1
|
Ozgür-Gunes Y, Le Stunff C, Bougnères P. Oligodendrocytes, the Forgotten Target of Gene Therapy. Cells 2024; 13:1973. [PMID: 39682723 DOI: 10.3390/cells13231973] [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: 10/29/2024] [Revised: 11/22/2024] [Accepted: 11/26/2024] [Indexed: 12/18/2024] Open
Abstract
If the billions of oligodendrocytes (OLs) populating the central nervous system (CNS) of patients could express their feelings, they would undoubtedly tell gene therapists about their frustration with the other neural cell populations, neurons, microglia, or astrocytes, which have been the favorite targets of gene transfer experiments. This review questions why OLs have been left out of most gene therapy attempts. The first explanation is that the pathogenic role of OLs is still discussed in most CNS diseases. Another reason is that the so-called ubiquitous CAG, CBA, CBh, or CMV promoters-widely used in gene therapy studies-are unable or poorly able to activate the transcription of episomal transgene copies brought by adeno-associated virus (AAV) vectors in OLs. Accordingly, transgene expression in OLs has either not been found or not been evaluated in most gene therapy studies in rodents or non-human primates. The aims of the current review are to give OLs their rightful place among the neural cells that future gene therapy could target and to encourage researchers to test the effect of OL transduction in various CNS diseases.
Collapse
Affiliation(s)
- Yasemin Ozgür-Gunes
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA
| | - Catherine Le Stunff
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l'Energie Atomique, 92260 Fontenay-aux-Roses, France
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- UMR1195 Inserm and University Paris Saclay, 94270 Le Kremlin-Bicêtre, France
| | - Pierre Bougnères
- MIRCen Institute, Laboratoire des Maladies Neurodégénératives, Commissariat à l'Energie Atomique, 92260 Fontenay-aux-Roses, France
- NEURATRIS at MIRCen, 92260 Fontenay-aux-Roses, France
- Therapy Design Consulting, 94300 Vincennes, France
| |
Collapse
|
2
|
Kofoed RH, Aubert I. Focused ultrasound gene delivery for the treatment of neurological disorders. Trends Mol Med 2024; 30:263-277. [PMID: 38216449 DOI: 10.1016/j.molmed.2023.12.006] [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: 10/27/2023] [Revised: 12/11/2023] [Accepted: 12/15/2023] [Indexed: 01/14/2024]
Abstract
The transformative potential of gene therapy has been demonstrated in humans. However, there is an unmet need for non-invasive targeted gene delivery and regulation in the treatment of brain disorders. Transcranial focused ultrasound (FUS) has gained tremendous momentum to address these challenges. FUS non-invasively modulates brain cells and their environment, and is a powerful tool to facilitate gene delivery across the blood-brain barrier (BBB) with millimeter precision and promptly regulate transgene expression. This review highlights technical aspects of FUS-mediated gene therapies for the central nervous system (CNS) and lessons learned from discoveries in other organs. Understanding the possibilities and remaining obstacles of FUS-mediated gene therapy will be necessary to harness remarkable technologies and create life-changing treatments for neurological disorders.
Collapse
Affiliation(s)
- Rikke Hahn Kofoed
- Department of Neurosurgery, Department of Clinical Medicine, Aarhus University, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Center for Experimental Neuroscience (CENSE), Department of Neurosurgery, Aarhus University Hospital, Palle Juul-Jensens Boulevard 165, 8200 Aarhus N, Denmark; Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada.
| | - Isabelle Aubert
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Toronto, ON, Canada; Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
3
|
Blaess S, Krabbe S. Cell type specificity for circuit output in the midbrain dopaminergic system. Curr Opin Neurobiol 2023; 83:102811. [PMID: 37972537 DOI: 10.1016/j.conb.2023.102811] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 09/14/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023]
Abstract
Midbrain dopaminergic neurons are a relatively small group of neurons in the mammalian brain controlling a wide range of behaviors. In recent years, increasingly sophisticated tracing, imaging, transcriptomic, and machine learning approaches have provided substantial insights into the anatomical, molecular, and functional heterogeneity of dopaminergic neurons. Despite this wealth of new knowledge, it remains unclear whether and how the diverse features defining dopaminergic subclasses converge to delineate functional ensembles within the dopaminergic system. Here, we review recent studies investigating various aspects of dopaminergic heterogeneity and discuss how development, behavior, and disease influence subtype characteristics. We then outline what further approaches could be pursued to gain a more inclusive picture of dopaminergic diversity, which could be crucial to understanding the functional architecture of this system.
Collapse
Affiliation(s)
- Sandra Blaess
- Neurodevelopmental Genetics, Institute of Reconstructive Neurobiology, Medical Faculty, University of Bonn, 53127 Bonn, Germany.
| | - Sabine Krabbe
- German Center for Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany.
| |
Collapse
|
4
|
Neuman SS, Metzger JM, Bondarenko V, Wang Y, Felton J, Levine JE, Saha K, Gong S, Emborg ME. Striatonigral distribution of a fluorescent reporter following intracerebral delivery of genome editors. Front Bioeng Biotechnol 2023; 11:1237613. [PMID: 37564994 PMCID: PMC10410562 DOI: 10.3389/fbioe.2023.1237613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Accepted: 07/18/2023] [Indexed: 08/12/2023] Open
Abstract
Introduction: Targeted gene editing is proposed as a therapeutic approach for numerous disorders, including neurological diseases. As the brain is organized into neural networks, it is critical to understand how anatomically connected structures are affected by genome editing. For example, neurons in the substantia nigra pars compacta (SNpc) project to the striatum, and the striatum contains neurons that project to the substantia nigra pars reticulata (SNpr). Methods: Here, we report the effect of injecting genome editors into the striatum of Ai14 reporter mice, which have a LoxP-flanked stop cassette that prevents expression of the red fluorescent protein tdTomato. Two weeks following intracerebral delivery of either synthetic nanocapsules (NCs) containing CRISPR ribonucleoprotein targeting the tdTomato stop cassette or adeno-associated virus (AAV) vectors expressing Cre recombinase, the brains were collected, and the presence of tdTomato was assessed in both the striatum and SN. Results: TdTomato expression was observed at the injection site in both the NC- and AAV-treated groups and typically colocalized with the neuronal marker NeuN. In the SN, tdTomato-positive fibers were present in the pars reticulata, and SNpr area expressing tdTomato correlated with the size of the striatal genome edited area. Conclusion: These results demonstrate in vivo anterograde axonal transport of reporter gene protein products to the SNpr following neuronal genome editing in the striatum.
