601
|
Deverman BE, Ravina BM, Bankiewicz KS, Paul SM, Sah DWY. Gene therapy for neurological disorders: progress and prospects. Nat Rev Drug Discov 2018; 17:641-659. [DOI: 10.1038/nrd.2018.110] [Citation(s) in RCA: 175] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
602
|
Brooks AK, Gaj T. Innovations in CRISPR technology. Curr Opin Biotechnol 2018; 52:95-101. [DOI: 10.1016/j.copbio.2018.03.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 03/20/2018] [Indexed: 12/25/2022]
|
603
|
Chen B, Li Y, Yu B, Zhang Z, Brommer B, Williams PR, Liu Y, Hegarty SV, Zhou S, Zhu J, Guo H, Lu Y, Zhang Y, Gu X, He Z. Reactivation of Dormant Relay Pathways in Injured Spinal Cord by KCC2 Manipulations. Cell 2018; 174:521-535.e13. [PMID: 30033363 PMCID: PMC6063786 DOI: 10.1016/j.cell.2018.06.005] [Citation(s) in RCA: 121] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2017] [Revised: 03/24/2018] [Accepted: 05/31/2018] [Indexed: 12/22/2022]
Abstract
Many human spinal cord injuries are anatomically incomplete but exhibit complete paralysis. It is unknown why spared axons fail to mediate functional recovery in these cases. To investigate this, we undertook a small-molecule screen in mice with staggered bilateral hemisections in which the lumbar spinal cord is deprived of all direct brain-derived innervation, but dormant relay circuits remain. We discovered that a KCC2 agonist restored stepping ability, which could be mimicked by selective expression of KCC2, or hyperpolarizing DREADDs, in the inhibitory interneurons between and around the staggered spinal lesions. Mechanistically, these treatments transformed this injury-induced dysfunctional spinal circuit to a functional state, facilitating the relay of brain-derived commands toward the lumbar spinal cord. Thus, our results identify spinal inhibitory interneurons as a roadblock limiting the integration of descending inputs into relay circuits after injury and suggest KCC2 agonists as promising treatments for promoting functional recovery after spinal cord injury.
Collapse
Affiliation(s)
- Bo Chen
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Yi Li
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Bin Yu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001 Jiangsu, China
| | - Zicong Zhang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Benedikt Brommer
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Philip Raymond Williams
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Yuanyuan Liu
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Shane Vincent Hegarty
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Songlin Zhou
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001 Jiangsu, China
| | - Junjie Zhu
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Hong Guo
- Department of Neurosurgery, Brigham and Women's Hospital, 60 Fenwood Road., BTM 4th Floor, Boston, MA 02115, USA
| | - Yi Lu
- Department of Neurosurgery, Brigham and Women's Hospital, 60 Fenwood Road., BTM 4th Floor, Boston, MA 02115, USA
| | - Yiming Zhang
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA
| | - Xiaosong Gu
- Key Laboratory of Neuroregeneration of Jiangsu and Ministry of Education, Co-innovation Center of Neuroregeneration, Nantong University, Nantong, 226001 Jiangsu, China.
| | - Zhigang He
- F.M. Kirby Neurobiology Center, Boston Children's Hospital, and Department of Neurology, Harvard Medical School, 300 Longwood Avenue, Boston, MA 02115, USA.
| |
Collapse
|
604
|
Cell reprogramming approaches in gene- and cell-based therapies for Parkinson's disease. J Control Release 2018; 286:114-124. [PMID: 30026082 DOI: 10.1016/j.jconrel.2018.07.017] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2018] [Revised: 06/26/2018] [Accepted: 07/10/2018] [Indexed: 12/17/2022]
Abstract
Degeneration of dopamine (DA) neurons in the substantia nigra pars compacta is the pathological hallmark of Parkinson's disease (PD). In PD multiple pathogenic mechanisms initiate and drive this neurodegenerative process, making the development of effective treatments challenging. To date, PD patients are primarily treated with dopaminergic drugs able to temporarily enhance DA levels, therefore relieving motor symptoms. However, the drawbacks of these therapies including the inability to alter disease progression are constantly supporting the search for alternative treatment approaches. Over the past years efforts have been put into the development of new therapeutic strategies based on the delivery of therapeutic genes using viral vectors or transplantation of DA neurons for cell-based DA replacement. Here, past achievements and recent advances in gene- and cell-based therapies for PD are outlined. We discuss how current gene and cell therapy strategies hold great promise for the treatment of PD and how the use of stem cells and recent developments in cellular reprogramming could contribute to open a new avenue in PD therapy.
Collapse
|
605
|
More expansive gene transfer to the rat CNS: AAV PHP.EB vector dose-response and comparison to AAV PHP.B. Gene Ther 2018; 25:392-400. [PMID: 30013186 DOI: 10.1038/s41434-018-0028-5] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Revised: 05/02/2018] [Accepted: 06/07/2018] [Indexed: 11/09/2022]
Abstract
Engineered recombinant adeno-associated virus (AAV) vectors have advanced the transduction of neurons in the CNS on an expansive, wide-scale basis since the papers first using AAV9 for this purpose. Wide-scale CNS expression is relevant to gene therapy as well as indispensable for basic studies such as disease modeling. For example, the wide-scale gene transfer approach could expedite hypothesis testing in vivo relative to the generation of germ-line transgenic mice for all of the genes of interest. Wide-scale gene transfer is more efficient in neonates than in adults, so improving gene transfer efficiency in adults is an important goal. Here we characterized the relatively novel AAV PHP.EB vector for expansive gene transfer in the CNS of adult rats at three doses. The dose-response data were consistent; expression levels can be controlled in a reproducible manner in the rat from moderate to robust levels. Within the CNS, the AAV PHP.EB-derived expression was neuron-selective to neuron-specific, while outside the CNS, organs such as the liver and heart were transduced by the parenteral gene delivery. Though we demonstrated graded expression levels, only the high dose, 1.2 × 1014 vector genomes/kg, yielded efficient expression in spinal cord motor neurons of the adult rat, so this vector dose would be required for models of spinal cord motor neuron disease. The neuronal expression in the rat CNS was greater with AAV PHP.EB than the previous engineered vector AAV PHP.B. AAV PHP.EB is thus one of the most efficient AAV vectors in the field for CNS gene transfer.
Collapse
|
606
|
Naidoo J, Stanek LM, Ohno K, Trewman S, Samaranch L, Hadaczek P, O'Riordan C, Sullivan J, San Sebastian W, Bringas JR, Snieckus C, Mahmoodi A, Mahmoodi A, Forsayeth J, Bankiewicz KS, Shihabuddin LS. Extensive Transduction and Enhanced Spread of a Modified AAV2 Capsid in the Non-human Primate CNS. Mol Ther 2018; 26:2418-2430. [PMID: 30057240 DOI: 10.1016/j.ymthe.2018.07.008] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2018] [Revised: 06/28/2018] [Accepted: 07/03/2018] [Indexed: 11/17/2022] Open
Abstract
The present study was designed to characterize transduction of non-human primate brain and spinal cord with a modified adeno-associated virus serotype 2, incapable of binding to the heparan sulfate proteoglycan receptor, referred to as AAV2-HBKO. AAV2-HBKO was infused into the thalamus, intracerebroventricularly or via a combination of both intracerebroventricular and thalamic delivery. Thalamic injection of this modified vector encoding GFP resulted in widespread CNS transduction that included neurons in deep cortical layers, deep cerebellar nuclei, several subcortical regions, and motor neuron transduction in the spinal cord indicative of robust bidirectional axonal transport. Intracerebroventricular delivery similarly resulted in widespread cortical transduction, with one striking distinction that oligodendrocytes within superficial layers of the cortex were the primary cell type transduced. Robust motor neuron transduction was also observed in all levels of the spinal cord. The combination of thalamic and intracerebroventricular delivery resulted in transduction of oligodendrocytes in superficial cortical layers and neurons in deeper cortical layers. Several subcortical regions were also transduced. Our data demonstrate that AAV2-HBKO is a powerful vector for the potential treatment of a wide number of neurological disorders, and highlight that delivery route can significantly impact cellular tropism and pattern of CNS transduction.
