leading to severe demyelination. Although there is currently no effective therapy for Krabbe disease, recent gene therapy-based approaches in animal models have indicated a promising outlook for clinical treatment. This review highlights recent findings in the pathogenesis of Krabbe disease, and evaluates AAV-based gene therapy as a promising strategy for treating this devastating pediatric disease.
Collapse
Affiliation(s)
- Gibran Nasir
- Department of Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA, United States
| | - Rajiv Chopra
- AllianThera Biopharma, Boston, MA, United States
| | - Fiona Elwood
- Department of Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA, United States
| | - Seemin S Ahmed
- Department of Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA, United States
| |
Collapse
|
11
|
Meneghini V, Peviani M, Luciani M, Zambonini G, Gritti A. Delivery Platforms for CRISPR/Cas9 Genome Editing of Glial Cells in the Central Nervous System. Front Genome Ed 2021; 3:644319. [PMID: 34713256 PMCID: PMC8525379 DOI: 10.3389/fgeed.2021.644319] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2020] [Accepted: 01/21/2021] [Indexed: 12/14/2022] Open
Abstract
Glial cells (astrocytes, oligodendrocytes, and microglia) are emerging as key players in several physiological and pathological processes of the central nervous system (CNS). Astrocytes and oligodendrocytes are not only supportive cells that release trophic factors or regulate energy metabolism, but they also actively modulate critical neuronal processes and functions in the tripartite synapse. Microglia are defined as CNS-resident cells that provide immune surveillance; however, they also actively contribute to shaping the neuronal microenvironment by scavenging cell debris or regulating synaptogenesis and pruning. Given the many interconnected processes coordinated by glial cells, it is not surprising that both acute and chronic CNS insults not only cause neuronal damage but also trigger complex multifaceted responses, including neuroinflammation, which can critically contribute to the disease progression and worsening of symptoms in several neurodegenerative diseases. Overall, this makes glial cells excellent candidates for targeted therapies to treat CNS disorders. In recent years, the application of gene editing technologies has redefined therapeutic strategies to treat genetic and age-related neurological diseases. In this review, we discuss the advantages and limitations of clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9-based gene editing in the treatment of neurodegenerative disorders, focusing on the development of viral- and nanoparticle-based delivery methods for in vivo glial cell targeting.
Collapse
Affiliation(s)
- Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Marco Peviani
- Department of Biology and Biotechnology "L. Spallanzani", University of Pavia, Pavia, Italy
| | - Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giada Zambonini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
12
|
Mangiameli E, Cecchele A, Morena F, Sanvito F, Matafora V, Cattaneo A, Della Volpe L, Gnani D, Paulis M, Susani L, Martino S, Di Micco R, Bachi A, Gritti A. Human iPSC-based neurodevelopmental models of globoid cell leukodystrophy uncover patient- and cell type-specific disease phenotypes. Stem Cell Reports 2021; 16:1478-1495. [PMID: 33989519 PMCID: PMC8190599 DOI: 10.1016/j.stemcr.2021.04.011] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 11/20/2022] Open
Abstract
Globoid cell leukodystrophy (GLD) is a rare neurodegenerative lysosomal storage disease caused by an inherited deficiency of β-galactocerebrosidase (GALC). GLD pathogenesis and therapeutic correction have been poorly studied in patient neural cells. Here, we investigated the impact of GALC deficiency and lentiviral vector-mediated GALC rescue/overexpression in induced pluripotent stem cell (iPSC)-derived neural progenitors and neuronal/glial progeny obtained from two GLD patients. GLD neural progeny displayed progressive psychosine storage, oligodendroglial and neuronal defects, unbalanced lipid composition, and early activation of cellular senescence, depending on the disease-causing mutation. The partial rescue of the neural differentiation program upon GALC reconstitution and psychosine clearance suggests multiple mechanisms contributing to neural pathology in GLD. Also, the pathological phenotype associated to supraphysiological GALC levels highlights the need of regulated GALC expression for proper human neural commitment/differentiation. These data have important implications for establishing safe therapeutic strategies to enhance disease correction of GLD.
Collapse
Affiliation(s)
- Elisabeth Mangiameli
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Anna Cecchele
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Francesca Sanvito
- Pathology Unit, IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Vittoria Matafora
- IFOM-FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Angela Cattaneo
- IFOM-FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Lucrezia Della Volpe
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Daniela Gnani
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Marianna Paulis
- Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy; National Research Council (CNR)-IRGB/UOS of Milan, Milan, Italy
| | - Lucia Susani
- Humanitas Clinical and Research Center-IRCCS, Rozzano, Milan, Italy; National Research Council (CNR)-IRGB/UOS of Milan, Milan, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Via del Giochetto, 06123 Perugia, Italy
| | - Raffaella Di Micco
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy
| | - Angela Bachi
- IFOM-FIRC Institute of Molecular Oncology, Via Adamello 16, 20139 Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Via Olgettina 60, 20132 Milan, Italy.
| |
Collapse
|
13
|
Lotun A, Gessler DJ, Gao G. Canavan Disease as a Model for Gene Therapy-Mediated Myelin Repair. Front Cell Neurosci 2021; 15:661928. [PMID: 33967698 PMCID: PMC8102781 DOI: 10.3389/fncel.2021.661928] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 03/23/2021] [Indexed: 11/13/2022] Open
Abstract
In recent years, the scientific and therapeutic fields for rare, genetic central nervous system (CNS) diseases such as leukodystrophies, or white matter disorders, have expanded significantly in part due to technological advancements in cellular and clinical screenings as well as remedial therapies using novel techniques such as gene therapy. However, treatments aimed at normalizing the pathological changes associated with leukodystrophies have especially been complicated due to the innate and variable effects of glial abnormalities, which can cause large-scale functional deficits in developmental myelination and thus lead to downstream neuronal impairment. Emerging research in the past two decades have depicted glial cells, particularly oligodendrocytes and astrocytes, as key, regulatory modulators in constructing and maintaining myelin function and neuronal viability. Given the significance of myelin formation in the developing brain, myelin repair in a time-dependent fashion is critical in restoring homeostatic functionality to the CNS of patients diagnosed with white matter disorders. Using Canavan Disease (CD) as a leukodystrophy model, here we review the hypothetical roles of N-acetylaspartate (NAA), one of the brain's most abundant amino acid derivatives, in Canavan disease's CNS myelinating pathology, as well as discuss the possible functions astrocytes serve in both CD and other leukodystrophies' time-sensitive disease correction. Through this analysis, we also highlight the potential remyelinating benefits of gene therapy for other leukodystrophies in which alternative CNS cell targeting for white matter disorders may be an applicable path for reparative treatment.
Collapse
Affiliation(s)
- Anoushka Lotun
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Neurosurgery, University of Minnesota, Minneapolis, MN, United States
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, United States.,Department of Microbiology and Physiological Systems, University of Massachusetts Medical School, Worcester, MA, United States.,Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA, United States
| |
Collapse
|
14
|
Wilson I, Vitelli C, Yu GK, Pacheco G, Vincelette J, Bunting S, Sisó S. Quantitative Assessment of Neuroinflammation, Myelinogenesis, Demyelination, and Nerve Fiber Regeneration in Immunostained Sciatic Nerves From Twitcher Mice With a Tissue Image Analysis Platform. Toxicol Pathol 2021; 49:950-962. [PMID: 33691530 DOI: 10.1177/0192623321991469] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Scoring demyelination and regeneration in hematoxylin and eosin-stained nerves poses a challenge even for the trained pathologist. This article demonstrates how combinatorial multiplex immunohistochemistry (IHC) and quantitative digital pathology bring new insights into the peripheral neuropathogenesis of the Twitcher mouse, a model of Krabbe disease. The goal of this investigational study was to integrate modern pathology tools to traditional anatomic pathology microscopy workflows, in order to generate quantitative data in a large number of samples, and aid the understanding of complex disease pathomechanisms. We developed a novel IHC toolkit using a combination of CD68, periaxin-1, phosphorylated neurofilaments and SOX-10 to interrogate inflammation, myelination, axonal size, and Schwann cell counts in sciatic nerves from 17-, 21-, 25-, and 35-day-old wild-type and Twitcher mice using self-customized digital image algorithms. Our quantitative analyses highlight that nerve macrophage infiltration and interstitial expansion are the earliest detectable changes in Twitcher nerves. By 17 days of age, while the diameter of axons is small, the number of myelinated axons is still normal. However, from 21 days onward Twitcher nerves contain 75% of wild-type myelinated nerve fiber numbers despite containing 3 times more Schwann cells. In 35-day-old Twitcher mice when demyelination is detectable, nerve myelination drops to 50%.
Collapse
Affiliation(s)
- Irene Wilson
- Research and Early Development, 10926BioMarin Pharmaceutical Inc., San Rafael, CA, USA.,Dominican University of California, San Rafael, CA, USA
| | - Cathy Vitelli
- Research and Early Development, 10926BioMarin Pharmaceutical Inc., San Rafael, CA, USA
| | - Guoying Karen Yu
- Research and Early Development, 10926BioMarin Pharmaceutical Inc., San Rafael, CA, USA
| | - Glenn Pacheco
- Research and Early Development, 10926BioMarin Pharmaceutical Inc., San Rafael, CA, USA
| | - Jon Vincelette
- Research and Early Development, 10926BioMarin Pharmaceutical Inc., San Rafael, CA, USA
| | - Stuart Bunting
- Research and Early Development, 10926BioMarin Pharmaceutical Inc., San Rafael, CA, USA
| | - Sílvia Sisó
- Research and Early Development, 10926BioMarin Pharmaceutical Inc., San Rafael, CA, USA.,Dominican University of California, San Rafael, CA, USA
| |
Collapse
|
15
|
Luciani M, Gritti A, Meneghini V. Human iPSC-Based Models for the Development of Therapeutics Targeting Neurodegenerative Lysosomal Storage Diseases. Front Mol Biosci 2020; 7:224. [PMID: 33062642 PMCID: PMC7530250 DOI: 10.3389/fmolb.2020.00224] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Accepted: 08/10/2020] [Indexed: 01/30/2023] Open
Abstract
Lysosomal storage diseases (LSDs) are a group of rare genetic conditions. The absence or deficiency of lysosomal proteins leads to excessive storage of undigested materials and drives secondary pathological mechanisms including autophagy, calcium homeostasis, ER stress, and mitochondrial abnormalities. A large number of LSDs display mild to severe central nervous system (CNS) involvement. Animal disease models and post-mortem tissues partially recapitulate the disease or represent the final stage of CNS pathology, respectively. In the last decades, human models based on induced pluripotent stem cells (hiPSCs) have been extensively applied to investigate LSD pathology in several tissues and organs, including the CNS. Neural stem/progenitor cells (NSCs) derived from patient-specific hiPSCs (hiPS-NSCs) are a promising tool to define the effects of the pathological storage on neurodevelopment, survival and function of neurons and glial cells in neurodegenerative LSDs. Additionally, the development of novel 2D co-culture systems and 3D hiPSC-based models is fostering the investigation of neuron-glia functional and dysfunctional interactions, also contributing to define the role of neurodevelopment and neuroinflammation in the onset and progression of the disease, with important implications in terms of timing and efficacy of treatments. Here, we discuss the advantages and limits of the application of hiPS-NSC-based models in the study and treatment of CNS pathology in different LSDs. Additionally, we review the state-of-the-art and the prospective applications of NSC-based therapy, highlighting the potential exploitation of hiPS-NSCs for gene and cell therapy approaches in the treatment of neurodegenerative LSDs.
