1
|
Thompson A, Arano R, Saleem U, Preciado R, Munoz L, Nelson I, Ramos K, Kim Y, Li Y, Xu W. Brain-wide circuit-specific targeting of astrocytes. CELL REPORTS METHODS 2023; 3:100653. [PMID: 38052209 PMCID: PMC10753298 DOI: 10.1016/j.crmeth.2023.100653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 10/04/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023]
Abstract
Astrocytes are integral components of brain circuitry. They enwrap synapses, react to neuronal activity, and regulate synaptic transmission. Astrocytes are heterogeneous and exhibit distinct features and functions in different circuits. Selectively targeting the astrocytes associated with a given neuronal circuit would enable elucidation of their circuit-specific functions but has been technically challenging to date. Recently, we constructed anterograde transneuronal viral vectors based on yellow fever vaccine YFV-17D. Among them, the replication-incompetent YFVΔNS1-Cre can selectively turn on reporter genes in postsynaptic neurons if the viral gene NS1 is expressed in postsynaptic neurons. Here we show that without exogenous expression of NS1 at the postsynaptic sites, locally injected YFVΔNS1-Cre selectively turns on reporter genes in astrocytes in downstream brain regions. The targeting of astrocytes can occur across the whole brain but is specific for the neuronal circuits traced. Therefore, YFVΔNS1-Cre provides a tool for selective genetic targeting of astrocytes to reveal their circuit-specific roles.
Collapse
Affiliation(s)
- Alyssa Thompson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rachel Arano
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Uzair Saleem
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rebecca Preciado
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lizbeth Munoz
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ian Nelson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katarina Ramos
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yerim Kim
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ying Li
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Wei Xu
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
2
|
Herdt AR, Peng H, Dickson DW, Golde TE, Eckman EA, Lee CW. Brain Targeted AAV1-GALC Gene Therapy Reduces Psychosine and Extends Lifespan in a Mouse Model of Krabbe Disease. Genes (Basel) 2023; 14:1517. [PMID: 37628569 PMCID: PMC10454254 DOI: 10.3390/genes14081517] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/14/2023] [Accepted: 07/21/2023] [Indexed: 08/27/2023] Open
Abstract
Krabbe disease (KD) is a progressive and devasting neurological disorder that leads to the toxic accumulation of psychosine in the white matter of the central nervous system (CNS). The condition is inherited via biallelic, loss-of-function mutations in the galactosylceramidase (GALC) gene. To rescue GALC gene function in the CNS of the twitcher mouse model of KD, an adeno-associated virus serotype 1 vector expressing murine GALC under control of a chicken β-actin promoter (AAV1-GALC) was administered to newborn mice by unilateral intracerebroventricular injection. AAV1-GALC treatment significantly improved body weight gain and survival of the twitcher mice (n = 8) when compared with untreated controls (n = 5). The maximum weight gain after postnatal day 10 was significantly increased from 81% to 217%. The median lifespan was extended from 43 days to 78 days (range: 74-88 days) in the AAV1-GALC-treated group. Widespread expression of GALC protein and alleviation of KD neuropathology were detected in the CNS of the treated mice when examined at the moribund stage. Functionally, elevated levels of psychosine were completely normalized in the forebrain region of the treated mice. In the posterior region, which includes the mid- and the hindbrain, psychosine was reduced by an average of 77% (range: 53-93%) compared to the controls. Notably, psychosine levels in this region were inversely correlated with body weight and lifespan of AAV1-GALC-treated mice, suggesting that the degree of viral transduction of posterior brain regions following ventricular injection determined treatment efficacy on growth and survivability, respectively. Overall, our results suggest that viral vector delivery via the cerebroventricular system can partially correct psychosine accumulation in brain that leads to slower disease progression in KD.
Collapse
Affiliation(s)
- Aimee R. Herdt
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Hui Peng
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Dennis W. Dickson
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Todd E. Golde
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA 30322, USA
- Department of Neurology, Emory University, Atlanta, GA 30322, USA
- Emory Center for Neurodegenerative Disease, Emory University, Atlanta, GA 30322, USA
| | - Elizabeth A. Eckman
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| | - Chris W. Lee
- Biomedical Research Institute of New Jersey, Cedar Knolls, NJ 07927, USA (E.A.E.)
- MidAtlantic Neonatology Associates (MANA), Morristown, NJ 07960, USA
- Atlantic Health System, Morristown, NJ 07960, USA
| |
Collapse
|
3
|
Campos LJ, Arokiaraj CM, Chuapoco MR, Chen X, Goeden N, Gradinaru V, Fox AS. Advances in AAV technology for delivering genetically encoded cargo to the nonhuman primate nervous system. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 4:100086. [PMID: 37397806 PMCID: PMC10313870 DOI: 10.1016/j.crneur.2023.100086] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/05/2023] [Accepted: 03/17/2023] [Indexed: 07/04/2023] Open
Abstract
Modern neuroscience approaches including optogenetics, calcium imaging, and other genetic manipulations have facilitated our ability to dissect specific circuits in rodent models to study their role in neurological disease. These approaches regularly use viral vectors to deliver genetic cargo (e.g., opsins) to specific tissues and genetically-engineered rodents to achieve cell-type specificity. However, the translatability of these rodent models, cross-species validation of identified targets, and translational efficacy of potential therapeutics in larger animal models like nonhuman primates remains difficult due to the lack of efficient primate viral vectors. A refined understanding of the nonhuman primate nervous system promises to deliver insights that can guide the development of treatments for neurological and neurodegenerative diseases. Here, we outline recent advances in the development of adeno-associated viral vectors for optimized use in nonhuman primates. These tools promise to help open new avenues for study in translational neuroscience and further our understanding of the primate brain.
Collapse
Affiliation(s)
- Lillian J. Campos
- Department of Psychology and the California National Primate Research Center, University of California, Davis, CA, 05616, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Cynthia M. Arokiaraj
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Miguel R. Chuapoco
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Nick Goeden
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Capsida Biotherapeutics, Thousand Oaks, CA, 91320, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| | - Andrew S. Fox
- Department of Psychology and the California National Primate Research Center, University of California, Davis, CA, 05616, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, 20815, USA
| |
Collapse
|
4
|
Zoabi S, Andreyanov M, Heinrich R, Ron S, Carmi I, Gutfreund Y, Berlin S. A custom-made AAV1 variant (AAV1-T593K) enables efficient transduction of Japanese quail neurons in vitro and in vivo. Commun Biol 2023; 6:337. [PMID: 36977781 PMCID: PMC10050006 DOI: 10.1038/s42003-023-04712-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 03/15/2023] [Indexed: 03/30/2023] Open
Abstract
The widespread use of rodents in neuroscience has prompted the development of optimized viral variants for transduction of brain cells, in vivo. However, many of the viruses developed are less efficient in other model organisms, with birds being among the most resistant to transduction by current viral tools. Resultantly, the use of genetically-encoded tools and methods in avian species is markedly lower than in rodents; likely holding the field back. We sought to bridge this gap by developing custom viruses towards the transduction of brain cells of the Japanese quail. We first develop a protocol for culturing primary neurons and glia from quail embryos, followed by characterization of cultures via immunostaining, single cell mRNA sequencing, patch clamp electrophysiology and calcium imaging. We then leveraged the cultures for the rapid screening of various viruses, only to find that all yielded poor to no infection of cells in vitro. However, few infected neurons were obtained by AAV1 and AAV2. Scrutiny of the sequence of the AAV receptor found in quails led us to rationally design a custom-made AAV variant (AAV1-T593K; AAV1*) that exhibits improved transduction efficiencies in vitro and in vivo (14- and five-fold, respectively). Together, we present unique culturing method, transcriptomic profiles of quail's brain cells and a custom-tailored AAV1 for transduction of quail neurons in vitro and in vivo.
