1
|
Witteveen I, Balmer T. Comparative Analysis of Six Adeno-Associated Viral Vector Serotypes in Mouse Inferior Colliculus and Cerebellum. eNeuro 2024; 11:ENEURO.0391-24.2024. [PMID: 39467650 DOI: 10.1523/eneuro.0391-24.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/17/2024] [Accepted: 10/22/2024] [Indexed: 10/30/2024] Open
Abstract
Adeno-associated viral vector (AAV) serotypes vary in how effectively they express genes across different cell types and brain regions. Here we report a systematic comparison of the AAV serotypes 1, 2, 5, 8, 9, and the directed evolution derived AAVrg, in the inferior colliculus (IC) and cerebellum. The AAVs were identical apart from their different serotypes, each having a synapsin promotor and expressing GFP (AAV-hSyn-GFP). Identical titers and volumes were injected into the IC and cerebellum of adult male and female mice, and brains were sectioned and imaged 2 weeks later. Transduction efficacy, anterograde labeling of axonal projections, and retrograde labeling of somata were characterized and compared across serotypes. Cell-type tropism was assessed by analyzing the morphology of the GFP-labeled neurons in the cerebellar cortex. In both the cerebellum and IC, AAV1 expressed GFP in more cells, labeled a larger volume, and produced significantly brighter labeling than all other serotypes, indicating superior transgene expression. AAV1 labeled more Purkinje cells, unipolar brush cells, and molecular layer interneurons than the other serotypes, while AAV2 labeled a greater number of granule cells. These results provide guidelines for the use of AAVs as gene delivery tools in these regions.
Collapse
Affiliation(s)
| | - Timothy Balmer
- School of Life Sciences, Arizona State University, Tempe, Arizona 85287
| |
Collapse
|
2
|
Witteveen I, Balmer T. Comparative analysis of six adeno-associated viral vector serotypes in mouse inferior colliculus and cerebellum. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.17.618966. [PMID: 39484622 PMCID: PMC11526941 DOI: 10.1101/2024.10.17.618966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Adeno-associated viral vector (AAV) serotypes vary in how effectively they express genes across different cell types and brain regions. Here we report a systematic comparison of the AAV serotypes 1, 2, 5, 8, 9, and the directed evolution derived AAVrg, in the inferior colliculus and cerebellum. The AAVs were identical apart from their different serotypes, each having a synapsin promotor and expressing GFP (AAV-hSyn-GFP). Identical titers and volumes were injected into the inferior colliculus and cerebellum of adult male and female mice and brains were sectioned and imaged 2 weeks later. Transduction efficacy, anterograde labeling of axonal projections, and retrograde labeling of somata, were characterized and compared across serotypes. Cell-type tropism was assessed by analyzing the morphology of the GFP-labeled neurons in the cerebellar cortex. In both the cerebellum and inferior colliculus, AAV1 expressed GFP in more cells, labeled a larger volume, and produced significantly brighter labeling than all other serotypes, indicating superior transgene expression. AAV1 labeled more Purkinje cells, unipolar brush cells, and molecular layer interneurons than the other serotypes, while AAV2 labeled a greater number of granule cells. These results provide guidelines for the use of AAVs as gene delivery tools in these regions.
Collapse
Affiliation(s)
- Isabelle Witteveen
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| | - Timothy Balmer
- School of Life Sciences, Arizona State University, Tempe, AZ, 85287, USA
| |
Collapse
|
3
|
López-Canul M, He Q, Sasson T, Ettaoussi M, Gregorio DD, Ochoa-Sanchez R, Catoire H, Posa L, Rouleau G, Beaulieu JM, Comai S, Gobbi G. Selective Enhancement of REM Sleep in Male Rats through Activation of Melatonin MT 1 Receptors Located in the Locus Ceruleus Norepinephrine Neurons. J Neurosci 2024; 44:e0914232024. [PMID: 38744530 PMCID: PMC11255427 DOI: 10.1523/jneurosci.0914-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 04/03/2024] [Accepted: 04/06/2024] [Indexed: 05/16/2024] Open
Abstract
Sleep disorders affect millions of people around the world and have a high comorbidity with psychiatric disorders. While current hypnotics mostly increase non-rapid eye movement sleep (NREMS), drugs acting selectively on enhancing rapid eye movement sleep (REMS) are lacking. This polysomnographic study in male rats showed that the first-in-class selective melatonin MT1 receptor partial agonist UCM871 increases the duration of REMS without affecting that of NREMS. The REMS-promoting effects of UCM871 occurred by inhibiting, in a dose-response manner, the firing activity of the locus ceruleus (LC) norepinephrine (NE) neurons, which express MT1 receptors. The increase of REMS duration and the inhibition of LC-NE neuronal activity by UCM871 were abolished by MT1 pharmacological antagonism and by an adeno-associated viral (AAV) vector, which selectively knocked down MT1 receptors in the LC-NE neurons. In conclusion, MT1 receptor agonism inhibits LC-NE neurons and triggers REMS, thus representing a novel mechanism and target for REMS disorders and/or psychiatric disorders associated with REMS impairments.
Collapse
Affiliation(s)
- Martha López-Canul
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Qianzi He
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Tania Sasson
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Mohamed Ettaoussi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Rafael Ochoa-Sanchez
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Helene Catoire
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Luca Posa
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Guy Rouleau
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jean Martin Beaulieu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5G 2C8, Canada
| | - Stefano Comai
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- Research Institute, McGill University Health Center, McGill University, Montreal, Quebec H3A 1A1, Canada
| |
Collapse
|
4
|
Looser ZJ, Faik Z, Ravotto L, Zanker HS, Jung RB, Werner HB, Ruhwedel T, Möbius W, Bergles DE, Barros LF, Nave KA, Weber B, Saab AS. Oligodendrocyte-axon metabolic coupling is mediated by extracellular K + and maintains axonal health. Nat Neurosci 2024; 27:433-448. [PMID: 38267524 PMCID: PMC10917689 DOI: 10.1038/s41593-023-01558-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 12/13/2023] [Indexed: 01/26/2024]
Abstract
The integrity of myelinated axons relies on homeostatic support from oligodendrocytes (OLs). To determine how OLs detect axonal spiking and how rapid axon-OL metabolic coupling is regulated in the white matter, we studied activity-dependent calcium (Ca2+) and metabolite fluxes in the mouse optic nerve. We show that fast axonal spiking triggers Ca2+ signaling and glycolysis in OLs. OLs detect axonal activity through increases in extracellular potassium (K+) concentrations and activation of Kir4.1 channels, thereby regulating metabolite supply to axons. Both pharmacological inhibition and OL-specific inactivation of Kir4.1 reduce the activity-induced axonal lactate surge. Mice lacking oligodendroglial Kir4.1 exhibit lower resting lactate levels and altered glucose metabolism in axons. These early deficits in axonal energy metabolism are associated with late-onset axonopathy. Our findings reveal that OLs detect fast axonal spiking through K+ signaling, making acute metabolic coupling possible and adjusting the axon-OL metabolic unit to promote axonal health.
Collapse
Affiliation(s)
- Zoe J Looser
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Zainab Faik
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Luca Ravotto
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Henri S Zanker
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Ramona B Jung
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Hauke B Werner
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Torben Ruhwedel
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Wiebke Möbius
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Dwight E Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University, Baltimore, MD, USA
| | - L Felipe Barros
- Centro de Estudios Científicos (CECs), Valdivia, Chile
- Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia, Chile
| | - Klaus-Armin Nave
- Department of Neurogenetics, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Bruno Weber
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Aiman S Saab
- Institute of Pharmacology and Toxicology, University of Zurich, Zurich, Switzerland.
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
5
|
Mueller SA, Oler JA, Roseboom PH, Aggarwal N, Kenwood MM, Riedel MK, Elam VR, Olsen ME, DiFilippo AH, Christian BT, Hu X, Galvan A, Boehm MA, Michaelides M, Kalin NH. DREADD-mediated amygdala activation is sufficient to induce anxiety-like responses in young nonhuman primates. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100111. [PMID: 38020807 PMCID: PMC10663133 DOI: 10.1016/j.crneur.2023.100111] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Anxiety disorders are among the most prevalent psychiatric disorders, with symptoms often beginning early in life. To model the pathophysiology of human pathological anxiety, we utilized Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) in a nonhuman primate model of anxious temperament to selectively increase neuronal activity of the amygdala. Subjects included 10 young rhesus macaques; 5 received bilateral infusions of AAV5-hSyn-HA-hM3Dq into the dorsal amygdala, and 5 served as controls. Subjects underwent behavioral testing in the human intruder paradigm following clozapine or vehicle administration, prior to and following surgery. Behavioral results indicated that clozapine treatment post-surgery increased freezing across different threat-related contexts in hM3Dq subjects. This effect was again observed approximately 1.9 years following surgery, indicating the long-term functional capacity of DREADD-induced neuronal activation. [11C]deschloroclozapine PET imaging demonstrated amygdala hM3Dq-HA specific binding, and immunohistochemistry revealed that hM3Dq-HA expression was most prominent in basolateral nuclei. Electron microscopy confirmed expression was predominantly on neuronal membranes. Together, these data demonstrate that activation of primate amygdala neurons is sufficient to induce increased anxiety-related behaviors, which could serve as a model to investigate pathological anxiety in humans.
Collapse
Affiliation(s)
- Sascha A.L. Mueller
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Jonathan A. Oler
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Patrick H. Roseboom
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Nakul Aggarwal
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Margaux M. Kenwood
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Marissa K. Riedel
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Victoria R. Elam
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Miles E. Olsen
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Alexandra H. DiFilippo
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Bradley T. Christian
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Xing Hu
- Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Adriana Galvan
- Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Matthew A. Boehm
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ned H. Kalin
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| |
Collapse
|
6
|
Mueller SAL, Oler JA, Roseboom PH, Aggarwal N, Kenwood MM, Riedel MK, Elam VR, Olsen ME, DiFilippo AH, Christian BT, Hu X, Galvan A, Boehm MA, Michaelides M, Kalin NH. DREADD-mediated amygdala activation is sufficient to induce anxiety-like responses in young nonhuman primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543911. [PMID: 37333300 PMCID: PMC10274719 DOI: 10.1101/2023.06.06.543911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Anxiety disorders are among the most prevalent psychiatric disorders, with symptoms often beginning early in life. To model the pathophysiology of human pathological anxiety, we utilized Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) in a nonhuman primate model of anxious temperament to selectively increase neuronal activity of the amygdala. Subjects included 10 young rhesus macaques; 5 received bilateral infusions of AAV5-hSyn-HA-hM3Dq into the dorsal amygdala, and 5 served as controls. Subjects underwent behavioral testing in the human intruder paradigm following clozapine or vehicle administration, prior to and following surgery. Behavioral results indicated that clozapine treatment post-surgery increased freezing across different threat-related contexts in hM3Dq subjects. This effect was again observed approximately 1.9 years following surgery, indicating the long-term functional capacity of DREADD-induced neuronal activation. [11C]deschloroclozapine PET imaging demonstrated amygdala hM3Dq-HA specific binding, and immunohistochemistry revealed that hM3Dq-HA expression was most prominent in basolateral nuclei. Electron microscopy confirmed expression was predominantly on neuronal membranes. Together, these data demonstrate that activation of primate amygdala neurons is sufficient to induce increased anxiety-related behaviors, which could serve as a model to investigate pathological anxiety in humans.