Collapse
Affiliation(s)
- Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
16
|
Ricca A, Cascino F, Morena F, Martino S, Gritti A. In vitro Validation of Chimeric β-Galactosylceramidase Enzymes With Improved Enzymatic Activity and Increased Secretion. Front Mol Biosci 2020; 7:167. [PMID: 32850960 PMCID: PMC7396597 DOI: 10.3389/fmolb.2020.00167] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Accepted: 07/01/2020] [Indexed: 12/27/2022] Open
Abstract
Globoid Cell Leukodystrophy (GLD) is a lysosomal storage disease (LSD) caused by inherited defects of the β-galactosylceramidase (GALC) gene. The infantile forms display a rapid and aggressive central and peripheral nervous system (CNS and PNS) dysfunction. No treatments are available for GLD patients. Effective gene therapy (GT) strategies for GLD require a safe and widespread delivery of the functional GALC enzyme to all affected tissues/organs, and particularly to the CNS. The use of chimeric lysosomal enzymes with increased secretion and enhanced transport across the blood-brain barrier (BBB) that boost the efficacy of GT approaches in pre-clinical models of similar neurodegenerative LSDs may benefit GLD as well. Here, we tested the safety and biological efficacy of chimeric GALC enzymes engineered to express an alternative signal peptide (iduronate-2-sulfatase - IDSsp) and the low-density lipoprotein receptor (LDLr)-binding domain from the Apolipoprotein E II (ApoE II) in GLD murine neural and hematopoietic stem/progenitor cells and progeny, which are relevant cells types in the context of in vivo and ex vivo GT platforms. We show that the lentiviral vector-mediated expression of the chimeric GALC enzymes is safe and leads to supranormal enzymatic activity in both neural and hematopoietic cells. The IDSsp.GALC shows enhanced expression and secretion in comparison to the unmodified GALC. The chimeric GALC enzymes produced by LV-transduced cells reduce intracellular galactosylceramide (GalCer) storage and effectively cross-correct GLD murine neurons and glial cells, indicating that the transgenic enzymes are delivered to lysosomes, efficiently secreted, and functional. Of note, the expression of LDLr and LDLr-related proteins in GLD neurons and glial cells supports the exploitation of this system to enhance the GALC supply in affected CNS cells and tissues. These in vitro studies support the use of chimeric GALC enzymes to develop novel and more effective GT approaches for GLD.
Collapse
Affiliation(s)
- Alessandra Ricca
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Federica Cascino
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
17
|
Novel bicistronic lentiviral vectors correct β-Hexosaminidase deficiency in neural and hematopoietic stem cells and progeny: implications for in vivo and ex vivo gene therapy of GM2 gangliosidosis. Neurobiol Dis 2019; 134:104667. [PMID: 31682993 DOI: 10.1016/j.nbd.2019.104667] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 10/28/2019] [Accepted: 10/31/2019] [Indexed: 01/03/2023] Open
Abstract
The favorable outcome of in vivo and ex vivo gene therapy approaches in several Lysosomal Storage Diseases suggests that these treatment strategies might equally benefit GM2 gangliosidosis. Tay-Sachs and Sandhoff disease (the main forms of GM2 gangliosidosis) result from mutations in either the HEXA or HEXB genes encoding, respectively, the α- or β-subunits of the lysosomal β-Hexosaminidase enzyme. In physiological conditions, α- and β-subunits combine to generate β-Hexosaminidase A (HexA, αβ) and β-Hexosaminidase B (HexB, ββ). A major impairment to establishing in vivo or ex vivo gene therapy for GM2 gangliosidosis is the need to synthesize the α- and β-subunits at high levels and with the correct stoichiometric ratio, and to safely deliver the therapeutic products to all affected tissues/organs. Here, we report the generation and in vitro validation of novel bicistronic lentiviral vectors (LVs) encoding for both the murine and human codon optimized Hexa and Hexb genes. We show that these LVs drive the safe and coordinate expression of the α- and β-subunits, leading to supranormal levels of β-Hexosaminidase activity with prevalent formation of a functional HexA in SD murine neurons and glia, murine bone marrow-derived hematopoietic stem/progenitor cells (HSPCs), and human SD fibroblasts. The restoration/overexpression of β-Hexosaminidase leads to the reduction of intracellular GM2 ganglioside storage in transduced and in cross-corrected SD murine neural progeny, indicating that the transgenic enzyme is secreted and functional. Importantly, bicistronic LVs safely and efficiently transduce human neurons/glia and CD34+ HSPCs, which are target and effector cells, respectively, in prospective in vivo and ex vivo GT approaches. We anticipate that these bicistronic LVs may overcome the current requirement of two vectors co-delivering the α- or β-subunits genes. Careful assessment of the safety and therapeutic potential of these bicistronic LVs in the SD murine model will pave the way to the clinical development of LV-based gene therapy for GM2 gangliosidosis.
Collapse
|
18
|
Pellegrini D, Del Grosso A, Angella L, Giordano N, Dilillo M, Tonazzini I, Caleo M, Cecchini M, McDonnell LA. Quantitative Microproteomics Based Characterization of the Central and Peripheral Nervous System of a Mouse Model of Krabbe Disease. Mol Cell Proteomics 2019; 18:1227-1241. [PMID: 30926673 PMCID: PMC6553931 DOI: 10.1074/mcp.ra118.001267] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2018] [Revised: 03/15/2019] [Indexed: 11/06/2022] Open
Abstract
Krabbe disease is a rare, childhood lysosomal storage disorder caused by a deficiency of galactosylceramide beta-galactosidase (GALC). The major effect of GALC deficiency is the accumulation of psychosine in the nervous system and widespread degeneration of oligodendrocytes and Schwann cells, causing rapid demyelination. The molecular mechanisms of Krabbe disease are not yet fully elucidated and a definite cure is still missing. Here we report the first in-depth characterization of the proteome of the Twitcher mouse, a spontaneous mouse model of Krabbe disease, to investigate the proteome changes in the Central and Peripheral Nervous System. We applied a TMT-based workflow to compare the proteomes of the corpus callosum, motor cortex and sciatic nerves of littermate homozygous Twitcher and wild-type mice. More than 400 protein groups exhibited differences in expression and included proteins involved in pathways that can be linked to Krabbe disease, such as inflammatory and defense response, lysosomal proteins accumulation, demyelination, reduced nervous system development and cell adhesion. These findings provide new insights on the molecular mechanisms of Krabbe disease, representing a starting point for future functional experiments to study the molecular pathogenesis of Krabbe disease. Data are available via ProteomeXchange with identifier PXD010594.
Collapse
Affiliation(s)
- Davide Pellegrini
- From ‡NEST, Scuola Normale Superiore, Pisa 56127, Italy
- §Fondazione Pisana per la Scienza ONLUS, 56107 San Giuliano Terme, Pisa, Italy
| | - Ambra Del Grosso
- From ‡NEST, Scuola Normale Superiore, Pisa 56127, Italy
- ¶NEST, Istituto Nanoscienze-CNR, Pisa, Italy
| | | | | | - Marialaura Dilillo
- §Fondazione Pisana per la Scienza ONLUS, 56107 San Giuliano Terme, Pisa, Italy
| | | | | | - Marco Cecchini
- From ‡NEST, Scuola Normale Superiore, Pisa 56127, Italy
- ¶NEST, Istituto Nanoscienze-CNR, Pisa, Italy
| | - Liam A McDonnell
- §Fondazione Pisana per la Scienza ONLUS, 56107 San Giuliano Terme, Pisa, Italy;
- **Center for Proteomics and Metabolomics, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
19
|
Argentati C, Morena F, Bazzucchi M, Armentano I, Emiliani C, Martino S. Adipose Stem Cell Translational Applications: From Bench-to-Bedside. Int J Mol Sci 2018; 19:E3475. [PMID: 30400641 PMCID: PMC6275042 DOI: 10.3390/ijms19113475] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2018] [Revised: 10/22/2018] [Accepted: 11/01/2018] [Indexed: 02/08/2023] Open
Abstract
During the last five years, there has been a significantly increasing interest in adult adipose stem cells (ASCs) as a suitable tool for translational medicine applications. The abundant and renewable source of ASCs and the relatively simple procedure for cell isolation are only some of the reasons for this success. Here, we document the advances in the biology and in the innovative biotechnological applications of ASCs. We discuss how the multipotential property boosts ASCs toward mesenchymal and non-mesenchymal differentiation cell lineages and how their character is maintained even if they are combined with gene delivery systems and/or biomaterials, both in vitro and in vivo.
Collapse
Affiliation(s)
- Chiara Argentati
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Martina Bazzucchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
| | - Ilaria Armentano
- Department of Ecological and Biological Sciences, Tuscia University Largo dell'Università, snc, 01100 Viterbo, Italy.
| | - Carla Emiliani
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy.
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Via del Giochetto, 06126 Perugia, Italy.