Collapse
Affiliation(s)
- Samuel S. Neuman
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Jeanette M. Metzger
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Viktoriya Bondarenko
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Yuyuan Wang
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI, United States
| | - Jesi Felton
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
| | - Jon E. Levine
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Neuroscience, University of Wisconsin–Madison, Madison, WI, United States
| | - Krishanu Saha
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI, United States
| | - Shaoqin Gong
- Department of Ophthalmology and Visual Sciences, University of Wisconsin–Madison, Madison, WI, United States
- Department of Biomedical Engineering, University of Wisconsin–Madison, Madison, WI, United States
- Wisconsin Institute for Discovery, University of Wisconsin–Madison, Madison, WI, United States
| | - Marina E. Emborg
- Wisconsin National Primate Research Center, University of Wisconsin-Madison, Madison, WI, United States
- Department of Medical Physics, University of Wisconsin-Madison, Madison, WI, United States
| |
Collapse
|
5
|
Kasper JM, Smith AE, Miller SN, Ara, Russell WK, Cunningham KA, Hommel JD. Role of neuropeptide neuromedin U in the nucleus accumbens shell in cocaine self-administration in male rats. Neuropsychopharmacology 2022; 47:1875-1882. [PMID: 34916591 PMCID: PMC9485260 DOI: 10.1038/s41386-021-01234-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Revised: 10/29/2021] [Accepted: 11/03/2021] [Indexed: 11/09/2022]
Abstract
The nucleus accumbens shell (NAcSh) and its afferent and efferent neuronal projections control key aspects of motivation for cocaine. A recently described regulator of γ-aminobutyric acid (GABA) projections from the dorsal raphe nucleus (DRN) to the NAcSh (DRN → NAcSh) is the neuropeptide neuromedin U (NMU). Here, we find that systemic administration of NMU decreases breakpoint for cocaine on a progressive ratio schedule of reinforcement in male rats. Employing a retrograde adeno-associated virus (AAV), we found that RNAi-mediated knockdown of the NMU receptor 2 (NMUR2) in afferent DRN projections to the NAcSh increases the breakpoint for cocaine. Our previous studies demonstrated that NMU regulates GABA release in the NAcSh, and our current investigation found that systemic NMU administration suppresses cocaine-evoked GABA release in the NAcSh and increases phosphorylated c-Fos expression in neurons projecting from the NAcSh to the ventral pallidum (VP). To further probe the impact of NMU/NMUR2 on neuroanatomical pathways regulating motivation for cocaine, we employed multi-viral transsynaptic studies. Using a combination of rabies virus and retrograde AAV helper virus, we mapped the impact of NMU across three distinct brain regions simultaneously and found a direct connection of GABAergic DRN neurons to the NAcSh → VP pathway. Together, these data reveal that NMU/NMUR2 modulates a direct connection within the GABAergic DRN → NAcSh → VP circuit that diminishes breakpoints for cocaine. These findings importantly advance our understanding of the neurochemical underpinnings of pathway-specific regulation of neurocircuitry that may regulate cocaine self-administration, providing a unique therapeutic perspective.
Collapse
Affiliation(s)
- James M Kasper
- Center for Addiction Research and Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Ashley E Smith
- Center for Addiction Research and Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Sierra N Miller
- Center for Addiction Research and Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Ara
- Center for Addiction Research and Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - William K Russell
- Department of Biochemistry and Molecular Biology, University of Texas Medical Branch, Galveston, TX, 77555, USA
| | - Kathryn A Cunningham
- Center for Addiction Research and Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| | - Jonathan D Hommel
- Center for Addiction Research and Department of Pharmacology and Toxicology, University of Texas Medical Branch, Galveston, TX, 77555, USA.
| |
Collapse
|
6
|
A Conditioned Place Preference for Heroin Is Signaled by Increased Dopamine and Direct Pathway Activity and Decreased Indirect Pathway Activity in the Nucleus Accumbens. J Neurosci 2022; 42:2011-2024. [PMID: 35031576 PMCID: PMC8916759 DOI: 10.1523/jneurosci.1451-21.2021] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 12/20/2021] [Accepted: 12/23/2021] [Indexed: 11/21/2022] Open
Abstract
Repeated pairing of a drug with a neutral stimulus, such as a cue or context, leads to the attribution of the drug's reinforcing properties to that stimulus, and exposure to that stimulus in the absence of the drug can elicit drug-seeking. A principal role for the NAc in the response to drug-associated stimuli has been well documented. Direct and indirect pathway medium spiny neurons (dMSNs and iMSNs) have been shown to bidirectionally regulate cue-induced heroin-seeking in rats expressing addiction-like phenotypes, and a shift in NAc activity toward the direct pathway has been shown in mice following cocaine conditioned place preference (CPP). However, how NAc signaling guides heroin CPP, and whether heroin alters the balance of signaling between dMSNs and iMSNs, remains unknown. Moreover, the role of NAc dopamine signaling in heroin reinforcement is unclear. Here, we integrate fiber photometry for in vivo monitoring of dopamine and dMSN/iMSN calcium activity with a heroin CPP procedure in rats to begin to address these questions. We identify a sensitization-like response to heroin in the NAc, with prominent iMSN activity during initial heroin exposure and prominent dMSN activity following repeated heroin exposure. We demonstrate a ramp in dopamine activity, dMSN activation, and iMSN inactivation preceding entry into a heroin-paired context, and a decrease in dopamine activity, dMSN inactivation, and iMSN activation preceding exit from a heroin-paired context. Finally, we show that buprenorphine is sufficient to prevent the development of heroin CPP and reduce Fos activation in the NAc after conditioning. Together, these data support the hypothesis that an imbalance in NAc activity contributes to the development of drug-cue associations that can drive addiction processes.SIGNIFICANCE STATEMENT The attribution of the reinforcing effects of drugs to neutral stimuli (e.g., cues and contexts) contributes to the long-standing nature of addiction, as re-exposure to drug-associated stimuli can reinstate drug-seeking and -taking even after long periods of abstinence. The NAc has an established role in encoding the value of drug-associated stimuli, and dopamine release into the NAc is known to modulate the reinforcing effects of drugs, including heroin. Using fiber photometry, we show that entering a heroin-paired context is driven by dopamine signaling and NAc direct pathway activation, whereas exiting a heroin-paired context is driven by NAc indirect pathway activation. This study provides further insight into the role of NAc microcircuitry in encoding the reinforcing properties of heroin.