Collapse
Affiliation(s)
- Shaden Zoabi
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Michael Andreyanov
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Ronit Heinrich
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Shaked Ron
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Ido Carmi
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Yoram Gutfreund
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel
| | - Shai Berlin
- Department of Neuroscience, Ruth and Bruce Rappaport Faculty of Medicine, Technion- Israel Institute of Technology, Haifa, Israel.
| |
Collapse
|
5
|
Dadarwal R, Ortiz-Rios M, Boretius S. Fusion of quantitative susceptibility maps and T1-weighted images improve brain tissue contrast in primates. Neuroimage 2022; 264:119730. [PMID: 36332851 DOI: 10.1016/j.neuroimage.2022.119730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/12/2022] [Accepted: 11/01/2022] [Indexed: 11/06/2022] Open
Abstract
Recent progress in quantitative susceptibility mapping (QSM) has enabled the accurate delineation of submillimeter-scale subcortical brain structures in humans. However, the simultaneous visualization of cortical, subcortical, and white matter structure remains challenging, utilizing QSM data solely. Here we present TQ-SILiCON, a fusion method that enhances the contrast of cortex and subcortical structures and provides an excellent white matter delineation by combining QSM and conventional T1-weighted (T1w) images. In this study, we first applied QSM in the macaque monkey to map iron-rich subcortical structures. Implementing the same QSM acquisition and analysis methods allowed a similar accurate delineation of subcortical structures in humans. However, the QSM contrast of white and cortical gray matter was not sufficient for appropriate segmentation. Applying automatic brain tissue segmentation to TQ-SILiCON images of the macaque improved the classification of subcortical brain structures as compared to the single T1 contrast by maintaining an excellent white to cortical gray matter contrast. Furthermore, we validated our dual-contrast fusion approach in humans and similarly demonstrated improvements in automated segmentation of the cortex and subcortical structures. We believe the proposed contrast will facilitate translational studies in nonhuman primates to investigate the pathophysiology of neurodegenerative diseases that affect subcortical structures such as the basal ganglia in humans.
Collapse
Affiliation(s)
- Rakshit Dadarwal
- Functional Imaging Laboratory, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany; Georg-August University of Göttingen, Göttingen, Germany.
| | - Michael Ortiz-Rios
- Functional Imaging Laboratory, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany; Leibniz Science Campus Primate Cognition, Göttingen, Germany
| | - Susann Boretius
- Functional Imaging Laboratory, German Primate Center - Leibniz Institute for Primate Research, Göttingen, Germany; Georg-August University of Göttingen, Göttingen, Germany; Leibniz Science Campus Primate Cognition, Göttingen, Germany
| |
Collapse
|
6
|
Srinivasan E, Ram V, Rajasekaran R. A review on Huntington protein Insight into protein aggregation and therapeutic interventions. Curr Drug Metab 2022; 23:260-282. [PMID: 35319359 DOI: 10.2174/1389200223666220321103942] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 12/13/2021] [Accepted: 01/15/2022] [Indexed: 11/22/2022]
Abstract
Huntington disease (HD) is a distressing, innate neurodegenerative disease that descends from CAG repeat expansion in the huntingtin gene causing behavioral changes, motor dysfunction, and dementia in children and adults. Mutation in huntingtin (HTT) protein has been suggested to cause neuron loss in the cortex and striatum through various mechanisms including abnormal regulation of transcription, proteasomal dysfunction, post-translational modification, and other events, regulating toxicity. Pathogenesis of HD involves cleavage of the huntingtin protein followed by the neuronal accumulation of its aggregated form. Several research groups made possible efforts to reduce huntingtin gene expression, protein accumulation, and protein aggregation using inhibitors and molecular chaperones as developing drugs against HD. Herein, we review the mechanism proposed towards the formation of HTT protein aggregation and the impact of therapeutic strategies for the treatment of HD.
Collapse
Affiliation(s)
- E Srinivasan
- Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore - 632014, Tamil Nadu, India
- Department of Bioinformatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai - 602105, Tamil Nadu, India
| | - Vavish Ram
- Bioinformatics Lab, Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore - 632014, Tamil Nadu, India
| | - R Rajasekaran
- Bioinformatics Lab, Department of Biotechnology, School of Bio Sciences and Technology, Vellore Institute of Technology, Vellore - 632014, Tamil Nadu, India
| |
Collapse
|
7
|
Menon S, Kofoed RH, Nabbouh F, Xhima K, Al-Fahoum Y, Langman T, Mount HTJ, Shihabuddin LS, Sardi SP, Fraser PE, Watts JC, Aubert I, Tandon A. Viral alpha-synuclein knockdown prevents spreading synucleinopathy. Brain Commun 2021; 3:fcab247. [PMID: 34761222 PMCID: PMC8576194 DOI: 10.1093/braincomms/fcab247] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 07/14/2021] [Accepted: 07/17/2021] [Indexed: 12/19/2022] Open
Abstract
The accumulation of aggregated alpha-synuclein (α-syn) in Parkinson's disease, dementia with Lewy bodies and multiple system atrophy is thought to involve a common prion-like mechanism, whereby misfolded α-syn provides a conformational template for further accumulation of pathological α-syn. We tested whether silencing α-syn gene expression could reduce native non-aggregated α-syn substrate and thereby disrupt the propagation of pathological α-syn initiated by seeding with synucleinopathy-affected mouse brain homogenates. Unilateral intracerebral injections of adeno-associated virus serotype-1 encoding microRNA targeting the α-syn gene reduced the extent and severity of both the α-syn pathology and motor deficits. Importantly, a moderate 50% reduction in α-syn was sufficient to prevent the spread of α-syn pathology to distal brain regions. Our study combines behavioural, immunohistochemical and biochemical data that strongly support α-syn knockdown gene therapy for synucleinopathies.
Collapse
Affiliation(s)
- Sindhu Menon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
| | - Rikke H Kofoed
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
| | - Fadl Nabbouh
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
| | - Kristiana Xhima
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Yasmeen Al-Fahoum
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
| | - Tammy Langman
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
| | - Howard T J Mount
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
- Department of Psychiatry, University of Toronto, Toronto, ON M5S 1A1, Canada
- Department of Physiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Lamya S Shihabuddin
- Sanofi, Framingham, MA 01701, USA
- Present address: 5AM Ventures, Boston, MA, USA
| | | | - Paul E Fraser
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Joel C Watts
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Isabelle Aubert
- Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A1, Canada
| | - Anurag Tandon
- Tanz Centre for Research in Neurodegenerative Diseases, Toronto, ON M5T 0S8, Canada
- Department of Medicine, University of Toronto, Toronto, ON M5S 1A1, Canada
- Correspondence to: Anurag Tandon, PhD Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Krembil Discovery Tower Rm. 4KD481, 60 Leonard Ave, Toronto, ON M5T 0S8, Canada E-mail:
| |
Collapse
|
8
|
Lonser RR, Akhter AS, Zabek M, Elder JB, Bankiewicz KS. Direct convective delivery of adeno-associated virus gene therapy for treatment of neurological disorders. J Neurosurg 2021; 134:1751-1763. [PMID: 32915526 DOI: 10.3171/2020.4.jns20701] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/16/2020] [Indexed: 11/06/2022]
Abstract
Molecular biological insights have led to a fundamental understanding of the underlying genomic mechanisms of nervous system disease. These findings have resulted in the identification of therapeutic genes that can be packaged in viral capsids for the treatment of a variety of neurological conditions, including neurodegenerative, metabolic, and enzyme deficiency disorders. Recent data have demonstrated that gene-carrying viral vectors (most often adeno-associated viruses) can be effectively distributed by convection-enhanced delivery (CED) in a safe, reliable, targeted, and homogeneous manner across the blood-brain barrier. Critically, these vectors can be monitored using real-time MRI of a co-infused surrogate tracer to accurately predict vector distribution and transgene expression at the perfused site. The unique properties of CED of adeno-associated virus vectors allow for cell-specific transgene manipulation of the infused anatomical site and/or widespread interconnected sites via antero- and/or retrograde transport. The authors review the convective properties of viral vectors, associated technology, and clinical applications.