Collapse
Affiliation(s)
- Sascha A L Mueller
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Jonathan A Oler
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Patrick H Roseboom
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Nakul Aggarwal
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Margaux M Kenwood
- Department of Psychiatry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Marissa K Riedel
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Victoria R Elam
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Miles E Olsen
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Alexandra H DiFilippo
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Bradley T Christian
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Xing Hu
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Adriana Galvan
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Matthew A Boehm
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ned H Kalin
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| |
Collapse
|
7
|
Eleftheriou A, Ravotto L, Wyss MT, Warnock G, Siebert A, Zaiss M, Weber B. Simultaneous dynamic glucose-enhanced (DGE) MRI and fiber photometry measurements of glucose in the healthy mouse brain. Neuroimage 2023; 265:119762. [PMID: 36427752 DOI: 10.1016/j.neuroimage.2022.119762] [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: 07/14/2022] [Revised: 10/27/2022] [Accepted: 11/21/2022] [Indexed: 11/25/2022] Open
Abstract
Glucose is the main energy source in the brain and its regulated uptake and utilization are important biomarkers of pathological brain function. Glucose Chemical Exchange Saturation Transfer (GlucoCEST) and its time-resolved version Dynamic Glucose-Enhanced MRI (DGE) are promising approaches to monitor glucose and detect tumors, since they are radioactivity-free, do not require 13C labeling and are is easily translatable to the clinics. The main principle of DGE is clear. However, what remains to be established is to which extent the signal reflects vascular, extracellular or intracellular glucose. To elucidate the compartmental contributions to the DGE signal, we coupled it with FRET-based fiber photometry of genetically encoded sensors, a technique that combines quantitative glucose readout with cellular specificity. The glucose sensor FLIIP was used with fiber photometry to measure astrocytic and neuronal glucose changes upon injection of D-glucose, 3OMG and L-glucose, in the anaesthetized murine brain. By correlating the kinetic profiles of the techniques, we demonstrate the presence of a vascular contribution to the signal, especially at early time points after injection. Furthermore, we show that, in the case of the commonly used contrast agent 3OMG, the DGE signal actually anticorrelates with the glucose concentration in neurons and astrocytes.
Collapse
Affiliation(s)
- Afroditi Eleftheriou
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Luca Ravotto
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Matthias T Wyss
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Geoffrey Warnock
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Anita Siebert
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland
| | - Moritz Zaiss
- Institute of Neuroradiology, University Hospital Erlangen, Friedrich-Alexander University Erlangen Nürnberg, Erlangen, Germany; High-field Magnetic Resonance Center, Max Planck Institute for Biological Cybernetics, Tübingen, Germany
| | - Bruno Weber
- University of Zurich, Institute of Pharmacology and Toxicology, Zurich, Switzerland; Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
8
|
Pietersz KL, Pouw S, Klima J, Ellederova Z, Bohuslavova B, Chrastina J, Liscak R, Urgosik D, Starek Z, Crha M, Lewis O, Wooley M, Johnson D, Brouwers CC, Evers M, Motlik J, Martens GJM, Konstantinova PS, Blits B. Transduction profiles in minipig following MRI guided delivery of AAV-5 into thalamic and corona radiata areas. J Neurosci Methods 2022; 365:109382. [PMID: 34637809 DOI: 10.1016/j.jneumeth.2021.109382] [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: 02/12/2021] [Revised: 10/03/2021] [Accepted: 10/05/2021] [Indexed: 11/17/2022]
Abstract
BACKGROUND As a step towards clinical use of AAV-mediated gene therapy, brains of large animals are used to settle delivery parameters as most brain connections, and relative sizes in large animals and primates, are reasonably common. Prior to application in the clinic, approaches that have shown to be successful in rodent models are tested in larger animal species, such as dogs, non-human primates, and in this case, minipigs. NEW METHOD We evaluated alternate delivery routes to target the basal ganglia by injections into the more superficial corona radiata, and, deeper into the brain, the thalamus. Anatomically known connections can be used to predict the expression of the transgene following infusion of AAV5. For optimal control over delivery of the vector with regards to anatomical location in the brain and spread in the tissue, we have used magnetic resonance image-guided convection-enhanced diffusion delivery. RESULTS While the transduction of the cortex was observed, only partial transduction of the basal ganglia was achieved via the corona radiata. Thalamic administration, on the other hand, resulted in widespread transduction from the midbrain to the frontal cortex COMPARISON WITH EXISTING METHODS: Compared to other methods, such as delivery directly to the striatum, thalamic injection may provide an alternative when for instance, injection into the basal ganglia directly is not feasible. CONCLUSIONS The study results suggest that thalamic administration of AAV5 has significant potential for indications where the transduction of specific areas of the brain is required.
Collapse
Affiliation(s)
- K L Pietersz
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands; Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - S Pouw
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands
| | - J Klima
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - Z Ellederova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - B Bohuslavova
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - J Chrastina
- Department of Neurosurgery, St. Anne's University Hospital, Brno, Czech Republic
| | - R Liscak
- Department of Stereotactic Radioneurosurgery, Na Homolce Hospital, Prague, Czech Republic
| | - D Urgosik
- Department of Stereotactic Radioneurosurgery, Na Homolce Hospital, Prague, Czech Republic
| | - Z Starek
- Interventional Cardiac Electrophysiology, St.' Anne's University Hospital, Brno, Czech Republic
| | - M Crha
- Small Animal Clinic, Veterinary and Pharmaceutical University, Brno, Czech Republic
| | - O Lewis
- Renishaw Neuro Solutions (RNS) ltd, Renishaw plc, Gloucestershire, UK
| | - M Wooley
- Renishaw Neuro Solutions (RNS) ltd, Renishaw plc, Gloucestershire, UK
| | - D Johnson
- Renishaw Neuro Solutions (RNS) ltd, Renishaw plc, Gloucestershire, UK
| | - C C Brouwers
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands
| | - M Evers
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands
| | - J Motlik
- Institute of Animal Physiology and Genetics, Libechov, Czech Republic
| | - G J M Martens
- Department of Molecular Animal Physiology, Donders Institute for Brain, Cognition and Behaviour, Centre for Neuroscience, Faculty of Science, Radboud University, Nijmegen, The Netherlands
| | - P S Konstantinova
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands
| | - B Blits
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, The Netherlands.
| |
Collapse
|
9
|
Adeno-Associated Vector-Delivered CRISPR/ SaCas9 System Reduces Feline Leukemia Virus Production In Vitro. Viruses 2021; 13:v13081636. [PMID: 34452500 PMCID: PMC8402633 DOI: 10.3390/v13081636] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2021] [Revised: 08/12/2021] [Accepted: 08/14/2021] [Indexed: 12/16/2022] Open
Abstract
Feline leukemia virus (FeLV) is a retrovirus of cats worldwide. High viral loads are associated with progressive infection and the death of the host, due to FeLV-associated disease. In contrast, low viral loads, an effective immune response, and a better clinical outcome can be observed in cats with regressive infection. We hypothesize that by lowering viral loads in progressively infected cats, using CRISPR/SaCas9-assisted gene therapy, the cat’s immune system may be permitted to direct the infection towards a regressive outcome. In a step towards this goal, the present study evaluates different adeno-associated vectors (AAVs) for their competence in delivering a gene editing system into feline cells, followed by investigations of the CRISPR/SaCas9 targeting efficiency for different sites within the FeLV provirus. Nine natural AAV serotypes, two AAV hybrid strains, and Anc80L65, an in silico predicted AAV ancestor, were tested for their potential to infect different feline cell lines and feline primary cells. AAV-DJ revealed superior infection efficiency and was thus employed in subsequent transduction experiments. The introduction of double-strand breaks, using the CRISPR/SaCas9 system targeting 12 selected FeLV provirus sites, was confirmed by T7 endonuclease 1 (T7E1), as well as Tracking of Indels by Decomposition (TIDE) analysis. The highest percentage (up to 80%) of nonhomologous end-joining (NHEJ) was found in the highly conserved gag and pol regions. Subsequent transduction experiments, using AAV-DJ, confirmed indel formation and showed a significant reduction in FeLV p27 antigen for some targets. The targeting of the FeLV provirus was efficient when using the CRISPR/SaCas9 approach in vitro. Whether the observed extent of provirus targeting will be sufficient to provide progressively FeLV-infected cats with the means to overcome the infection needs to be further investigated in vivo.
Collapse
|
10
|
Wang J, Zhang L. Retrograde Axonal Transport Property of Adeno-Associated Virus and Its Possible Application in Future. Microbes Infect 2021; 23:104829. [PMID: 33878458 DOI: 10.1016/j.micinf.2021.104829] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 03/30/2021] [Accepted: 04/05/2021] [Indexed: 12/19/2022]
Abstract
Gene therapy has become a treatment method for many diseases. Adeno-associated virus (AAV) is one of the most common virus vectors, is also widely used in the gene therapy field. During the past 2 decades, the retrograde axonal transportability of AAV has been discovered and utilized. Many studies have worked on the retrograde axonal transportability of AAV, and more and more people are interested in this field. This review described the current application, influence factors, and mechanism of retrograde axonal transportability of AAV and predicted its potential use in disease treatment in near future.
Collapse
Affiliation(s)
- Jingjing Wang
- Department of Gastroenterology, The Third Central Hospital of Tianjin, 83 Jintang Road, Hedong District, Tianjin, 300170, China
| | - Liqin Zhang
- Department of Otolaryngology, Peking Union Medical College Hospital, Dongcheng Qu, Beijing, 100730, China.
| |
Collapse
|
11
|
Petrucci AN, Joyal KG, Chou JW, Li R, Vencer KM, Buchanan GF. Post-ictal Generalized EEG Suppression is reduced by Enhancing Dorsal Raphe Serotonergic Neurotransmission. Neuroscience 2020; 453:206-221. [PMID: 33242541 DOI: 10.1016/j.neuroscience.2020.11.029] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 11/14/2020] [Accepted: 11/16/2020] [Indexed: 01/02/2023]
Abstract
Sudden unexpected death in epilepsy (SUDEP) is the leading cause of death in patients with refractory epilepsy. A proposed risk marker for SUDEP is the duration of post-ictal generalized EEG suppression (PGES). The mechanisms underlying PGES are unknown. Serotonin (5-HT) has been implicated in SUDEP pathophysiology. Seizures suppress activity of 5-HT neurons in the dorsal raphe nucleus (DRN). We hypothesized that suppression of DRN 5-HT neuron activity contributes to PGES and increasing 5-HT neurotransmission or stimulating the DRN before a seizure would decrease PGES duration. Adult C57BL/6J and Pet1-Cre mice received EEG/EMG electrodes, a bipolar stimulating/recording electrode in the right basolateral amygdala, and either a microdialysis guide cannula or an injection of adeno-associated virus (AAV) allowing expression of channelrhodopsin2 plus an optic fiber into the DRN. Systemic application of the selective 5-HT reuptake inhibitor citalopram (20 mg/kg) decreased PGES duration from seizures induced during wake (n = 23) and non-rapid eye movement (NREM) sleep (n = 13) whereas fluoxetine (10 mg/kg) pretreatment decreased PGES duration following seizures induced from wake (n = 11), but not NREM sleep (n = 9). Focal chemical (n = 6) or optogenetic (n = 8) stimulation of the DRN reduced PGES duration following seizures in kindled mice induced during wake. During PGES, animals exhibited immobility and suppression of EEG activity that was reduced by citalopram pretreatment. These results suggest 5-HT and the DRN may regulate PGES.
Collapse
Affiliation(s)
- Alexandra N Petrucci
- Interdisciplinary Graduate Program in Neuroscience, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States; Department of Neurology, Carver College of Medicine, Carver College of Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States.
| | - Katelyn G Joyal
- Interdisciplinary Graduate Program in Neuroscience, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States; Department of Neurology, Carver College of Medicine, Carver College of Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States.
| | - Jonathan W Chou
- Department of Neurology, Carver College of Medicine, Carver College of Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States; Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, United States.
| | - Rui Li
- Department of Neurology, Carver College of Medicine, Carver College of Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States.
| | - Kimberly M Vencer
- Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States; Department of Health and Human Physiology, College of Liberal Arts and Sciences, University of Iowa, Iowa City, IA 52242, United States
| | - Gordon F Buchanan
- Interdisciplinary Graduate Program in Neuroscience, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States; Department of Neurology, Carver College of Medicine, Carver College of Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States; Iowa Neuroscience Institute, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, United States.
| |
Collapse
|
12
|
Düring DN, Dittrich F, Rocha MD, Tachibana RO, Mori C, Okanoya K, Boehringer R, Ehret B, Grewe BF, Gerber S, Ma S, Rauch M, Paterna JC, Kasper R, Gahr M, Hahnloser RHR. Fast Retrograde Access to Projection Neuron Circuits Underlying Vocal Learning in Songbirds. Cell Rep 2020; 33:108364. [PMID: 33176132 PMCID: PMC8236207 DOI: 10.1016/j.celrep.2020.108364] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Revised: 07/29/2020] [Accepted: 10/19/2020] [Indexed: 02/06/2023] Open
Abstract
Understanding the structure and function of neural circuits underlying speech and language is a vital step toward better treatments for diseases of these systems. Songbirds, among the few animal orders that share with humans the ability to learn vocalizations from a conspecific, have provided many insights into the neural mechanisms of vocal development. However, research into vocal learning circuits has been hindered by a lack of tools for rapid genetic targeting of specific neuron populations to meet the quick pace of developmental learning. Here, we present a viral tool that enables fast and efficient retrograde access to projection neuron populations. In zebra finches, Bengalese finches, canaries, and mice, we demonstrate fast retrograde labeling of cortical or dopaminergic neurons. We further demonstrate the suitability of our construct for detailed morphological analysis, for in vivo imaging of calcium activity, and for multi-color brainbow labeling. Düring et al. describe a fast and efficient viral vector to dissect structure and function of neural circuits underlying learned vocalizations in songbirds. The AAV variant provides retrograde access to projection neuron circuits, including dopaminergic pathways in songbirds and additionally in mice, and allows for retrograde calcium imaging and multispectral brainbow labeling.