- CEMIN, Center of Excellence on Nanostructured Innovative Materials, Via del Giochetto, 06126 Perugia, Italy.
| |
Collapse
|
20
|
Abstract
Leukodystrophies are a heterogeneous class of genetic diseases affecting the white matter in the central nervous system with a broad range of clinical manifestations and a frequently progressive course. An interest in precision medicine has emerged over the last several decades, and biomedical research in leukodystrophies has made exciting advances along this front through therapeutic target discovery and novel disease model systems. In this review, we discuss current and emerging therapeutic approaches in leukodystrophies, including gene therapy, antisense oligonucleotide therapy, CRISPR/CAS-based gene editing, and cell and stem cell based therapies.
Collapse
Affiliation(s)
- Eliza Gordon-Lipkin
- 1 Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA .,2 Departments of Neurology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ali Fatemi
- 1 Department of Neurology and Developmental Medicine, Kennedy Krieger Institute, Baltimore, MD, USA .,2 Departments of Neurology and Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD, USA.,3 Moser Center for Leukodystrophies and Neurogenetics, Kennedy Krieger Institute, Baltimore, MD, USA
| |
Collapse
|
21
|
Luddi A, Crifasi L, Capaldo A, Piomboni P, Costantino-Ceccarini E. Suppression of galactocerebrosidase premature termination codon and rescue of galactocerebrosidase activity in twitcher cells. J Neurosci Res 2017; 94:1273-83. [PMID: 27638609 DOI: 10.1002/jnr.23790] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2016] [Revised: 05/06/2016] [Accepted: 05/23/2016] [Indexed: 11/10/2022]
Abstract
Krabbe's disease (KD) is a degenerative lysosomal storage disease resulting from deficiency of β-galactocerebrosidase activity. Over 100 mutations are known to cause the disease, and these usually occur in compound heterozygote patterns. In affected patients, nonsense mutations leading to a nonfunctional enzyme are often found associated with other mutations. The twitcher mouse is a naturally occurring model of KD, containing in β-galactocerebrosidase a premature stop codon, W339X. Recent studies have shown that selected compounds may induce the ribosomal bypass of premature stop codons without affecting the normal termination codons. The rescue of β-galactocerebrosidase activity induced by treatment with premature termination codon (PTC) 124, a well-characterized compound known to induce ribosomal read-through, was investigated on oligodendrocytes prepared from twitcher mice and on human fibroblasts from patients bearing nonsense mutations. The effectiveness of the nonsense-mediated mRNA decay (NMD) inhibitor 1 (NMDI1), a newly identified inhibitor of NMD, was also tested. Incubation of these cell lines with PTC124 and NMDI1 increased the levels of mRNA and rescued galactocerebrosidase enzymatic activity in a dose-dependent manner. The low but sustained expression of β-galactocerebrosidase in oligodendrocytes was sufficient to improve the morphology of the differentiated cells. Our in vitro approach provides the basis for further investigation of ribosomal read-through as an alternative therapeutic strategy to ameliorate the quality of life in selected KD patients. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alice Luddi
- Department of Molecular and Developmental Medicine, Siena University, Siena, Italy.
| | - Laura Crifasi
- Department of Molecular and Developmental Medicine, Siena University, Siena, Italy
| | - Angela Capaldo
- Department of Molecular and Developmental Medicine, Siena University, Siena, Italy
| | - Paola Piomboni
- Department of Molecular and Developmental Medicine, Siena University, Siena, Italy
| | | |
Collapse
|
22
|
Ricca A, Gritti A. Perspective on innovative therapies for globoid cell leukodystrophy. J Neurosci Res 2017; 94:1304-17. [PMID: 27638612 DOI: 10.1002/jnr.23752] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2016] [Revised: 03/25/2016] [Accepted: 03/30/2016] [Indexed: 12/24/2022]
Abstract
Globoid cell leukodystrophy (GLD), or Krabbe's disease, is a lysosomal storage disorder resulting from deficiency of the lysosomal hydrolase galactosylceramidase. The infantile forms are characterized by a unique relentless and aggressive progression with a wide range of neurological symptoms and complications. Here we review and discuss the basic concepts and the novel mechanisms identified as key contributors to the peculiar GLD pathology, highlighting their therapeutic implications. Then, we evaluate evidence from extensive experimental studies on GLD animal models that have highlighted fundamental requirements to obtain substantial therapeutic benefit, including early and timely intervention, high levels of enzymatic reconstitution, and global targeting of affected tissues. Continuous efforts in understanding GLD pathophysiology, the interplay between various therapies, and the mechanisms of disease correction upon intervention may allow advancing research with innovative approaches and prioritizing treatment strategies to develop more efficacious treatments. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alessandra Ricca
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-Tiget), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
23
|
Nicaise AM, Bongarzone ER, Crocker SJ. A microglial hypothesis of globoid cell leukodystrophy pathology. J Neurosci Res 2017; 94:1049-61. [PMID: 27638591 DOI: 10.1002/jnr.23773] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2016] [Revised: 05/05/2016] [Accepted: 05/06/2016] [Indexed: 11/09/2022]
Abstract
Globoid cell leukodystrophy (GLD), also known as Krabbe's disease, is a fatal demyelinating disease accompanied by the formation of giant, multinucleated cells called globoid cells. Previously believed to be a byproduct of inflammation, these cells can be found early in disease before evidence of any damage. The precise mechanism by which these globoid cells cause oligodendrocyte dysfunction is not completely understood, nor is their cell type defined. This Review outlines the idea that microglial cells are transformed into an unknown and undefined novel M3 phenotype in GLD, which is cytotoxic to oligodendrocytes, leading to disease progression. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Alexandra M Nicaise
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut
| | - Ernesto R Bongarzone
- Department of Anatomy and Cell Biology, University of Illinois at Chicago, Chicago, Illinois
| | - Stephen J Crocker
- Department of Neuroscience, University of Connecticut School of Medicine, Farmington, Connecticut.
| |
Collapse
|
24
|
Maher KR, Yeager AM. Cellular transplant therapies for globoid cell leukodystrophy: Preclinical and clinical observations. J Neurosci Res 2017; 94:1180-8. [PMID: 27638602 DOI: 10.1002/jnr.23782] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Revised: 05/11/2016] [Accepted: 05/11/2016] [Indexed: 12/21/2022]
Abstract
Globoid cell leukodystrophy (GLD) is a progressive neurodegenerative disorder caused by the deficiency of galactocerebrosidase (GALC), resulting in accumulation of toxic metabolites in neural tissues. Clinically variable based on age of onset, infantile GLD is generally a rapidly fatal syndrome of progressive neurologic and cognitive decline, whereas later-onset GLD has a more indolent, protracted clinical course. Animal models, particularly the twitcher mouse, have allowed investigation of both the pathophysiology of and the potential treatment modalities for GLD. Cellular therapy for GLD, notably hematopoietic cell transplantation (HCT; transplantation of bone marrow, peripheral blood stem cells, or umbilical cord blood cells) from a normal related or unrelated allogeneic donor provides a self-renewing source of GALC in donor-derived cells. The only currently available treatment option in human GLD, allogeneic HCT, can slow the progression of the disease and improve survival, especially when performed in presymptomatic infants. Because persistent neurologic dysfunction still occurs after HCT in GLD, preclinical studies are evaluating combinations of HCT with other treatment modalities. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Keri R Maher
- University of Arizona Cancer Center, Tucson, Arizona.,Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona
| | - Andrew M Yeager
- University of Arizona Cancer Center, Tucson, Arizona. .,Department of Medicine, University of Arizona College of Medicine, Tucson, Arizona. .,Department of Pediatrics, University of Arizona College of Medicine, Tucson, Arizona.
| |
Collapse
|
25
|
Penati R, Fumagalli F, Calbi V, Bernardo ME, Aiuti A. Gene therapy for lysosomal storage disorders: recent advances for metachromatic leukodystrophy and mucopolysaccaridosis I. J Inherit Metab Dis 2017; 40:543-554. [PMID: 28560469 PMCID: PMC5500670 DOI: 10.1007/s10545-017-0052-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/03/2017] [Revised: 04/15/2017] [Accepted: 04/27/2017] [Indexed: 01/10/2023]
Abstract
Lysosomal storage diseases (LSDs) are rare inherited metabolic disorders characterized by a dysfunction in lysosomes, leading to waste material accumulation and severe organ damage. Enzyme replacement therapy (ERT) and haematopoietic stem cell transplant (HSCT) have been exploited as potential treatments for LSDs but pre-clinical and clinical studies have shown in some cases limited efficacy. Intravenous ERT is able to control the damage of visceral organs but cannot prevent nervous impairment. Depending on the disease type, HSCT has important limitations when performed for early variants, unless treatment occurs before disease onset. In the attempt to overcome these issues, gene therapy has been proposed as a valuable therapeutic option, either ex vivo, with target cells genetically modified in vitro, or in vivo, by inserting the genetic material with systemic or intra-parenchymal, in situ administration. In particular, the use of autologous haematopoietic stem cells (HSC) transduced with a viral vector containing a healthy copy of the mutated gene would allow supra-normal production of the defective enzyme and cross correction of target cells in multiple tissues, including the central nervous system. This review will provide an overview of the most recent scientific advances in HSC-based gene therapy approaches for the treatment of LSDs with particular focus on metachromatic leukodystrophy (MLD) and mucopolysaccharidosis type I (MPS-I).
Collapse
Affiliation(s)
- Rachele Penati
- Unit of Pediatric Immunohematology and Stem Cell Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Fumagalli
- Unit of Pediatric Immunohematology and Stem Cell Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Department of Neurology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Valeria Calbi
- Unit of Pediatric Immunohematology and Stem Cell Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Maria Ester Bernardo
- Unit of Pediatric Immunohematology and Stem Cell Program, IRCCS San Raffaele Scientific Institute, Milan, Italy
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Aiuti
- Unit of Pediatric Immunohematology and Stem Cell Program, IRCCS San Raffaele Scientific Institute, Milan, Italy.
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy.