Collapse
Affiliation(s)
- Jerusha Naidoo
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Lisa M Stanek
- CNS Genetic Diseases, Neuroscience Research TA, Sanofi, Framingham, MA, USA
| | - Kousaku Ohno
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Savanah Trewman
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Lluis Samaranch
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Piotr Hadaczek
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | | | - Jennifer Sullivan
- CNS Genetic Diseases, Neuroscience Research TA, Sanofi, Framingham, MA, USA
| | - Waldy San Sebastian
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - John R Bringas
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Christopher Snieckus
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Amin Mahmoodi
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Amir Mahmoodi
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - John Forsayeth
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA
| | - Krystof S Bankiewicz
- Interventional Neuro Center, Department of Neurological Surgery, University of California, San Francisco, San Francisco, CA, USA.
| | | |
Collapse
|
607
|
Drinnenberg A, Franke F, Morikawa RK, Jüttner J, Hillier D, Hantz P, Hierlemann A, Azeredo da Silveira R, Roska B. How Diverse Retinal Functions Arise from Feedback at the First Visual Synapse. Neuron 2018; 99:117-134.e11. [PMID: 29937281 PMCID: PMC6101199 DOI: 10.1016/j.neuron.2018.06.001] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 03/20/2018] [Accepted: 06/01/2018] [Indexed: 11/21/2022]
Abstract
Many brain regions contain local interneurons of distinct types. How does an interneuron type contribute to the input-output transformations of a given brain region? We addressed this question in the mouse retina by chemogenetically perturbing horizontal cells, an interneuron type providing feedback at the first visual synapse, while monitoring the light-driven spiking activity in thousands of ganglion cells, the retinal output neurons. We uncovered six reversible perturbation-induced effects in the response dynamics and response range of ganglion cells. The effects were enhancing or suppressive, occurred in different response epochs, and depended on the ganglion cell type. A computational model of the retinal circuitry reproduced all perturbation-induced effects and led us to assign specific functions to horizontal cells with respect to different ganglion cell types. Our combined experimental and theoretical work reveals how a single interneuron type can differentially shape the dynamical properties of distinct output channels of a brain region.
Collapse
Affiliation(s)
- Antonia Drinnenberg
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Faculty of Natural Sciences, University of Basel, 4003 Basel, Switzerland
| | - Felix Franke
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering of ETH Zurich, 4058 Basel, Switzerland
| | - Rei K Morikawa
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland
| | - Josephine Jüttner
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland
| | - Daniel Hillier
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland
| | - Peter Hantz
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Andreas Hierlemann
- Bio Engineering Laboratory, Department of Biosystems Science and Engineering of ETH Zurich, 4058 Basel, Switzerland
| | - Rava Azeredo da Silveira
- Department of Physics, Ecole Normale Supérieure, 75005 Paris, France; Laboratoire de Physique Statistique, École Normale Supérieure, PSL Research University; Université Paris Diderot Sorbonne Paris-Cité; Sorbonne Universités UPMC Univ Paris 06; CNRS, 75005 Paris, France; Princeton Neuroscience Institute, Princeton University, Princeton, NJ 08544, USA.
| | - Botond Roska
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; Institute of Molecular and Clinical Ophthalmology Basel, 4031 Basel, Switzerland; Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland.
| |
Collapse
|
608
|
AAVvector-mediated in vivo reprogramming into pluripotency. Nat Commun 2018; 9:2651. [PMID: 29985406 PMCID: PMC6037684 DOI: 10.1038/s41467-018-05059-x] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2017] [Accepted: 05/14/2018] [Indexed: 12/19/2022] Open
Abstract
In vivo reprogramming of somatic cells into induced pluripotent stem cells (iPSC) holds vast potential for basic research and regenerative medicine. However, it remains hampered by a need for vectors to express reprogramming factors (Oct-3/4, Klf4, Sox2, c-Myc; OKSM) in selected organs. Here, we report OKSM delivery vectors based on pseudotyped Adeno-associated virus (AAV). Using the AAV-DJ capsid, we could robustly reprogram mouse embryonic fibroblasts with low vector doses. Swapping to AAV8 permitted to efficiently reprogram somatic cells in adult mice by intravenous vector delivery, evidenced by hepatic or extra-hepatic teratomas and iPSC in the blood. Notably, we accomplished full in vivo reprogramming without c-Myc. Most iPSC generated in vitro or in vivo showed transcriptionally silent, intronic or intergenic vector integration, likely reflecting the increased host genome accessibility during reprogramming. Our approach crucially advances in vivo reprogramming technology, and concurrently facilitates investigations into the mechanisms and consequences of AAV persistence. In vivo reprogramming of somatic cells is hampered by the need for vectors to express the OKSM factors in selected organs. Here the authors report new AAV-based vectors capable of in vivo reprogramming at low doses.
Collapse
|
609
|
Bedbrook CN, Deverman BE, Gradinaru V. Viral Strategies for Targeting the Central and Peripheral Nervous Systems. Annu Rev Neurosci 2018; 41:323-348. [DOI: 10.1146/annurev-neuro-080317-062048] [Citation(s) in RCA: 94] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recombinant viruses allow for targeted transgene expression in specific cell populations throughout the nervous system. The adeno-associated virus (AAV) is among the most commonly used viruses for neuroscience research. Recombinant AAVs (rAAVs) are highly versatile and can package most cargo composed of desired genes within the capsid's ∼5-kb carrying capacity. Numerous regulatory elements and intersectional strategies have been validated in rAAVs to enable cell type–specific expression. rAAVs can be delivered to specific neuronal populations or globally throughout the animal. The AAV capsids have natural cell type or tissue tropism and trafficking that can be modified for increased specificity. Here, we describe recently engineered AAV capsids and associated cargo that have extended the utility of AAVs in targeting molecularly defined neurons throughout the nervous system, which will further facilitate neuronal circuit interrogation and discovery.
Collapse
Affiliation(s)
- Claire N. Bedbrook
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Benjamin E. Deverman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California 91125, USA
| |
Collapse
|
610
|
Szablowski JO, Lee-Gosselin A, Lue B, Malounda D, Shapiro MG. Acoustically targeted chemogenetics for the non-invasive control of neural circuits. Nat Biomed Eng 2018; 2:475-484. [PMID: 30948828 DOI: 10.1038/s41551-018-0258-2] [Citation(s) in RCA: 83] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2017] [Accepted: 06/05/2018] [Indexed: 01/22/2023]
Abstract
Neurological and psychiatric disorders are often characterized by dysfunctional neural circuits in specific regions of the brain. Existing treatment strategies, including the use of drugs and implantable brain stimulators, aim to modulate the activity of these circuits. However, they are not cell-type-specific, lack spatial targeting or require invasive procedures. Here, we report a cell-type-specific and non-invasive approach based on acoustically targeted chemogenetics that enables the modulation of neural circuits with spatiotemporal specificity. The approach uses ultrasound waves to transiently open the blood-brain barrier and transduce neurons at specific locations in the brain with virally encoded engineered G-protein-coupled receptors. The engineered neurons subsequently respond to systemically administered designer compounds to activate or inhibit their activity. In a mouse model of memory formation, the approach can modify and subsequently activate or inhibit excitatory neurons within the hippocampus, with selective control over individual brain regions. This technology overcomes some of the key limitations associated with conventional brain therapies.
Collapse
Affiliation(s)
- Jerzy O Szablowski
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Audrey Lee-Gosselin
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Brian Lue
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Dina Malounda
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Mikhail G Shapiro
- Division of Chemistry and Chemical Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
611
|
Pope CN, Brimijoin S. Cholinesterases and the fine line between poison and remedy. Biochem Pharmacol 2018; 153:205-216. [PMID: 29409903 PMCID: PMC5959757 DOI: 10.1016/j.bcp.2018.01.044] [Citation(s) in RCA: 82] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Accepted: 01/26/2018] [Indexed: 12/20/2022]
Abstract
Acetylcholinesterase (AChE, EC 3.1.1.7) and butyrylcholinesterase (BChE, EC 3.1.1.8) are related enzymes found across the animal kingdom. The critical role of acetylcholinesterase in neurotransmission has been known for almost a century, but a physiological role for butyrylcholinesterase is just now emerging. The cholinesterases have been deliberately targeted for both therapy and toxicity, with cholinesterase inhibitors being used in the clinic for a variety of disorders and conversely for their toxic potential as pesticides and chemical weapons. Non-catalytic functions of the cholinesterases (ChEs) participate in both neurodevelopment and disease. Manipulating either the catalytic activities or the structure of these enzymes can potentially shift the balance between beneficial and adverse effect in a wide number of physiological processes.