Collapse
Affiliation(s)
- Daniel N Düring
- Institute of Neuroinformatics, University of Zurich/ETH Zurich, Zurich, Switzerland; Neuroscience Center Zurich, Zurich, Switzerland; Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany.
| | - Falk Dittrich
- Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| | - Mariana D Rocha
- Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| | | | - Chihiro Mori
- Department of Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuo Okanoya
- Department of Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Roman Boehringer
- Institute of Neuroinformatics, University of Zurich/ETH Zurich, Zurich, Switzerland; Neuroscience Center Zurich, Zurich, Switzerland
| | - Benjamin Ehret
- Institute of Neuroinformatics, University of Zurich/ETH Zurich, Zurich, Switzerland; Neuroscience Center Zurich, Zurich, Switzerland
| | - Benjamin F Grewe
- Institute of Neuroinformatics, University of Zurich/ETH Zurich, Zurich, Switzerland; Neuroscience Center Zurich, Zurich, Switzerland
| | - Stefan Gerber
- Institute of Neuroinformatics, University of Zurich/ETH Zurich, Zurich, Switzerland
| | - Shouwen Ma
- Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| | - Melanie Rauch
- Viral Vector Facility, Neuroscience Center Zurich, Zurich, Switzerland
| | | | - Robert Kasper
- Imaging Facility at the Max Planck Institute of Neurobiology, Munich, Germany
| | - Manfred Gahr
- Department of Behavioural Neurobiology, Max Planck Institute for Ornithology, Seewiesen, Germany
| | - Richard H R Hahnloser
- Institute of Neuroinformatics, University of Zurich/ETH Zurich, Zurich, Switzerland; Neuroscience Center Zurich, Zurich, Switzerland
| |
Collapse
|
13
|
Pietersz KL, Martier RM, Baatje MS, Liefhebber JM, Brouwers CC, Pouw SM, Fokkert L, Lubelski J, Petry H, Martens GJM, van Deventer SJ, Konstantinova P, Blits B. Transduction patterns in the CNS following various routes of AAV-5-mediated gene delivery. Gene Ther 2020; 28:435-446. [PMID: 32801344 DOI: 10.1038/s41434-020-0178-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/12/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023]
Abstract
Various administration routes of adeno-associated virus (AAV)-based gene therapy have been examined to target the central nervous system to answer the question what the most optimal delivery route is for treatment of the brain with certain indications. In this study, we evaluated AAV5 vector system for its capability to target the central nervous system via intrastriatal, intrathalamic or intracerebroventricular delivery routes in rats. AAV5 is an ideal candidate for gene therapy because of its relatively low level of existing neutralizing antibodies compared to other serotypes, and its broad tissue and cell tropism. Intrastriatal administration of AAV5-GFP resulted in centralized localized vector distribution and expression in the frontal part of the brain. Intrathalamic injection showed transduction and gradient expression from the rostral brain into lumbar spinal cord, while intracerebroventricular administration led to a more evenly, albeit relatively superficially distributed, transduction and expression throughout the central nervous system. To visualize the differences between localized and intra-cerebral spinal fluid administration routes, we compared intrastriatal to intracerebroventricular and intrathecal administration of AAV5-GFP. Together, our results demonstrate that for efficient transgene expression, various administration routes can be applied.
Collapse
|
14
|
Sevoflurane anesthesia-mediated oxidative stress and cognitive impairment in hippocampal neurons of old rats can be ameliorated by expression of brain derived neurotrophic factor. Neurosci Lett 2020; 721:134785. [PMID: 32027953 DOI: 10.1016/j.neulet.2020.134785] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Accepted: 01/23/2020] [Indexed: 01/19/2023]
Abstract
Postoperative cognitive dysfunction in elderly patients has been related to neurodegenerative disorders and mortality. Sevoflurane anesthesia has been implicated in both postoperative cognitive dysfunction and neurotoxicity. Given the advantages of using inhaled anesthetics like sevoflurane, it is important to understand how their usage results in neurotoxicity and subsequently devise ways to circumvent or attenuate the anesthetic-mediated induction in neurotoxicity. We have used an aged rat model to investigate the molecular mechanisms by which sevoflurane inhalation results in neurotoxicity and whether modulation of these molecular mechanisms can inhibit or attenuate neurotoxicity and cognitive learning and memory impairment in these animals. Low- or high-dose of sevoflurane resulted in reactive oxygen species generation, increased NADPH oxidase protein expression, apoptosis and autophagy. Sevoflurane inhalation resulted in significant inhibition of brain derived neurotrophic factor (BDNF) and cognitive impairment. And the activation of PI3K/Akt/mTOR signaling pathways are attenuated in sevoflurane-mediated anesthesia. Adeno-associated virus (AAV)-mediated expression of Bdnf, but not controls EGFP, attenuated sevoflurane-induced oxidative stress and cognitive impairment in the rats. Our results highlight that AAV-mediated gene therapy might offer a potential therapeutic opportunity to treat post-operative cognitive impairment resulting from inhaled anesthetics.
Collapse
|
15
|
Dhanushkodi A, Xue Y, Roguski EE, Ding Y, Matta SG, Heck D, Fan GH, McDonald MP. Lentiviral-mediated knock-down of GD3 synthase protects against MPTP-induced motor deficits and neurodegeneration. Neurosci Lett 2018; 692:53-63. [PMID: 30391320 DOI: 10.1016/j.neulet.2018.10.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 10/05/2018] [Accepted: 10/16/2018] [Indexed: 11/16/2022]
Abstract
Converging evidence demonstrates an important role for gangliosides in brain function and neurodegenerative diseases. Exogenous GM1 is broadly neuroprotective, including in rodent, feline, and primate models of Parkinson's disease, and has shown positive effects in clinical trials. We and others have shown that inhibition of the ganglioside biosynthetic enzyme GD3 synthase (GD3S) increases endogenous levels GM1 ganglioside. We recently reported that targeted deletion of St8sia1, the gene that codes for GD3S, prevents motor impairments and significantly attenuates neurodegeneration induced by 1-methy-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP). The current study investigated the effects of GD3S inhibition on the neurotoxicity and parkinsonism induced by MPTP. Mice were injected intrastriatally with a lentiviral-vector-mediated shRNA construct targeting GD3S (shGD3S) or a scrambled-sequence control (scrRNA). An MPTP regimen of 18 mg/kg x 5 days reduced tyrosine-hydroxylase-positive neurons in the substantia nigra pars compacta of scrRNA-treated mice by nearly two-thirds. In mice treated with shGD3S the MPTP-induced lesion was approximately half that size. MPTP induced bradykinesia and deficits in fine motor skills in mice treated with scrRNA. These deficits were absent in shGD3S-treated mice. These results suggest that inhibition of GD3S protects against the nigrostriatal damage, bradykinesia, and fine-motor-skill deficits associated with MPTP administration.
Collapse
Affiliation(s)
- Anandh Dhanushkodi
- Department of Neurology, University of Tennessee Health Science Center Memphis, TN 38163, United States
| | - Yi Xue
- Department of Neurology, University of Tennessee Health Science Center Memphis, TN 38163, United States
| | - Emily E Roguski
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Yun Ding
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Shannon G Matta
- Department of Pharmacology, University of Tennessee Health Science Center, Memphis, TN 38163, United States
| | - Detlef Heck
- Department of Anatomy & Neurobiology, University of Tennessee Health Science Center Memphis, TN 38163, United States
| | - Guo-Huang Fan
- Department of Pharmacology & Toxicology, Virginia Commonwealth University, Richmond, VA 23284, United States
| | - Michael P McDonald
- Department of Neurology, University of Tennessee Health Science Center Memphis, TN 38163, United States; Department of Anatomy & Neurobiology, University of Tennessee Health Science Center Memphis, TN 38163, United States.
| |
Collapse
|
16
|
Looser ZJ, Barrett MJP, Hirrlinger J, Weber B, Saab AS. Intravitreal AAV-Delivery of Genetically Encoded Sensors Enabling Simultaneous Two-Photon Imaging and Electrophysiology of Optic Nerve Axons. Front Cell Neurosci 2018; 12:377. [PMID: 30405358 PMCID: PMC6205974 DOI: 10.3389/fncel.2018.00377] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 10/03/2018] [Indexed: 11/21/2022] Open
Abstract
Myelination of axons by oligodendrocytes is a key feature of the remarkably fast operating CNS. Oligodendrocytes not only tune axonal conduction speed but are also suggested to maintain long-term axonal integrity by providing metabolic support to the axons they ensheath. However, how myelinating oligodendrocytes impact axonal energy homeostasis remains poorly understood and difficult to investigate. Here, we provide a method of how to study electrically active myelinated axons expressing genetically encoded sensors by combining electrophysiology and two-photon imaging of acutely isolated optic nerves. We show that intravitreal adeno-associated viral (AAV) vector delivery is an efficient tool to achieve functional sensor expression in optic nerve axons, which is demonstrated by measuring axonal ATP dynamics following AAV-mediated sensor expression. This novel approach allows for fast expression of any optical sensor of interest to be studied in optic nerve axons without the need to go through the laborious process of producing new transgenic mouse lines. Viral-mediated biosensor expression in myelinated axons and the subsequent combination of nerve recordings and sensor imaging outlines a powerful method to investigate oligodendroglial support functions and to further interrogate cellular mechanisms governing axonal energy homeostasis under physiological and pathological conditions.
Collapse
Affiliation(s)
- Zoe J. Looser
- Institute of Pharmacology & Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Matthew J. P. Barrett
- Institute of Pharmacology & Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Johannes Hirrlinger
- Carl-Ludwig-Institute for Physiology, University of Leipzig, Leipzig, Germany
- Department of Neurogenetics, Max-Planck-Institute of Experimental Medicine, Göttingen, Germany
| | - Bruno Weber
- Institute of Pharmacology & Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| | - Aiman S. Saab
- Institute of Pharmacology & Toxicology, University of Zurich, Zurich, Switzerland
- Neuroscience Center Zurich, University and ETH Zurich, Zurich, Switzerland
| |
Collapse
|
17
|
Mendez-Gomez HR, Singh J, Meyers C, Chen W, Gorbatyuk OS, Muzyczka N. The Lipase Activity of Phospholipase D2 is Responsible for Nigral Neurodegeneration in a Rat Model of Parkinson's Disease. Neuroscience 2018. [PMID: 29526688 DOI: 10.1016/j.neuroscience.2018.02.047] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Phospholipase D2 (PLD2), an enzyme involved in vesicle trafficking and membrane signaling, interacts with α-synuclein, a protein known to contribute in the development of Parkinson disease (PD). We previously reported that PLD2 overexpression in rat substantia nigra pars compacta (SNc) causes a rapid neurodegeneration of dopamine neurons, and that α-synuclein suppresses PLD2-induced nigral degeneration (Gorbatyuk et al., 2010). Here, we report that PLD2 toxicity is due to its lipase activity. Overexpression of a catalytically inactive mutant (K758R) of PLD2 prevents the loss of dopaminergic neurons in the SNc and does not show signs of toxicity after 10 weeks of overexpression. Further, mutant K758R does not affect dopamine levels in the striatum. In contrast, mutants that prevent PLD2 interaction with dynamin or growth factor receptor bound protein 2 (Grb2) but retained lipase activity, continued to show rapid neurodegeneration. These findings suggest that neither the interaction of PLD2 with dynamin, which has a role in vesicle trafficking, nor the PLD2 interaction with Grb2, which has multiple roles in cell cycle control, chemotaxis and activation of tyrosine kinase complexes, are the primary cause of neurodegeneration. Instead, the synthesis of phosphatidic acid (the product of PLD2), which is a second messenger in multiple cellular pathways, appears to be the key to PLD2 induced neurodegeneration. The fact that α-synuclein is a regulator of PLD2 activity suggests that regulation of PLD2 activity could be important in the progression of PD.