- Vita Salute San Raffaele University, Milan, Italy.
| |
Collapse
|
26
|
Ashrafi MR, Tavasoli AR. Childhood leukodystrophies: A literature review of updates on new definitions, classification, diagnostic approach and management. Brain Dev 2017; 39:369-385. [PMID: 28117190 DOI: 10.1016/j.braindev.2017.01.001] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Revised: 12/29/2016] [Accepted: 01/04/2017] [Indexed: 12/29/2022]
Abstract
Childhood leukodystrophies are a growing category of neurological disorders in pediatric neurology practice. With the help of new advanced genetic studies such as whole exome sequencing (WES) and whole genome sequencing (WGS), the list of childhood heritable white matter disorders has been increased to more than one hundred disorders. During the last three decades, the basic concepts and definitions, classification, diagnostic approach and medical management of these disorders much have changed. Pattern recognition based on brain magnetic resonance imaging (MRI), has played an important role in this process. We reviewed the last Global Leukodystrophy Initiative (GLIA) expert opinions in definition, new classification, diagnostic approach and medical management including emerging treatments for pediatric leukodystrophies.
Collapse
Affiliation(s)
- Mahmoud Reza Ashrafi
- Department of Child Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ali Reza Tavasoli
- Department of Child Neurology, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
27
|
de Vito G, Cappello V, Tonazzini I, Cecchini M, Piazza V. RP-CARS reveals molecular spatial order anomalies in myelin of an animal model of Krabbe disease. JOURNAL OF BIOPHOTONICS 2017; 10:385-393. [PMID: 26990139 DOI: 10.1002/jbio.201500305] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/24/2015] [Revised: 01/29/2016] [Accepted: 02/21/2016] [Indexed: 05/05/2023]
Abstract
Krabbe disease (KD) is a rare demyelinating sphingolipidosis, often fatal in the first years of life. It is caused by the inactivation of the galactocerebrosidase (GALC) enzyme that causes an increase in the cellular levels of psychosine considered to be at the origin of the tissue-level effects. GALC is inactivated also in the Twitcher (TWI) mouse: a genetic model of KD that is providing important insights into the understating of the pathogenetic process and the development of possible treatments. In this article an innovative optical technique, RP-CARS, is proposed as a tool to study the degree of order of the CH2 bonds inside the myelin sheaths of TWI-mice sciatic-nerve fibres. RP-CARS, a recently developed variation of CARS microscopy, is able to combine the intrinsic chemical selectivity of CARS microscopy with molecular-bond-spatial-orientation sensibility. This is the first time RP-CARS is applied to the study of a genetic model of a pathology, leading to the demonstration of a post-onset progressive spatial disorganization of the myelin CH2 bonds. The presented result could be of great interest for a deeper understanding of the pathogenic mechanisms underlying the human KD and, moreover, it is an additional proof of the experimental validity of this microscopy technique. RP-CARS image (2850 cm-1 , CH2 bonds) of a sciatic-nerve optical longitudinal section from a Twitcher P23 (symptomatic) mouse. Scale bar: 10 microns. The image was constructed by colour-mapping the degree of molecular order of the CH2 bonds inside the myelin walls, as displayed in the colour bar on the right.
Collapse
Affiliation(s)
- Giuseppe de Vito
- NEST, Scuola Normale Superiore, Piazza San Silvestro 12, I-56127, Pisa, Italy
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, I-56127, Pisa, Italy
| | - Valentina Cappello
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, I-56127, Pisa, Italy
| | - Ilaria Tonazzini
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Piazza San Silvestro 12, Pisa, I-56127, Italy
| | - Marco Cecchini
- NEST, Scuola Normale Superiore and Istituto Nanoscienze-CNR, Piazza San Silvestro 12, Pisa, I-56127, Italy
| | - Vincenzo Piazza
- Center for Nanotechnology Innovation @NEST, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, I-56127, Pisa, Italy
| |
Collapse
|
28
|
Georgiou E, Sidiropoulou K, Richter J, Papaneophytou C, Sargiannidou I, Kagiava A, von Jonquieres G, Christodoulou C, Klugmann M, Kleopa KA. Gene therapy targeting oligodendrocytes provides therapeutic benefit in a leukodystrophy model. Brain 2017; 140:599-616. [PMID: 28100454 PMCID: PMC5837386 DOI: 10.1093/brain/aww351] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Revised: 11/22/2016] [Accepted: 11/28/2016] [Indexed: 12/20/2022] Open
Abstract
Pelizaeus-Merzbacher-like disease or hypomyelinating leukodystrophy-2 is an autosomal recessively inherited leukodystrophy with childhood onset resulting from mutations in the gene encoding the gap junction protein connexin 47 (Cx47, encoded by GJC2). Cx47 is expressed specifically in oligodendrocytes and is crucial for gap junctional communication throughout the central nervous system. Previous studies confirmed that a cell autonomous loss-of-function mechanism underlies hypomyelinating leukodystrophy-2 and that transgenic oligodendrocyte-specific expression of another connexin, Cx32 (GJB1), can restore gap junctions in oligodendrocytes to achieve correction of the pathology in a disease model. To develop an oligodendrocyte-targeted gene therapy, we cloned the GJC2/Cx47 gene under the myelin basic protein promoter and used an adeno-associated viral vector (AAV.MBP.Cx47myc) to deliver the gene to postnatal Day 10 mice via a single intracerebral injection in the internal capsule area. Lasting Cx47 expression specifically in oligodendrocytes was detected in Cx47 single knockout and Cx32/Cx47 double knockout mice up to 12 weeks post-injection, including the corpus callosum and the internal capsule but also in more distant areas of the cerebrum and in the spinal cord. Application of this oligodendrocyte-targeted somatic gene therapy at postnatal Day 10 in groups of double knockout mice, a well characterized model of hypomyelinating leukodystrophy-2, resulted in significant improvement in motor performance and coordination at 1 month of age in treated compared to mock-treated mice, as well as prolonged survival. Furthermore, immunofluorescence and morphological analysis revealed improvement in demyelination, oligodendrocyte apoptosis, inflammation, and astrogliosis, all typical features of this leukodystrophy model in both brain and spinal cord. Functional dye transfer analysis confirmed the re-establishment of oligodendrocyte gap junctional connectivity in treated as opposed to untreated mice. These results provide a significant advance in the development of oligodendrocyte-cell specific gene therapy. Adeno-associated viral vectors can be used to target therapeutic expression of a myelin gene to oligodendrocytes. We show evidence for the first somatic gene therapy approach to treat hypomyelinating leukodystrophy-2 preclinically, providing a potential treatment for this and similar forms of leukodystrophies.
Collapse
Affiliation(s)
- Elena Georgiou
- 1 Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | | | - Jan Richter
- 3 Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Christos Papaneophytou
- 1 Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Irene Sargiannidou
- 1 Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Alexia Kagiava
- 1 Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Georg von Jonquieres
- 4 Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Christina Christodoulou
- 3 Department of Molecular Virology, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Matthias Klugmann
- 4 Translational Neuroscience Facility and Department of Physiology, School of Medical Sciences, University of New South Wales, Sydney, Australia
| | - Kleopas A. Kleopa
- 1 Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
- 5 Neurology Clinics, The Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| |
Collapse
|
29
|
Two factor-based reprogramming of rodent and human fibroblasts into Schwann cells. Nat Commun 2017; 8:14088. [PMID: 28169300 PMCID: PMC5309703 DOI: 10.1038/ncomms14088] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/18/2016] [Indexed: 11/29/2022] Open
Abstract
Schwann cells (SCs) generate the myelin wrapping of peripheral nerve axons and are promising candidates for cell therapy. However, to date a renewable source of SCs is lacking. In this study, we show the conversion of skin fibroblasts into induced Schwann cells (iSCs) by driving the expression of two transcription factors, Sox10 and Egr2. iSCs resembled primary SCs in global gene expression profiling and PNS identity. In vitro, iSCs wrapped axons generating compact myelin sheaths with regular nodal structures. Conversely, iSCs from Twitcher mice showed a severe loss in their myelinogenic potential, demonstrating that iSCs can be an attractive system for in vitro modelling of PNS diseases. The same two factors were sufficient to convert human fibroblasts into iSCs as defined by distinctive molecular and functional traits. Generating iSCs through direct conversion of somatic cells offers opportunities for in vitro disease modelling and regenerative therapies. Schwann cells (SCs) myelinate peripheral nerve axons and offer opportunities for the treatment of injuries and demyelinating diseases but reliable and renewable sources of these cells are hard to come by. Here the authors reprogram rat, mouse and human fibroblasts into Schwann cells using two transcription factors.
Collapse
|
30
|
Gonzalez EA, Baldo G. Gene Therapy for Lysosomal Storage Disorders. JOURNAL OF INBORN ERRORS OF METABOLISM AND SCREENING 2017. [DOI: 10.1177/2326409816689786] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Affiliation(s)
- Esteban Alberto Gonzalez
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Genetic and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| | - Guilherme Baldo
- Gene Therapy Center, Hospital de Clínicas de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
- Genetic and Molecular Biology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
- Physiology, Universidade Federal do Rio Grande do Sul, Porto Alegre, Rio Grande do Sul, Brazil
| |
Collapse
|
31
|
Sauer AV, Hernandez RJ, Fumagalli F, Bianchi V, Poliani PL, Dallatomasina C, Riboni E, Politi LS, Tabucchi A, Carlucci F, Casiraghi M, Carriglio N, Cominelli M, Forcellini CA, Barzaghi F, Ferrua F, Minicucci F, Medaglini S, Leocani L, la Marca G, Notarangelo LD, Azzari C, Comi G, Baldoli C, Canale S, Sessa M, D’Adamo P, Aiuti A. Alterations in the brain adenosine metabolism cause behavioral and neurological impairment in ADA-deficient mice and patients. Sci Rep 2017; 7:40136. [PMID: 28074903 PMCID: PMC5225479 DOI: 10.1038/srep40136] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2016] [Accepted: 11/23/2016] [Indexed: 02/03/2023] Open
Abstract
Adenosine Deaminase (ADA) deficiency is an autosomal recessive variant of severe combined immunodeficiency (SCID) caused by systemic accumulation of ADA substrates. Neurological and behavioral abnormalities observed in ADA-SCID patients surviving after stem cell transplantation or gene therapy represent an unresolved enigma in the field. We found significant neurological and cognitive alterations in untreated ADA-SCID patients as well as in two groups of patients after short- and long-term enzyme replacement therapy with PEG-ADA. These included motor dysfunction, EEG alterations, sensorineural hypoacusia, white matter and ventricular alterations in MRI as well as a low mental development index or IQ. Ada-deficient mice were significantly less active and showed anxiety-like behavior. Molecular and metabolic analyses showed that this phenotype coincides with metabolic alterations and aberrant adenosine receptor signaling. PEG-ADA treatment corrected metabolic adenosine-based alterations, but not cellular and signaling defects, indicating an intrinsic nature of the neurological and behavioral phenotype in ADA deficiency.