Collapse
Affiliation(s)
- Russell R Lonser
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Asad S Akhter
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Mirosław Zabek
- 2Department of Neurological Surgery, Bródno Hospital, Warsaw, Poland
| | - J Bradley Elder
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Krystof S Bankiewicz
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| |
Collapse
|
9
|
Piguet F, de Saint Denis T, Audouard E, Beccaria K, André A, Wurtz G, Schatz R, Alves S, Sevin C, Zerah M, Cartier N. The Challenge of Gene Therapy for Neurological Diseases: Strategies and Tools to Achieve Efficient Delivery to the Central Nervous System. Hum Gene Ther 2021; 32:349-374. [PMID: 33167739 DOI: 10.1089/hum.2020.105] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
For more than 10 years, gene therapy for neurological diseases has experienced intensive research growth and more recently therapeutic interventions for multiple indications. Beneficial results in several phase 1/2 clinical studies, together with improved vector technology have advanced gene therapy for the central nervous system (CNS) in a new era of development. Although most initial strategies have focused on orphan genetic diseases, such as lysosomal storage diseases, more complex and widespread conditions like Alzheimer's disease, Parkinson's disease, epilepsy, or chronic pain are increasingly targeted for gene therapy. Increasing numbers of applications and patients to be treated will require improvement and simplification of gene therapy protocols to make them accessible to the largest number of affected people. Although vectors and manufacturing are a major field of academic research and industrial development, there is a growing need to improve, standardize, and simplify delivery methods. Delivery is the major issue for CNS therapies in general, and particularly for gene therapy. The blood-brain barrier restricts the passage of vectors; strategies to bypass this obstacle are a central focus of research. In this study, we present the different ways that can be used to deliver gene therapy products to the CNS. We focus on results obtained in large animals that have allowed the transfer of protocols to human patients and have resulted in the generation of clinical data. We discuss the different routes of administration, their advantages, and their limitations. We describe techniques, equipment, and protocols and how they should be selected for safe delivery and improved efficiency for the next generation of gene therapy trials for CNS diseases.
Collapse
Affiliation(s)
- Françoise Piguet
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Timothée de Saint Denis
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Emilie Audouard
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Kevin Beccaria
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Arthur André
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Neurosurgery, Hôpitaux Universitaires La Pitié-Salpêtrière, Sorbonne Universités, UPMC Univ Paris 6, Paris, France
| | - Guillaume Wurtz
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Raphael Schatz
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| | - Sandro Alves
- BrainVectis-Askbio France, iPeps Paris Brain Institute, Paris, France
| | - Caroline Sevin
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,BrainVectis-Askbio France, iPeps Paris Brain Institute, Paris, France.,APHP, Department of Neurology, Hopital le Kremlin Bicetre, Paris, France
| | - Michel Zerah
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France.,APHP, Department of Pediatric Neurosurgery, Hôpital Necker-Enfants Malades, APHP Centre. Université de Paris, Paris, France
| | - Nathalie Cartier
- NeuroGenCell, INSERM U1127, Paris Brain Institute (ICM), Sorbonne University, CNRS, AP-HP, University Hospital Pitié-Salpêtrière, Paris, France
| |
Collapse
|
10
|
Spronck EA, Vallès A, Lampen MH, Montenegro-Miranda PS, Keskin S, Heijink L, Evers MM, Petry H, van Deventer SJ, Konstantinova P, de Haan M. Intrastriatal Administration of AAV5-miHTT in Non-Human Primates and Rats Is Well Tolerated and Results in miHTT Transgene Expression in Key Areas of Huntington Disease Pathology. Brain Sci 2021; 11:brainsci11020129. [PMID: 33498212 PMCID: PMC7908995 DOI: 10.3390/brainsci11020129] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/10/2021] [Accepted: 01/17/2021] [Indexed: 02/04/2023] Open
Abstract
Huntington disease (HD) is a fatal, neurodegenerative genetic disorder with aggregation of mutant Huntingtin protein (mutHTT) in the brain as a key pathological mechanism. There are currently no disease modifying therapies for HD; however, HTT-lowering therapies hold promise. Recombinant adeno-associated virus serotype 5 expressing a microRNA that targets HTT mRNA (AAV5-miHTT) is in development for the treatment of HD with promising results in rodent and minipig HD models. To support a clinical trial, toxicity studies were performed in non-human primates (NHP, Macaca fascicularis) and Sprague-Dawley rats to evaluate the safety of AAV5-miHTT, the neurosurgical administration procedure, vector delivery and expression of the miHTT transgene during a 6-month observation period. For accurate delivery of AAV5-miHTT to the striatum, real-time magnetic resonance imaging (MRI) with convection-enhanced delivery (CED) was used in NHP. Catheters were successfully implanted in 24 NHP, without neurological symptoms, and resulted in tracer signal in the target areas. Widespread vector DNA and miHTT transgene distribution in the brain was found, particularly in areas associated with HD pathology. Intrastriatal administration of AAV5-miHTT was well tolerated with no clinically relevant changes in either species. These studies demonstrate the excellent safety profile of AAV5-miHTT, the reproducibility and tolerability of intrastriatal administration, and the delivery of AAV5-miHTT to the brain, which support the transition of AAV5-miHTT into clinical studies.
Collapse
Affiliation(s)
- Elisabeth A. Spronck
- uniQure biopharma B.V., 1105 BP Amsterdam, The Netherlands; (A.V.); (M.H.L.); (P.S.M.-M.); (S.K.); (L.H.); (M.M.E.); (H.P.); (P.K.)
- Correspondence: ; Tel.: +31-(0)20-240-6091
| | - Astrid Vallès
- uniQure biopharma B.V., 1105 BP Amsterdam, The Netherlands; (A.V.); (M.H.L.); (P.S.M.-M.); (S.K.); (L.H.); (M.M.E.); (H.P.); (P.K.)
| | - Margit H. Lampen
- uniQure biopharma B.V., 1105 BP Amsterdam, The Netherlands; (A.V.); (M.H.L.); (P.S.M.-M.); (S.K.); (L.H.); (M.M.E.); (H.P.); (P.K.)
| | - Paula S. Montenegro-Miranda
- uniQure biopharma B.V., 1105 BP Amsterdam, The Netherlands; (A.V.); (M.H.L.); (P.S.M.-M.); (S.K.); (L.H.); (M.M.E.); (H.P.); (P.K.)
| | - Sonay Keskin
- uniQure biopharma B.V., 1105 BP Amsterdam, The Netherlands; (A.V.); (M.H.L.); (P.S.M.-M.); (S.K.); (L.H.); (M.M.E.); (H.P.); (P.K.)
| | - Liesbeth Heijink
- uniQure biopharma B.V., 1105 BP Amsterdam, The Netherlands; (A.V.); (M.H.L.); (P.S.M.-M.); (S.K.); (L.H.); (M.M.E.); (H.P.); (P.K.)