Collapse
Affiliation(s)
- Hector R Mendez-Gomez
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA; UF Genetics Institute and Powell Gene Therapy Center, USA.
| | - Jasbir Singh
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA; UF Genetics Institute and Powell Gene Therapy Center, USA
| | - Craig Meyers
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA; UF Genetics Institute and Powell Gene Therapy Center, USA
| | - Weijun Chen
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA; UF Genetics Institute and Powell Gene Therapy Center, USA
| | - Oleg S Gorbatyuk
- Department of Vision Sciences, Center for Neurodegeneration and Experimental Therapy, University of Alabama at Birmingham, AL, USA
| | - Nicholas Muzyczka
- Department of Molecular Genetics and Microbiology, University of Florida College of Medicine, Gainesville, FL, USA; UF Genetics Institute and Powell Gene Therapy Center, USA
| |
Collapse
|
18
|
Dastor M, Schreiber J, Prochazka L, Angelici B, Kleinert J, Klebba I, Doshi J, Shen L, Benenson Y. A Workflow for In Vivo Evaluation of Candidate Inputs and Outputs for Cell Classifier Gene Circuits. ACS Synth Biol 2018; 7:474-489. [PMID: 29257672 DOI: 10.1021/acssynbio.7b00303] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Cell classifier gene circuits that integrate multiple molecular inputs to restrict the expression of therapeutic outputs to cancer cells have the potential to result in efficacious and safe cancer therapies. Preclinical translation of the hitherto developments requires creating the conditions where the animal model, the delivery platform, in vivo expression levels of the inputs, and the efficacy of the output, all come together to enable detailed evaluation of the fully assembled circuits. Here we show an integrated workflow that addresses these issues and builds the framework for preclinical classifier studies using the design framework of microRNA (miRNA, miR)-based classifier gene circuits. Specifically, we employ HCT-116 colorectal cancer cell xenograft in an experimental mouse metastatic liver tumor model together with Adeno-associated virus (AAV) vector delivery platform. Novel engineered AAV-based constructs are used to validate in vivo the candidate inputs miR-122 and miR-7 and, separately, the cytotoxic output HSV-TK/ganciclovir. We show that while the data are largely consistent with expectations, crucial insights are gained that could not have been obtained in vitro. The results highlight the importance of detailed stepwise interrogation of the experimental parameters as a necessary step toward clinical translation of synthetic gene circuits.
Collapse
Affiliation(s)
- Margaux Dastor
- Department of Biosystems
Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Joerg Schreiber
- Department of Biosystems
Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Laura Prochazka
- Department of Biosystems
Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Bartolomeo Angelici
- Department of Biosystems
Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Jonathan Kleinert
- Department of Biosystems
Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Ina Klebba
- Department of Biosystems
Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Jiten Doshi
- Department of Biosystems
Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Linling Shen
- Department of Biosystems
Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| | - Yaakov Benenson
- Department of Biosystems
Science and Engineering, ETH Zurich, Mattenstrasse 26, 4058 Basel, Switzerland
| |
Collapse
|
19
|
Yao X, Liu X, Zhang Y, Li Y, Zhao C, Yao S, Wei Y. Gene Therapy of Adult Neuronal Ceroid Lipofuscinoses with CRISPR/Cas9 in Zebrafish. Hum Gene Ther 2017; 28:588-597. [PMID: 28478735 DOI: 10.1089/hum.2016.190] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Adult-onset neuronal ceroid lipofuscinosis (ANCL), one of the neuronal ceroid lipofuscinosis (NCLs), is an inherited neurodegenerative disorder with progressive neuronal dysfunction. Recently, mutations in the DNAJC5 gene that encodes cysteine-string protein alpha (CSPα) have been reported to be associated with familial autosomal-dominant ANCL (AD-ANCL). This study constructed an ANCL transgenic zebrafish model expressing the human mutant DNAJC5 (mDNAJC5) gene under the control of a zebrafish neuron-specific promoter. To investigate whether gene therapy based on genome-editing technology could treat ANCL, a panel of TALEN and Cas9 nucleases was designed to disrupt the mDNAJC5 gene in this transgenic animal model. By screening these nucleases, it was found that one nuclease that targeted the 5' coding region efficiently alleviated mDNAJC5 protein aggregates in the affected neurons. Therefore, this study provides a gene therapy strategy via the use of the CRISPR/Cas9 system to treat neural genetic diseases.
Collapse
Affiliation(s)
- Xiaomin Yao
- 1 State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Xiaowei Liu
- 1 State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yaguang Zhang
- 1 State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yuhao Li
- 2 Key Laboratory of Tumor Microenvironment and Neurovascular Regulation, Nankai University School of Medicine , Tianjin, People's Republic of China
| | - Chenjian Zhao
- 1 State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Shaohua Yao
- 1 State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| | - Yuquan Wei
- 1 State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, People's Republic of China
| |
Collapse
|
20
|
Klaus F, Paterna JC, Marzorati E, Sigrist H, Götze L, Schwendener S, Bergamini G, Jehli E, Azzinnari D, Fuertig R, Fontana A, Seifritz E, Pryce CR. Differential effects of peripheral and brain tumor necrosis factor on inflammation, sickness, emotional behavior and memory in mice. Brain Behav Immun 2016; 58:310-326. [PMID: 27515532 DOI: 10.1016/j.bbi.2016.08.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2016] [Revised: 07/22/2016] [Accepted: 08/01/2016] [Indexed: 12/21/2022] Open
Abstract
Tumor necrosis factor alpha (TNF) is increased in depression and clinical-trial evidence indicates that blocking peripheral TNF has some antidepressant efficacy. In rodents, peripheral or intracerebroventricular TNF results in sickness e.g. reduced body weight, altered emotional behavior and impaired memory. However, the underlying pathways and responsible brain regions are poorly understood. The aim of this mouse study was to increase understanding by comparing the effects of sustained increases in TNF in the circulation, in brain regions impacted by increased circulating TNF, or specific brain regions. Increased peripheral TNF achieved by repeated daily injection (IP-TNF) or osmotic pump resulted in decreased body weight, decreased saccharin (reward) consumption, and increased memory of an aversive conditioned stimulus. These effects co-occurred with increased plasma interleukin-6 and increased IP-derived TNF in brain peri-ventricular regions. An adenovirus-associated viral TNF vector (AAV-TNF) was constructed, brain injection of which resulted in dose-dependent, sustained and region-specific TNF expression, and was without effect on blood cytokine levels. Lateral ventricle AAV-TNF yielded increased TNF in the same brain regions as IP-TNF. In contrast to IP-TNF it was without effect on body weight, saccharin consumption and fear memory, although it did increase anxiety. Hippocampal AAV-TNF led to decreased body weight. It increased conditioning to but not subsequent memory of an aversive context, suggesting impaired consolidation; it also increased anxiety. Amygdala AAV-TNF was without effect on body weight and aversive stimulus learning-memory, but reduced saccharin consumption and increased anxiety. This study adds significantly to the evidence that both peripheral and brain region-specific increases in TNF lead to both sickness and depression- and anxiety disorder-relevant behavior and do so via different pathways. It thereby highlights the complexity in terms of indirect and direct pathways via which increased TNF can act and which need to be taken into account when considering it as a therapeutic target.
Collapse
Affiliation(s)
- Federica Klaus
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland; Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Switzerland
| | - Jean-Charles Paterna
- Viral Vector Facility, Neuroscience Center Zurich, University of Zurich and ETH Zurich, Switzerland
| | - Elisa Marzorati
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland
| | - Hannes Sigrist
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland
| | - Lea Götze
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland
| | | | - Giorgio Bergamini
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Switzerland
| | - Elisabeth Jehli
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland
| | - Damiano Azzinnari
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Switzerland
| | - René Fuertig
- CNS Diseases Research Germany, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Adriano Fontana
- Institute of Experimental Immunology, Inflammation and Sickness Behaviour, University of Zurich, Switzerland
| | - Erich Seifritz
- Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Switzerland
| | - Christopher R Pryce
- Preclinical Laboratory for Translational Research into Affective Disorders, Department of Psychiatry, Psychotherapy and Psychosomatics, Psychiatric Hospital, University of Zurich, Switzerland; Neuroscience Center Zurich, University of Zurich and ETH Zurich, Switzerland.
| |
Collapse
|
21
|
Kosillo P, Zhang YF, Threlfell S, Cragg SJ. Cortical Control of Striatal Dopamine Transmission via Striatal Cholinergic Interneurons. Cereb Cortex 2016; 26:4160-4169. [PMID: 27566978 PMCID: PMC5066833 DOI: 10.1093/cercor/bhw252] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2016] [Revised: 07/01/2016] [Accepted: 07/19/2016] [Indexed: 12/25/2022] Open
Abstract
Corticostriatal regulation of striatal dopamine (DA) transmission has long been postulated, but ionotropic glutamate receptors have not been localized directly to DA axons. Striatal cholinergic interneurons (ChIs) are emerging as major players in striatal function, and can govern DA transmission by activating nicotinic receptors (nAChRs) on DA axons. Cortical inputs to ChIs have historically been perceived as sparse, but recent evidence indicates that they strongly activate ChIs. We explored whether activation of M1/M2 corticostriatal inputs can consequently gate DA transmission, via ChIs. We reveal that optogenetic activation of channelrhodopsin-expressing corticostriatal axons can drive striatal DA release detected with fast-scan cyclic voltammetry and requires activation of nAChRs on DA axons and AMPA receptors on ChIs that promote short-latency action potentials. By contrast, DA release driven by optogenetic activation of intralaminar thalamostriatal inputs involves additional activation of NMDA receptors on ChIs and action potential generation over longer timescales. Therefore, cortical and thalamic glutamate inputs can modulate DA transmission by regulating ChIs as gatekeepers, through ionotropic glutamate receptors. The different use of AMPA and NMDA receptors by cortical versus thalamic inputs might lead to distinct input integration strategies by ChIs and distinct modulation of the function of DA and striatum.
Collapse
Affiliation(s)
- Polina Kosillo
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK.,Current address: Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | - Yan-Feng Zhang
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK
| | - Sarah Threlfell
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK.,Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3QX, UK
| | - Stephanie J Cragg
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, OX1 3PT, UK.,Oxford Parkinson's Disease Centre, University of Oxford, Oxford, OX1 3QX, UK
| |
Collapse
|
22
|
Gerits A, Vancraeyenest P, Vreysen S, Laramée ME, Michiels A, Gijsbers R, Van den Haute C, Moons L, Debyser Z, Baekelandt V, Arckens L, Vanduffel W. Serotype-dependent transduction efficiencies of recombinant adeno-associated viral vectors in monkey neocortex. NEUROPHOTONICS 2015; 2:031209. [PMID: 26839901 PMCID: PMC4729112 DOI: 10.1117/1.nph.2.3.031209] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Accepted: 08/25/2015] [Indexed: 06/05/2023]
Abstract
Viral vector-mediated expression of genes (e.g., coding for opsins and designer receptors) has grown increasingly popular. Cell-type specific expression is achieved by altering viral vector tropism through crosspackaging or by cell-specific promoters driving gene expression. Detailed information about transduction properties of most recombinant adeno-associated viral vector (rAAV) serotypes in macaque cortex is gradually becoming available. Here, we compare transduction efficiencies and expression patterns of reporter genes in two macaque neocortical areas employing different rAAV serotypes and promoters. A short version of the calmodulin-kinase-II (CaMKIIα0.4) promoter resulted in reporter gene expression in cortical neurons for all tested rAAVs, albeit with different efficiencies for spread: rAAV2/5>>rAAV2/7>rAAV2/8>rAAV2/9>>rAAV2/1 and proportion of transduced cells: rAAV2/1>rAAV2/5>rAAV2/7=rAAV2/9>rAAV2/8. In contrast to rodent studies, the cytomegalovirus (CMV) promoter appeared least efficient in macaque cortex. The human synapsin-1 promoter preceded by the CMV enhancer (enhSyn1) produced homogeneous reporter gene expression across all layers, while two variants of the CaMKIIα promoter resulted in different laminar transduction patterns and cell specificities. Finally, differences in expression patterns were observed when the same viral vector was injected in two neocortical areas. Our results corroborate previous findings that reporter-gene expression patterns and efficiency of rAAV transduction depend on serotype, promoter, cortical layer, and area.