Collapse
Affiliation(s)
- Aisha V. Sauer
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Raisa Jofra Hernandez
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesca Fumagalli
- Neurology Unit, Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Veronica Bianchi
- Dulbecco Telethon Institute at Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Chiara Dallatomasina
- Psychological Service, Neurological Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Elisa Riboni
- Psychological Service, Neurological Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Letterio S. Politi
- Imaging Core and Neuroradiology Unit, Head and Neck Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Antonella Tabucchi
- Department of Medical Biotechnologies, University of Siena, Italy
- U.O.C. Clinical Pathology, AOUS, Siena, Italy
| | - Filippo Carlucci
- Department of Medical Biotechnologies, University of Siena, Italy
- U.O.C. Clinical Pathology, AOUS, Siena, Italy
| | - Miriam Casiraghi
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Nicola Carriglio
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Carlo Alberto Forcellini
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Hospital, Milan, Italy
| | - Federica Barzaghi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Francesca Ferrua
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| | - Fabio Minicucci
- Neurophysiology Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Stefania Medaglini
- Neurophysiology Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Letizia Leocani
- Neurophysiology Center, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Giancarlo la Marca
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Lucia D. Notarangelo
- Department of Molecular and Translational Medicine, Pathology Unit, University of Brescia, Brescia, Italy
| | - Chiara Azzari
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Giancarlo Comi
- Psychological Service, Neurological Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Cristina Baldoli
- Neuroradiology Unit, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Sabrina Canale
- Multimedica hospital, Neurological Rehabilitation, Limbiate, Italy
| | - Maria Sessa
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Neurology Unit, Neurology Department, IRCCS San Raffaele Hospital, Milan, Italy
| | - Patrizia D’Adamo
- Dulbecco Telethon Institute at Division of Neuroscience, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Alessandro Aiuti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), IRCCS San Raffaele Scientific Institute, Milan, Italy
- Pediatric Immunohematology and Bone Marrow Transplantation Unit, IRCCS San Raffaele Hospital, Milan, Italy
- Vita-Salute San Raffaele University, Milan, Italy
| |
Collapse
|
32
|
Lysosomal Re-acidification Prevents Lysosphingolipid-Induced Lysosomal Impairment and Cellular Toxicity. PLoS Biol 2016; 14:e1002583. [PMID: 27977664 PMCID: PMC5169359 DOI: 10.1371/journal.pbio.1002583] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 11/11/2016] [Indexed: 12/20/2022] Open
Abstract
Neurodegenerative lysosomal storage disorders (LSDs) are severe and untreatable, and mechanisms underlying cellular dysfunction are poorly understood. We found that toxic lipids relevant to three different LSDs disrupt multiple lysosomal and other cellular functions. Unbiased drug discovery revealed several structurally distinct protective compounds, approved for other uses, that prevent lysosomal and cellular toxicities of these lipids. Toxic lipids and protective agents show unexpected convergence on control of lysosomal pH and re-acidification as a critical component of toxicity and protection. In twitcher mice (a model of Krabbe disease [KD]), a central nervous system (CNS)-penetrant protective agent rescued myelin and oligodendrocyte (OL) progenitors, improved motor behavior, and extended lifespan. Our studies reveal shared principles relevant to several LSDs, in which diverse cellular and biochemical disruptions appear to be secondary to disruption of lysosomal pH regulation by specific lipids. These studies also provide novel protective strategies that confer therapeutic benefits in a mouse model of a severe LSD.
Collapse
|
33
|
Alton EWFW, Beekman JM, Boyd AC, Brand J, Carlon MS, Connolly MM, Chan M, Conlon S, Davidson HE, Davies JC, Davies LA, Dekkers JF, Doherty A, Gea-Sorli S, Gill DR, Griesenbach U, Hasegawa M, Higgins TE, Hironaka T, Hyndman L, McLachlan G, Inoue M, Hyde SC, Innes JA, Maher TM, Moran C, Meng C, Paul-Smith MC, Pringle IA, Pytel KM, Rodriguez-Martinez A, Schmidt AC, Stevenson BJ, Sumner-Jones SG, Toshner R, Tsugumine S, Wasowicz MW, Zhu J. Preparation for a first-in-man lentivirus trial in patients with cystic fibrosis. Thorax 2016; 72:137-147. [PMID: 27852956 PMCID: PMC5284333 DOI: 10.1136/thoraxjnl-2016-208406] [Citation(s) in RCA: 99] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2016] [Revised: 06/21/2016] [Accepted: 06/28/2016] [Indexed: 01/03/2023]
Abstract
We have recently shown that non-viral gene therapy can stabilise the decline of lung function in patients with cystic fibrosis (CF). However, the effect was modest, and more potent gene transfer agents are still required. Fuson protein (F)/Hemagglutinin/Neuraminidase protein (HN)-pseudotyped lentiviral vectors are more efficient for lung gene transfer than non-viral vectors in preclinical models. In preparation for a first-in-man CF trial using the lentiviral vector, we have undertaken key translational preclinical studies. Regulatory-compliant vectors carrying a range of promoter/enhancer elements were assessed in mice and human air–liquid interface (ALI) cultures to select the lead candidate; cystic fibrosis transmembrane conductance receptor (CFTR) expression and function were assessed in CF models using this lead candidate vector. Toxicity was assessed and ‘benchmarked’ against the leading non-viral formulation recently used in a Phase IIb clinical trial. Integration site profiles were mapped and transduction efficiency determined to inform clinical trial dose-ranging. The impact of pre-existing and acquired immunity against the vector and vector stability in several clinically relevant delivery devices was assessed. A hybrid promoter hybrid cytosine guanine dinucleotide (CpG)- free CMV enhancer/elongation factor 1 alpha promoter (hCEF) consisting of the elongation factor 1α promoter and the cytomegalovirus enhancer was most efficacious in both murine lungs and human ALI cultures (both at least 2-log orders above background). The efficacy (at least 14% of airway cells transduced), toxicity and integration site profile supports further progression towards clinical trial and pre-existing and acquired immune responses do not interfere with vector efficacy. The lead rSIV.F/HN candidate expresses functional CFTR and the vector retains 90–100% transduction efficiency in clinically relevant delivery devices. The data support the progression of the F/HN-pseudotyped lentiviral vector into a first-in-man CF trial in 2017.
Collapse
Affiliation(s)
- Eric W F W Alton
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Jeffery M Beekman
- Department of Pediatric Pulmonology, Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
| | - A Christopher Boyd
- Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - June Brand
- Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK.,Lung Pathology Unit, Department of Airway Disease Infection, NHLI, Imperial College London, London, UK
| | - Marianne S Carlon
- Laboratory for Molecular Virology and Gene Therapy, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Brussels, Belgium
| | - Mary M Connolly
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Mario Chan
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Sinead Conlon
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Heather E Davidson
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Jane C Davies
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Lee A Davies
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Johanna F Dekkers
- Department of Pediatric Pulmonology, Laboratory of Translational Immunology, Wilhelmina Children's Hospital, University Medical Centre, Utrecht, The Netherlands
| | - Ann Doherty
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Sabrina Gea-Sorli
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Deborah R Gill
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Uta Griesenbach
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | | | - Tracy E Higgins
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | | | - Laura Hyndman
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Gerry McLachlan
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Roslin Institute & R(D)SVS, University of Edinburgh, Midlothian, UK
| | - Makoto Inoue
- ID Pharme Co. Ltd. (DNAVEC Center), Tsukuba, Japan
| | - Stephen C Hyde
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - J Alastair Innes
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Toby M Maher
- Fibrosis Research Group, Inflammation, Repair & Development Section, National Heart and Lung Institute, Sir Alexander Fleming Building, Imperial College, London, UK
| | - Caroline Moran
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Cuixiang Meng
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Michael C Paul-Smith
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Ian A Pringle
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Kamila M Pytel
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Andrea Rodriguez-Martinez
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | | | - Barbara J Stevenson
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Centre for Genomic and Experimental Medicine, IGMM, University of Edinburgh, Edinburgh, UK
| | - Stephanie G Sumner-Jones
- UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK.,Gene Medicine Research Group, NDCLS, John Radcliffe Hospital, Oxford, UK
| | - Richard Toshner
- Fibrosis Research Group, Inflammation, Repair & Development Section, National Heart and Lung Institute, Sir Alexander Fleming Building, Imperial College, London, UK
| | | | - Marguerite W Wasowicz
- Department of Gene Therapy, National Heart and Lung Institute, Imperial College London, London, UK.,UK Cystic Fibrosis Gene Therapy Consortium, Oxford, UK
| | - Jie Zhu
- Lung Pathology Unit, Department of Airway Disease Infection, NHLI, Imperial College London, London, UK
| |
Collapse
|
34
|
Meneghini V, Frati G, Sala D, De Cicco S, Luciani M, Cavazzin C, Paulis M, Mentzen W, Morena F, Giannelli S, Sanvito F, Villa A, Bulfone A, Broccoli V, Martino S, Gritti A. Generation of Human Induced Pluripotent Stem Cell-Derived Bona Fide Neural Stem Cells for Ex Vivo Gene Therapy of Metachromatic Leukodystrophy. Stem Cells Transl Med 2016; 6:352-368. [PMID: 28191778 PMCID: PMC5442804 DOI: 10.5966/sctm.2015-0414] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2015] [Accepted: 08/09/2016] [Indexed: 12/12/2022] Open
Abstract
Allogeneic fetal‐derived human neural stem cells (hfNSCs) that are under clinical evaluation for several neurodegenerative diseases display a favorable safety profile, but require immunosuppression upon transplantation in patients. Neural progenitors derived from patient‐specific induced pluripotent stem cells (iPSCs) may be relevant for autologous ex vivo gene‐therapy applications to treat genetic diseases with unmet medical need. In this scenario, obtaining iPSC‐derived neural stem cells (NSCs) showing a reliable “NSC signature” is mandatory. Here, we generated human iPSC (hiPSC) clones via reprogramming of skin fibroblasts derived from normal donors and patients affected by metachromatic leukodystrophy (MLD), a fatal neurodegenerative lysosomal storage disease caused by genetic defects of the arylsulfatase A (ARSA) enzyme. We differentiated hiPSCs into NSCs (hiPS‐NSCs) sharing molecular, phenotypic, and functional identity with hfNSCs, which we used as a “gold standard” in a side‐by‐side comparison when validating the phenotype of hiPS‐NSCs and predicting their performance after intracerebral transplantation. Using lentiviral vectors, we efficiently transduced MLD hiPSCs, achieving supraphysiological ARSA activity that further increased upon neural differentiation. Intracerebral transplantation of hiPS‐NSCs into neonatal and adult immunodeficient MLD mice stably restored ARSA activity in the whole central nervous system. Importantly, we observed a significant decrease of sulfatide storage when ARSA‐overexpressing cells were used, with a clear advantage in those mice receiving neonatal as compared with adult intervention. Thus, we generated a renewable source of ARSA‐overexpressing iPSC‐derived bona fide hNSCs with improved features compared with clinically approved hfNSCs. Patient‐specific ARSA‐overexpressing hiPS‐NSCs may be used in autologous ex vivo gene therapy protocols to provide long‐lasting enzymatic supply in MLD‐affected brains. Stem Cells Translational Medicine2017;6:352–368