Collapse
Affiliation(s)
- Carey N Pope
- Department of Physiological Sciences, Interdisciplinary Toxicology Program, Oklahoma State University, Stillwater, OK 74078, USA.
| | - Stephen Brimijoin
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic, Rochester, MN 55902, USA
| |
Collapse
|
612
|
Evers MM, Miniarikova J, Juhas S, Vallès A, Bohuslavova B, Juhasova J, Skalnikova HK, Vodicka P, Valekova I, Brouwers C, Blits B, Lubelski J, Kovarova H, Ellederova Z, van Deventer SJ, Petry H, Motlik J, Konstantinova P. AAV5-miHTT Gene Therapy Demonstrates Broad Distribution and Strong Human Mutant Huntingtin Lowering in a Huntington's Disease Minipig Model. Mol Ther 2018; 26:2163-2177. [PMID: 30007561 PMCID: PMC6127509 DOI: 10.1016/j.ymthe.2018.06.021] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 06/18/2018] [Accepted: 06/20/2018] [Indexed: 02/07/2023] Open
Abstract
Huntington’s disease (HD) is a fatal neurodegenerative disorder caused by a CAG trinucleotide repeat expansion in the huntingtin gene. Previously, we showed strong huntingtin reduction and prevention of neuronal dysfunction in HD rodents using an engineered microRNA targeting human huntingtin, delivered via adeno-associated virus (AAV) serotype 5 vector with a transgene encoding an engineered miRNA against HTT mRNA (AAV5-miHTT). One of the challenges of rodents as a model of neurodegenerative diseases is their relatively small brain, making successful translation to the HD patient difficult. This is particularly relevant for gene therapy approaches, where distribution achieved upon local administration into the parenchyma is likely dependent on brain size and structure. Here, we aimed to demonstrate the translation of huntingtin-lowering gene therapy to a large-animal brain. We investigated the feasibility, efficacy, and tolerability of one-time intracranial administration of AAV5-miHTT in the transgenic HD (tgHD) minipig model. We detected widespread dose-dependent distribution of AAV5-miHTT throughout the tgHD minipig brain that correlated with the engineered microRNA expression. Both human mutant huntingtin mRNA and protein were significantly reduced in all brain regions transduced by AAV5-miHTT. The combination of widespread vector distribution and extensive huntingtin lowering observed with AAV5-miHTT supports the translation of a huntingtin-lowering gene therapy for HD from preclinical studies into the clinic.
Collapse
Affiliation(s)
- Melvin M Evers
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands.
| | - Jana Miniarikova
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Stefan Juhas
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Astrid Vallès
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | | | - Jana Juhasova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | | | - Petr Vodicka
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Ivona Valekova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Cynthia Brouwers
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Bas Blits
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Jacek Lubelski
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Hana Kovarova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Zdenka Ellederova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Sander J van Deventer
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Harald Petry
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| | - Jan Motlik
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Pavlina Konstantinova
- Department of Research & Development, uniQure biopharma B.V., Amsterdam, the Netherlands
| |
Collapse
|
613
|
Benger M, Kinali M, Mazarakis ND. Autism spectrum disorder: prospects for treatment using gene therapy. Mol Autism 2018; 9:39. [PMID: 29951185 PMCID: PMC6011246 DOI: 10.1186/s13229-018-0222-8] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/07/2018] [Indexed: 01/01/2023] Open
Abstract
Autism spectrum disorder (ASD) is characterised by the concomitant occurrence of impaired social interaction; restricted, perseverative and stereotypical behaviour; and abnormal communication skills. Recent epidemiological studies have reported a dramatic increase in the prevalence of ASD with as many as 1 in every 59 children being diagnosed with ASD. The fact that ASD appears to be principally genetically driven, and may be reversible postnatally, has raised the exciting possibility of using gene therapy as a disease-modifying treatment. Such therapies have already started to seriously impact on human disease and particularly monogenic disorders (e.g. metachromatic leukodystrophy, SMA type 1). In regard to ASD, technical advances in both our capacity to model the disorder in animals and also our ability to deliver genes to the central nervous system (CNS) have led to the first preclinical studies in monogenic ASD, involving both gene replacement and silencing. Furthermore, our increasing awareness and understanding of common dysregulated pathways in ASD have broadened gene therapy's potential scope to include various polygenic ASDs. As this review highlights, despite a number of outstanding challenges, gene therapy has excellent potential to address cognitive dysfunction in ASD.
Collapse
Affiliation(s)
- Matthew Benger
- Gene Therapy, Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, W12 0NN, London, UK
| | - Maria Kinali
- Present address: The Portland Hospital, 205-209 Great Portland Street, London, W1W 5AH UK
| | - Nicholas D. Mazarakis
- Gene Therapy, Centre for Neuroinflammation and Neurodegeneration, Division of Brain Sciences, Faculty of Medicine, Imperial College London, Hammersmith Hospital Campus, W12 0NN, London, UK
| |
Collapse
|
614
|
The State of the NIH BRAIN Initiative. J Neurosci 2018; 38:6427-6438. [PMID: 29921715 DOI: 10.1523/jneurosci.3174-17.2018] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2018] [Revised: 06/01/2018] [Accepted: 06/04/2018] [Indexed: 12/30/2022] Open
Abstract
The BRAIN Initiative arose from a grand challenge to "accelerate the development and application of new technologies that will enable researchers to produce dynamic pictures of the brain that show how individual brain cells and complex neural circuits interact at the speed of thought." The BRAIN Initiative is a public-private effort focused on the development and use of powerful tools for acquiring fundamental insights about how information processing occurs in the central nervous system (CNS). As the Initiative enters its fifth year, NIH has supported >500 principal investigators, who have answered the Initiative's challenge via hundreds of publications describing novel tools, methods, and discoveries that address the Initiative's seven scientific priorities. We describe scientific advances produced by individual laboratories, multi-investigator teams, and entire consortia that, over the coming decades, will produce more comprehensive and dynamic maps of the brain, deepen our understanding of how circuit activity can produce a rich tapestry of behaviors, and lay the foundation for understanding how its circuitry is disrupted in brain disorders. Much more work remains to bring this vision to fruition, and the National Institutes of Health continues to look to the diverse scientific community, from mathematics, to physics, chemistry, engineering, neuroethics, and neuroscience, to ensure that the greatest scientific benefit arises from this unique research Initiative.
Collapse
|
615
|
|
616
|
Sun S, Schaffer DV. Engineered viral vectors for functional interrogation, deconvolution, and manipulation of neural circuits. Curr Opin Neurobiol 2018; 50:163-170. [PMID: 29614429 PMCID: PMC5984719 DOI: 10.1016/j.conb.2017.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/27/2017] [Accepted: 12/16/2017] [Indexed: 12/19/2022]
Abstract
Optimization of traditional replication-competent viral tracers has granted access to immediate synaptic partners of target neuronal populations, enabling the dissection of complex brain circuits into functional neural pathways. The excessive virulence of most conventional tracers, however, impedes their utility in revealing and genetically perturbing cellular function on long time scales. As a promising alternative, the natural capacity of adeno-associated viral (AAV) vectors to safely mediate persistent and robust gene expression has stimulated strong interest in adapting them for sparse neuronal labeling and physiological studies. Furthermore, increasingly refined engineering strategies have yielded novel AAV variants with enhanced target specificity, transduction, and retrograde trafficking in the CNS. These potent vectors offer new opportunities for characterizing the identity and connectivity of single neurons within immense networks and modulating their activity via robust delivery of functional genetic tools.
Collapse
Affiliation(s)
- Sabrina Sun
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA; Department of Bioengineering, University of California, Berkeley, CA, USA; The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
617
|
Abstract
Brain cells communicate with one another via local and long-range synaptic connections. Structural connectivity is the foundation for neural function. Brain-wide connectivity can be described at macroscopic, mesoscopic and microscopic levels. The mesoscale connectome represents connections between neuronal types across different brain regions. Building a mesoscale connectome requires a detailed understanding of the cell type composition of different brain regions and the patterns of inputs and outputs that each of these cell types receives and forms, respectively. In this review, I discuss historical and contemporary tracing techniques in both anterograde and retrograde directions to map the input and output connections at population and individual cell levels, as well as imaging and network analysis approaches to build mesoscale connectomes for mammalian brains.
Collapse
Affiliation(s)
- Hongkui Zeng
- Allen Institute for Brain Science, Seattle, WA 98109, USA.
| |
Collapse
|
618
|
DiCarlo JE, Mahajan VB, Tsang SH. Gene therapy and genome surgery in the retina. J Clin Invest 2018; 128:2177-2188. [PMID: 29856367 DOI: 10.1172/jci120429] [Citation(s) in RCA: 95] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
Precision medicine seeks to treat disease with molecular specificity. Advances in genome sequence analysis, gene delivery, and genome surgery have allowed clinician-scientists to treat genetic conditions at the level of their pathology. As a result, progress in treating retinal disease using genetic tools has advanced tremendously over the past several decades. Breakthroughs in gene delivery vectors, both viral and nonviral, have allowed the delivery of genetic payloads in preclinical models of retinal disorders and have paved the way for numerous successful clinical trials. Moreover, the adaptation of CRISPR-Cas systems for genome engineering have enabled the correction of both recessive and dominant pathogenic alleles, expanding the disease-modifying power of gene therapies. Here, we highlight the translational progress of gene therapy and genome editing of several retinal disorders, including RPE65-, CEP290-, and GUY2D-associated Leber congenital amaurosis, as well as choroideremia, achromatopsia, Mer tyrosine kinase- (MERTK-) and RPGR X-linked retinitis pigmentosa, Usher syndrome, neovascular age-related macular degeneration, X-linked retinoschisis, Stargardt disease, and Leber hereditary optic neuropathy.