Collapse
Affiliation(s)
- Annelies Gerits
- KU Leuven, Laboratory of Neuro- and Psychophysiology, Department of Neurosciences, O&N2 Herestraat 49 bus 10.21, 3000 Leuven, Belgium
| | - Pascaline Vancraeyenest
- KU Leuven, Laboratory of Neuro- and Psychophysiology, Department of Neurosciences, O&N2 Herestraat 49 bus 10.21, 3000 Leuven, Belgium
| | - Samme Vreysen
- KU Leuven, Laboratory of Neuroplasticity and Neuroproteomics, Faculty of Science, Naamsestraat 59, 3000 Leuven, Belgium
| | - Marie-Eve Laramée
- KU Leuven, Laboratory of Neuroplasticity and Neuroproteomics, Faculty of Science, Naamsestraat 59, 3000 Leuven, Belgium
| | - Annelies Michiels
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Kapucijnenvoer 33, VCTB +5, 3000 Leuven, Belgium
- KU Leuven, Leuven Viral Vector Core, Kapucijnenvoer 33, VCTB +5, 3000 Leuven, Belgium
| | - Rik Gijsbers
- KU Leuven, Leuven Viral Vector Core, Kapucijnenvoer 33, VCTB +5, 3000 Leuven, Belgium
- KU Leuven, Laboratory of Molecular Virology and Gene Therapy, Department of Neurosciences, Kapucijnenvoer 33, VCTB +5, 3000 Leuven, Flanders, Belgium
| | - Chris Van den Haute
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Kapucijnenvoer 33, VCTB +5, 3000 Leuven, Belgium
- KU Leuven, Leuven Viral Vector Core, Kapucijnenvoer 33, VCTB +5, 3000 Leuven, Belgium
| | - Lieve Moons
- KU Leuven, Laboratory of Neural Circuit Development and Regeneration, Faculty of Science, Naamsestraat 61, 3000 Leuven, Belgium
| | - Zeger Debyser
- KU Leuven, Laboratory of Molecular Virology and Gene Therapy, Department of Neurosciences, Kapucijnenvoer 33, VCTB +5, 3000 Leuven, Flanders, Belgium
| | - Veerle Baekelandt
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Kapucijnenvoer 33, VCTB +5, 3000 Leuven, Belgium
| | - Lutgarde Arckens
- KU Leuven, Laboratory of Neuroplasticity and Neuroproteomics, Faculty of Science, Naamsestraat 59, 3000 Leuven, Belgium
| | - Wim Vanduffel
- KU Leuven, Laboratory of Neuro- and Psychophysiology, Department of Neurosciences, O&N2 Herestraat 49 bus 10.21, 3000 Leuven, Belgium
- Massachusetts General Hospital, Athinoula A. Martinos Center for Biomedical Imaging, 149 13th street, Charlestown, Massachusetts 02129, United States
- Harvard Medical School, Department of Radiology, 149 13th street, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
23
|
Scheyltjens I, Laramée ME, Van den Haute C, Gijsbers R, Debyser Z, Baekelandt V, Vreysen S, Arckens L. Evaluation of the expression pattern of rAAV2/1, 2/5, 2/7, 2/8, and 2/9 serotypes with different promoters in the mouse visual cortex. J Comp Neurol 2015; 523:2019-42. [PMID: 26012540 DOI: 10.1002/cne.23819] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 12/18/2014] [Accepted: 05/21/2015] [Indexed: 12/12/2022]
Abstract
This study compared the expression pattern, laminar distribution, and cell specificity of several rAAV serotypes (2/1, 2/5, 2/7, 2/8, and 2/9) injected in the primary visual cortex (V1) of adult C57Bl/6J mice. In order to obtain specific expression in certain neuron subtypes, different promoter sequences were evaluated for excitatory cell specificity: a universal cytomegalovirus (CMV) promoter, and two versions of the excitatory neuron-specific Ca(2+) /calmodulin-dependent kinase subunit α (CaMKIIα) promoter, CaMKIIα 0.4 and CaMKIIα 1.3. The spatial distribution as well as the cell type specificity was immunohistochemically verified. Depending on the rAAV serotype used, the transduced volume expressing reporter protein differed substantially (rAAV2/5 ≫ 2/7 ≈ 2/9 ≈ 2/8 ≫ 2/1). Excitatory neuron-specific targeting was promoter-dependent, with a surprising difference between the 1.3 kb and 0.4 kb CaMKIIα promoters. While CaMKIIα 1.3 and CMV carrying vectors were comparable, with 78% of the transduced neurons being excitatory for CMV and 82% for CaMKIIα 1.3, the shorter CaMKIIα 0.4 version resulted in 95% excitatory specificity. This study therefore puts forward the CaMKIIα 0.4 promoter as the best choice to target excitatory neurons with rAAVs. Together, these results can be used as an aid to select the most optimal vector system to deliver transgenes into specific rodent neocortical circuits, allowing further elucidation of their functions.
Collapse
Affiliation(s)
- Isabelle Scheyltjens
- KU Leuven, Laboratory of Neuroplasticity and Neuroproteomics, Department of Biology, Leuven, Belgium
| | - Marie-Eve Laramée
- KU Leuven, Laboratory of Neuroplasticity and Neuroproteomics, Department of Biology, Leuven, Belgium
| | - Chris Van den Haute
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven, Belgium.,KU Leuven, Leuven Viral Vector Core, Leuven, Belgium
| | - Rik Gijsbers
- KU Leuven, Leuven Viral Vector Core, Leuven, Belgium.,KU Leuven, Laboratory for Molecular Virology and Gene Therapy, Leuven, Belgium
| | - Zeger Debyser
- KU Leuven, Laboratory for Molecular Virology and Gene Therapy, Leuven, Belgium
| | - Veerle Baekelandt
- KU Leuven, Laboratory for Neurobiology and Gene Therapy, Department of Neurosciences, Leuven, Belgium
| | - Samme Vreysen
- KU Leuven, Laboratory of Neuroplasticity and Neuroproteomics, Department of Biology, Leuven, Belgium
| | - Lutgarde Arckens
- KU Leuven, Laboratory of Neuroplasticity and Neuroproteomics, Department of Biology, Leuven, Belgium
| |
Collapse
|
24
|
BAG1 is Neuroprotective in In Vivo and In Vitro Models of Parkinson’s Disease. J Mol Neurosci 2014; 55:587-95. [DOI: 10.1007/s12031-014-0396-2] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Accepted: 07/29/2014] [Indexed: 11/27/2022]
|
25
|
Castle MJ, Gershenson ZT, Giles AR, Holzbaur ELF, Wolfe JH. Adeno-associated virus serotypes 1, 8, and 9 share conserved mechanisms for anterograde and retrograde axonal transport. Hum Gene Ther 2014; 25:705-20. [PMID: 24694006 DOI: 10.1089/hum.2013.189] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Adeno-associated virus (AAV) vectors often undergo long-distance axonal transport after brain injection. This leads to transduction of brain regions distal to the injection site, although the extent of axonal transport and distal transduction varies widely among AAV serotypes. The mechanisms driving this variability are poorly understood. This is a critical problem for applications that require focal gene expression within a specific brain region, and also impedes the utilization of vector transport for applications requiring widespread delivery of transgene to the brain. Here, we compared AAV serotypes 1 and 9, which frequently demonstrate distal transduction, with serotype 8, which rarely spreads beyond the injection site. To examine directional AAV transport in vitro, we used a microfluidic chamber to apply dye-labeled AAV to the axon termini or to the cell bodies of primary rat embryonic cortical neurons. All three serotypes were actively transported along axons, with transport characterized by high velocities and prolonged runs in both the anterograde and retrograde directions. Coinfection with pairs of serotypes indicated that AAV1, 8, and 9 share the same intracellular compartments for axonal transport. In vivo, both AAV8 and 9 demonstrated anterograde and retrograde transport within a nonreciprocal circuit after injection into adult mouse brain, with highly similar distributions of distal transduction. However, in mass-cultured neurons, we found that AAV1 was more frequently transported than AAV8 or 9, and that the frequency of AAV9 transport could be enhanced by increasing receptor availability. Thus, while these serotypes share conserved mechanisms for axonal transport both in vitro and in vivo, the frequency of transport can vary among serotypes, and axonal transport can be markedly increased by enhancing vector uptake. This suggests that variability in distal transduction in vivo likely results from differential uptake at the plasma membrane, rather than fundamental differences in transport mechanisms among AAV serotypes.
Collapse
Affiliation(s)
- Michael J Castle
- 1 Research Institute of the Children's Hospital of Philadelphia , Philadelphia, PA 19104
| | | | | | | | | |
Collapse
|
26
|
Abstract
In order to study the molecular pathways of Parkinson's disease (PD) and to develop novel therapeutic strategies, scientific investigators rely on animal models. The identification of PD-associated genes has led to the development of genetic PD models as an alternative to toxin-based models. Viral vector-mediated loco-regional gene delivery provides an attractive way to express transgenes in the central nervous system. Several vector systems based on various viruses have been developed. In this chapter, we give an overview of the different viral vector systems used for targeting the CNS. Further, we describe the different viral vector-based PD models currently available based on overexpression strategies for autosomal dominant genes such as α-synuclein and LRRK2, and knockout or knockdown strategies for autosomal recessive genes, such as parkin, DJ-1, and PINK1. Models based on overexpression of α-synuclein are the most prevalent and extensively studied, and therefore the main focus of this chapter. Many efforts have been made to increase the expression levels of α-synuclein in the dopaminergic neurons. The best α-synuclein models currently available have been developed from a combined approach using newer AAV serotypes and optimized vector constructs, production, and purification methods. These third-generation α-synuclein models show improved face and predictive validity, and therefore offer the possibility to reliably test novel therapeutics.
Collapse
|
27
|
Transgene expression in target-defined neuron populations mediated by retrograde infection with adeno-associated viral vectors. J Neurosci 2013; 33:15195-206. [PMID: 24048849 DOI: 10.1523/jneurosci.1618-13.2013] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Tools enabling the manipulation of well defined neuronal subpopulations are critical for probing complex neuronal networks. Cre recombinase (Cre) mouse driver lines in combination with the Cre-dependent expression of proteins using viral vectors--in particular, recombinant adeno-associated viral vectors (rAAVs)--have emerged as a widely used platform for achieving transgene expression in specified neural populations. However, the ability of rAAVs to further specify neuronal subsets on the basis of their anatomical connectivity has been reported as limited or inconsistent. Here, we systematically tested a variety of widely used neurotropic rAAVs for their ability to mediate retrograde gene transduction in the mouse brain. We tested pseudotyped rAAVs of several common serotypes (rAAV 2/1, 2/5, and 2/9) as well as constructs both with and without Cre-dependent expression switches. Many of the rAAVs tested--in particular, though not exclusively, Cre-dependent vectors--showed a robust capacity for retrograde infection and transgene expression. Retrograde expression was successful over distances as large as 6 mm and in multiple neuron types, including olfactory projection neurons, neocortical pyramidal cells projecting to distinct targets, and corticofugal and modulatory projection neurons. Retrograde infection using transgenes such as ChR2 allowed for optical control or optically assisted electrophysiological identification of neurons defined genetically as well as by their projection target. These results establish a widely accessible tool for achieving combinatorial specificity and stable, long-term transgene expression to isolate precisely defined neuron populations in the intact animal.