Collapse
|
7
|
Wohlwend M, Laurila PP, Williams K, Romani M, Lima T, Pattawaran P, Benegiamo G, Salonen M, Schneider BL, Lahti J, Eriksson JG, Barrès R, Wisløff U, Moreira JBN, Auwerx J. The exercise-induced long noncoding RNA CYTOR promotes fast-twitch myogenesis in aging. Sci Transl Med 2021; 13:eabc7367. [PMID: 34878822 DOI: 10.1126/scitranslmed.abc7367] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Martin Wohlwend
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.,Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway.,Clinic of Cardiology, St. Olavs Hospital, Torgarden, NO-3250 Trondheim, Norway
| | - Pirkka-Pekka Laurila
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Kristine Williams
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Mario Romani
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Tanes Lima
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Pattamaprapanont Pattawaran
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Giorgia Benegiamo
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Minna Salonen
- Chronic Disease Prevention Unit, National Institute for Health and Welfare, FI-00271 Helsinki, Finland
| | - Bernard L Schneider
- Bertarelli Foundation Gene Therapy Platform, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1202 Geneva, Switzerland
| | - Jari Lahti
- Turku Institute for Advanced Studies, University of Turku, FI-20014 Turku, Finland.,Department of Psychology and Logopedics, University of Helsinki, FI-00014 Helsinki, Finland
| | - Johan G Eriksson
- Department of General Practice and Primary Health Care, University of Helsinki and Helsinki University Hospital, FI-00014 Helsinki, Finland.,Folkhälsan Research Center, University of Helsinki, FI-00014 Helsinki, Finland.,Department of Obstetrics and Gynecology, Yong Loo Lin School of Medicine, National University of Singapore, SG-119228 Singapore, Singapore.,Singapore Institute for Clinical Sciences (SICS), Agency for Science, Technology and Research, SG-117609 Singapore, Singapore
| | - Romain Barrès
- Novo Nordisk Foundation Center for Basic Metabolic Research, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2200 Copenhagen, Denmark
| | - Ulrik Wisløff
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - José B N Moreira
- Department of Circulation and Medical Imaging, Faculty of Medicine and Health Sciences, Norwegian University of Science and Technology (NTNU), NO-7491 Trondheim, Norway
| | - Johan Auwerx
- Laboratory of Integrative Systems Physiology, École Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| |
Collapse
|
8
|
Adeno-Associated Viral Vectors as Versatile Tools for Parkinson's Research, Both for Disease Modeling Purposes and for Therapeutic Uses. Int J Mol Sci 2021; 22:ijms22126389. [PMID: 34203739 PMCID: PMC8232322 DOI: 10.3390/ijms22126389] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/17/2022] Open
Abstract
It is without any doubt that precision medicine therapeutic strategies targeting neurodegenerative disorders are currently witnessing the spectacular rise of newly designed approaches based on the use of viral vectors as Trojan horses for the controlled release of a given genetic payload. Among the different types of viral vectors, adeno-associated viruses (AAVs) rank as the ones most commonly used for the purposes of either disease modeling or for therapeutic strategies. Here, we reviewed the current literature dealing with the use of AAVs within the field of Parkinson’s disease with the aim to provide neuroscientists with the advice and background required when facing a choice on which AAV might be best suited for addressing a given experimental challenge. Accordingly, here we will be summarizing some insights on different AAV serotypes, and which would be the most appropriate AAV delivery route. Next, the use of AAVs for modeling synucleinopathies is highlighted, providing potential readers with a landscape view of ongoing pre-clinical and clinical initiatives pushing forward AAV-based therapeutic approaches for Parkinson’s disease and related synucleinopathies.
Collapse
|
9
|
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
|
10
|
Caló L, Hidari E, Wegrzynowicz M, Dalley JW, Schneider BL, Podgajna M, Anichtchik O, Carlson E, Klenerman D, Spillantini MG. CSPα reduces aggregates and rescues striatal dopamine release in α-synuclein transgenic mice. Brain 2021; 144:1661-1669. [PMID: 33760024 PMCID: PMC8320296 DOI: 10.1093/brain/awab076] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 12/24/2020] [Accepted: 12/29/2020] [Indexed: 02/02/2023] Open
Abstract
α-Synuclein aggregation at the synapse is an early event in Parkinson's disease and is associated with impaired striatal synaptic function and dopaminergic neuronal death. The cysteine string protein (CSPα) and α-synuclein have partially overlapping roles in maintaining synaptic function and mutations in each cause neurodegenerative diseases. CSPα is a member of the DNAJ/HSP40 family of co-chaperones and like α-synuclein, chaperones the SNARE complex assembly and controls neurotransmitter release. α-Synuclein can rescue neurodegeneration in CSPαKO mice. However, whether α-synuclein aggregation alters CSPα expression and function is unknown. Here we show that α-synuclein aggregation at the synapse is associated with a decrease in synaptic CSPα and a reduction in the complexes that CSPα forms with HSC70 and STGa. We further show that viral delivery of CSPα rescues in vitro the impaired vesicle recycling in PC12 cells with α-synuclein aggregates and in vivo reduces synaptic α-synuclein aggregates increasing monomeric α-synuclein and restoring normal dopamine release in 1-120hαSyn mice. These novel findings reveal a mechanism by which α-synuclein aggregation alters CSPα at the synapse, and show that CSPα rescues α-synuclein aggregation-related phenotype in 1-120hαSyn mice similar to the effect of α-synuclein in CSPαKO mice. These results implicate CSPα as a potential therapeutic target for the treatment of early-stage Parkinson's disease.