Collapse
Affiliation(s)
- James E DiCarlo
- Jonas Children's Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, New York, USA
| | - Vinit B Mahajan
- Omics Laboratory, Byers Eye Institute, Department of Ophthalmology, Stanford University, Palo Alto, California, USA.,Veterans Affairs Palo Alto Health Care System, Palo Alto, California, USA
| | - Stephen H Tsang
- Jonas Children's Vision Care and Bernard and Shirlee Brown Glaucoma Laboratory, Columbia Stem Cell Initiative, Departments of Ophthalmology, Pathology and Cell Biology, Institute of Human Nutrition, College of Physicians and Surgeons, Columbia University, New York, New York, USA.,Edward S. Harkness Eye Institute, New York-Presbyterian Hospital, New York, New York, USA
| |
Collapse
|
619
|
Falcicchia C, Simonato M, Verlengia G. New Tools for Epilepsy Therapy. Front Cell Neurosci 2018; 12:147. [PMID: 29896092 PMCID: PMC5986878 DOI: 10.3389/fncel.2018.00147] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 05/14/2018] [Indexed: 12/19/2022] Open
Abstract
One third of the epilepsies are refractory to conventional antiepileptic drugs (AEDs) and, therefore, identification of new therapies is highly needed. Here, we briefly describe two approaches, direct cell grafting and gene therapy, that may represent alternatives to conventional drugs for the treatment of focal epilepsies. In addition, we discuss more in detail some new tools, cell based-biodelivery systems (encapsulated cell biodelivery (ECB) devices) and new generation gene therapy vectors, which may help in the progress toward clinical translation. The field is advancing rapidly, and there is optimism that cell and/or gene therapy strategies will soon be ready for testing in drug-resistant epileptic patients.
Collapse
Affiliation(s)
- Chiara Falcicchia
- Department of Medical Sciences, Section of Pharmacology, and Neuroscience Center, University of Ferrara and National Institute of Neuroscience, Ferrara, Italy
| | - Michele Simonato
- Department of Medical Sciences, Section of Pharmacology, and Neuroscience Center, University of Ferrara and National Institute of Neuroscience, Ferrara, Italy.,School of Medicine, University Vita-Salute San Raffaele, Milan, Italy
| | - Gianluca Verlengia
- Department of Medical Sciences, Section of Pharmacology, and Neuroscience Center, University of Ferrara and National Institute of Neuroscience, Ferrara, Italy.,School of Medicine, University Vita-Salute San Raffaele, Milan, Italy
| |
Collapse
|
620
|
Zhang X, He T, Chai Z, Samulski RJ, Li C. Blood-brain barrier shuttle peptides enhance AAV transduction in the brain after systemic administration. Biomaterials 2018; 176:71-83. [PMID: 29860139 DOI: 10.1016/j.biomaterials.2018.05.041] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 05/23/2018] [Accepted: 05/24/2018] [Indexed: 12/20/2022]
Abstract
The adeno-associated virus (AAV) vector has been used in preclinical and clinical trials of gene therapy for central nervous system (CNS) diseases. One of the biggest challenges of effectively delivering AAV to the brain is to surmount the blood-brain barrier (BBB). Herein, we identified several potential BBB shuttle peptides that significantly enhanced AAV8 transduction in the brain after a systemic administration, the best of which was the THR peptide. The enhancement of AAV8 brain transduction by THR is dose-dependent, and neurons are the primary THR targets. Mechanism studies revealed that THR directly bound to the AAV8 virion, increasing its ability to cross the endothelial cell barrier. Further experiments showed that binding of THR to the AAV virion did not interfere with AAV8 infection biology, and that THR competitively blocked transferrin from binding to AAV8. Taken together, our results demonstrate, for the first time, that BBB shuttle peptides are able to directly interact with AAV and increase the ability of the AAV vectors to cross the BBB for transduction enhancement in the brain. These results will shed important light on the potential applications of BBB shuttle peptides for enhancing brain transduction with systemic administration of AAV vectors.
Collapse
Affiliation(s)
- Xintao Zhang
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Ting He
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Zheng Chai
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - R Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27599, USA; Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC, 27510, USA.
| |
Collapse
|
621
|
Kaemmerer WF. How will the field of gene therapy survive its success? Bioeng Transl Med 2018; 3:166-177. [PMID: 30065971 PMCID: PMC6063870 DOI: 10.1002/btm2.10090] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Revised: 04/16/2018] [Accepted: 04/17/2018] [Indexed: 02/01/2023] Open
Abstract
In August 2017, for the first time, a gene therapy was approved for market release in the United States. That approval was followed by two others before the end of the year. This article cites primary literature, review articles concerning particular biotechnologies, and press releases by the FDA and others in order to provide an overview of the current status of the field of gene therapy with respect to its translation into practice. Technical hurdles that have been overcome in the past decades are summarized, as are hurdles that need to be the subject of continued research. Then, some social and practical challenges are identified that must be overcome if the field of gene therapy, having survived past failures, is to achieve not only technical and clinical but also market success. One of these, the need for an expanded capacity for the manufacturing of viral vectors to be able to meet the needs of additional gene therapies that will be coming soon, is a challenge that the talents of current and future bioengineers may help address.
Collapse
|
622
|
New approaches for brain repair—from rescue to reprogramming. Nature 2018; 557:329-334. [DOI: 10.1038/s41586-018-0087-1] [Citation(s) in RCA: 118] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Accepted: 03/15/2018] [Indexed: 01/05/2023]
|
623
|
Chemical Processing of Brain Tissues for Large-Volume, High-Resolution Optical Imaging. ACTA ACUST UNITED AC 2018. [DOI: 10.1007/978-981-10-9020-2_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
624
|
Vectored gene delivery for lifetime animal contraception: Overview and hurdles to implementation. Theriogenology 2018; 112:63-74. [DOI: 10.1016/j.theriogenology.2017.11.003] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2017] [Revised: 10/25/2017] [Accepted: 11/02/2017] [Indexed: 12/24/2022]
|
625
|
Davidsson M, Díaz-Fernández P, Torroba M, Schwich OD, Aldrin-Kirk P, Quintino L, Heuer A, Wang G, Lundberg C, Björklund T. Molecular barcoding of viral vectors enables mapping and optimization of mRNA trans-splicing. RNA (NEW YORK, N.Y.) 2018; 24:673-687. [PMID: 29386333 PMCID: PMC5900565 DOI: 10.1261/rna.063925.117] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2017] [Accepted: 01/25/2018] [Indexed: 05/04/2023]
Abstract
Genome editing has proven to be highly potent in the generation of functional gene knockouts in dividing cells. In the CNS however, efficient technologies to repair sequences are yet to materialize. Reprogramming on the mRNA level is an attractive alternative as it provides means to perform in situ editing of coding sequences without nuclease dependency. Furthermore, de novo sequences can be inserted without the requirement of homologous recombination. Such reprogramming would enable efficient editing in quiescent cells (e.g., neurons) with an attractive safety profile for translational therapies. In this study, we applied a novel molecular-barcoded screening assay to investigate RNA trans-splicing in mammalian neurons. Through three alternative screening systems in cell culture and in vivo, we demonstrate that factors determining trans-splicing are reproducible regardless of the screening system. With this screening, we have located the most permissive trans-splicing sequences targeting an intron in the Synapsin I gene. Using viral vectors, we were able to splice full-length fluorophores into the mRNA while retaining very low off-target expression. Furthermore, this approach also showed evidence of functionality in the mouse striatum. However, in its current form, the trans-splicing events are stochastic and the overall activity lower than would be required for therapies targeting loss-of-function mutations. Nevertheless, the herein described barcode-based screening assay provides a unique possibility to screen and map large libraries in single animals or cell assays with very high precision.
Collapse
Affiliation(s)
- Marcus Davidsson
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Paula Díaz-Fernández
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Marcos Torroba
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Oliver D Schwich
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Patrick Aldrin-Kirk
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Luis Quintino
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Andreas Heuer
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Gang Wang
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Cecilia Lundberg
- CNS Gene Therapy, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| | - Tomas Björklund
- Molecular Neuromodulation, Department of Experimental Medical Science, Lund University, 221 84 Lund, Sweden
| |
Collapse
|
626
|
Current state of in vivo panning technologies: Designing specificity and affinity into the future of drug targeting. Adv Drug Deliv Rev 2018; 130:39-49. [PMID: 29964079 DOI: 10.1016/j.addr.2018.06.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Revised: 05/24/2018] [Accepted: 06/22/2018] [Indexed: 11/20/2022]
Abstract
Targeting ligands are used in drug delivery to improve drug distribution to desired cells or tissues and to facilitate cellular entry. In vivo biopanning, whereby billions of potential ligand sequences are screened in biologically-relevant and complex conditions, is a powerful method for identification of novel target ligands. This tool has impacted drug delivery technologies and expanded our arsenal of therapeutics and diagnostics. Within this review we will discuss current in vivo panning technologies and ways that these technologies can be improved to advance next-generation drug delivery strategies.