Collapse
|
28
|
Coune PG, Schneider BL, Aebischer P. Parkinson's disease: gene therapies. Cold Spring Harb Perspect Med 2013; 2:a009431. [PMID: 22474617 DOI: 10.1101/cshperspect.a009431] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
With the recent development of effective gene delivery systems, gene therapy for the central nervous system is finding novel applications. Here, we review existing viral vectors and discuss gene therapy strategies that have been proposed for Parkinson's disease. To date, most of the clinical trials were based on viral vectors to deliver therapeutic transgenes to neurons within the basal ganglia. Initial trials used genes to relieve the major motor symptoms caused by nigrostriatal degeneration. Although these new genetic approaches still need to prove more effective than existing symptomatic treatments, there is a need for disease-modifying strategies. The investigation of the genetic factors implicated in Parkinson's disease is providing precious insights in disease pathology that, combined with innovative gene delivery systems, will hopefully offer novel opportunities for gene therapy interventions to slow down, or even halt disease progression.
Collapse
Affiliation(s)
- Philippe G Coune
- Neurodegenerative Studies Laboratory, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | | | | |
Collapse
|
29
|
Bäck S, Peränen J, Galli E, Pulkkila P, Lonka-Nevalaita L, Tamminen T, Voutilainen MH, Raasmaja A, Saarma M, Männistö PT, Tuominen RK. Gene therapy with AAV2-CDNF provides functional benefits in a rat model of Parkinson's disease. Brain Behav 2013; 3:75-88. [PMID: 23532969 PMCID: PMC3607149 DOI: 10.1002/brb3.117] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2012] [Revised: 11/29/2012] [Accepted: 12/16/2012] [Indexed: 11/10/2022] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) protein has been shown to protect the nigrostriatal dopaminergic pathway when given as intrastriatal infusions in rat and mouse models of Parkinson's disease (PD). In this study, we assessed the neuroprotective effect of CDNF delivered with a recombinant adeno-associated viral (AAV) serotype 2 vector in a rat 6-hydroxydopamine (6-OHDA) model of PD. AAV2 vectors encoding CDNF, glial cell line-derived neurotrophic factor (GDNF), or green fluorescent protein were injected into the rat striatum. Protein expression analysis showed that our AAV2 vector efficiently delivered the neurotrophic factor genes into the brain and gave rise to a long-lasting expression of the proteins. Two weeks after AAV2 vector injection, 6-OHDA was injected into the rat striatum, creating a progressive degeneration of the nigrostriatal dopaminergic system. Treatment with AAV2-CDNF resulted in a marked decrease in amphetamine-induced ipsilateral rotations while it provided only partial protection of tyrosine hydroxylase (TH)-immunoreactive cells in the rat substantia nigra pars compacta and TH-reactive fibers in the striatum. Results from this study provide additional evidence that CDNF can be considered a potential treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Susanne Bäck
- Division of Pharmacology and Toxicology Faculty of Pharmacy, University of Helsinki Helsinki, Finland
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Molecular and behavioral changes associated with adult hippocampus-specific SynGAP1 knockout. Learn Mem 2012; 19:268-81. [PMID: 22700469 DOI: 10.1101/lm.026351.112] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The synaptic Ras/Rap-GTPase-activating protein (SynGAP1) plays a unique role in regulating specific downstream intracellular events in response to N-methyl-D-aspartate receptor (NMDAR) activation. Constitutive heterozygous loss of SynGAP1 disrupts NMDAR-mediated physiological and behavioral processes, but the disruptions might be of developmental origin. Therefore, the precise role of SynGAP1 in the adult brain, including its relative functional significance within specific brain regions, remains unexplored. The present study constitutes the first attempt in achieving adult hippocampal-specific SynGAP1 knockout using the Cre/loxP approach. Here, we report that this manipulation led to a significant numerical increase in both small and large GluA1 and NR1 immunoreactive clusters, many of which were non-opposed to presynaptic terminals. In parallel, the observed marked decline in the amplitude of spontaneous excitatory currents (sEPSCs) and inter-event intervals supported the impression that SynGAP1 loss might facilitate the accumulation of extrasynaptic glutamatergic receptors. In addition, SynGAP1-mediated signaling appears to be critical for the proper integration and survival of newborn neurons. The manipulation impaired reversal learning in the probe test of the water maze and induced a delay-dependent impairment in spatial recognition memory. It did not significantly affect anxiety or reference memory acquisition but induced a substantial elevation in spontaneous locomotor activity in the open field test. Thus, the present study demonstrates the functional significance of SynGAP1 signaling in the adult brain by capturing several changes that are dependent on NMDAR and hippocampal integrity.
Collapse
|
31
|
Weinberg MS, Samulski RJ, McCown TJ. Adeno-associated virus (AAV) gene therapy for neurological disease. Neuropharmacology 2012; 69:82-8. [PMID: 22465202 DOI: 10.1016/j.neuropharm.2012.03.004] [Citation(s) in RCA: 109] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2011] [Revised: 03/06/2012] [Accepted: 03/08/2012] [Indexed: 12/09/2022]
Abstract
Diseases of the central nervous system (CNS) have provided enormous opportunities for the therapeutic application of viral vector gene transfer. Adeno-associated virus (AAV) has been the vector of choice in recent clinical trials of neurological disease, including Parkinson's and Alzheimer's disease, due to the safety, efficacy, and stability of AAV gene transfer to the CNS. This review highlights the strategies employed for improving direct and peripheral targeting of therapeutic vectors to CNS tissue, and considers the significance of cellular and tissue transduction specificity, transgene regulation, and other variables that influence achievement of successful therapeutic goals. This article is part of the Special Issue entitled 'New Targets and Approaches to the Treatment of Epilepsy'.
Collapse
Affiliation(s)
- Marc S Weinberg
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | | | | |
Collapse
|
32
|
Zhang Y, Friedlander RM. Using non-coding small RNAs to develop therapies for Huntington's disease. Gene Ther 2012; 18:1139-49. [PMID: 22158031 DOI: 10.1038/gt.2011.170] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Huntington's disease (HD) is caused by an expansion of CAG triplets at the 5' end of the HD gene, which encodes a pathologically elongated polyglutamine stretch near the N-terminus of huntingtin. HD is an incurable autosomal-dominant neurodegenerative disease characterized by movement disorder, as well as emotional distress and dementia. The newly discovered roles of the non-coding small RNAs in specific degradation or translational suppression of the targeted mRNAs suggest a potential therapeutic approach of post-transcriptional gene silencing that targets the underlying disease etiology rather than the downstream pathological consequences. From pre-clinical trials in different HD animal models to cells from HD patients, small RNA interference has been applied to 'allele-non-specifically or allele-specifically' silence the mutant HD transgene or endogenous mutant HD allele. Silencing the mutant HD transgene significantly inhibits neurodegeneration, improves motor control, and extends survival of HD mice. With future improvement of mutant allele selectivity (preserving the expression of the neuroprotective wild-type allele), target specificity, efficacy and safety, as well as optimization of delivery methods, small non-coding RNA-based therapeutic applications will be a promising approach to treat HD.
Collapse
Affiliation(s)
- Y Zhang
- Department of Neurological Surgery, UPMC Presbyterian Hospital, University of Pittsburgh Medical Center, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | |
Collapse
|
33
|
Kalmbach A, Hedrick T, Waters J. Selective optogenetic stimulation of cholinergic axons in neocortex. J Neurophysiol 2012; 107:2008-19. [PMID: 22236708 DOI: 10.1152/jn.00870.2011] [Citation(s) in RCA: 76] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Acetylcholine profoundly affects neocortical function, being involved in arousal, attention, learning, memory, sensory and motor function, and plasticity. The majority of cholinergic afferents to neocortex are from neurons in nucleus basalis. Nucleus basalis also contains projecting neurons that release other transmitters, including GABA and possibly glutamate. Hence, electrical stimulation of nucleus basalis evokes the release of a mixture of neurotransmitters in neocortex, and this lack of selectivity has impeded research on cholinergic signaling in neocortex. We describe a method for the selective stimulation of cholinergic axons in neocortex. We used the Cre-lox system and a viral vector to express the light-activated protein channelrhodopsin-2 in cholinergic neurons in nucleus basalis and their axons in neocortex. Labeled neurons depolarized on illumination with blue light but were otherwise unchanged. In anesthetized mice, illumination of neocortex desynchronized the local field potential, indicating that light evoked release of ACh. This novel technique will enable many new studies of the cellular, network, and behavioral physiology of ACh in neocortex.
Collapse
Affiliation(s)
- Abigail Kalmbach
- Department of Physiology, Feinberg School of Medicine, Northwestern Univ., Chicago, IL 60611, USA
| | | | | |
Collapse
|
34
|
Abstract
The recent development of optogenetics, a revolutionary research tool in neuroscience, portends an evolution of current clinical neuromodulation tools. A form of gene therapy, optogenetics makes possible highly precise spatial and temporal control of specific neuronal populations. This technique has already provided several new insights relevant to clinical neuroscience, from the physiological substrate of functional magnetic resonance imaging to the mechanism of deep brain stimulation in Parkinson's disease. The increased precision of optogenetic techniques also raises the possibility of eventual human use. Translational efforts have begun in primates, with success reported from multiple labs in rhesus macaques. These developments will remain of ongoing interest to neurologists and neurosurgeons.
Collapse
Affiliation(s)
- Paul S A Kalanithi
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
| | | |
Collapse
|
35
|
Frontotemporal lobar degeneration-related proteins induce only subtle memory-related deficits when bilaterally overexpressed in the dorsal hippocampus. Exp Neurol 2011; 233:807-14. [PMID: 22177996 DOI: 10.1016/j.expneurol.2011.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2011] [Revised: 11/01/2011] [Accepted: 12/01/2011] [Indexed: 02/08/2023]
Abstract
Frontotemporal lobar degeneration (FTLD) is a neurodegenerative disease that involves cognitive decline and dementia. To model the hippocampal neurodegeneration and memory-related behavioral impairment that occurs in FTLD and other tau and TDP-43 proteinopathy diseases, we used an adeno-associated virus serotype 9 (AAV9) vector to induce bilateral expression of either microtubule-associated protein tau or transactive response DNA binding protein 43 kDa (TDP-43) in adult rat dorsal hippocampus. Human wild-type forms of tau or TDP-43 were expressed. The vectors/doses were designed for moderate expression levels within neurons. Rats were evaluated for acquisition and retention in the Morris water task over 12 weeks after gene transfer. Neither vector altered acquisition performance compared to controls. In measurements of retention, there was impairment in the TDP-43 group. Histological examination revealed specific loss of dentate gyrus granule cells and concomitant gliosis proximal to the injection site in the TDP-43 group, with shrinkage of the dorsal hippocampus. Despite specific tau pathology, the tau gene transfer surprisingly did not cause obvious neuronal loss or behavioral impairment. The data demonstrate that TDP-43 produced mild behavioral impairment and hippocampal neurodegeneration in rats, whereas tau did not. The models could be of value for studying mechanisms of FTLD and other diseases with tau and TDP-43 pathology in the hippocampus including Alzheimer's disease, with relevance to early stage mild impairment.
Collapse
|
36
|
Abstract
Both observational and perturbational technologies are essential for advancing the understanding of brain function and dysfunction. But while observational techniques have greatly advanced in the last century, techniques for perturbation that are matched to the speed and heterogeneity of neural systems have lagged behind. The technology of optogenetics represents a step toward addressing this disparity. Reliable and targetable single-component tools (which encompass both light sensation and effector function within a single protein) have enabled versatile new classes of investigation in the study of neural systems. Here we provide a primer on the application of optogenetics in neuroscience, focusing on the single-component tools and highlighting important problems, challenges, and technical considerations.