Collapse
Affiliation(s)
- Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| | - Giacomo Frati
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| | - Davide Sala
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| | - Silvia De Cicco
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| | - Marco Luciani
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| | - Chiara Cavazzin
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| | - Marianna Paulis
- National Research Council, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Italy
| | | | - Francesco Morena
- Biochemistry and Molecular Biology Unit, Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Serena Giannelli
- Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| | - Francesca Sanvito
- Anatomy and Histopathology Department, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| | - Anna Villa
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
- National Research Council, Milan, Italy
- Humanitas Clinical and Research Center, Rozzano, Italy
| | | | - Vania Broccoli
- Division of Neuroscience, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| | - Sabata Martino
- Biochemistry and Molecular Biology Unit, Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy, Division of Regenerative Medicine, Stem Cells and Gene Therapy, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele, Milan, Italy
| |
Collapse
|
35
|
Meneghini V, Lattanzi A, Tiradani L, Bravo G, Morena F, Sanvito F, Calabria A, Bringas J, Fisher-Perkins JM, Dufour JP, Baker KC, Doglioni C, Montini E, Bunnell BA, Bankiewicz K, Martino S, Naldini L, Gritti A. Pervasive supply of therapeutic lysosomal enzymes in the CNS of normal and Krabbe-affected non-human primates by intracerebral lentiviral gene therapy. EMBO Mol Med 2016; 8:489-510. [PMID: 27025653 PMCID: PMC5128736 DOI: 10.15252/emmm.201505850] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) and globoid cell leukodystrophy (GLD or Krabbe disease) are severe neurodegenerative lysosomal storage diseases (LSD) caused by arylsulfatase A (ARSA) and galactosylceramidase (GALC) deficiency, respectively. Our previous studies established lentiviral gene therapy (GT) as a rapid and effective intervention to provide pervasive supply of therapeutic lysosomal enzymes in CNS tissues of MLD and GLD mice. Here, we investigated whether this strategy is similarly effective in juvenile non-human primates (NHP). To provide proof of principle for tolerability and biological efficacy of the strategy, we established a comprehensive study in normal NHP delivering a clinically relevant lentiviral vector encoding for the human ARSA transgene. Then, we injected a lentiviral vector coding for the human GALC transgene in Krabbe-affected rhesus macaques, evaluating for the first time the therapeutic potential of lentiviral GT in this unique LSD model. We showed favorable safety profile and consistent pattern of LV transduction and enzyme biodistribution in the two models, supporting the robustness of the proposed GT platform. We documented moderate inflammation at the injection sites, mild immune response to vector particles in few treated animals, no indication of immune response against transgenic products, and no molecular evidence of insertional genotoxicity. Efficient gene transfer in neurons, astrocytes, and oligodendrocytes close to the injection sites resulted in robust production and extensive spreading of transgenic enzymes in the whole CNS and in CSF, leading to supraphysiological ARSA activity in normal NHP and close to physiological GALC activity in the Krabbe NHP, in which biological efficacy was associated with preliminary indication of therapeutic benefit. These results support the rationale for the clinical translation of intracerebral lentiviral GT to address CNS pathology in MLD, GLD, and other neurodegenerative LSD.
Collapse
Affiliation(s)
- Vasco Meneghini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Annalisa Lattanzi
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luigi Tiradani
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Gabriele Bravo
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, Biochemistry and Molecular Biology Unit, University of Perugia, Perugia, Italy
| | - Francesca Sanvito
- Anatomy and Histopathology Department, San Raffaele Scientific Institute, Milano, Italy
| | - Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - John Bringas
- University of California San Francisco (UCSF), San Francisco, CA, USA
| | - Jeanne M Fisher-Perkins
- Division of Regenerative Medicine, Tulane National Primate Research Center, Covington, LA, USA
| | - Jason P Dufour
- Division of Regenerative Medicine, Tulane National Primate Research Center, Covington, LA, USA
| | - Kate C Baker
- Division of Regenerative Medicine, Tulane National Primate Research Center, Covington, LA, USA
| | - Claudio Doglioni
- Anatomy and Histopathology Department, San Raffaele Scientific Institute, Milano, Italy Vita-Salute San Raffaele University, Milan, Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Bruce A Bunnell
- Division of Regenerative Medicine, Tulane National Primate Research Center, Covington, LA, USA
| | | | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, Biochemistry and Molecular Biology Unit, University of Perugia, Perugia, Italy
| | - Luigi Naldini
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy Vita-Salute San Raffaele University, Milan, Italy
| | - Angela Gritti
- San Raffaele Telethon Institute for Gene Therapy (SR-TIGET), Division of Regenerative Medicine, Stem Cells and Gene Therapy, IRCCS San Raffaele Scientific Institute, Milan, Italy
| |
Collapse
|
36
|
|
37
|
Intrathecal gene therapy rescues a model of demyelinating peripheral neuropathy. Proc Natl Acad Sci U S A 2016; 113:E2421-9. [PMID: 27035961 DOI: 10.1073/pnas.1522202113] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023] Open
Abstract
Inherited demyelinating peripheral neuropathies are progressive incurable diseases without effective treatment. To develop a gene therapy approach targeting myelinating Schwann cells that can be translatable, we delivered a lentiviral vector using a single lumbar intrathecal injection and a myelin-specific promoter. The human gene of interest, GJB1, which is mutated in X-linked Charcot-Marie-Tooth Disease (CMT1X), was delivered intrathecally into adult Gjb1-null mice, a genetically authentic model of CMT1X that develops a demyelinating peripheral neuropathy. We obtained widespread, stable, and cell-specific expression of connexin32 in up to 50% of Schwann cells in multiple lumbar spinal roots and peripheral nerves. Behavioral and electrophysiological analysis revealed significantly improved motor performance, quadriceps muscle contractility, and sciatic nerve conduction velocities. Furthermore, treated mice exhibited reduced numbers of demyelinated and remyelinated fibers and fewer inflammatory cells in lumbar motor roots, as well as in the femoral motor and sciatic nerves. This study demonstrates that a single intrathecal lentiviral gene delivery can lead to Schwann cell-specific expression in spinal roots extending to multiple peripheral nerves. This clinically relevant approach improves the phenotype of an inherited neuropathy mouse model and provides proof of principle for treating inherited demyelinating neuropathies.
Collapse
|
38
|
Design of a regulated lentiviral vector for hematopoietic stem cell gene therapy of globoid cell leukodystrophy. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2015; 2:15038. [PMID: 26509184 PMCID: PMC4605225 DOI: 10.1038/mtm.2015.38] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 08/26/2015] [Indexed: 12/21/2022]
Abstract
Globoid cell leukodystrophy (GLD) is a demyelinating lysosomal storage disease due to the deficiency of the galactocerebrosidase (GALC) enzyme. The favorable outcome of hematopoietic stem and progenitor cell (HSPC)-based approaches in GLD and other similar diseases suggests HSPC gene therapy as a promising therapeutic option for patients. The path to clinical development of this strategy was hampered by a selective toxicity of the overexpressed GALC in the HSPC compartment. Here, we presented the optimization of a lentiviral vector (LV) in which miR-126 regulation was coupled to codon optimization of the human GALC cDNA to obtain a selective and enhanced enzymatic activity only upon transduced HSPCs differentiation. The safety of human GALC overexpression driven by this LV was extensively demonstrated in vitro and in vivo on human HSPCs from healthy donors. No perturbation in the content of proapoptotic sphingolipids, gene expression profile, and capability of engraftment and mutlilineage differentiation in chimeric mice was observed. The therapeutic potential of this LV was then assessed in a severe GLD murine model that benefited from transplantation of corrected HSPCs with longer survival and ameliorated phenotype as compared to untreated siblings. This construct has thus been selected as a candidate for clinical translation.
Collapse
|
39
|
Guo XZ, Ye XL, Xiao WZ, Wei XN, You QH, Che XH, Cai YJ, Chen F, Yuan H, Liu XJ, Yu MH. Downregulation of VMP1 confers aggressive properties to colorectal cancer. Oncol Rep 2015; 34:2557-66. [PMID: 26328607 DOI: 10.3892/or.2015.4240] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Accepted: 08/07/2015] [Indexed: 11/05/2022] Open
Abstract
Vacuole membrane protein 1 (VMP1) was recently found to be involved in the process of tumor metastasis and is also considered to play a vital role in balancing apoptosis and autophagy. In the present study, the expression of VMP1 in colorectal cancer and matched adjacent non‑cancerous tissues was evaluated by immunohistochemistry (IHC) for studying the role of VMP1 in the process of colorectal cancer. Kaplan‑Meier analysis and the log-rank test were used to calculate the correlation of classic clinicopathological characteristics related to survival and the expression of VMP1. In vitro, a VMP1 stable gene silencing cell model was constructed using a lentiviral vector. The invasive ability and proliferation of colorectal cancer cells were evaluated by Transwell and MTT assays, respectively, and the underlying signaling pathway was explored by western blotting. Additionally, drug susceptibility to cisplatin, oxaliplatin and 5-FU was tested before and after VMP1 knockout. Finally, an animal model was constructed to explore the role of VMP1 in the physiopathologic process of colorectal cancer. Our results indicated that VMP1 showed increased expression in the adjacent non-cancer tissues compared with that in the colorectal cancer tissues. For different stages of colorectal cancer, expression of VMP1 had a negative correlation with the malignancy of the cancer. In clinical research, we also found that the median survival of patients with low VMP1 expression was much shorter than the survival of patients with high expression. In vitro, after infection with the lentivirus, cells with VMP1 knockout gained significant aggressive properties in regards to invasion and proliferation, and the mechanisms may be related to the activation of the PI3K/Akt/ZO-1/E-cadherin pathway. We also found that shVMP1 cells were more sensitive to 5-FU, but not cisplatin and oxaliplatin. Finally, we found a higher number of formed nodules in nude mice after intraperitoneal injection with shVMP1 cells in the in vivo study.