| | - Melvin M. Evers
- uniQure biopharma B.V., 1105 BP Amsterdam, The Netherlands; (A.V.); (M.H.L.); (P.S.M.-M.); (S.K.); (L.H.); (M.M.E.); (H.P.); (P.K.)
| | - Harald Petry
- uniQure biopharma B.V., 1105 BP Amsterdam, The Netherlands; (A.V.); (M.H.L.); (P.S.M.-M.); (S.K.); (L.H.); (M.M.E.); (H.P.); (P.K.)
| | - Sander J. van Deventer
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands;
| | - Pavlina Konstantinova
- uniQure biopharma B.V., 1105 BP Amsterdam, The Netherlands; (A.V.); (M.H.L.); (P.S.M.-M.); (S.K.); (L.H.); (M.M.E.); (H.P.); (P.K.)
| | - Martin de Haan
- Madeha Management & Consultancy, 1222 LM Nederhorst den Berg, The Netherlands;
| |
Collapse
|
11
|
Barker RA, Fujimaki M, Rogers P, Rubinsztein DC. Huntingtin-lowering strategies for Huntington's disease. Expert Opin Investig Drugs 2020; 29:1125-1132. [PMID: 32745442 DOI: 10.1080/13543784.2020.1804552] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Huntington's disease (HD) is an incurable, autosomal dominant neurodegenerative disease caused by an abnormally long polyglutamine tract in the huntingtin protein. Because this mutation causes disease via gain-of-function, lowering huntingtin levels represents a rational therapeutic strategy. AREAS COVERED We searched MEDLINE, CENTRAL, and other trial databases, and relevant company and HD funding websites for press releases until April 2020 to review strategies for huntingtin lowering, including autophagy and PROTACs, which have been studied in preclinical models. We focussed our analyses on oligonucleotide (ASOs) and miRNA approaches, which have entered or are about to enter clinical trials. EXPERT OPINION ASO and mRNA approaches for lowering mutant huntingtin protein production and strategies for increasing mutant huntingtin clearance are attractive because they target the cause of disease. However, questions concerning the optimal mode of delivery and associated safety issues remain. It is unclear if the human CNS coverage with intrathecal or intraparenchymal delivery will be sufficient for efficacy. The extent that one must lower mutant huntingtin levels for it to be therapeutic is uncertain and the extent to which CNS lowering of wild-type huntingtin is safe is unclear. Polypharmacy may be an effective approach for ameliorating signs and symptoms and for preventing/delaying onset and progression.
Collapse
Affiliation(s)
- Roger A Barker
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge , Cambridge, UK
| | - Motoki Fujimaki
- Department of Medical Genetics, Cambridge Institute for Medical Research , Cambridge, UK.,UK Dementia Research Institute , Cambridge, UK
| | - Priya Rogers
- Department of Clinical Neurosciences, John Van Geest Centre for Brain Repair, and MRC-WT Cambridge Stem Cell Institute, University of Cambridge , Cambridge, UK
| | - David C Rubinsztein
- Department of Medical Genetics, Cambridge Institute for Medical Research , Cambridge, UK.,UK Dementia Research Institute , Cambridge, UK
| |
Collapse
|
12
|
Tabrizi SJ, Flower MD, Ross CA, Wild EJ. Huntington disease: new insights into molecular pathogenesis and therapeutic opportunities. Nat Rev Neurol 2020; 16:529-546. [PMID: 32796930 DOI: 10.1038/s41582-020-0389-4] [Citation(s) in RCA: 241] [Impact Index Per Article: 60.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/29/2020] [Indexed: 12/11/2022]
Abstract
Huntington disease (HD) is a neurodegenerative disease caused by CAG repeat expansion in the huntingtin gene (HTT) and involves a complex web of pathogenic mechanisms. Mutant HTT (mHTT) disrupts transcription, interferes with immune and mitochondrial function, and is aberrantly modified post-translationally. Evidence suggests that the mHTT RNA is toxic, and at the DNA level, somatic CAG repeat expansion in vulnerable cells influences the disease course. Genome-wide association studies have identified DNA repair pathways as modifiers of somatic instability and disease course in HD and other repeat expansion diseases. In animal models of HD, nucleocytoplasmic transport is disrupted and its restoration is neuroprotective. Novel cerebrospinal fluid (CSF) and plasma biomarkers are among the earliest detectable changes in individuals with premanifest HD and have the sensitivity to detect therapeutic benefit. Therapeutically, the first human trial of an HTT-lowering antisense oligonucleotide successfully, and safely, reduced the CSF concentration of mHTT in individuals with HD. A larger trial, powered to detect clinical efficacy, is underway, along with trials of other HTT-lowering approaches. In this Review, we discuss new insights into the molecular pathogenesis of HD and future therapeutic strategies, including the modulation of DNA repair and targeting the DNA mutation itself.
Collapse
Affiliation(s)
- Sarah J Tabrizi
- Huntington's Disease Centre, University College London, London, UK. .,Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK. .,UK Dementia Research Institute, University College London, London, UK.
| | - Michael D Flower
- Huntington's Disease Centre, University College London, London, UK.,Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK.,UK Dementia Research Institute, University College London, London, UK
| | - Christopher A Ross
- Departments of Neurology, Neuroscience and Pharmacology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Edward J Wild
- Huntington's Disease Centre, University College London, London, UK.,Department of Neurodegenerative Disease, Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
13
|
Cushnie AK, El-Nahal HG, Bohlen MO, May PJ, Basso MA, Grimaldi P, Wang MZ, de Velasco Ezequiel MF, Sommer MA, Heilbronner SR. Using rAAV2-retro in rhesus macaques: Promise and caveats for circuit manipulation. J Neurosci Methods 2020; 345:108859. [PMID: 32668316 DOI: 10.1016/j.jneumeth.2020.108859] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Revised: 07/01/2020] [Accepted: 07/10/2020] [Indexed: 12/21/2022]
Abstract
BACKGROUND Recent genetic technologies such as opto- and chemogenetics allow for the manipulation of brain circuits with unprecedented precision. Most studies employing these techniques have been undertaken in rodents, but a more human-homologous model for studying the brain is the nonhuman primate (NHP). Optimizing viral delivery of transgenes encoding actuator proteins could revolutionize the way we study neuronal circuits in NHPs. NEW METHOD: rAAV2-retro, a popular new capsid variant, produces robust retrograde labeling in rodents. Whether rAAV2-retro's highly efficient retrograde transport would translate to NHPs was unknown. Here, we characterized the anatomical distribution of labeling following injections of rAAV2-retro encoding opsins or DREADDs in the cortico-basal ganglia and oculomotor circuits of rhesus macaques. RESULTS rAAV2-retro injections in striatum, frontal eye field, and superior colliculus produced local labeling at injection sites and robust retrograde labeling in many afferent regions. In every case, however, a few brain regions with well-established projections to the injected structure lacked retrogradely labeled cells. We also observed robust terminal field labeling in downstream structures. COMPARISON WITH EXISTING METHOD(S) Patterns of labeling were similar to those obtained with traditional tract-tracers, except for some afferent labeling that was noticeably absent. CONCLUSIONS rAAV2-retro promises to be useful for circuit manipulation via retrograde transduction in NHPs, but caveats were revealed by our findings. Some afferently connected regions lacked retrogradely labeled cells, showed robust axon terminal labeling, or both. This highlights the importance of anatomically characterizing rAAV2-retro's expression in target circuits in NHPs before moving to manipulation studies.