Collapse
Affiliation(s)
- Ofer Yizhar
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|
37
|
Comparison of the efficacy of four viral vectors for transducing hypothalamic magnocellular neurosecretory neurons in the rat supraoptic nucleus. J Neurosci Methods 2011; 197:238-48. [PMID: 21392530 DOI: 10.1016/j.jneumeth.2011.02.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2010] [Revised: 02/09/2011] [Accepted: 02/24/2011] [Indexed: 02/08/2023]
Abstract
Since transgenes were first cloned into recombinant adenoviruses almost 30 years ago, a variety of viral vectors have become important tools in genetic research. Viruses adeptly transport genetic material into eukaryotic cells, and replacing all or part of the viral genome with genes of interest or silencing sequences creates a method of gene expression modulation in which the timing and location of manipulations can be specific. The hypothalamo-neurohypophyseal system (HNS), consisting of the paraventricular (PVN) and supraoptic (SON) nuclei in the hypothalamus, regulates fluid balance homeostasis and is highly plastic, yet tightly regulated by extracellular fluid (ECF) osmolality and volume. Its reversible plasticity and physiological relevance make it a good system for studying interactions between gene expression and physiology. Here, four viral vectors were compared for their ability to transduce magnocellular neurosecretory neurons (MNCs) of the SON in adult rats. The vectors included an adenovirus, a lentivirus (HIV) and two serotypes of adeno-associated viruses (AAV5 and AAV2). Though adenovirus and AAV2 vectors have previously been used to transduce SON neurons, HIV and AAV5 have not. All four vectors transduced MNCs, but the AAV vectors were the most effective, transducing large numbers of MNCs, with minimal or no glial transduction. The AAV vectors were injected using a convection enhanced delivery protocol to maximize dispersal through the tissue, resulting in the transduction of neurons throughout the anterior to posterior length of the SON (∼1.5mm). AAV5, but not AAV2, showed some selectivity for SON neurons relative to those in the surrounding hypothalamus.
Collapse
|
38
|
Efficient and stable transduction of dopaminergic neurons in rat substantia nigra by rAAV 2/1, 2/2, 2/5, 2/6.2, 2/7, 2/8 and 2/9. Gene Ther 2011; 18:517-27. [PMID: 21326331 DOI: 10.1038/gt.2010.179] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Dysfunction of the nigrostriatal system is the major cause of Parkinson's disease (PD). This brain region is therefore an important target for gene delivery aiming at disease modeling and gene therapy. Recombinant adeno-associated viral (rAAV) vectors have been developed as efficient vehicles for gene transfer into the central nervous system. Recently, several serotypes have been described, with varying tropism for brain transduction. In light of the further development of a viral vector-mediated rat model for PD, we performed a comprehensive comparison of the transduction and tropism for dopaminergic neurons (DNs) in the adult Wistar rat substantia nigra (SN) of seven rAAV vector serotypes (rAAV 2/1, 2/2, 2/5, 2/6.2, 2/7, 2/8 and 2/9). All vectors were normalized by titer and volume, and stereotactically injected into the SN. Gene expression was assessed non-invasively and quantitatively in vivo by bioluminescence imaging at 2 and 5 weeks after injection, and was found to be stable over time. Immunohistochemistry at 6 weeks following injection revealed the most widespread enhanced green fluorescence protein expression and the highest number of positive nigral cells using rAAV 2/7, 2/9 and 2/1. The area transduced by rAAV 2/8 was smaller, but nevertheless almost equal numbers of nigral cells were targeted. Detailed confocal analysis revealed that serotype 2/7, 2/9, 2/1 and 2/8 transduced at least 70% of the DNs. In conclusion, these results show that various rAAV serotypes efficiently transduce nigral DNs, but significant differences in transgene expression pattern and level were observed.
Collapse
|
39
|
Phenotypic correction of a mouse model for primary hyperoxaluria with adeno-associated virus gene transfer. Mol Ther 2010; 19:870-5. [PMID: 21119625 DOI: 10.1038/mt.2010.270] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Primary hyperoxaluria type I (PH1) is an inborn error of metabolism caused by deficiency of the hepatic enzyme alanine-glyoxylate aminotransferase (AGXT or AGT) which leads to overproduction of oxalate by the liver and subsequent urolithiasis and renal failure. The current therapy largely depends on liver transplantation, which is associated with significant morbidity and mortality. To explore an alternative treatment, we used somatic gene transfer in a mouse genetic model for PH1 (Agxt1KO). Recombinant adeno-associated virus (AAV) vectors containing the human AGXT complementary DNA (cDNA) were pseudotyped with capsids from either serotype 8 or 5, and delivered to the livers of Agxt1KO mice via the tail vein. Both AAV8-AGXT and AAV5-AGXT vectors were able to reduce oxaluria to normal levels. In addition, treated mice showed blunted increase of oxaluria after challenge with ethylene glycol (EG), a glyoxylate precursor. In mice, AGT enzyme activity in whole liver extracts were restored to normal without hepatic toxicity nor immunogenicity for the 50 day follow-up. In summary, this study demonstrates the correction of primary hyperoxaluria in mice treated with either AAV5 or AAV8 vectors.
Collapse
|
40
|
Sanchez CE, Tierney TS, Gale JT, Alavian KN, Sahin A, Lee JS, Mulligan RC, Carter BS. Recombinant adeno-associated virus type 2 pseudotypes: comparing safety, specificity, and transduction efficiency in the primate striatum. Laboratory investigation. J Neurosurg 2010; 114:672-80. [PMID: 20950087 DOI: 10.3171/2010.8.jns091583] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Although several clinical trials utilizing the adeno-associated virus (AAV) type 2 serotype 2 (2/2) are now underway, it is unclear whether this particular serotype offers any advantage over others in terms of safety or efficiency when delivered directly to the CNS. METHODS Recombinant AAV2-green fluorescent protein (GFP) serotypes 2/1, 2/2, 2/5, and 2/8 were generated following standard triple transfection protocols (final yield 5.4 × 10(12) particles/ml). A total of 180 μl of each solution was stereotactically infused, covering the entire rostrocaudal extent of the caudoputamen in 4 rhesus monkeys (Macaca mulatta) (3.0 ± 0.5 kg). After 6 weeks' survival, the brain was formalin fixed, cut at 40 μm, and stained with standard immunohistochemistry for anti-GFP, anticaspase-2, and cell-specific markers (anti-microtubule-associated protein-2 for neurons and anti-glial fibrillary acidic protein for glia). Unbiased stereological counting methods were used to determine cell number and striatal volume. RESULTS The entire striatum of each animal contained GFP-positive cells with significant labeling extending beyond the borders of the basal ganglia. No ischemic/necrotic, hemorrhagic, or neoplastic change was observed in any brain. Total infusate volumes were similar across the 4 serotypes. However, GFP-labeled cell density was markedly different. Adeno-associated virus 2/1, 2/2, and 2/5 each labeled < 8000 cells/mm(3), whereas serotype 8 labeled > 21,000 cells, a 3- to 4-fold higher transduction efficiency. On the other hand, serotype 8 also labeled neurons and glia with equal affinity compared with neuronal specificities > 89% for the other serotypes. Moderate caspase-2 colabeling was noted in neurons immediately around the AAV2/1 injection tracts, but was not seen above the background anywhere in the brain following injections with serotypes 2, 5, or 8. CONCLUSIONS Intrastriatal delivery of AAV2 yields the highest cell transduction efficiencies but lowest neuronal specificity for serotype 8 when compared with serotypes 1, 2, and 5. Only AAV2/1 revealed significant caspase-2 activation. Careful consideration of serotype-specific differences in AAV2 neurotropism, transduction efficiency, and potential toxicity may affect future human trials.
Collapse
Affiliation(s)
- Carlos E Sanchez
- Department of Neurosurgery, Massachusetts General Hospital, Boston, USA
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Fitzsimons HL, Riban V, Bland RJ, Wendelken JL, Sapan CV, During MJ. Biodistribution and safety assessment of AAV2-GAD following intrasubthalamic injection in the rat. J Gene Med 2010; 12:385-98. [PMID: 20352617 DOI: 10.1002/jgm.1449] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND The steps necessary to translate promising new biological therapies to the clinic are poorly documented. For gene therapy, there are unique aspects that need to be addressed in biodistribution studies. Notably, the spread of the vector beyond the intended target cells or tissue may result in persistent unwanted biological activity or unpredictable biological events; thus, it is critical to evaluate the risks associated with viral vector-mediated gene transfer prior to embarking on human clinical trials. METHODS In the present study, we conducted a comprehensive assessment of vector biodistribution throughout the brain, blood and major organs of rats that had been injected via the subthalamic nucleus with recombinant adeno-associated virus (AAV) expressing glutamic acid decarboxylase (GAD). In addition, behavioral and histological analyses were also performed. RESULTS AAV genomes were not detected in blood or cerebrospinal fluid, and did not disseminate to organs outside of the brain in the majority of animals. In the brain, an average of 97.3% of AAV2-GAD genomes were restricted to the area of the ipsilateral subthalamic nucleus (STN). There were no discernable effects of AAV2-GAD on general health, and a behavioral assessment of the animals did not reveal any alteration in general behavior, exploration, locomotion or motor symmetry. CONCLUSIONS The present study met Food and Drug Administration requirements, in addition to efficacy and toxicity studies in rodents and nonhuman primates, to support and supplement a Phase II clinical trial invloving the gene transfer of AAV2-GAD to the human STN for the potential therapy of Parkinson's disease.
Collapse
|
42
|
Gorbatyuk OS, Li S, Nguyen FN, Manfredsson FP, Kondrikova G, Sullivan LF, Meyers C, Chen W, Mandel RJ, Muzyczka N. α-Synuclein expression in rat substantia nigra suppresses phospholipase D2 toxicity and nigral neurodegeneration. Mol Ther 2010; 18:1758-68. [PMID: 20664530 DOI: 10.1038/mt.2010.137] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Abstract
We present genetic evidence that an in vivo role of α-synuclein (α-syn) is to inhibit phospholipase D2 (PLD2), an enzyme that is believed to participate in vesicle trafficking, membrane signaling, and both endo- and exocytosis. Overexpression of PLD2 in rat substantia nigra pars compacta (SNc) caused severe neurodegeneration of dopamine (DA) neurons, loss of striatal DA, and an associated ipsilateral amphetamine-induced rotational asymmetry. Coexpression of human wild type α-syn suppressed PLD2 neurodegeneration, DA loss, and amphetamine-induced rotational asymmetry. However, an α-syn mutant defective for inhibition of PLD2 in vitro also failed to inhibit PLD toxicity in vivo. Further, reduction of PLD2 activity in SNc, either by siRNA knockdown of PLD2 or overexpression of α-syn, both produced an unusual contralateral amphetamine-induced rotational asymmetry, opposite to that seen with overexpression of PLD2, suggesting that PLD2 and α-syn were both involved in DA release or reuptake. Finally, α-syn coimmunoprecipitated with PLD2 from extracts prepared from striatal tissues. Taken together, our data demonstrate that α-syn is an inhibitor of PLD2 in vivo, and confirm earlier reports that α-syn inhibits PLD2 in vitro. Our data also demonstrate that it is possible to use viral-mediated gene transfer to study gene interactions in vivo.
Collapse
Affiliation(s)
- Oleg S Gorbatyuk
- Department of Molecular Genetics and Microbiology, College of Medicine, University of Florida, Gainesville, Florida, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Krzyżosiak A, Szyszka-Niagolov M, Wietrzych M, Gobaille S, Muramatsu SI, Krężel W. Retinoid X Receptor Gamma Control of Affective Behaviors Involves Dopaminergic Signaling in Mice. Neuron 2010; 66:908-20. [DOI: 10.1016/j.neuron.2010.05.004] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/30/2010] [Indexed: 10/19/2022]
|
44
|
Rebuffat A, Harding CO, Ding Z, Thöny B. Comparison of adeno-associated virus pseudotype 1, 2, and 8 vectors administered by intramuscular injection in the treatment of murine phenylketonuria. Hum Gene Ther 2010; 21:463-77. [PMID: 19916803 PMCID: PMC2865356 DOI: 10.1089/hum.2009.127] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2009] [Accepted: 11/15/2009] [Indexed: 12/20/2022] Open
Abstract
Phenylketonuria (PKU) is caused by hepatic phenylalanine hydroxylase (PAH) deficiency and is associated with systemic accumulation of phenylalanine (Phe). Previously we demonstrated correction of murine PKU after intravenous injection of a recombinant type 2 adeno-associated viral vector pseudotyped with type 8 capsid (rAAV2/8), which successfully directed hepatic transduction and Pah gene expression. Here, we report that liver PAH activity and phenylalanine clearance were also restored in PAH-deficient mice after simple intramuscular injection of either AAV2 pseudotype 1 (rAAV2/1) or rAAV2/8 vectors. Serotype 2 AAV vector (rAAV2/2) was also investigated, but long-term phenylalanine clearance has been observed only for pseudotypes 1 and 8. Therapeutic correction was shown in both male and female mice, albeit more effectively in males, in which correction lasted for the entire period of the experiment (>1 year). Although phenylalanine levels began to rise in female mice at about 8-10 months after rAAV2/8 injection they remained only mildly hyperphenylalaninemic thereafter and subsequent supplementation with synthetic tetrahydrobiopterin resulted in a transient decrease in blood phenylalanine. Alternatively, subsequent administration of a second vector with a different AAV pseudotype to avoid immunity against the previously administrated vector was also successful for long-term treatment of female PKU mice. Overall, this relatively less invasive gene transfer approach completes our previous studies and allows comparison of complementary strategies in the development of efficient PKU gene therapy protocols.