Collapse
Affiliation(s)
- Xian-Zhi Guo
- Department of Medical Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, P.R. China
| | - Xiao-Lei Ye
- Drugs and Pharmacology Laboratory, Ningbo Institute of Medical Sciences, Ningbo, Zhejiang 315020, P.R. China
| | - Wei-Zhong Xiao
- Department of Internal Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, P.R. China
| | - Xue-Ni Wei
- Department of Pharmacology, China Pharmaceutical University, Nanjing, Jiangsu 211198, P.R. China
| | - Qing-Hua You
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, P.R. China
| | - Xiao-Hang Che
- Drugs and Pharmacology Laboratory, Ningbo Institute of Medical Sciences, Ningbo, Zhejiang 315020, P.R. China
| | - Yan-Jun Cai
- Department of Internal Neurology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, P.R. China
| | - Fang Chen
- Department of Hematology, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P.R. China
| | - Hao Yuan
- Department of General Surgery, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, P.R. China
| | - Xiao-Jian Liu
- Department of Medical Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, P.R. China
| | - Ming-Hua Yu
- Department of Medical Oncology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Pudong, Shanghai 201399, P.R. China
| |
Collapse
|
40
|
Isaacs CJ, Shinnick JE, Schadt K, Lynch DR, Lin KY. Prospects of gene and cell therapy for managing cardiac complications in Friedreich ataxia. Expert Opin Orphan Drugs 2015. [DOI: 10.1517/21678707.2015.1083854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
41
|
Kagiava A, Sargiannidou I, Bashiardes S, Richter J, Schiza N, Christodoulou C, Gritti A, Kleopa KA. Gene delivery targeted to oligodendrocytes using a lentiviral vector. J Gene Med 2015; 16:364-73. [PMID: 25394283 DOI: 10.1002/jgm.2813] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Revised: 10/22/2014] [Accepted: 11/07/2014] [Indexed: 12/29/2022] Open
Abstract
BACKGROUND Most leukodystrophies result from mutations in genes expressed in oligodendrocytes that may cause autonomous loss of function of cell structural proteins. Therefore, effective gene delivery to oligodendrocytes is necessary to develop future treatments. MATERIALS To achieve this, we cloned a lentiviral vector in which the enhanced green fluorescent protein (EGFP) expression was driven by the oligodendrocyte specific 2,3-cyclic nucleotide 3-phosphodiesterase promoter. The vector was inserted into C57BL/6 neonatal mouse brain by combined intraventricular and parenchymal injections. RESULTS Assessment of EGFP expression revealed a widespread distribution, specifically in cells of the oligodendrocyte linage, starting from postnatal day 6 (P6) in the subventricular zone and spreading through migrating oligodendrocyte precursors. By P30, it was detectable throughout the brain and persisted for at least 3 months, showing an increase both in the number of expressing cells and in intensity over time. EGFP expression was restricted to oligodendrocyte linage cells. On average, 20.3 ± 2.56% of all oligodendrocytes in different central nervous system areas were EGFP-positive, with regional variations. CONCLUSIONS Lentiviral gene delivery using an oligodendrocyte-specific promoter may achieve widespread and long-lasting expression selectively in oligodendrocytes, offering a possibility for gene therapy in certain leukodystrophies, although the relatively low rates of oligodendrocyte transduction are a limitation that remains to be overcome.
Collapse
Affiliation(s)
- Alexia Kagiava
- Neuroscience Laboratory, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Sargiannidou I, Kagiava A, Bashiardes S, Richter J, Christodoulou C, Scherer SS, Kleopa KA. Intraneural GJB1 gene delivery improves nerve pathology in a model of X-linked Charcot-Marie-Tooth disease. Ann Neurol 2015; 78:303-16. [PMID: 26010264 DOI: 10.1002/ana.24441] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 11/11/2022]
Abstract
OBJECTIVE X-linked Charcot-Marie-Tooth disease (CMT1X) is a common inherited neuropathy caused by mutations in the GJB1 gene encoding the gap junction protein connexin32 (Cx32). Clinical studies and disease models indicate that neuropathy mainly results from Schwann cell autonomous, loss-of-function mechanisms; therefore, CMT1X may be treatable by gene replacement. METHODS A lentiviral vector LV.Mpz-GJB1 carrying the GJB1 gene under the Schwann cell-specific myelin protein zero (Mpz) promoter was generated and delivered into the mouse sciatic nerve by a single injection immediately distal to the sciatic notch. Enhanced green fluorescent protein (EGFP) reporter gene expression was quantified and Cx32 expression was examined on a Cx32 knockout (KO) background. A gene therapy trial was performed in a Cx32 KO model of CMT1X. RESULTS EGFP was expressed throughout the length of the sciatic nerve in up to 50% of Schwann cells starting 2 weeks after injection and remaining stable for up to 16 weeks. Following LV.Mpz-GJB1 injection into Cx32 KO nerves, we detected Cx32 expression and correct localization in non-compact myelin areas where gap junctions are normally formed. Gene therapy trial by intraneural injection in groups of 2-month-old Cx32 KO mice, before demyelination onset, significantly reduced the ratio of abnormally myelinated fibers (p = 0.00148) and secondary inflammation (p = 0.0178) at 6 months of age compared to mock-treated animals. INTERPRETATION Gene delivery using a lentiviral vector leads to efficient gene expression specifically in Schwann cells. Restoration of Cx32 expression ameliorates nerve pathology in a disease model and provides a promising approach for future treatments of CMT1X and other inherited neuropathies.
Collapse
Affiliation(s)
| | | | - Stavros Bashiardes
- Department of Molecular Virology, Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Jan Richter
- Department of Molecular Virology, Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Christina Christodoulou
- Department of Molecular Virology, Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| | - Steven S Scherer
- Department of Neurology, University of Pennsylvania, Philadelphia, PA
| | - Kleopas A Kleopa
- Neuroscience Laboratory
- Neurology Clinics, Cyprus Institute of Neurology and Genetics and Cyprus School of Molecular Medicine, Nicosia, Cyprus
| |
Collapse
|
43
|
Bicchi I, Emiliani C, Vescovi A, Martino S. The Big Bluff of Amyotrophic Lateral Sclerosis Diagnosis: The Role of Neurodegenerative Disease Mimics. NEURODEGENER DIS 2015; 15:313-21. [PMID: 26227992 DOI: 10.1159/000435917] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 06/12/2015] [Indexed: 11/19/2022] Open
Abstract
Neurodegenerative diseases include a significant number of pathologies affecting the nervous system. Generally, the primary cause of each disease is specific; however, recently, it was shown that they may be correlated at molecular level. This aspect, together with the exhibition of similar symptoms, renders the diagnosis of these disorders difficult. Amyotrophic lateral sclerosis is one of these pathologies. Herein, we report several cases of amyotrophic lateral sclerosis misdiagnosed as a consequence of features that are common to several neurodegenerative diseases, such as Parkinson's, Huntington's and Alzheimer's disease, spinal muscular atrophy, progressive bulbar palsy, spastic paraplegia and frontotemporal dementia, and mostly with the lysosomal storage disorder GM2 gangliosidosis. Overall reports highlight that the differential diagnosis for amyotrophic lateral sclerosis should include correlated mechanisms.
Collapse
Affiliation(s)
- Ilaria Bicchi
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, Perugia, Italy
| | | | | | | |
Collapse
|
44
|
Evaluation of transduction properties of an adenovirus vector in neonatal mice. BIOMED RESEARCH INTERNATIONAL 2015; 2015:685374. [PMID: 26075257 PMCID: PMC4444570 DOI: 10.1155/2015/685374] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/03/2015] [Accepted: 04/30/2015] [Indexed: 12/05/2022]
Abstract
In gene therapy for congenital disorders, treatments during neonate and infant stages are promising. Replication-incompetent adenovirus (Ad) vectors have been used in gene therapy studies of genetic disorders; however, the transduction properties of Ad vectors in neonates and infants have not been fully examined. Accordingly, this study examined the properties of Ad vector-mediated transduction in neonatal mice. A first-generation Ad vector containing a cytomegalovirus (CMV) promoter-driven luciferase expression cassette was administered to neonatal mice on the second day of life via retro-orbital sinus. The highest Ad vector genome copy numbers and transgene expression were found in the neonatal liver. The neonatal heart exhibited the second highest levels of transgene expression among the organs examined. There was an approximately 1500-fold difference in the transgene expression levels between the adult liver and heart, while the neonatal liver exhibited only an approximately 30-fold higher level of transgene expression than the neonatal heart. A liver-specific promoter for firefly luciferase expression conferred a more than 100-fold higher luciferase expression in the liver relative to the other organs. No apparent hepatotoxicity was observed in neonatal mice following Ad vector administration. These findings should provide valuable information for gene therapy using Ad vectors in neonates and infants.