Collapse
Affiliation(s)
- Adriana K Cushnie
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | - Hala G El-Nahal
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States
| | - Martin O Bohlen
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States
| | - Paul J May
- Department of Neurobiology and Anatomical Sciences, University of Mississippi Medical Center, Jackson, 39216, United States
| | - Michele A Basso
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences and Neurobiology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, Univ. of California Los Angeles, Los Angeles, CA 90095, United States
| | - Piercesare Grimaldi
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences and Neurobiology, Jane and Terry Semel Institute for Neuroscience and Human Behavior, David Geffen School of Medicine, Univ. of California Los Angeles, Los Angeles, CA 90095, United States
| | - Maya Zhe Wang
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States
| | | | - Marc A Sommer
- Department of Biomedical Engineering, Duke University, Durham, NC 27708, United States; Department of Neurobiology, Duke University School of Medicine, Durham, NC 27708, United States; Center for Cognitive Neuroscience, Duke University, Durham, NC 27708, United States
| | - Sarah R Heilbronner
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55455, United States.
| |
Collapse
|
14
|
Nieuwenhuis B, Haenzi B, Hilton S, Carnicer-Lombarte A, Hobo B, Verhaagen J, Fawcett JW. Optimization of adeno-associated viral vector-mediated transduction of the corticospinal tract: comparison of four promoters. Gene Ther 2020; 28:56-74. [PMID: 32576975 PMCID: PMC7902269 DOI: 10.1038/s41434-020-0169-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 06/01/2020] [Accepted: 06/11/2020] [Indexed: 12/22/2022]
Abstract
Adeno-associated viral vectors are widely used as vehicles for gene transfer to the nervous system. The promoter and viral vector serotype are two key factors that determine the expression dynamics of the transgene. A previous comparative study has demonstrated that AAV1 displays efficient transduction of layer V corticospinal neurons, but the optimal promoter for transgene expression in corticospinal neurons has not been determined yet. In this paper, we report a side-by-side comparison between four commonly used promoters: the short CMV early enhancer/chicken β actin (sCAG), human cytomegalovirus (hCMV), mouse phosphoglycerate kinase (mPGK) and human synapsin (hSYN) promoter. Reporter constructs with each of these promoters were packaged in AAV1, and were injected in the sensorimotor cortex of rats and mice in order to transduce the corticospinal tract. Transgene expression levels and the cellular transduction profile were examined after 6 weeks. The AAV1 vectors harbouring the hCMV and sCAG promoters resulted in transgene expression in neurons, astrocytes and oligodendrocytes. The mPGK and hSYN promoters directed the strongest transgene expression. The mPGK promoter did drive expression in cortical neurons and oligodendrocytes, while transduction with AAV harbouring the hSYN promoter resulted in neuron-specific expression, including perineuronal net expressing interneurons and layer V corticospinal neurons. This promoter comparison study contributes to improve transgene delivery into the brain and spinal cord. The optimized transduction of the corticospinal tract will be beneficial for spinal cord injury research.
Collapse
Affiliation(s)
- Bart Nieuwenhuis
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK. .,Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.
| | - Barbara Haenzi
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Sam Hilton
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Alejandro Carnicer-Lombarte
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK
| | - Barbara Hobo
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands
| | - Joost Verhaagen
- Laboratory for Regeneration of Sensorimotor Systems, Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences (KNAW), Meibergdreef 47, 1105 BA, Amsterdam, The Netherlands.,Centre for Neurogenomics and Cognitive Research, Amsterdam Neuroscience, Vrije Universiteit Amsterdam, De Boelelaan 1085, 1081 HV, Amsterdam, The Netherlands
| | - James W Fawcett
- John van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Forvie Site, Robinson Way, Cambridge, CB2 0PY, UK.,Centre of Reconstructive Neuroscience, Institute of Experimental Medicine, Vídeňská 1083, 142 20, Prague 4, Czech Republic
| |
Collapse
|
15
|
Maxan A, Sciacca G, Alpaugh M, Tao Z, Breger L, Dehay B, Ling Z, Chuan Q, Cisbani G, Masnata M, Salem S, Lacroix S, Oueslati A, Bezard E, Cicchetti F. Use of adeno-associated virus-mediated delivery of mutant huntingtin to study the spreading capacity of the protein in mice and non-human primates. Neurobiol Dis 2020; 141:104951. [PMID: 32439599 DOI: 10.1016/j.nbd.2020.104951] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2019] [Revised: 03/20/2020] [Accepted: 05/16/2020] [Indexed: 01/27/2023] Open
Abstract
In order to model various aspects of Huntington's disease (HD) pathology, in particular protein spread, we administered adeno-associated virus (AAV) expressing green fluorescent protein (GFP) or GFP coupled to HTT-Exon1 (19Q or 103Q) to the central nervous system of adult wild-type (WT) mice and non-human primates. All animals underwent behavioral testing and post-mortem analyses to determine the long-term consequences of AAV injection. Both mice and non-human primates demonstrated behavioral changes at 2-3 weeks post-surgery. In mice, these changes were absent after 3 months while in non-human primates, they persisted in the majority of tested animals. Post-mortem analysis revealed that spreading of the aggregates was limited, although the virus did spread between synaptically-connected brain regions. Despite circumscribed spreading, the presence of mHTT generated changes in endogenous huntingtin (HTT) levels in both models. Together, these results suggest that viral expression of mHTTExon1 can induce spreading and seeding of HTT in both mice and non-human primates.
Collapse
Affiliation(s)
- Alexander Maxan
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada
| | - Giacomo Sciacca
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada
| | - Melanie Alpaugh
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada
| | - Zhu Tao
- Institute of Laboratory Animal Sciences, China Academy of Medical Sciences, Beijing, China
| | - Ludivine Breger
- Université de Bordeaux, Institut des maladies neurodégénératives, UMR 5293, CNRS UMR 5293, Bordeaux, France; Centre National de la Recherche Scientifique, Institut des maladies neurodégénératives, UMR 5293, 33076 Bordeaux, France
| | - Benjamin Dehay
- Université de Bordeaux, Institut des maladies neurodégénératives, UMR 5293, CNRS UMR 5293, Bordeaux, France; Centre National de la Recherche Scientifique, Institut des maladies neurodégénératives, UMR 5293, 33076 Bordeaux, France
| | - Zhang Ling
- Institute of Laboratory Animal Sciences, China Academy of Medical Sciences, Beijing, China
| | - Qin Chuan
- Institute of Laboratory Animal Sciences, China Academy of Medical Sciences, Beijing, China.
| | - Giulia Cisbani
- University of Toronto, Department of Nutritional Sciences, Toronto, ON M5S 1A8, Canada
| | - Maria Masnata
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada
| | - Shireen Salem
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada
| | - Steve Lacroix
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Médicine Moléculaire, Université Laval, Québec, QC G1K 0A6, Canada
| | - Abid Oueslati
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Médicine Moléculaire, Université Laval, Québec, QC G1K 0A6, Canada
| | - Erwan Bezard
- Institute of Laboratory Animal Sciences, China Academy of Medical Sciences, Beijing, China; Université de Bordeaux, Institut des maladies neurodégénératives, UMR 5293, CNRS UMR 5293, Bordeaux, France; Centre National de la Recherche Scientifique, Institut des maladies neurodégénératives, UMR 5293, 33076 Bordeaux, France
| | - Francesca Cicchetti
- Centre de Recherche du CHU de Québec - Université Laval, Axe Neurosciences, Québec, QC G1V 4G2, Canada; Département de Psychiatrie & Neurosciences, Université Laval, Québec, QC G1K 0A6, Canada.