Collapse
Affiliation(s)
- Laura Caló
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK,Dementia Research Institute, University of Cambridge, Cambridge, UK,Correspondence may also be addressed to: Dr Laura Caló E-mail:
| | - Eric Hidari
- Dementia Research Institute, University of Cambridge, Cambridge, UK,Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Michal Wegrzynowicz
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK,Laboratory of Molecular Basis of Neurodegeneration, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Jeffrey W Dalley
- Department of Psychology, University of Cambridge, Cambridge, UK,Department of Psychiatry, Hershel Smith Building for Brain and Mind Sciences, University of Cambridge, Cambridge, UK
| | - Bernard L Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland,Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1202 Geneva, Switzerland
| | - Martyna Podgajna
- Laboratory of Molecular Basis of Neurodegeneration, Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Oleg Anichtchik
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK
| | - Emma Carlson
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK
| | - David Klenerman
- Dementia Research Institute, University of Cambridge, Cambridge, UK,Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Maria Grazia Spillantini
- Department of Clinical Neurosciences, Clifford Allbutt Building, University of Cambridge, Cambridge, UK,Correspondence to: Prof. Maria Grazia Spillantini Department of Clinical Neurosciences Clifford Allbutt Building, Hills Road, Cambridge CB2 0AH, UK E-mail:
| |
Collapse
|
11
|
AMPA and NMDA Receptor Trafficking at Cocaine-Generated Synapses. J Neurosci 2021; 41:1996-2011. [PMID: 33436529 DOI: 10.1523/jneurosci.1918-20.2021] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Revised: 12/24/2020] [Accepted: 01/04/2021] [Indexed: 11/21/2022] Open
Abstract
Cocaine experience generates AMPA receptor (AMPAR)-silent synapses in the nucleus accumbens (NAc), which are thought to be new synaptic contacts enriched in GluN2B-containing NMDA receptors (NMDARs). After drug withdrawal, some of these synapses mature by recruiting AMPARs, strengthening the newly established synaptic transmission. Silent synapse generation and maturation are two consecutive cellular steps through which NAc circuits are profoundly remodeled to promote cue-induced cocaine seeking after drug withdrawal. However, the basic cellular processes that mediate these two critical steps remains underexplored. Using a combination of electrophysiology, viral-mediated gene transfer, and confocal imaging in male rats as well as knock-in (KI) mice of both sexes, our current study characterized the dynamic roles played by AMPARs and NMDARs in generation and maturation of silent synapses on NAc medium spiny neurons after cocaine self-administration and withdrawal. We report that cocaine-induced generation of silent synapses not only required synaptic insertion of GluN2B-containing NMDARs, but also, counterintuitively, involved insertion of AMPARs, which subsequently internalized, resulting in the AMPAR-silent state on withdrawal day 1. Furthermore, GluN2B NMDARs functioned to maintain these cocaine-generated synapses in the AMPAR-silent state during drug withdrawal, until they were replaced by nonGluN2B NMDARs, a switch that allowed AMPAR recruitment and maturation of silent synapses. These results reveal dynamic interactions between AMPARs and NMDARs during the generation and maturation of silent synapses after cocaine experience and provide a mechanistic basis through which new synaptic contacts and possibly new neural network patterns created by these synapses can be manipulated for therapeutic benefit.SIGNIFICANCE STATEMENT Studies over the past decade reveal a critical role of AMPA receptor-silent, NMDA receptor-containing synapses in forming cocaine-related memories that drive cocaine relapse. However, it remains incompletely understood how AMPA and NMDA receptors traffic at these synapses during their generation and maturation. The current study characterizes a two-step AMPA receptor trafficking cascade that contributes to the generation of silent synapses in response to cocaine experience, and a two-step NMDA receptor trafficking cascade that contributes to the maturation of these synapses after cocaine withdrawal. These results depict a highly regulated cellular procedure through which nascent glutamatergic synapses are generated in the adult brain after drug experience and provide significant insight into the roles of glutamate receptors in synapse formation and maturation.
Collapse
|
12
|
Deng P, Halmai J, Waldo JJ, Fink KD. Cell-Based Delivery Approaches for DNA-Binding Domains to the Central Nervous System. Curr Neuropharmacol 2021; 19:2125-2140. [PMID: 33998992 PMCID: PMC9185769 DOI: 10.2174/1570159x19666210517144044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 04/16/2021] [Accepted: 05/05/2021] [Indexed: 11/22/2022] Open
Abstract
Advancements in programmable DNA-Binding Proteins (DBDs) that target the genome, such as zinc fingers, transcription activator-like effectors, and Cas9, have broadened drug target design beyond traditional protein substrates. Effective delivery methodologies remain a major barrier in targeting the central nervous system. Currently, adeno-associated virus is the most wellvalidated delivery system for the delivery of DBDs towards the central nervous with multiple, ongoing clinical trials. While effective in transducing neuronal cells, viral delivery systems for DBDs remain problematic due to inherent viral packaging limits or immune responses that hinder translational potential. Direct administration of DBDs or encapsulation in lipid nanoparticles may provide alternative means towards delivering gene therapies into the central nervous system. This review will evaluate the strengths and limitations of current DBD delivery strategies in vivo. Furthermore, this review will discuss the use of adult stem cells as a putative delivery vehicle for DBDs and the potential advantages that these systems have over previous methodologies.
Collapse
Affiliation(s)
- Peter Deng
- Department of Neurology, Stem Cell Program and Gene Therapy Center, UC Davis Medical Center, Sacramento, CA, USA
| | - Julian Halmai
- Department of Neurology, Stem Cell Program and Gene Therapy Center, UC Davis Medical Center, Sacramento, CA, USA
| | - Jennifer J. Waldo
- Department of Neurology, Stem Cell Program and Gene Therapy Center, UC Davis Medical Center, Sacramento, CA, USA
| | - Kyle D. Fink
- Department of Neurology, Stem Cell Program and Gene Therapy Center, UC Davis Medical Center, Sacramento, CA, USA
| |
Collapse
|
13
|
Albaugh DL, Smith Y, Galvan A. Comparative analyses of transgene expression patterns after intra-striatal injections of rAAV2-retro in rats and rhesus monkeys: A light and electron microscopic study. Eur J Neurosci 2020; 52:4824-4839. [PMID: 33113247 PMCID: PMC7902345 DOI: 10.1111/ejn.15027] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Revised: 09/30/2020] [Accepted: 10/21/2020] [Indexed: 12/18/2022]
Abstract
Retrogradely-transducing viral vectors are versatile tools for anatomical and functional interrogations of neural circuits. These vectors can be applied in nonhuman primates (NHPs), powerful model species for neuroscientific studies with limited genetic tractability, but limited data are available regarding the tropism and transgene expression patterns of such viruses after injections in NHP brains. Consequently, NHP researchers must often rely on related data available from other species for experimental planning. To evaluate the suitability of rAAV2-retro in the NHP basal ganglia, we studied the transgene expression patterns at the light and electron microscope level after injections of rAAV2-retro vector encoding the opsin Jaws conjugated to a green fluorescent protein (GFP) in the putamen of rhesus macaques. For inter-species comparison, we injected the same vector in the rat dorsal striatum. In both species, GFP expression was observed in numerous cortical and subcortical regions with known striatal projections. However, important inter-species differences in pathway transduction were seen, including labeling of the intralaminar thalamostriatal projection in rats, but not monkeys. Electron microscopic ultrastructural observations within the basal ganglia revealed GFP labeling in both postsynaptic dendrites and presynaptic axonal terminals; the latter likely derived from anterograde transgene transport in neurons that project to the striatum, and from collaterals of these neurons. Our results suggest that certain neural pathways may be refractory to transduction by retrograde vectors in a species-specific manner, highlighting the need for caution when determining the suitability of a retrograde vector for NHP studies based solely on rodent data.