Collapse
|
45
|
Kuai XL, Ni RZ, Zhou GX, Mao ZB, Zhang JF, Yi N, Liu ZX, Shao N, Ni WK, Wang ZW. Transplantation of mouse embryonic stem cell-derived oligodendrocytes in the murine model of globoid cell leukodystrophy. Stem Cell Res Ther 2015; 6:30. [PMID: 25888852 PMCID: PMC4413525 DOI: 10.1186/s13287-015-0024-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2014] [Revised: 02/26/2015] [Accepted: 02/26/2015] [Indexed: 01/18/2023] Open
Abstract
Introduction Globoid cell leukodystrophy (GLD) is a severe disorder of the central and peripheral nervous system caused by the absence of galactocerebrosidase (GALC) activity. Cell-based therapies are highly promising strategies for GLD. In this study, G-Olig2 mouse embryonic stem cells (ESCs) were induced into oligodendrocyte progenitor cells (OPCs) and were implanted into the brains of twitcher mice, an animal model of GLD, to explore the therapeutic potential of the cells. Methods The G-Olig2 ESCs were induced into OPCs by using cytokines and a multi-step differentiation procedure. Oligodendrocyte markers were detected by reverse transcription-polymerase chain reaction (RT-PCR) and immunocytochemistry. The toxicity of psychosine to OPCs was determined by a cell proliferation assay kit. The GALC level of OPCs was also examined. OPCs were labeled with Dir and transplanted into the brains of twitcher mice. The transplanted cells were detected by in-Vivo Multispectral Imaging System and real-time PCR. The physiological effects of twitcher mice were assessed. Results Oligodendrocyte markers were expressed in OPCs, and 76% ± 5.76% of the OPCs were enhanced green fluorescent protein (eGFP)-positive, eGFP was driven by the Olig2 promoter. The effect of psychosine on cell viability indicated that OPCs were more resistant to psychosine toxicity. The GALC level of OPCs was 10.0 ± 1.23 nmol/hour per mg protein, which was significantly higher than other cells. Dir-labeled OPCs were injected into the forebrain of post-natal day 10 twitcher mice. The transplanted OPCs were myelin basic protein (MBP)-positive and remained along the injection tract as observed by fluorescent microscopy. The level of the Dir fluorescent signal and eGFP mRNA significantly decreased at days 10 and 20 after injection, as indicated by in-Vivo Multispectral Imaging System and real-time PCR. Because of poor cell survival and limited migration ability, there was no significant improvement in brain GALC activity, MBP level, life span, body weight, and behavioral deficits of twitcher mice. Conclusions ESC-derived OPC transplantation was not sufficient to reverse the clinical course of GLD in twitcher mice.
Collapse
Affiliation(s)
- Xiao Ling Kuai
- Department of Gastroenterology, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| | - Run Zhou Ni
- Department of Gastroenterology, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| | - Guo Xiong Zhou
- Department of Gastroenterology, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| | - Zheng Biao Mao
- Department of Gastroenterology, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| | - Jian Feng Zhang
- Department of Gastroenterology, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| | - Nan Yi
- Department of Gastroenterology, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| | - Zhao Xiu Liu
- Department of Gastroenterology, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| | - Nan Shao
- Department of Gastroenterology, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| | - Wen Kai Ni
- Department of Gastroenterology, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| | - Zhi Wei Wang
- Department of General Surgery, Nantong University Affiliated Hospital, 20 Xi Si Road, Nantong, Jiangsu, 226001, China.
| |
Collapse
|
46
|
Ricca A, Rufo N, Ungari S, Morena F, Martino S, Kulik W, Alberizzi V, Bolino A, Bianchi F, Del Carro U, Biffi A, Gritti A. Combined gene/cell therapies provide long-term and pervasive rescue of multiple pathological symptoms in a murine model of globoid cell leukodystrophy. Hum Mol Genet 2015; 24:3372-89. [PMID: 25749991 PMCID: PMC4498152 DOI: 10.1093/hmg/ddv086] [Citation(s) in RCA: 58] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2015] [Accepted: 03/04/2015] [Indexed: 01/11/2023] Open
Abstract
Globoid cell leukodystrophy (GLD) is a lysosomal storage disease caused by deficient activity of β-galactocerebrosidase (GALC). The infantile forms manifest with rapid and progressive central and peripheral demyelination, which represent a major hurdle for any treatment approach. We demonstrate here that neonatal lentiviral vector-mediated intracerebral gene therapy (IC GT) or transplantation of GALC-overexpressing neural stem cells (NSC) synergize with bone marrow transplant (BMT) providing dramatic extension of lifespan and global clinical–pathological rescue in a relevant GLD murine model. We show that timely and long-lasting delivery of functional GALC in affected tissues ensured by the exclusive complementary mode of action of the treatments underlies the outstanding benefit. In particular, the contribution of neural stem cell transplantation and IC GT during the early asymptomatic stage of the disease is instrumental to enhance long-term advantage upon BMT. We clarify the input of central nervous system, peripheral nervous system and periphery to the disease, and the relative contribution of treatments to the final therapeutic outcome, with important implications for treatment strategies to be tried in human patients. This study gives proof-of-concept of efficacy, tolerability and clinical relevance of the combined gene/cell therapies proposed here, which may constitute a feasible and effective therapeutic opportunity for children affected by GLD.
Collapse
Affiliation(s)
- Alessandra Ricca
- San Raffaele Scientific Institute, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (TIGET), Via Olgettina 58, Milano 20132, Italy
| | - Nicole Rufo
- San Raffaele Scientific Institute, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (TIGET), Via Olgettina 58, Milano 20132, Italy
| | - Silvia Ungari
- San Raffaele Scientific Institute, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (TIGET), Via Olgettina 58, Milano 20132, Italy
| | - Francesco Morena
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, via del Giochetto, Perugia, Italy
| | - Sabata Martino
- Department of Chemistry, Biology and Biotechnologies, University of Perugia, via del Giochetto, Perugia, Italy
| | - Wilem Kulik
- Laboratory of Genetic Metabolic Diseases, Academic Medical Center AMC, University of Amsterdam, Meibergdreef 9, Amsterdam, The Netherlands and
| | - Valeria Alberizzi
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, Milano, Italy
| | - Alessandra Bolino
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, Milano, Italy
| | - Francesca Bianchi
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, Milano, Italy
| | - Ubaldo Del Carro
- Division of Neuroscience, San Raffaele Scientific Institute, INSPE, Via Olgettina 58, Milano, Italy
| | - Alessandra Biffi
- San Raffaele Scientific Institute, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (TIGET), Via Olgettina 58, Milano 20132, Italy
| | - Angela Gritti
- San Raffaele Scientific Institute, Division of Regenerative Medicine, Stem Cells and Gene Therapy, San Raffaele Telethon Institute for Gene Therapy (TIGET), Via Olgettina 58, Milano 20132, Italy,
| |
Collapse
|
47
|
Graziano ACE, Cardile V. History, genetic, and recent advances on Krabbe disease. Gene 2014; 555:2-13. [PMID: 25260228 DOI: 10.1016/j.gene.2014.09.046] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2014] [Revised: 09/18/2014] [Accepted: 09/19/2014] [Indexed: 12/20/2022]
Abstract
Krabbe disease or globoid cell leukodystrophy is one of the classic genetic lysosomal storage diseases with autosomal recessive inheritance that affects both central and peripheral nervous systems in several species including humans, rhesus macaques, dogs, mice, and sheep. Since its identification in 1916, lots of scientific investigations were made to define the cause, to evaluate the molecular mechanisms of the damage and to develop more efficient therapies inducing clinical benefit and ameliorating the patients' quality of life. This manuscript gives a historical overview and summarizes the new recent findings about Krabbe disease. Human symptoms and phenotypes, gene encoding for β-galactocerebrosidase and encoded protein were described. Indications about the classical mutations were reported and some specific mutations in restricted geographical area, like the north of Catania City (Italy), were added. Briefly, here we present a mix of past and present investigations on Krabbe disease in order to update the knowledge on its genetic history and molecular mechanisms and to move new scientific investigations.
Collapse
Affiliation(s)
| | - Venera Cardile
- Department of Bio-Medical Science - Physiology Section, University of Catania, Catania, Italy.
| |
Collapse
|
48
|
Calabria A, Leo S, Benedicenti F, Cesana D, Spinozzi G, Orsini M, Merella S, Stupka E, Zanetti G, Montini E. VISPA: a computational pipeline for the identification and analysis of genomic vector integration sites. Genome Med 2014; 6:67. [PMID: 25342980 PMCID: PMC4169225 DOI: 10.1186/s13073-014-0067-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 08/22/2014] [Indexed: 11/10/2022] Open
Abstract
The analysis of the genomic distribution of viral vector genomic integration sites is a key step in hematopoietic stem cell-based gene therapy applications, allowing to assess both the safety and the efficacy of the treatment and to study the basic aspects of hematopoiesis and stem cell biology. Identifying vector integration sites requires ad-hoc bioinformatics tools with stringent requirements in terms of computational efficiency, flexibility, and usability. We developed VISPA (Vector Integration Site Parallel Analysis), a pipeline for automated integration site identification and annotation based on a distributed environment with a simple Galaxy web interface. VISPA was successfully used for the bioinformatics analysis of the follow-up of two lentiviral vector-based hematopoietic stem-cell gene therapy clinical trials. Our pipeline provides a reliable and efficient tool to assess the safety and efficacy of integrating vectors in clinical settings.
Collapse
Affiliation(s)
- Andrea Calabria
- San Raffaele Telethon Institute for Gene Therapy (TIGET), San Raffaele Scientific Institute, 20132 Milano, Italy
| | - Simone Leo
- Center for Advanced Studies, Research and Development in Sardinia (CRS4), 09010 Pula, CA Italy ; Università degli Studi di Cagliari, 09124 Cagliari, Italy
| | - Fabrizio Benedicenti
- San Raffaele Telethon Institute for Gene Therapy (TIGET), San Raffaele Scientific Institute, 20132 Milano, Italy
| | - Daniela Cesana
- San Raffaele Telethon Institute for Gene Therapy (TIGET), San Raffaele Scientific Institute, 20132 Milano, Italy
| | - Giulio Spinozzi
- San Raffaele Telethon Institute for Gene Therapy (TIGET), San Raffaele Scientific Institute, 20132 Milano, Italy ; Department of Informatics, Systems and Communication (DISCo) - University of Milano-Bicocca, Milano, Italy
| | - Massimilano Orsini
- Center for Advanced Studies, Research and Development in Sardinia (CRS4), 09010 Pula, CA Italy
| | - Stefania Merella
- Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy
| | - Elia Stupka
- Center for Translational Genomics and Bioinformatics, San Raffaele Scientific Institute, Via Olgettina 58, 20132 Milano, Italy
| | - Gianluigi Zanetti
- Center for Advanced Studies, Research and Development in Sardinia (CRS4), 09010 Pula, CA Italy
| | - Eugenio Montini
- San Raffaele Telethon Institute for Gene Therapy (TIGET), San Raffaele Scientific Institute, 20132 Milano, Italy
| |
Collapse
|