| |
Collapse
|
16
|
Weiss AR, Liguore WA, Domire JS, Button D, McBride JL. Intra-striatal AAV2.retro administration leads to extensive retrograde transport in the rhesus macaque brain: implications for disease modeling and therapeutic development. Sci Rep 2020; 10:6970. [PMID: 32332773 PMCID: PMC7181773 DOI: 10.1038/s41598-020-63559-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2020] [Accepted: 04/01/2020] [Indexed: 11/09/2022] Open
Abstract
Recently, AAV2.retro, a new capsid variant capable of efficient retrograde transport in brain, was generated in mice using a directed evolution approach. However, it remains unclear to what degree transport will be recapitulated in the substantially larger and more complex nonhuman primate (NHP) brain. Here, we compared the biodistribution of AAV2.retro with its parent serotype, AAV2, in adult macaques following delivery into the caudate and putamen, brain regions which comprise the striatum. While AAV2 transduction was primarily limited to the injected brain regions, AAV2.retro transduced cells in the striatum and in dozens of cortical and subcortical regions with known striatal afferents. We then evaluated the capability of AAV2.retro to deliver disease-related gene cargo to biologically-relevant NHP brain circuits by packaging a fragment of human mutant HTT, the causative gene mutation in Huntington’s disease. Following intra-striatal delivery, pathological mHTT-positive protein aggregates were distributed widely among cognitive, motor, and limbic cortico-basal ganglia circuits. Together, these studies demonstrate strong retrograde transport of AAV2.retro in NHP brain, highlight its utility in developing novel NHP models of brain disease and suggest its potential for querying circuit function and delivering therapeutic genes in the brain, particularly where treating dysfunctional circuits, versus single brain regions, is warranted.
Collapse
Affiliation(s)
- Alison R Weiss
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA
| | - William A Liguore
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA
| | - Jacqueline S Domire
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA
| | - Dana Button
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA
| | - Jodi L McBride
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, USA. .,Department of Behavioral Neuroscience, Oregon Health and Science University, Portland, USA. .,Department of Neurology, Oregon Health and Science University, Portland, USA.
| |
Collapse
|
17
|
Gonzalez-Alegre P. Recent advances in molecular therapies for neurological disease: triplet repeat disorders. Hum Mol Genet 2020; 28:R80-R87. [PMID: 31227833 DOI: 10.1093/hmg/ddz138] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 04/03/2019] [Accepted: 06/11/2019] [Indexed: 02/06/2023] Open
Abstract
Triplet repeat diseases (TRDs) are caused by pathogenic expansions of trinucleotide sequence repeats within coding and non-coding regions of different genes. They are typically progressive, very disabling and frequently involve the nervous system. Currently available symptomatic therapies provide modest benefit at best. The development of interventions that interfere with the natural history of these diseases is a priority. A common pathogenic process shared by most TRDs is the presence of toxicity from the messenger RNA or protein encoded by the gene harboring the abnormal expansion. Strategies to interfere with the expression of these genes using different molecular approaches are being pursued and have reached the clinical stage. This review will summarize the significant progress made in this field in the last few years, focusing on three main areas: the discovery of biomarkers of disease progression and target engagement, advances in preclinical studies for the polyglutamine ataxias and the initial clinical application in myotonic dystrophy type 1 and Huntington's disease.
Collapse
Affiliation(s)
- Pedro Gonzalez-Alegre
- Department of Neurology, the University of Pennsylvania, Philadelphia, PA 19104, USA.,Raymond G. Perelman Center for Cellular and Molecular Therapy, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| |
Collapse
|
18
|
Hirbec H, Déglon N, Foo LC, Goshen I, Grutzendler J, Hangen E, Kreisel T, Linck N, Muffat J, Regio S, Rion S, Escartin C. Emerging technologies to study glial cells. Glia 2020; 68:1692-1728. [PMID: 31958188 DOI: 10.1002/glia.23780] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 12/20/2019] [Accepted: 12/23/2019] [Indexed: 12/11/2022]
Abstract
Development, physiological functions, and pathologies of the brain depend on tight interactions between neurons and different types of glial cells, such as astrocytes, microglia, oligodendrocytes, and oligodendrocyte precursor cells. Assessing the relative contribution of different glial cell types is required for the full understanding of brain function and dysfunction. Over the recent years, several technological breakthroughs were achieved, allowing "glio-scientists" to address new challenging biological questions. These technical developments make it possible to study the roles of specific cell types with medium or high-content workflows and perform fine analysis of their mutual interactions in a preserved environment. This review illustrates the potency of several cutting-edge experimental approaches (advanced cell cultures, induced pluripotent stem cell (iPSC)-derived human glial cells, viral vectors, in situ glia imaging, opto- and chemogenetic approaches, and high-content molecular analysis) to unravel the role of glial cells in specific brain functions or diseases. It also illustrates the translation of some techniques to the clinics, to monitor glial cells in patients, through specific brain imaging methods. The advantages, pitfalls, and future developments are discussed for each technique, and selected examples are provided to illustrate how specific "gliobiological" questions can now be tackled.
Collapse
Affiliation(s)
- Hélène Hirbec
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Nicole Déglon
- Laboratory of Neurotherapies and Neuromodulation, Department of Clinical Neuroscience, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Lynette C Foo
- Neuroimmunology and Neurodegeneration Section, The Neuroscience and Rare Diseases Discovery and Translational Area, F. Hoffman-La Roche, Basel, Switzerland
| | - Inbal Goshen
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Jaime Grutzendler
- Department of Neurology, Yale University School of Medicine, New Haven, Connecticut, USA.,Department of Neuroscience, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Emilie Hangen
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Neurodegenerative Diseases Laboratory, Université Paris-Sud, Université Paris-Saclay, UMR 9199, Fontenay-aux-Roses, France
| | - Tirzah Kreisel
- Edmond and Lily Safra Center for Brain Sciences, The Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nathalie Linck
- Institute for Functional Genomics (IGF), University of Montpellier, CNRS, INSERM, Montpellier, France
| | - Julien Muffat
- Program in Neurosciences and Mental Health, The Hospital for Sick Children, and Department of Molecular Genetics, The University of Toronto, Toronto, Canada
| | - Sara Regio
- Laboratory of Neurotherapies and Neuromodulation, Department of Clinical Neuroscience, Lausanne University Hospital, University of Lausanne, Lausanne, Switzerland.,Laboratory of Neurotherapies and Neuromodulation, Neuroscience Research Center, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sybille Rion
- Neuroimmunology and Neurodegeneration Section, The Neuroscience and Rare Diseases Discovery and Translational Area, F. Hoffman-La Roche, Basel, Switzerland
| | - Carole Escartin
- Commissariat à l'Energie Atomique et aux Energies Alternatives, Département de la Recherche Fondamentale, Institut de Biologie François Jacob, MIRCen, Fontenay-aux-Roses, France.,Centre National de la Recherche Scientifique, Neurodegenerative Diseases Laboratory, Université Paris-Sud, Université Paris-Saclay, UMR 9199, Fontenay-aux-Roses, France
| |
Collapse
|
19
|
Ekman FK, Ojala DS, Adil MM, Lopez PA, Schaffer DV, Gaj T. CRISPR-Cas9-Mediated Genome Editing Increases Lifespan and Improves Motor Deficits in a Huntington's Disease Mouse Model. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:829-839. [PMID: 31465962 PMCID: PMC6717077 DOI: 10.1016/j.omtn.2019.07.009] [Citation(s) in RCA: 89] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/29/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022]
Abstract
Huntington's disease (HD) is a currently incurable and, ultimately, fatal neurodegenerative disorder caused by a CAG trinucleotide repeat expansion within exon 1 of the huntingtin (HTT) gene, which results in the production of a mutant protein that forms inclusions and selectively destroys neurons in the striatum and other adjacent structures. The RNA-guided Cas9 endonuclease from CRISPR-Cas9 systems is a versatile technology for inducing DNA double-strand breaks that can stimulate the introduction of frameshift-inducing mutations and permanently disable mutant gene function. Here, we show that the Cas9 nuclease from Staphylococcus aureus, a small Cas9 ortholog that can be packaged alongside a single guide RNA into a single adeno-associated virus (AAV) vector, can be used to disrupt the expression of the mutant HTT gene in the R6/2 mouse model of HD following its in vivo delivery to the striatum. Specifically, we found that CRISPR-Cas9-mediated disruption of the mutant HTT gene resulted in a ∼50% decrease in neuronal inclusions and significantly improved lifespan and certain motor deficits. These results thus illustrate the potential for CRISPR-Cas9 technology to treat HD and other autosomal dominant neurodegenerative disorders caused by a trinucleotide repeat expansion via in vivo genome editing.