Collapse
|
627
|
Yttri EA, Dudman JT. A Proposed Circuit Computation in Basal Ganglia: History-Dependent Gain. Mov Disord 2018; 33:704-716. [PMID: 29575303 PMCID: PMC6001446 DOI: 10.1002/mds.27321] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 12/21/2017] [Accepted: 01/08/2018] [Indexed: 12/24/2022] Open
Abstract
In this Scientific Perspectives we first review the recent advances in our understanding of the functional architecture of basal ganglia circuits. Then we argue that these data can best be explained by a model in which basal ganglia act to control the gain of movement kinematics to shape performance based on prior experience, which we refer to as a history-dependent gain computation. Finally, we discuss how insights from the history-dependent gain model might translate from the bench to the bedside, primarily the implications for the design of adaptive deep brain stimulation. Thus, we explicate the key empirical and conceptual support for a normative, computational model with substantial explanatory power for the broad role of basal ganglia circuits in health and disease. © 2018 The Authors. Movement Disorders published by Wiley Periodicals, Inc. on behalf of International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Eric Allen Yttri
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnVirginiaUSA
- Present address:
Department of Biological SciencesCarnegie Mellon UniversityPittsburghPennsylvaniaUSA
| | - Joshua Tate Dudman
- Janelia Research Campus, Howard Hughes Medical InstituteAshburnVirginiaUSA
| |
Collapse
|
628
|
|
629
|
Valenzuela V, Jackson KL, Sardi SP, Hetz C. Gene Therapy Strategies to Restore ER Proteostasis in Disease. Mol Ther 2018; 26:1404-1413. [PMID: 29728295 DOI: 10.1016/j.ymthe.2018.04.004] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2018] [Revised: 04/01/2018] [Accepted: 04/04/2018] [Indexed: 12/11/2022] Open
Abstract
Proteostasis alterations are proposed as a transversal hallmark of several pathological conditions, including metabolic disorders, mechanical injury, cardiac malfunction, neurodegeneration, and cancer. Strategies to improve proteostasis aim to reduce the accumulation of specific disease-related misfolded proteins or bolster the endogenous mechanisms to fold and degrade abnormal proteins. Endoplasmic reticulum (ER) stress is a common pathological signature of a variety of diseases, which engages the unfolded protein response (UPR) as a cellular reaction to mitigate ER stress. Pharmacological modulation of the UPR is challenging considering the physiological importance of the pathway in various organs. However, local targeting of ER stress responses in the affected tissue using gene therapy is emerging as a possible solution to overcome side effects. The delivery of ER chaperones or active UPR components using adeno-associated virus (AAV) has demonstrated outstanding beneficial effects in several disease models (e.g., neurodegenerative conditions, eye disorders, and metabolic diseases). Here, we discuss current efforts to design and optimize gene therapy strategies to improve ER proteostasis in different disease contexts.
Collapse
Affiliation(s)
- Vicente Valenzuela
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile
| | - Kasey L Jackson
- Neuroscience Therapeutic Area, Sanofi, Framingham, MA 01701, USA
| | - Sergio P Sardi
- Neuroscience Therapeutic Area, Sanofi, Framingham, MA 01701, USA
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile; Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Buck Institute for Research on Aging, Novato, CA 94945, USA; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Harvard University, Boston, MA 02115, USA.
| |
Collapse
|
630
|
Pardieck J, Sakiyama-Elbert S. Genome engineering for CNS injury and disease. Curr Opin Biotechnol 2018; 52:89-94. [PMID: 29597076 DOI: 10.1016/j.copbio.2018.03.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2018] [Revised: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 01/01/2023]
Abstract
Recent developments in genome engineering methods have advanced our knowledge of central nervous system (CNS) function in both normal health and following disease or injury. This review discusses current literature using gene editing tools in CNS disease and injury research, such as improving viral-mediated targeting of cell populations, generating new methods for genome editing, reprogramming cells into CNS cell types, and using organoids as models of development and disease. Readers may gain inspiration for continuing research into new genome engineering methods and for therapies for CNS applications.
Collapse
Affiliation(s)
- Jennifer Pardieck
- Department of Biomedical Engineering, University of Texas at Austin, Austin, TX, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO, USA
| | | |
Collapse
|
631
|
Abstract
In recent years, the number of clinical trials in which adeno-associated virus (AAV) vectors have been used for in vivo gene transfer has steadily increased. The excellent safety profile, together with the high efficiency of transduction of a broad range of target tissues, has established AAV vectors as the platform of choice for in vivo gene therapy. Successful application of the AAV technology has also been achieved in the clinic for a variety of conditions, including coagulation disorders, inherited blindness, and neurodegenerative diseases, among others. Clinical translation of novel and effective "therapeutic products" is, however, a long process that involves several cycles of iterations from bench to bedside that are required to address issues encountered during drug development. For the AAV vector gene transfer technology, several hurdles have emerged in both preclinical studies and clinical trials; addressing these issues will allow in the future to expand the scope of AAV gene transfer as a therapeutic modality for a variety of human diseases. In this review, we will give an overview on the biology of AAV vector, discuss the design of AAV-based gene therapy strategies for in vivo applications, and present key achievements and emerging issues in the field. We will use the liver as a model target tissue for gene transfer based on the large amount of data available from preclinical and clinical studies.
Collapse
Affiliation(s)
- Pasqualina Colella
- Genethon, INSERM U951 INTEGRARE, University of Evry, University Paris-Saclay, 91001 Evry, France
| | - Giuseppe Ronzitti
- Genethon, INSERM U951 INTEGRARE, University of Evry, University Paris-Saclay, 91001 Evry, France
| | - Federico Mingozzi
- Genethon, INSERM U951 INTEGRARE, University of Evry, University Paris-Saclay, 91001 Evry, France
- University Pierre and Marie Curie-Paris 6 and INSERM U974, 75651 Paris, France
| |
Collapse
|
632
|
Abstract
In recent years, the role of autophagy in the pathogenesis of most neurodegenerative diseases has transitioned into a limbo of protective or detrimental effects. Genetic evidence indicates that mutations in autophagy-regulatory genes can result in the occurrence of amyotrophic lateral sclerosis (ALS), suggesting a physiological role of the pathway to motoneuron function. However, experimental manipulation of autophagy in ALS models led to conflicting results depending on the intervention strategy and the disease model used. A recent work by the Maniatis group systematically explored the role of cell-specific autophagy in motoneurons at different disease stages, revealing surprising and unexpected findings. Autophagy activity at early stages may contribute to maintaining the structure and function of neuromuscular junctions, whereas at later steps of the disease it has a pathogenic activity possibly involving cell-nonautonomous mechanisms related to glial activation. This new study adds a new layer of complexity in the field, suggesting an intricate interplay between proteostasis alterations, the time-differential function of autophagy in neurons, and muscle innervation in ALS.