Collapse
Affiliation(s)
- Daniel L Albaugh
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Udall Center of Excellence for Parkinson's Disease Research, Atlanta, GA, USA
| | - Yoland Smith
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Udall Center of Excellence for Parkinson's Disease Research, Atlanta, GA, USA
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| | - Adriana Galvan
- Division of Neuropharmacology and Neurological Disorders, Yerkes National Primate Research Center, Emory University, Atlanta, GA, USA
- Udall Center of Excellence for Parkinson's Disease Research, Atlanta, GA, USA
- Department of Neurology, School of Medicine, Emory University, Atlanta, GA, USA
| |
Collapse
|
14
|
Sano H, Kobayashi K, Yoshioka N, Takebayashi H, Nambu A. Retrograde gene transfer into neural pathways mediated by adeno-associated virus (AAV)-AAV receptor interaction. J Neurosci Methods 2020; 345:108887. [DOI: 10.1016/j.jneumeth.2020.108887] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2020] [Revised: 07/17/2020] [Accepted: 07/27/2020] [Indexed: 12/21/2022]
|
15
|
Engmann AK, Bizzozzero F, Schneider MP, Pfyffer D, Imobersteg S, Schneider R, Hofer AS, Wieckhorst M, Schwab ME. The Gigantocellular Reticular Nucleus Plays a Significant Role in Locomotor Recovery after Incomplete Spinal Cord Injury. J Neurosci 2020; 40:8292-8305. [PMID: 32978289 PMCID: PMC7577599 DOI: 10.1523/jneurosci.0474-20.2020] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2020] [Revised: 09/14/2020] [Accepted: 09/18/2020] [Indexed: 11/21/2022] Open
Abstract
Traditionally, the brainstem has been seen as hardwired and poorly capable of plastic adaptations following spinal cord injury (SCI). Data acquired over the past decades, however, suggest differently: following SCI in various animal models (lamprey, chick, rodents, nonhuman primates), different forms of spontaneous anatomic plasticity of reticulospinal projections, many of them originating from the gigantocellular reticular nucleus (NRG), have been observed. In line with these anatomic observations, animals and humans with incomplete SCI often show various degrees of spontaneous motor recovery of hindlimb/leg function. Here, we investigated the functional relevance of two different modes of reticulospinal fiber growth after cervical hemisection, local rewiring of axotomized projections at the lesion site versus compensatory outgrowth of spared axons, using projection-specific, adeno-associated virus-mediated chemogenetic neuronal silencing. Detailed assessment of joint movements and limb kinetics during overground locomotion in female adult rats showed that locally rewired as well as compensatory NRG fibers were responsible for different aspects of recovered forelimb and hindlimb functions (i.e., stability, strength, coordination, speed, or timing). During walking and swimming, both locally rewired as well as compensatory NRG plasticity were crucial for recovered function, while the contribution of locally rewired NRG plasticity to wading performance was limited. Our data demonstrate comprehensively that locally rewired as well as compensatory plasticity of reticulospinal axons functionally contribute to the observed spontaneous improvement of stepping performance after incomplete SCI and are at least partially causative to the observed recovery of function, which can also be observed in human patients with spinal hemisection lesions.SIGNIFICANCE STATEMENT Following unilateral hemisection of the spinal cord, reticulospinal projections are destroyed on the injured side, resulting in impaired locomotion. Over time, a high degree of recovery can be observed in lesioned animals, like in human hemicord patients. In the rat, recovery is accompanied by pronounced spontaneous plasticity of axotomized and spared reticulospinal axons. We demonstrate the causative relevance of locally rewired as well as compensatory reticulospinal plasticity for the recovery of locomotor functions following spinal hemisection, using chemogenetic tools to selectively silence newly formed connections in behaviorally recovered animals. Moving from a correlative to a causative understanding of the role of neuroanatomical plasticity for functional recovery is fundamental for successful translation of treatment approaches from experimental studies to the clinics.
Collapse
Affiliation(s)
- Anne K Engmann
- Department of Health Sciences and Technology, ETH Zurich, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Flavio Bizzozzero
- Department of Health Sciences and Technology, ETH Zurich, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Marc P Schneider
- Department of Health Sciences and Technology, ETH Zurich, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Dario Pfyffer
- Department of Health Sciences and Technology, ETH Zurich, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Stefan Imobersteg
- Department of Health Sciences and Technology, ETH Zurich, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Regula Schneider
- Department of Health Sciences and Technology, ETH Zurich, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Anna-Sophie Hofer
- Department of Health Sciences and Technology, ETH Zurich, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Martin Wieckhorst
- Department of Health Sciences and Technology, ETH Zurich, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| | - Martin E Schwab
- Department of Health Sciences and Technology, ETH Zurich, Brain Research Institute, University of Zurich, 8057 Zurich, Switzerland
| |
Collapse
|
16
|
Lin K, Zhong X, Li L, Ying M, Yang T, Zhang Z, He X, Xu F. AAV9-Retro mediates efficient transduction with axon terminal absorption and blood-brain barrier transportation. Mol Brain 2020; 13:138. [PMID: 33054827 PMCID: PMC7556953 DOI: 10.1186/s13041-020-00679-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Accepted: 09/30/2020] [Indexed: 12/18/2022] Open
Abstract
Recombinant adeno-associated viruses (rAAVs), particularly those that permit efficient gene transfer to neurons from axonal terminals or across the blood-brain barrier, are useful vehicles for structural and functional studies of the neural circuit and for the treatment of many gene-deficient brain diseases that need to compensate for the correct genes in every cell in the whole brain. However, AAVs with these two advantages have not been reported. Here, we describe a new capsid engineering method, which exploits the combination of different capsids and aims to yield a capsid that can provide more alternative routes of administration that are more suitable for the wide-scale transduction of the central nervous system (CNS). A new AAV variant, AAV9-Retro, was developed by inserting the 10-mer peptide fragment from AAV2-Retro into the capsid of AAV9, and the biodistribution properties were evaluated in mice. By intracranial and intravenous injection in the mice, we found that AAV9-Retro can retrogradely infect projection neurons with an efficiency comparable to that of AAV2-Retro and retains the characteristic of AAV9, which can be transported across the nervous system. Our strategy provides a new tool for the manipulation of neural circuits and future preclinical and clinical treatment of some neurological and neurodegenerative disorders.