Collapse
Affiliation(s)
- Freja K Ekman
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA
| | - David S Ojala
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Maroof M Adil
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Paola A Lopez
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; The Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA.
| | - Thomas Gaj
- Department of Bioengineering, University of Illinois, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
20
|
Stanek LM, Bu J, Shihabuddin LS. Astrocyte transduction is required for rescue of behavioral phenotypes in the YAC128 mouse model with AAV-RNAi mediated HTT lowering therapeutics. Neurobiol Dis 2019; 129:29-37. [DOI: 10.1016/j.nbd.2019.04.015] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 03/14/2019] [Accepted: 04/24/2019] [Indexed: 12/11/2022] Open
|
21
|
Lau CH, Ho JWT, Lo PK, Tin C. Targeted Transgene Activation in the Brain Tissue by Systemic Delivery of Engineered AAV1 Expressing CRISPRa. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 16:637-649. [PMID: 31108320 PMCID: PMC6526230 DOI: 10.1016/j.omtn.2019.04.015] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 04/14/2019] [Accepted: 04/14/2019] [Indexed: 01/10/2023]
Abstract
Targeted transcriptional modulation in the central nervous system (CNS) can be achieved by adeno-associated virus (AAV) delivery of CRISPR activation (CRISPRa) and interference (CRISPRi) transgenes. To enable AAV packaging, we constructed minimal CRISPRa and CRISPRi transgenes by fusing catalytically inactive Staphylococcus aureus Cas9 (dSaCas9) to the transcriptional activator (VP64 and VP160) and repressor (KRAB and SID4X) domains along with truncated regulatory elements. We then evaluated the performance of these constructs in two reporter assays (bioluminescent and fluorescent), five endogenous genes (Camk2a, Mycn, Nrf2, Keap1, and PDGFRA), and two cell lines (neuro-2a [N2a] and U87) by targeting the promoter and/or enhancer regions. To enable systemic delivery of AAVs to the CNS, we have also generated an AAV1-PHP.B by inserting a 7-mer PHP.B peptide on AAV1 capsid. We showed that AAV1-PHP.B can efficiently cross the blood-brain barrier (BBB) and be taken up by the brain tissue upon lateral tail vein injection in mice. Importantly, a single-dose intravenous administration of AAV1-PHP.B expressing CRISPRa was shown to achieve targeted transgene activation in the mouse brain. This proof-of-concept study will contribute to the development of a non-invasive, specific and potent AAV-CRISPR system for correcting transcriptional misregulation in broad brain areas and multiple neuroanatomical structures.
Collapse
Affiliation(s)
- Cia-Hin Lau
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Jonathan Weng-Thim Ho
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong
| | - Pik Kwan Lo
- Department of Chemistry, City University of Hong Kong, Kowloon Tong, Hong Kong; CityU Shenzhen Research Institute, Shenzhen, China
| | - Chung Tin
- Department of Biomedical Engineering, City University of Hong Kong, Kowloon Tong, Hong Kong; CityU Shenzhen Research Institute, Shenzhen, China.
| |
Collapse
|
22
|
Abstract
Huntington's disease (HD) is characterized by a significant loss of striatal neurons that project to the globus pallidus and substantia nigra, together with loss of cortical projection neurons in varying regions. Mutant huntingtin is suggested to drive the pathogenesis partially by downregulating corticostriatal brain-derived neurotrophic factor (BDNF) levels and signaling. Neurotrophic factors are endogenous peptides that promote the survival and maintenance of neurons. BDNF and other neurotrophic factors have shown neuroprotective benefits in various animal models of neurodegeneration, and are interesting candidates to protect the cell populations that are destined to die in HD. In an attempt to enhance the delivery of neurotrophic factors, several methods have been established to deliver long-term neurotrophic factor gene therapy to human target tissues. This chapter discusses two alternative approaches that have been shown to have potential to deliver neurotrophic factors as a neuroprotective gene therapy for HD. The methods are (1) ex vivo approach where encapsulated cells engineered to express neurotrophic factor are inserted into brain parenchyma or ventricle, and (2) in vivo viral vector therapy, in which viral vector is injected into desired brain area to express gene of interest in the host cells.
Collapse
|
23
|
Abstract
The 25 years since the identification of the gene responsible for Huntington disease (HD) have stood witness to profound discoveries about the nature of the disease and its pathogenesis. Despite this progress, however, the development of disease-modifying therapies has thus far been slow. Preclinical validation of the therapeutic potential of disrupted pathways in HD has led to the advancement of pharmacological agents, both novel and repurposed, for clinical evaluation. The most promising therapeutic approaches include huntingtin (HTT) lowering and modification as well as modulation of neuroinflammation and synaptic transmission. With clinical trials for many of these approaches imminent or currently ongoing, the coming years are promising not only for HD but also for more prevalent neurodegenerative disorders, such as Alzheimer and Parkinson disease, in which many of these pathways have been similarly implicated.
Collapse
|
24
|
Fuentes CM, Schaffer DV. Adeno-associated virus-mediated delivery of CRISPR-Cas9 for genome editing in the central nervous system. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2018; 7:33-41. [PMID: 34046535 PMCID: PMC8153090 DOI: 10.1016/j.cobme.2018.08.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The emergence of CRISPR-Cas9 as a powerful genome editing tool has led to several studies exploring its potential to treat neurological disorders. Cas9 and its sgRNA can be readily engineered to target any gene and can be multiplexed to target several genes at once. Furthermore, the use of adeno-associated virus (AAV) to deliver with Cas9 and its sgRNA is a promising therapeutic combination with strong potential to reach the clinic. Here we discuss how Cas9 editing has been utilized for gene insertion, knockout, and deletion in vivo for applications in the central nervous system (CNS). Furthermore, we highlight major challenges that remain for AAV-Cas9-sgRNA clinical translation.