Collapse
Affiliation(s)
- Vicente Valenzuela
- a Biomedical Neuroscience Institute (BNI), Faculty of Medicine , University of Chile , Santiago , Chile.,b Center for Geroscience , Brain Health and Metabolism (GERO) , Santiago , Chile.,c Program of Cellular and Molecular Biology , Institute of Biomedical Sciences, University of Chile , Santiago , Chile
| | - Melissa Nassif
- d Center for Integrative Biology (CIB), Faculty of Sciences , Universidad Mayor , Santiago , Chile
| | - Claudio Hetz
- a Biomedical Neuroscience Institute (BNI), Faculty of Medicine , University of Chile , Santiago , Chile.,b Center for Geroscience , Brain Health and Metabolism (GERO) , Santiago , Chile.,c Program of Cellular and Molecular Biology , Institute of Biomedical Sciences, University of Chile , Santiago , Chile.,e Buck Institute for Research on Aging , Novato , CA , USA.,f Department of Immunology and Infectious Diseases , Harvard School of Public Health , Boston MA , USA
| |
Collapse
|
633
|
Rincon MY, de Vin F, Duqué SI, Fripont S, Castaldo SA, Bouhuijzen-Wenger J, Holt MG. Widespread transduction of astrocytes and neurons in the mouse central nervous system after systemic delivery of a self-complementary AAV-PHP.B vector. Gene Ther 2018. [PMID: 29523880 DOI: 10.1038/s41434-018-0005-z] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Until recently, adeno-associated virus 9 (AAV9) was considered the AAV serotype most effective in crossing the blood-brain barrier (BBB) and transducing cells of the central nervous system (CNS), following systemic injection. However, a newly engineered capsid, AAV-PHP.B, is reported to cross the BBB at even higher efficiency. We investigated how much we could boost CNS transgene expression by using AAV-PHP.B carrying a self-complementary (sc) genome. To allow comparison, 6 weeks old C57BL/6 mice received intravenous injections of scAAV2/9-GFP or scAAV2/PHP.B-GFP at equivalent doses. Three weeks postinjection, transgene expression was assessed in brain and spinal cord. We consistently observed more widespread CNS transduction and higher levels of transgene expression when using the scAAV2/PHP.B-GFP vector. In particular, we observed an unprecedented level of astrocyte transduction in the cortex, when using a ubiquitous CBA promoter. In comparison, neuronal transduction was much lower than previously reported. However, strong neuronal expression (including spinal motor neurons) was observed when the human synapsin promoter was used. These findings constitute the first reported use of an AAV-PHP.B capsid, encapsulating a scAAV genome, for gene transfer in adult mice. Our results underscore the potential of this AAV construct as a platform for safer and more efficacious gene therapy vectors for the CNS.
Collapse
Affiliation(s)
- Melvin Y Rincon
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,KU Leuven, Department of Neuroscience, Leuven, Belgium
| | - Filip de Vin
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,KU Leuven, Department of Neuroscience, Leuven, Belgium
| | - Sandra I Duqué
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,KU Leuven, Department of Neuroscience, Leuven, Belgium
| | - Shelly Fripont
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,KU Leuven, Department of Neuroscience, Leuven, Belgium
| | - Stephanie A Castaldo
- VIB-KU Leuven Center for Cancer Biology, Leuven, Belgium.,KU Leuven, Department of Oncology, Leuven, Belgium
| | - Jessica Bouhuijzen-Wenger
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium.,KU Leuven, Department of Neuroscience, Leuven, Belgium
| | - Matthew G Holt
- VIB-KU Leuven Center for Brain & Disease Research, Leuven, Belgium. .,KU Leuven, Department of Neuroscience, Leuven, Belgium.
| |
Collapse
|
634
|
Grames MS, Dayton RD, Jackson KL, Richard AD, Lu X, Klein RL. Cre‐dependent AAV vectors for highly targeted expression of disease‐related proteins and neurodegeneration in the substantia nigra. FASEB J 2018. [DOI: 10.1096/fj.201701529rr] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Affiliation(s)
- Mychal S. Grames
- Department of Pharmacology, Toxicology, and NeuroscienceLouisiana State University (LSU) Health ShreveportShreveportLouisianaUSA
| | - Robert D. Dayton
- Department of Pharmacology, Toxicology, and NeuroscienceLouisiana State University (LSU) Health ShreveportShreveportLouisianaUSA
| | - Kasey L. Jackson
- Department of Pharmacology, Toxicology, and NeuroscienceLouisiana State University (LSU) Health ShreveportShreveportLouisianaUSA
| | - Adam D. Richard
- Department of Pharmacology, Toxicology, and NeuroscienceLouisiana State University (LSU) Health ShreveportShreveportLouisianaUSA
| | - Xiaohong Lu
- Department of Pharmacology, Toxicology, and NeuroscienceLouisiana State University (LSU) Health ShreveportShreveportLouisianaUSA
| | - Ronald L. Klein
- Department of Pharmacology, Toxicology, and NeuroscienceLouisiana State University (LSU) Health ShreveportShreveportLouisianaUSA
| |
Collapse
|
635
|
Safety First: Perspective on Patient-Centered Development of AAV Gene Therapy Products. Mol Ther 2018; 26:669-671. [PMID: 29503193 DOI: 10.1016/j.ymthe.2018.02.009] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
|
636
|
Giannelli SG, Luoni M, Castoldi V, Massimino L, Cabassi T, Angeloni D, Demontis GC, Leocani L, Andreazzoli M, Broccoli V. Cas9/sgRNA selective targeting of the P23H Rhodopsin mutant allele for treating retinitis pigmentosa by intravitreal AAV9.PHP.B-based delivery. Hum Mol Genet 2018; 27:761-779. [PMID: 29281027 DOI: 10.1093/hmg/ddx438] [Citation(s) in RCA: 93] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2017] [Accepted: 12/18/2017] [Indexed: 01/05/2024] Open
Abstract
P23H is the most common mutation in the RHODOPSIN (RHO) gene leading to a dominant form of retinitis pigmentosa (RP), a rod photoreceptor degeneration that invariably causes vision loss. Specific disruption of the disease P23H RHO mutant while preserving the wild-type (WT) functional allele would be an invaluable therapy for this disease. However, various technologies tested in the past failed to achieve effective changes and consequently therapeutic benefits. We validated a CRISPR/Cas9 strategy to specifically inactivate the P23H RHO mutant, while preserving the WT allele in vitro. We, then, translated this approach in vivo by delivering the CRISPR/Cas9 components in murine Rho+/P23H mutant retinae. Targeted retinae presented a high rate of cleavage in the P23H but not WT Rho allele. This gene manipulation was sufficient to slow photoreceptor degeneration and improve retinal functions. To improve the translational potential of our approach, we tested intravitreal delivery of this system by means of adeno-associated viruses (AAVs). To this purpose, the employment of the AAV9-PHP.B resulted the most effective in disrupting the P23H Rho mutant. Finally, this approach was translated successfully in human cells engineered with the homozygous P23H RHO gene mutation. Overall, this is a significant proof-of-concept that gene allele specific targeting by CRISPR/Cas9 technology is specific and efficient and represents an unprecedented tool for treating RP and more broadly dominant genetic human disorders affecting the eye, as well as other tissues.
Collapse
Affiliation(s)
- Serena G Giannelli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Mirko Luoni
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Valerio Castoldi
- Experimental Neurophysiology Unit, Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute, 20132 Milan, Italy
| | - Luca Massimino
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
- Institute of Neuroscience, National Research Council (CNR), 20129 Milan, Italy
| | - Tommaso Cabassi
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
- Institute of Neuroscience, National Research Council (CNR), 20129 Milan, Italy
| | - Debora Angeloni
- Institute of Life Sciences, Scuola Superiore Sant'Anna, 56124 Pisa, Italy
| | | | - Letizia Leocani
- Experimental Neurophysiology Unit, Institute of Experimental Neurology (INSPE), San Raffaele Scientific Institute, 20132 Milan, Italy
| | | | - Vania Broccoli
- Stem Cell and Neurogenesis Unit, Division of Neuroscience, San Raffaele Scientific Institute, 20132 Milan, Italy
- Institute of Neuroscience, National Research Council (CNR), 20129 Milan, Italy
| |
Collapse
|
637
|
Katrekar D, Moreno AM, Chen G, Worlikar A, Mali P. Oligonucleotide conjugated multi-functional adeno-associated viruses. Sci Rep 2018; 8:3589. [PMID: 29483550 PMCID: PMC5827683 DOI: 10.1038/s41598-018-21742-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2017] [Accepted: 02/07/2018] [Indexed: 02/06/2023] Open
Abstract
Recombinant adeno-associated viruses (AAVs) are among the most commonly used vehicles for in vivo gene delivery. However, their tropism is limited, and additionally their efficacy can be negatively affected by prevalence of neutralizing antibodies in sera. Methodologies to systematically engineer AAV capsid properties would thus be of great relevance. In this regard, we develop here multi-functional AAVs by engineering precision tethering of oligonucleotides onto the AAV surface, and thereby enabling a spectrum of nucleic-acid programmable functionalities. Towards this, we engineered genetically encoded incorporation of unnatural amino acids (UAA) bearing bio-orthogonal chemical handles onto capsid proteins. Via these we enabled site-specific coupling of oligonucleotides onto the AAV capsid surface using facile click chemistry. The resulting oligo-AAVs could be sequence specifically labeled, and also patterned in 2D using DNA array substrates. Additionally, we utilized these oligo conjugations to engineer viral shielding by lipid-based cloaks that efficaciously protected the AAV particles from neutralizing serum. We confirmed these 'cloaked AAVs' retained full functionality via their ability to transduce a range of cell types, and also enable robust delivery of CRISPR-Cas9 effectors. Taken together, we anticipate this programmable oligo-AAV system will have broad utility in synthetic biology and AAV engineering applications.