Collapse
Affiliation(s)
- Kunzhang Lin
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xin Zhong
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Lei Li
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Min Ying
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, PR China
| | - Tian Yang
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Zhijian Zhang
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
| | - Xiaobin He
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China.
- University of Chinese Academy of Sciences, Beijing, 100049, PR China.
| | - Fuqiang Xu
- Wuhan National Laboratory for Optoelectronics, Huazhong University of Science and Technology, Wuhan, 430074, China.
- Center for Brain Science, State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Wuhan Institute of Physics and Mathematics, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China.
- University of Chinese Academy of Sciences, Beijing, 100049, PR China.
- Shenzhen Key Lab of Neuropsychiatric Modulation, Guangdong Provincial Key Laboratory of Brain Connectome and Behavior, CAS Key Laboratory of Brain Connectome and Manipulation, The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen, Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, 518055, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
| |
Collapse
|
17
|
Medial Nucleus Accumbens Projections to the Ventral Tegmental Area Control Food Consumption. J Neurosci 2020; 40:4727-4738. [PMID: 32354856 DOI: 10.1523/jneurosci.3054-18.2020] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2018] [Revised: 04/15/2020] [Accepted: 04/22/2020] [Indexed: 12/12/2022] Open
Abstract
Decades of research have shown that the NAc is a critical region influencing addiction, mood, and food consumption through its effects on reinforcement learning, motivation, and hedonic experience. Pharmacological studies have demonstrated that inhibition of the NAc shell induces voracious feeding, leading to the hypothesis that the inhibitory projections that emerge from the NAc normally act to restrict feeding. While much of this work has focused on projections to the lateral hypothalamus, the role of NAc projections to the VTA in the control food intake has been largely unexplored. Using a retrograde viral labeling technique and real-time monitoring of neural activity with fiber photometry, we find that medial NAc shell projections to the VTA (mNAc→VTA) are inhibited during food-seeking and food consumption in male mice. We also demonstrate that this circuit bidirectionally controls feeding: optogenetic activation of NAc projections to the VTA inhibits food-seeking and food intake (in both sexes), while optogenetic inhibition of this circuit potentiates food-seeking behavior. Additionally, we show that activity of the NAc to VTA pathway is necessary for adaptive inhibition of food intake in response to external cues. These data provide new insight into NAc control over feeding in mice, and contribute to an emerging literature elucidating the role of inhibitory midbrain feedback within the mesolimbic circuit.SIGNIFICANCE STATEMENT The medial NAc has long been known to control consummatory behavior, with particular focus on accumbens projections to the lateral hypothalamus. Conversely, NAc projections to the VTA have mainly been studied in the context of drug reward. We show that NAc projections to the VTA bidirectionally control food intake, consistent with a permissive role in feeding. Additionally, we show that this circuit is normally inactivated during consumption and food-seeking. Together, these findings elucidate how mesolimbic circuits control food consumption.
Collapse
|
18
|
Jan A, Richner M, Vægter CB, Nyengaard JR, Jensen PH. Gene Transfer in Rodent Nervous Tissue Following Hindlimb Intramuscular Delivery of Recombinant Adeno-Associated Virus Serotypes AAV2/6, AAV2/8, and AAV2/9. Neurosci Insights 2019; 14:1179069519889022. [PMID: 32363345 PMCID: PMC7176396 DOI: 10.1177/1179069519889022] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Accepted: 10/15/2019] [Indexed: 01/15/2023] Open
Abstract
Recombinant adeno-associated virus (rAAV) vectors have emerged as the safe vehicles of choice for long-term gene transfer in mammalian nervous system. Recombinant adeno-associated virus–mediated localized gene transfer in adult nervous system following direct inoculation, that is, intracerebral or intrathecal, is well documented. However, recombinant adeno-associated virus delivery in defined neuronal populations in adult animals using less-invasive methods as well as avoiding ectopic gene expression following systemic inoculation remain challenging. Harnessing the capability of some recombinant adeno-associated virus serotypes for retrograde transduction may potentially address such limitations (Note: The term retrograde transduction in this manuscript refers to the uptake of injected recombinant adeno-associated virus particles at nerve terminals, retrograde transport, and subsequent transduction of nerve cell soma). In some studies, recombinant adeno-associated virus serotypes 2/6, 2/8, and 2/9 have been shown to exhibit transduction of connected neuroanatomical tracts in adult animals following lower limb intramuscular recombinant adeno-associated virus delivery in a pattern suggestive of retrograde transduction. However, an extensive side-by-side comparison of these serotypes following intramuscular delivery regarding tissue viral load, and the effect of promoter on transgene expression, has not been performed. Hence, we delivered recombinant adeno-associated virus serotypes 2/6, 2/8, or 2/9 encoding enhanced green fluorescent protein (eGFP), under the control of either cytomegalovirus (CMV) or human synapsin (hSyn) promoter, via a single unilateral hindlimb intramuscular injection in the bicep femoris of adult C57BL/6J mice. Four weeks post injection, we quantified viral load and transgene (enhanced green fluorescent protein) expression in muscle and related nervous tissues. Our data show that the select recombinant adeno-associated virus serotypes transduce sciatic nerve and groups of neurons in the dorsal root ganglia on the injected side, indicating that the intramuscular recombinant adeno-associated virus delivery is useful for achieving gene transfer in local neuroanatomical tracts. We also observed sparse recombinant adeno-associated virus viral delivery or eGFP transduction in lumbar spinal cord and a noticeable lack thereof in brain. Therefore, further improvements in recombinant adeno-associated virus design are warranted to achieve efficient widespread retrograde transduction following intramuscular and possibly other peripheral routes of delivery.