Collapse
Affiliation(s)
- Christina M. Fuentes
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - David V. Schaffer
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
- Department of Chemical and Biolomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA
- The Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA
| |
Collapse
|
25
|
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: 29.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
|
26
|
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: 36] [Impact Index Per Article: 6.0] [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
|
27
|
Barnett SC, Perry BAL, Dalrymple-Alford JC, Parr-Brownlie LC. Optogenetic stimulation: Understanding memory and treating deficits. Hippocampus 2018; 28:457-470. [DOI: 10.1002/hipo.22960] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2018] [Revised: 04/24/2018] [Accepted: 05/02/2018] [Indexed: 01/01/2023]
Affiliation(s)
- S. C. Barnett
- Department of Psychology; University of Canterbury; Christchurch 8041 New Zealand
- Brain Research New Zealand; New Zealand
| | - B. A. L. Perry
- Department of Psychology; University of Canterbury; Christchurch 8041 New Zealand
- Brain Research New Zealand; New Zealand
| | - J. C. Dalrymple-Alford
- Department of Psychology; University of Canterbury; Christchurch 8041 New Zealand
- Brain Research New Zealand; New Zealand
- New Zealand Brain Research Institute; Christchurch New Zealand
| | - L. C. Parr-Brownlie
- Brain Research New Zealand; New Zealand
- Department of Anatomy, School of Biomedical Science; Brain Health Research Centre, University of Otago; Dunedin New Zealand
| |
Collapse
|
28
|
Sullivan JA, Stanek LM, Lukason MJ, Bu J, Osmond SR, Barry EA, O'Riordan CR, Shihabuddin LS, Cheng SH, Scaria A. Rationally designed AAV2 and AAVrh8R capsids provide improved transduction in the retina and brain. Gene Ther 2018; 25:205-219. [PMID: 29785047 DOI: 10.1038/s41434-018-0017-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Revised: 03/25/2018] [Accepted: 03/27/2018] [Indexed: 01/18/2023]
Abstract
The successful application of adeno-associated virus (AAV) gene delivery vectors as a therapeutic paradigm will require efficient gene delivery to the appropriate cells in affected organs. In this study, we utilized a rational design approach to introduce modifications to the AAV2 and AAVrh8R capsids and the resulting variants were evaluated for transduction activity in the retina and brain. The modifications disrupted either capsid/receptor binding or altered capsid surface charge. Specifically, we mutated AAV2 amino acids R585A and R588A, which are required for binding to its receptor, heparan sulfate proteoglycans, to generate a variant referred to as AAV2-HBKO. In contrast to parental AAV2, the AAV2-HBKO vector displayed low-transduction activity following intravitreal delivery to the mouse eye; however, following its subretinal delivery, AAV2-HBKO resulted in significantly greater photoreceptor transduction. Intrastriatal delivery of AAV2-HBKO to mice facilitated widespread striatal and cortical expression, in contrast to the restricted transduction pattern of the parental AAV2 vector. Furthermore, we found that altering the surface charge on the AAVrh8R capsid by modifying the number of arginine residues on the capsid surface had a profound impact on subretinal transduction. The data further validate the potential of capsid engineering to improve AAV gene therapy vectors for clinical applications.
Collapse
Affiliation(s)
| | - Lisa M Stanek
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| | | | - Jie Bu
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| | | | | | | | | | - Seng H Cheng
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| | - Abraham Scaria
- Sanofi, 49 New York Avenue, Framingham, MA, 01701-9322, USA
| |
Collapse
|
29
|
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: 4.0] [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
|
30
|
Keeler AM, Sapp E, Chase K, Sottosanti E, Danielson E, Pfister E, Stoica L, DiFiglia M, Aronin N, Sena-Esteves M. Cellular Analysis of Silencing the Huntington's Disease Gene Using AAV9 Mediated Delivery of Artificial Micro RNA into the Striatum of Q140/Q140 Mice. J Huntingtons Dis 2017; 5:239-248. [PMID: 27689620 DOI: 10.3233/jhd-160215] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND The genetic mutation in Huntington's disease (HD) is a CAG repeat expansion in the coding region of the huntingtin (Htt) gene. RNAi strategies have proven effective in substantially down-regulating Htt mRNA in the striatum through delivery of siRNAs or viral vectors based on whole tissue assays, but the extent of htt mRNA lowering in individual neurons is unknown. OBJECTIVE Here we characterize the effect of an AAV9-GFP-miRHtt vector on Htt mRNA levels in striatal neurons of Q140/Q140 knock-in mice. METHODS HD mice received bilateral striatal injections of AAV9-GFP-miRHtt or AAV9-GFP at 6 or 12 weeks and striata were evaluated at 6 months of age for levels of Htt mRNA and protein and for mRNA signal within striatal neurons using RNAscope multiplex fluorescence in situ hybridization. RESULTS Compared to controls, the striatum of 6-month old mice treated at 6 or 12 weeks of age with AAV9-GFP-miRHtt showed a reduction of 40-50% in Htt mRNA and lowering of 25-40% in protein levels. The number of Htt mRNA foci in medium spiny neurons (MSNs) of untreated Q140/Q140 mice varied widely per cell (0 to 34 per cell), with ∼10% of MSNs devoid of foci. AAV9-GFP-miRHtt treatment shifted the distribution toward lower numbers and the percentage of cells without foci increased to 14-20%. The average number of Htt mRNA foci per MSN was reduced by 43%. CONCLUSIONS The findings here show that intrastriatal infusion of an AAV9-GFP-miRHtt vector lowers mRNA expression of Htt in striatum by ∼50%, through a partial reduction in the number of copies of mutant Htt mRNAs per cell. These findings demonstrate at the neuronal level the variable levels of Htt mRNA expression in MSNs and the neuronal heterogeneity of RNAi dependent Htt mRNA knockdown.
Collapse
Affiliation(s)
- Allison M Keeler
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Ellen Sapp
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA
| | - Kathryn Chase
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Emily Sottosanti
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Eric Danielson
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | - Edith Pfister
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Lorelei Stoica
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marian DiFiglia
- Department of Neurology, Massachusetts General Hospital, Charlestown, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Neil Aronin
- Department of Medicine, University of Massachusetts Medical School, Worcester, MA, USA.,RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Miguel Sena-Esteves
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA.,Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| |
Collapse
|
31
|
Samaranch L, Blits B, San Sebastian W, Hadaczek P, Bringas J, Sudhakar V, Macayan M, Pivirotto PJ, Petry H, Bankiewicz KS. MR-guided parenchymal delivery of adeno-associated viral vector serotype 5 in non-human primate brain. Gene Ther 2017; 24:253-261. [PMID: 28300083 PMCID: PMC5404203 DOI: 10.1038/gt.2017.14] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/19/2017] [Accepted: 02/01/2017] [Indexed: 12/19/2022]
Abstract
The present study was designed to characterize transduction of non-human primate brain and spinal cord with AAV5 viral vector after parenchymal delivery. AAV5-CAG-GFP (1 × 1013 vector genomes per milliliter (vg ml−1)) was bilaterally infused either into putamen, thalamus or with the combination left putamen and right thalamus. Robust expression of GFP was seen throughout infusion sites and also in other distal nuclei. Interestingly, thalamic infusion of AAV5 resulted in the transduction of the entire corticospinal axis, indicating transport of AAV5 over long distances. Regardless of site of injection, AAV5 transduced both neurons and astrocytes equally. Our data demonstrate that AAV5 is a very powerful vector for the central nervous system and has potential for treatment of a wide range of neurological pathologies with cortical, subcortical and/or spinal cord affection.
Collapse
Affiliation(s)
- L Samaranch
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - B Blits
- Neurobiology, Research and Development, UniQure NV, Amsterdam 1105BA, The Netherlands
| | - W San Sebastian
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - P Hadaczek
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - J Bringas
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - V Sudhakar
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - M Macayan
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - P J Pivirotto
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - H Petry
- Neurobiology, Research and Development, UniQure NV, Amsterdam 1105BA, The Netherlands
| | - K S Bankiewicz
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| |
Collapse
|