Collapse
Affiliation(s)
- Dhruva Katrekar
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Ana M Moreno
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Genghao Chen
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Atharv Worlikar
- Department of Bioengineering, University of California, San Diego, CA, USA
| | - Prashant Mali
- Department of Bioengineering, University of California, San Diego, CA, USA.
| |
Collapse
|
638
|
Cavalca E, Cesani M, Gifford JC, Sena-Esteves M, Terreni MR, Leoncini G, Peviani M, Biffi A. Metallothioneins are neuroprotective agents in lysosomal storage disorders. Ann Neurol 2018; 83:418-432. [DOI: 10.1002/ana.25161] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2017] [Revised: 11/07/2017] [Accepted: 01/24/2018] [Indexed: 12/23/2022]
Affiliation(s)
- Eleonora Cavalca
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center; Boston MA
- Vita Salute San Raffaele University; Milan Italy
- San Raffaele Telethon Institute for Gene Therapy; San Raffaele Scientific Institute; Milan Italy
| | - Martina Cesani
- San Raffaele Telethon Institute for Gene Therapy; San Raffaele Scientific Institute; Milan Italy
| | - Jennifer C. Gifford
- Department of Neurology and Horae Gene Therapy Center; University of Massachusetts Medical School; Worcester MA
| | - Miguel Sena-Esteves
- Department of Neurology and Horae Gene Therapy Center; University of Massachusetts Medical School; Worcester MA
| | | | - Giuseppe Leoncini
- Pathology Department; San Raffaele Scientific Institute; Milan Italy
| | - Marco Peviani
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center; Boston MA
| | - Alessandra Biffi
- Gene Therapy Program, Dana-Farber/Boston Children's Cancer and Blood Disorders Center; Boston MA
- San Raffaele Telethon Institute for Gene Therapy; San Raffaele Scientific Institute; Milan Italy
- Harvard Medical School; Boston MA
| |
Collapse
|
639
|
Miniarikova J, Evers MM, Konstantinova P. Translation of MicroRNA-Based Huntingtin-Lowering Therapies from Preclinical Studies to the Clinic. Mol Ther 2018; 26:947-962. [PMID: 29503201 DOI: 10.1016/j.ymthe.2018.02.002] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Revised: 01/30/2018] [Accepted: 02/05/2018] [Indexed: 12/21/2022] Open
Abstract
The single mutation underlying the fatal neuropathology of Huntington's disease (HD) is a CAG triplet expansion in exon 1 of the huntingtin (HTT) gene, which gives rise to a toxic mutant HTT protein. There have been a number of not yet successful therapeutic advances in the treatment of HD. The current excitement in the HD field is due to the recent development of therapies targeting the culprit of HD either at the DNA or RNA level to reduce the overall mutant HTT protein. In this review, we briefly describe short-term and long-term HTT-lowering strategies targeting HTT transcripts. One of the most advanced HTT-lowering strategies is a microRNA (miRNA)-based gene therapy delivered by a single administration of an adeno-associated viral (AAV) vector to the HD patient. We outline the outcome measures for the miRNA-based HTT-lowering therapy in the context of preclinical evaluation in HD animal and cell models. We highlight the strengths and ongoing queries of the HTT-lowering gene therapy as an HD intervention with a potential disease-modifying effect. This review provides a perspective on the fast-developing HTT-lowering therapies for HD and their translation to the clinic based on existing knowledge in preclinical models.
Collapse
Affiliation(s)
- Jana Miniarikova
- Department of Research and Development, uniQure, Amsterdam, the Netherlands; Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Melvin M Evers
- Department of Research and Development, uniQure, Amsterdam, the Netherlands
| | | |
Collapse
|
640
|
Flotte TR, Büning H. Severe Toxicity in Nonhuman Primates and Piglets with Systemic High-Dose Administration of Adeno-Associated Virus Serotype 9-Like Vectors: Putting Patients First. Hum Gene Ther 2018; 29:283-284. [PMID: 29378415 DOI: 10.1089/hum.2018.021] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
|
641
|
Hordeaux J, Wang Q, Katz N, Buza EL, Bell P, Wilson JM. The Neurotropic Properties of AAV-PHP.B Are Limited to C57BL/6J Mice. Mol Ther 2018; 26:664-668. [PMID: 29428298 DOI: 10.1016/j.ymthe.2018.01.018] [Citation(s) in RCA: 289] [Impact Index Per Article: 41.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2018] [Accepted: 01/17/2018] [Indexed: 10/18/2022] Open
Abstract
Improved delivery of adeno-associated virus (AAV) vectors to the CNS will greatly enhance their clinical utility. Selection of AAV9 variants in a mouse model led to the isolation of a capsid called PHP.B, which resulted in remarkable transduction of the CNS following intravenous infusion. However, we now show here that this enhanced CNS tropism is restricted to the model in which it was selected, i.e., a Cre transgenic mouse in a C57BL/6J background, and was not found in nonhuman primates or the other commonly used mouse strain BALB/cJ. We also report the potential for serious acute toxicity in NHP after systemic administration of high dose of AAV.
Collapse
Affiliation(s)
- Juliette Hordeaux
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Qiang Wang
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nathan Katz
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth L Buza
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Peter Bell
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - James M Wilson
- Gene Therapy Program, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
642
|
Robinson JE, Gradinaru V. Dopaminergic dysfunction in neurodevelopmental disorders: recent advances and synergistic technologies to aid basic research. Curr Opin Neurobiol 2018; 48:17-29. [PMID: 28850815 PMCID: PMC5825239 DOI: 10.1016/j.conb.2017.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2017] [Accepted: 08/03/2017] [Indexed: 12/19/2022]
Abstract
Neurodevelopmental disorders (NDDs) represent a diverse group of syndromes characterized by abnormal development of the central nervous system and whose symptomatology includes cognitive, emotional, sensory, and motor impairments. The identification of causative genetic defects has allowed for creation of transgenic NDD mouse models that have revealed pathophysiological mechanisms of disease phenotypes in a neural circuit- and cell type-specific manner. Mouse models of several syndromes, including Rett syndrome, Fragile X syndrome, Angelman syndrome, Neurofibromatosis type 1, etc., exhibit abnormalities in the structure and function of dopaminergic circuitry, which regulates motivation, motor behavior, sociability, attention, and executive function. Recent advances in technologies for functional circuit mapping, including tissue clearing, viral vector-based tracing methods, and optical readouts of neural activity, have refined our knowledge of dopaminergic circuits in unperturbed states, yet these tools have not been widely applied to NDD research. Here, we will review recent findings exploring dopaminergic function in NDD models and discuss the promise of new tools to probe NDD pathophysiology in these circuits.
Collapse
Affiliation(s)
- J Elliott Robinson
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
| |
Collapse
|
643
|
In vivo simultaneous transcriptional activation of multiple genes in the brain using CRISPR-dCas9-activator transgenic mice. Nat Neurosci 2018; 21:440-446. [PMID: 29335603 DOI: 10.1038/s41593-017-0060-6] [Citation(s) in RCA: 200] [Impact Index Per Article: 28.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 12/07/2017] [Indexed: 12/22/2022]
Abstract
Despite rapid progresses in the genome-editing field, in vivo simultaneous overexpression of multiple genes remains challenging. We generated a transgenic mouse using an improved dCas9 system that enables simultaneous and precise in vivo transcriptional activation of multiple genes and long noncoding RNAs in the nervous system. As proof of concept, we were able to use targeted activation of endogenous neurogenic genes in these transgenic mice to directly and efficiently convert astrocytes into functional neurons in vivo. This system provides a flexible and rapid screening platform for studying complex gene networks and gain-of-function phenotypes in the mammalian brain.
Collapse
|
644
|
Astrocytes and presynaptic plasticity in the striatum: Evidence and unanswered questions. Brain Res Bull 2018; 136:17-25. [DOI: 10.1016/j.brainresbull.2017.01.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2016] [Revised: 12/30/2016] [Accepted: 01/02/2017] [Indexed: 02/03/2023]
|
645
|
Pyles B, Bailus BJ, O'Geen H, Segal DJ. Purified Protein Delivery to Activate an Epigenetically Silenced Allele in Mouse Brain. Methods Mol Biol 2018; 1767:227-239. [PMID: 29524138 PMCID: PMC6281562 DOI: 10.1007/978-1-4939-7774-1_12] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The ability to activate or repress specific genes in the brain could have a tremendous impact for understanding and treating neurological disorders. Artificial transcription factors based on zinc finger, TALE, and CRISPR/Cas9 programmable DNA-binding platforms have been widely used to regulate the expression of specific genes in cultured cells, but their delivery into the brain represents a critical challenge to apply such tools in live animals. In previous work, we developed a purified, zinc finger-based artificial transcription factor that could be injected systemically, cross the blood-brain barrier, and alter expression of a specific gene in the brain of an adult mouse model of Angelman syndrome. Importantly, our mode of delivery produced widespread distribution throughout the brain. Here we describe our most current methods for the production and purification of the factor, dosage optimization, and use of live animal fluorescence imaging to visualize the kinetics of distribution.