Collapse
Affiliation(s)
- Asad Jan
- Aarhus Institute of Advanced Studies and Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Mette Richner
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Christian B Vægter
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark
| | - Jens R Nyengaard
- Core Center for Molecular Morphology, Section for Stereology and Microscopy, Department of Clinical Medicine, and Centre for Stochastic Geometry and Advanced Bioimaging, Aarhus University, Aarhus, Denmark
| | - Poul H Jensen
- Danish Research Institute of Translational Neuroscience (DANDRITE), Department of Biomedicine, Aarhus University, Aarhus, Denmark
| |
Collapse
|
19
|
Williams ET, Glauser L, Tsika E, Jiang H, Islam S, Moore DJ. Parkin mediates the ubiquitination of VPS35 and modulates retromer-dependent endosomal sorting. Hum Mol Genet 2019; 27:3189-3205. [PMID: 29893854 DOI: 10.1093/hmg/ddy224] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/04/2018] [Indexed: 12/27/2022] Open
Abstract
Mutations in a number of genes cause familial forms of Parkinson's disease (PD), including mutations in the vacuolar protein sorting 35 ortholog (VPS35) and parkin genes. In this study, we identify a novel functional interaction between parkin and VPS35. We demonstrate that parkin interacts with and robustly ubiquitinates VPS35 in human neural cells. Familial parkin mutations are impaired in their ability to ubiquitinate VPS35. Parkin mediates the attachment of an atypical poly-ubiquitin chain to VPS35 with three lysine residues identified within the C-terminal region of VPS35 that are covalently modified by ubiquitin. Notably, parkin-mediated VPS35 ubiquitination does not promote the proteasomal degradation of VPS35. Furthermore, parkin does not influence the steady-state levels or turnover of VPS35 in neural cells and VPS35 levels are normal in the brains of parkin knockout mice. These data suggest that ubiquitination of VPS35 by parkin may instead serve a non-degradative cellular function potentially by regulating retromer-dependent sorting. Accordingly, we find that components of the retromer-associated WASH complex are markedly decreased in the brain of parkin knockout mice, suggesting that parkin may modulate WASH complex-dependent retromer sorting. Parkin gene silencing in primary cortical neurons selectively disrupts the vesicular sorting of the autophagy receptor ATG9A, a WASH-dependent retromer cargo. Parkin is not required for dopaminergic neurodegeneration induced by the expression of PD-linked D620N VPS35 in mice, consistent with VPS35 being located downstream of parkin function. Our data reveal a novel functional interaction of parkin with VPS35 that may be important for retromer-mediated endosomal sorting and PD.
Collapse
Affiliation(s)
- Erin T Williams
- Van Andel Institute Graduate School, Van Andel Research Institute, Grand Rapids, MI, USA.,Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Liliane Glauser
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland
| | - Elpida Tsika
- Brain Mind Institute, Swiss Federal Institute of Technology (EPFL), Lausanne, Switzerland.,AC Immune SA, EPFL Innovation Park, Lausanne, Switzerland
| | - Haisong Jiang
- Institute for Cell Engineering and Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Shariful Islam
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Darren J Moore
- Center for Neurodegenerative Science, Van Andel Research Institute, Grand Rapids, MI, USA
| |
Collapse
|
20
|
Hudry E, Vandenberghe LH. Therapeutic AAV Gene Transfer to the Nervous System: A Clinical Reality. Neuron 2019; 101:839-862. [PMID: 30844402 PMCID: PMC11804970 DOI: 10.1016/j.neuron.2019.02.017] [Citation(s) in RCA: 223] [Impact Index Per Article: 37.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 02/07/2019] [Accepted: 02/11/2019] [Indexed: 02/07/2023]
Abstract
Gene transfer has long been a compelling yet elusive therapeutic modality. First mainly considered for rare inherited disorders, gene therapy may open treatment opportunities for more challenging and complex diseases such as Alzheimer's or Parkinson's disease. Today, examples of striking clinical proof of concept, the first gene therapy drugs coming onto the market, and the emergence of powerful new molecular tools have broadened the number of avenues to target neurological disorders but have also highlighted safety concerns and technology gaps. The vector of choice for many nervous system targets currently is the adeno-associated viral (AAV) vector due to its desirable safety profile and strong neuronal tropism. In aggregate, the clinical success, the preclinical potential, and the technological innovation have made therapeutic AAV drug development a reality, particularly for nervous system disorders. Here, we discuss the rationale, opportunities, limitations, and progress in clinical AAV gene therapy.
Collapse
Affiliation(s)
- Eloise Hudry
- Department of Neurology, The Massachusetts General Hospital and NeuroDiscovery Center, Harvard Medical School, Boston, MA, USA.
| | - Luk H Vandenberghe
- Grousbeck Gene Therapy Center, Schepens Eye Research Institute and Massachusetts Eye and Ear Infirmary, Boston, MA, USA; Ocular Genomics Institute, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA; Harvard Program in Therapeutic Science, Harvard University, Cambridge, MA, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA, USA; The Broad Institute of Harvard and MIT, Cambridge, MA, USA.
| |
Collapse
|
21
|
|
22
|
D'Amico D, Mottis A, Potenza F, Sorrentino V, Li H, Romani M, Lemos V, Schoonjans K, Zamboni N, Knott G, Schneider BL, Auwerx J. The RNA-Binding Protein PUM2 Impairs Mitochondrial Dynamics and Mitophagy During Aging. Mol Cell 2019; 73:775-787.e10. [PMID: 30642763 DOI: 10.1016/j.molcel.2018.11.034] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2018] [Revised: 10/23/2018] [Accepted: 11/27/2018] [Indexed: 12/11/2022]
Abstract
Little information is available about how post-transcriptional mechanisms regulate the aging process. Here, we show that the RNA-binding protein Pumilio2 (PUM2), which is a translation repressor, is induced upon aging and acts as a negative regulator of lifespan and mitochondrial homeostasis. Multi-omics and cross-species analyses of PUM2 function show that it inhibits the translation of the mRNA encoding for the mitochondrial fission factor (Mff), thereby impairing mitochondrial fission and mitophagy. This mechanism is conserved in C. elegans by the PUM2 ortholog PUF-8. puf-8 knock-down in old nematodes and Pum2 CRISPR/Cas9-mediated knockout in the muscles of elderly mice enhances mitochondrial fission and mitophagy in both models, hence improving mitochondrial quality control and tissue homeostasis. Our data reveal how a PUM2-mediated layer of post-transcriptional regulation links altered Mff translation to mitochondrial dynamics and mitophagy, thereby mediating age-related mitochondrial dysfunctions.