Collapse
Affiliation(s)
- Alexandre Rebuffat
- Division of Clinical Chemistry and Biochemistry, Department of Pediatrics, University of Zürich, CH-8032 Zürich, Switzerland
| | - Cary O. Harding
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97201, USA
| | - Zhaobing Ding
- Division of Clinical Chemistry and Biochemistry, Department of Pediatrics, University of Zürich, CH-8032 Zürich, Switzerland
- Present address: Institute of Bioengineering and Nanotechnology, The Nanos, 138669, Singapore
| | - Beat Thöny
- Division of Clinical Chemistry and Biochemistry, Department of Pediatrics, University of Zürich, CH-8032 Zürich, Switzerland
| |
Collapse
|
45
|
Molecular and cellular approaches for diversifying and extending optogenetics. Cell 2010; 141:154-165. [PMID: 20303157 PMCID: PMC4160532 DOI: 10.1016/j.cell.2010.02.037] [Citation(s) in RCA: 749] [Impact Index Per Article: 49.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2009] [Revised: 01/11/2010] [Accepted: 02/18/2010] [Indexed: 12/17/2022]
Abstract
Optogenetic technologies employ light to control biological processes within targeted cells in vivo with high temporal precision. Here, we show that application of molecular trafficking principles can expand the optogenetic repertoire along several long-sought dimensions. Subcellular and transcellular trafficking strategies now permit (1) optical regulation at the far-red/infrared border and extension of optogenetic control across the entire visible spectrum, (2) increased potency of optical inhibition without increased light power requirement (nanoampere-scale chloride-mediated photocurrents that maintain the light sensitivity and reversible, step-like kinetic stability of earlier tools), and (3) generalizable strategies for targeting cells based not only on genetic identity, but also on morphology and tissue topology, to allow versatile targeting when promoters are not known or in genetically intractable organisms. Together, these results illustrate use of cell-biological principles to enable expansion of the versatile fast optogenetic technologies suitable for intact-systems biology and behavior.
Collapse
|
46
|
Colle MA, Piguet F, Bertrand L, Raoul S, Bieche I, Dubreil L, Sloothaak D, Bouquet C, Moullier P, Aubourg P, Cherel Y, Cartier N, Sevin C. Efficient intracerebral delivery of AAV5 vector encoding human ARSA in non-human primate. Hum Mol Genet 2010; 19:147-58. [PMID: 19837699 DOI: 10.1093/hmg/ddp475] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
Metachromatic leukodystrophy (MLD) is a lethal neurodegenerative disease caused by a deficiency in the lysosomal arylsulfatase A (ARSA) enzyme leading to the accumulation of sulfatides in glial and neuronal cells. We previously demonstrated in ARSA-deficient mice that intracerebral injection of a serotype 5 adeno-associated vector (AAV) encoding human ARSA corrects the biochemical, neuropathological and behavioral abnormalities. However, before considering a potential clinical application, scaling-up issues should be addressed in large animals. Therefore, we performed intracerebral injection of the same AAV vector (total dose of 3.8 x 10(11) or 1.9 x 10(12) vector genome, three sites of injection in the right hemisphere, two deposits per site of injection) into three selected areas of the centrum semiovale white matter, or in the deep gray matter nuclei (caudate nucleus, putamen, thalamus) of six non-human primates to evaluate vector distribution, as well as expression and activity of human ARSA. The procedure was perfectly tolerated, without any adverse effect or change in neurobehavioral examination. AAV vector was detected in a brain volume of 12-15 cm(3) that corresponded to 37-46% of the injected hemisphere. ARSA enzyme was expressed in multiple interconnected brain areas over a distance of 22-33 mm. ARSA activity was increased by 12-38% in a brain volume that corresponded to 50-65% of injected hemisphere. These data provide substantial evidence for potential benefits of brain gene therapy in patients with MLD.
Collapse
|
47
|
Mason MRJ, Ehlert EME, Eggers R, Pool CW, Hermening S, Huseinovic A, Timmermans E, Blits B, Verhaagen J. Comparison of AAV serotypes for gene delivery to dorsal root ganglion neurons. Mol Ther 2010; 18:715-24. [PMID: 20179682 DOI: 10.1038/mt.2010.19] [Citation(s) in RCA: 133] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
For many experiments in the study of the peripheral nervous system, it would be useful to genetically manipulate primary sensory neurons. We have compared vectors based on adeno-associated virus (AAV) serotypes 1, 2, 3, 4, 5, 6, and 8, and lentivirus (LV), all expressing green fluorescent protein (GFP), for efficiency of transduction of sensory neurons, expression level, cellular tropism, and persistence of transgene expression following direct injection into the dorsal root ganglia (DRG), using histological quantification and qPCR. Two weeks after injection, AAV1, AAV5, and AAV6 had transduced the most neurons. The time course of GFP expression from these three vectors was studied from 1 to 12 weeks after injection. AAV5 was the most effective serotype overall, followed by AAV1. Both these serotypes showed increasing neuronal transduction rates at later time points, with some injections of AAV5 yielding over 90% of DRG neurons GFP(+) at 12 weeks. AAV6 performed well initially, but transduction rates declined dramatically between 4 and 12 weeks. AAV1 and AAV5 both transduced large-diameter neurons, IB4(+) neurons, and CGRP(+) neurons. In conclusion, AAV5 is a highly effective gene therapy vector for primary sensory neurons following direct injection into the DRG.
Collapse
Affiliation(s)
- Matthew R J Mason
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, Institute of the Royal Academy of Arts and Sciences, Amsterdam, the Netherlands.
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Bockstael O, Chtarto A, Wakkinen J, Yang X, Melas C, Levivier M, Brotchi J, Tenenbaum L. Differential transgene expression profiles in rat brain, using rAAV2/1 vectors with tetracycline-inducible and cytomegalovirus promoters. Hum Gene Ther 2009; 19:1293-305. [PMID: 19866492 DOI: 10.1089/hum.2008.099] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The biodistribution of transgene expression in the CNS after localized stereotaxic vector delivery is an important issue for the safety of gene therapy for neurological diseases. The cellular specificity of transgene expression from rAAV2/1 vectors (recombinant adeno-associated viral vectors pseudotyped with viral capsids from serotype 1) using the tetracycline-inducible (TetON) expression cassette in comparison with the cytomegalovirus (CMV) promoter was investigated in the rat nigrostriatal pathway. After intrastriatal injection, although green fluorescent protein (GFP) was expressed mainly in neurons with both vectors, the relative proportions of DARPP-32-positive projection neurons and parvalbumin-positive interneurons were, respectively, 13:1 and 2:1 for the CMV and TetON vectors. DARP32-positive neurons projecting to the globus pallidus were strongly GFP positive with both vectors, whereas those projecting to the substantia nigra pars reticulata (SNpr) were efficiently labeled by the CMV vector but poorly by the TetON vector. Numerous GFP-positive cells were evidenced in the subventricular zone with both vectors. However, in the olfactory bulb (OB), GFP-positive neurons were observed with the CMV vector but not the TetON vector. We conclude that the absence of significant amounts of transgene product in distant regions (SN and OB) constitutes a safety advantage of the AAV2/1-TetON vector for striatal gene therapy. Midbrain injections resulted in selective GFP expression in tyrosine hydroxylase-positive neurons by the TetON vector whereas with the CMV vector, GFP-positive cells covered a widespread area of the midbrain. The biodistribution of GFP protein corresponded to that of the transcripts and not of the viral genomes. We conclude that the rAAV2/1-TetON vector constitutes an interesting tool for specific transgene expression in midbrain dopaminergic neurons.
Collapse
Affiliation(s)
- Olivier Bockstael
- Multidisciplinary Research Institute (IRIBHM), Université Libre de Bruxelles, Hôpital Erasme, Brussels, Belgium
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Lim ST, Airavaara M, Harvey BK. Viral vectors for neurotrophic factor delivery: a gene therapy approach for neurodegenerative diseases of the CNS. Pharmacol Res 2009; 61:14-26. [PMID: 19840853 DOI: 10.1016/j.phrs.2009.10.002] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2009] [Revised: 10/11/2009] [Accepted: 10/11/2009] [Indexed: 01/11/2023]
Abstract
The clinical manifestation of most diseases of the central nervous system results from neuronal dysfunction or loss. Diseases such as stroke, epilepsy and neurodegeneration (e.g. Alzheimer's disease and Parkinson's disease) share common cellular and molecular mechanisms (e.g. oxidative stress, endoplasmic reticulum stress, mitochondrial dysfunction) that contribute to the loss of neuronal function. Neurotrophic factors (NTFs) are secreted proteins that regulate multiple aspects of neuronal development including neuronal maintenance, survival, axonal growth and synaptic plasticity. These properties of NTFs make them likely candidates for preventing neurodegeneration and promoting neuroregeneration. One approach to delivering NTFs to diseased cells is through viral vector-mediated gene delivery. Viral vectors are now routinely used as tools for studying gene function as well as developing gene-based therapies for a variety of diseases. Currently, many clinical trials using viral vectors in the nervous system are underway or completed, and seven of these trials involve NTFs for neurodegeneration. In this review, we discuss viral vector-mediated gene transfer of NTFs to treat neurodegenerative diseases of the central nervous system.
Collapse
Affiliation(s)
- Seung T Lim
- Department of Neuroscience, Georgetown University Medical Center, Washington, DC 20057, United States
| | | | | |
Collapse
|
50
|
Dodiya HB, Bjorklund T, Stansell J, Mandel RJ, Kirik D, Kordower JH. Differential transduction following basal ganglia administration of distinct pseudotyped AAV capsid serotypes in nonhuman primates. Mol Ther 2009; 18:579-87. [PMID: 19773746 DOI: 10.1038/mt.2009.216] [Citation(s) in RCA: 73] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
We examined the transduction efficiency of different adeno-associated virus (AAV) capsid serotypes encoding for green fluorescent protein (GFP) flanked by AAV2 inverted terminal repeats in the nonhuman primate basal ganglia as a prelude to translational studies, as well as clinical trials in patients with Parkinson's disease (PD). Six intact young adult cynomolgus monkeys received a single 10 microl injection of AAV2/1-GFP, AAV2/5-GFP, or AAV2/8-GFP pseudotyped vectors into the caudate nucleus and putamen bilaterally in a pattern that resulted in each capsid serotype being injected into at least four striatal sites. GFP immunohistochemistry revealed excellent transduction rates for each AAV pseudotype. Stereological estimates of GFP+ cells within the striatum revealed that AAV2/5-GFP transduces significantly higher number of cells than AAV2/8-GFP (P < 0.05) and there was no significant difference between AAV2/5-GFP and AAV2/1-GFP (P = 0.348). Consistent with this result, Cavalieri estimates revealed that AAV2/5-GFP resulted in a significantly larger transduction volume than AAV2/8-GFP (P < 0.05). Each pseudotype transduced striatal neurons effectively [>95% GFP+ cells colocalized neuron-specific nuclear protein (NeuN)]. The current data suggest that AAV2/5 and AAV2/1 are superior to AAV2/8 for gene delivery to the nonhuman primate striatum and therefore better candidates for therapeutic applications targeting this structure.
Collapse
Affiliation(s)
- Hemraj B Dodiya
- Department of Neurological Sciences, Rush University Medical Center, Chicago, Illinois 60612, USA
| | | | | | | | | | | |
Collapse
|