Collapse
Affiliation(s)
- Benjamin Pyles
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, USA
| | - Barbara J Bailus
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, USA
- The Buck Institute for Research on Aging, Novato, CA, USA
| | - Henriette O'Geen
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, USA
| | - David J Segal
- Genome Center and Department of Biochemistry and Molecular Medicine, University of California, Davis, Davis, CA, USA.
| |
Collapse
|
646
|
Raikwar SP, Thangavel R, Dubova I, Ahmed ME, Selvakumar PG, Kempuraj D, Zaheer S, Iyer S, Zaheer A. Neuro-Immuno-Gene- and Genome-Editing-Therapy for Alzheimer's Disease: Are We There Yet? J Alzheimers Dis 2018; 65:321-344. [PMID: 30040732 PMCID: PMC6130335 DOI: 10.3233/jad-180422] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/26/2018] [Indexed: 12/16/2022]
Abstract
Alzheimer's disease (AD) is a highly complex neurodegenerative disorder and the current treatment strategies are largely ineffective thereby leading to irreversible and progressive cognitive decline in AD patients. AD continues to defy successful treatment despite significant advancements in the field of molecular medicine. Repeatedly, early promising preclinical and clinical results have catapulted into devastating setbacks leading to multi-billion dollar losses not only to the top pharmaceutical companies but also to the AD patients and their families. Thus, it is very timely to review the progress in the emerging fields of gene therapy and stem cell-based precision medicine. Here, we have made sincere efforts to feature the ongoing progress especially in the field of AD gene therapy and stem cell-based regenerative medicine. Further, we also provide highlights in elucidating the molecular mechanisms underlying AD pathogenesis and describe novel AD therapeutic targets and strategies for the new drug discovery. We hope that the quantum leap in the scientific advancements and improved funding will bolster novel concepts that will propel the momentum toward a trajectory leading to a robust AD patient-specific next generation precision medicine with improved cognitive function and excellent life quality.
Collapse
Affiliation(s)
- Sudhanshu P. Raikwar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Ramasamy Thangavel
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Iuliia Dubova
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Pushpavathi Govindhasamy Selvakumar
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Smita Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
| | - Shankar Iyer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| | - Asgar Zaheer
- Department of Neurology, Center for Translational Neuroscience, School of Medicine, University of Missouri, Columbia, MO, USA
- U.S. Department of Veterans Affairs, Harry S. Truman Memorial Veteran’s Hospital, Columbia, MO, USA
| |
Collapse
|
647
|
Cachón-González MB, Zaccariotto E, Cox TM. Genetics and Therapies for GM2 Gangliosidosis. Curr Gene Ther 2018; 18:68-89. [PMID: 29618308 PMCID: PMC6040173 DOI: 10.2174/1566523218666180404162622] [Citation(s) in RCA: 71] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2017] [Revised: 01/10/2018] [Accepted: 01/27/2018] [Indexed: 12/30/2022]
Abstract
Tay-Sachs disease, caused by impaired β-N-acetylhexosaminidase activity, was the first GM2 gangliosidosis to be studied and one of the most severe and earliest lysosomal diseases to be described. The condition, associated with the pathological build-up of GM2 ganglioside, has acquired almost iconic status and serves as a paradigm in the study of lysosomal storage diseases. Inherited as a classical autosomal recessive disorder, this global disease of the nervous system induces developmental arrest with regression of attained milestones; neurodegeneration progresses rapidly to cause premature death in young children. There is no effective treatment beyond palliative care, and while the genetic basis of GM2 gangliosidosis is well established, the molecular and cellular events, from diseasecausing mutations and glycosphingolipid storage to disease manifestations, remain to be fully delineated. Several therapeutic approaches have been attempted in patients, including enzymatic augmentation, bone marrow transplantation, enzyme enhancement, and substrate reduction therapy. Hitherto, none of these stratagems has materially altered the course of the disease. Authentic animal models of GM2 gangliodidosis have facilitated in-depth evaluation of innovative applications such as gene transfer, which in contrast to other interventions, shows great promise. This review outlines current knowledge pertaining the pathobiology as well as potential innovative treatments for the GM2 gangliosidoses.
Collapse
Affiliation(s)
| | - Eva Zaccariotto
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
648
|
Lau CH, Suh Y. In vivo genome editing in animals using AAV-CRISPR system: applications to translational research of human disease. F1000Res 2017; 6:2153. [PMID: 29333255 PMCID: PMC5749125 DOI: 10.12688/f1000research.11243.1] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 12/14/2017] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated virus (AAV) has shown promising therapeutic efficacy with a good safety profile in a wide range of animal models and human clinical trials. With the advent of clustered regulatory interspaced short palindromic repeat (CRISPR)-based genome-editing technologies, AAV provides one of the most suitable viral vectors to package, deliver, and express CRISPR components for targeted gene editing. Recent discoveries of smaller Cas9 orthologues have enabled the packaging of Cas9 nuclease and its chimeric guide RNA into a single AAV delivery vehicle for robust
in vivo genome editing. Here, we discuss how the combined use of small Cas9 orthologues, tissue-specific minimal promoters, AAV serotypes, and different routes of administration has advanced the development of efficient and precise
in vivo genome editing and comprehensively review the various AAV-CRISPR systems that have been effectively used in animals. We then discuss the clinical implications and potential strategies to overcome off-target effects, immunogenicity, and toxicity associated with CRISPR components and AAV delivery vehicles. Finally, we discuss ongoing non-viral-based
ex vivo gene therapy clinical trials to underscore the current challenges and future prospects of CRISPR/Cas9 delivery for human therapeutics.
Collapse
Affiliation(s)
- Cia-Hin Lau
- Department of Mechanical and Biomedical Engineering, City University of Hong Kong, Hong Kong, SAR, China
| | - Yousin Suh
- Department of Genetics, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Ophthalmology and Visual Sciences, Albert Einstein College of Medicine, Bronx, New York, USA.,Department of Medicine, Albert Einstein College of Medicine, Bronx, New York, USA.,Institute for Aging Research, Albert Einstein College of Medicine, Bronx, New York, USA
| |
Collapse
|
649
|
Schubert R, Trenholm S, Balint K, Kosche G, Cowan CS, Mohr MA, Munz M, Martinez-Martin D, Fläschner G, Newton R, Krol J, Scherf BG, Yonehara K, Wertz A, Ponti A, Ghanem A, Hillier D, Conzelmann KK, Müller DJ, Roska B. Virus stamping for targeted single-cell infection in vitro and in vivo. Nat Biotechnol 2017; 36:81-88. [DOI: 10.1038/nbt.4034] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 11/13/2017] [Indexed: 11/09/2022]
|
650
|
Evolution of a designed protein assembly encapsulating its own RNA genome. Nature 2017; 552:415-420. [PMID: 29236688 DOI: 10.1038/nature25157] [Citation(s) in RCA: 145] [Impact Index Per Article: 18.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Accepted: 11/21/2017] [Indexed: 12/20/2022]
Abstract
The challenges of evolution in a complex biochemical environment, coupling genotype to phenotype and protecting the genetic material, are solved elegantly in biological systems by the encapsulation of nucleic acids. In the simplest examples, viruses use capsids to surround their genomes. Although these naturally occurring systems have been modified to change their tropism and to display proteins or peptides, billions of years of evolution have favoured efficiency at the expense of modularity, making viral capsids difficult to engineer. Synthetic systems composed of non-viral proteins could provide a 'blank slate' to evolve desired properties for drug delivery and other biomedical applications, while avoiding the safety risks and engineering challenges associated with viruses. Here we create synthetic nucleocapsids, which are computationally designed icosahedral protein assemblies with positively charged inner surfaces that can package their own full-length mRNA genomes. We explore the ability of these nucleocapsids to evolve virus-like properties by generating diversified populations using Escherichia coli as an expression host. Several generations of evolution resulted in markedly improved genome packaging (more than 133-fold), stability in blood (from less than 3.7% to 71% of packaged RNA protected after 6 hours of treatment), and in vivo circulation time (from less than 5 minutes to approximately 4.5 hours). The resulting synthetic nucleocapsids package one full-length RNA genome for every 11 icosahedral assemblies, similar to the best recombinant adeno-associated virus vectors. Our results show that there are simple evolutionary paths through which protein assemblies can acquire virus-like genome packaging and protection. Considerable effort has been directed at 'top-down' modification of viruses to be safe and effective for drug delivery and vaccine applications; the ability to design synthetic nanomaterials computationally and to optimize them through evolution now enables a complementary 'bottom-up' approach with considerable advantages in programmability and control.
Collapse
|