Collapse
Affiliation(s)
- Davide D'Amico
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Adrienne Mottis
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Francesca Potenza
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Vincenzo Sorrentino
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Hao Li
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Mario Romani
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Vera Lemos
- Laboratory of Metabolic Signaling, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Kristina Schoonjans
- Laboratory of Metabolic Signaling, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne, CH-1015, Lausanne, Switzerland
| | - Nicola Zamboni
- Department of Biology, Institute of Molecular Systems Biology, Eidgenössische Technische Hochschule Zürich (ETHZ), CH-8093, Zurich, Switzerland
| | - Graham Knott
- BioEM Facility, Faculty of Life Sciences Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Bernard L Schneider
- Brain Mind Insitute, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory for Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), CH-1015 Lausanne, Switzerland.
| |
Collapse
|
23
|
Blessing D, Vachey G, Pythoud C, Rey M, Padrun V, Wurm FM, Schneider BL, Déglon N. Scalable Production of AAV Vectors in Orbitally Shaken HEK293 Cells. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2018; 13:14-26. [PMID: 30591923 PMCID: PMC6305802 DOI: 10.1016/j.omtm.2018.11.004] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Accepted: 11/16/2018] [Indexed: 01/30/2023]
Abstract
Adeno-associated virus (AAV) vectors are currently among the most commonly applied for in vivo gene therapy approaches. The evaluation of vectors during clinical development requires the production of considerable amounts of highly pure and potent vectors. Here, we set up a scalable process for AAV production, using orbitally shaken bioreactors and a fully characterized suspension-adapted cell line, HEKExpress. We conducted a proof-of-concept production of AAV2/8 and AAV2/9 vectors using HEKExpress cells. Furthermore, we compared the production of AAV2/9 vectors using this suspension cell line to classical protocols based on adherent HEK293 cells to demonstrate bioequivalence in vitro and in vivo. Following upstream processing, we purified vectors via gradient centrifugation and immunoaffinity chromatography. The in vitro characterization revealed differences due to the purification method, as well as the transfection protocol and the corresponding HEK293 cell line. The purification method and cell line used also affected in vivo transduction efficiency after bilateral injection of AAV2/9 vectors expressing a GFP reporter fused with a nuclear localization signal (AAV2/9-CBA-nlsGFP) into the striatum of adult mice. These results show that AAV vectors deriving from suspension HEKExpress cells are bioequivalent and may exhibit higher potency than vectors produced with adherent HEK293 cells.
Collapse
Affiliation(s)
- Daniel Blessing
- Department of Clinical Neurosciences, Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
- Neurosciences Research Center (CRN), Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Gabriel Vachey
- Department of Clinical Neurosciences, Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
- Neurosciences Research Center (CRN), Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Catherine Pythoud
- Department of Clinical Neurosciences, Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
- Neurosciences Research Center (CRN), Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Maria Rey
- Department of Clinical Neurosciences, Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
- Neurosciences Research Center (CRN), Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital, 1011 Lausanne, Switzerland
| | - Vivianne Padrun
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Florian M. Wurm
- ExcellGene SA, 1870 Monthey, Switzerland
- Faculty of Life Science, Ecole Polytechnique Fédérale de Lausanne (EFPL), 1015 Lausanne, Switzerland
| | - Bernard L. Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
- Corresponding author: Bernard Schneider, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| | - Nicole Déglon
- Department of Clinical Neurosciences, Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital (CHUV), 1011 Lausanne, Switzerland
- Neurosciences Research Center (CRN), Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital, 1011 Lausanne, Switzerland
- Corresponding author: Nicole Déglon, Department of Clinical Neurosciences, Laboratory of Neurotherapies and Neuromodulation (LNTM), Lausanne University Hospital (CHUV), Avenue de Beaumont, Pavillon 3, 1011 Lausanne, Switzerland.
| |
Collapse
|
24
|
Optogenetic Tractography for anatomo-functional characterization of cortico-subcortical neural circuits in non-human primates. Sci Rep 2018; 8:3362. [PMID: 29463867 PMCID: PMC5820256 DOI: 10.1038/s41598-018-21486-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2017] [Accepted: 01/29/2018] [Indexed: 11/08/2022] Open
Abstract
Dissecting neural circuitry in non-human primates (NHP) is crucial to identify potential neuromodulation anatomical targets for the treatment of pharmacoresistant neuropsychiatric diseases by electrical neuromodulation. How targets of deep brain stimulation (DBS) and cortical targets of transcranial magnetic stimulation (TMS) compare and might complement one another is an important question. Combining optogenetics and tractography may enable anatomo-functional characterization of large brain cortico-subcortical neural pathways. For the proof-of-concept this approach was used in the NHP brain to characterize the motor cortico-subthalamic pathway (m_CSP) which might be involved in DBS action mechanism in Parkinson’s disease (PD). Rabies-G-pseudotyped and Rabies-G-VSVg-pseudotyped EIAV lentiviral vectors encoding the opsin ChR2 gene were stereotaxically injected into the subthalamic nucleus (STN) and were retrogradely transported to the layer of the motor cortex projecting to STN. A precise anatomical mapping of this pathway was then performed using histology-guided high angular resolution MRI tractography guiding accurately cortical photostimulation of m_CSP origins. Photoexcitation of m_CSP axon terminals or m_CSP cortical origins modified the spikes distribution for photosensitive STN neurons firing rate in non-equivalent ways. Optogenetic tractography might help design preclinical neuromodulation studies in NHP models of neuropsychiatric disease choosing the most appropriate target for the tested hypothesis.
Collapse
|
25
|
Robins MT, Chiang T, Mores KL, Alongkronrusmee D, van Rijn RM. Critical Role for G i/o-Protein Activity in the Dorsal Striatum in the Reduction of Voluntary Alcohol Intake in C57Bl/6 Mice. Front Psychiatry 2018; 9:112. [PMID: 29686629 PMCID: PMC5900748 DOI: 10.3389/fpsyt.2018.00112] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/19/2018] [Indexed: 12/21/2022] Open
Abstract
The transition from non-dependent alcohol use to alcohol dependence involves increased activity of the dorsal striatum. Interestingly, the dorsal striatum expresses a large number of inhibitory G-protein-coupled receptors (GPCRs), which when activated may inhibit alcohol-induced increased activity and can decrease alcohol consumption. Here, we explore the hypothesis that dorsal striatal Gi/o-protein activation is sufficient to reduce voluntary alcohol intake. Using a voluntary, limited-access, two-bottle choice, drink-in-the-dark model of alcohol (10%) consumption, we validated the importance of Gi/o signaling in this region by locally expressing neuron-specific, adeno-associated-virus encoded Gi/o-coupled muscarinic M4 designer receptors exclusively activated by designer drugs (DREADD) in the dorsal striatum and observed a decrease in alcohol intake upon DREADD activation. We validated our findings by activating Gi/o-coupled delta-opioid receptors (DORs), which are natively expressed in the dorsal striatum, using either a G-protein biased agonist or a β-arrestin-biased agonist. Local infusion of TAN-67, an in vitro-determined Gi/o-protein biased DOR agonist, decreased voluntary alcohol intake in wild-type and β-arrestin-2 knockout (KO) mice. SNC80, a β-arrestin-2 biased DOR agonist, increased alcohol intake in wild-type mice; however, SNC80 decreased alcohol intake in β-arrestin-2 KO mice, thus resulting in a behavioral outcome generally observed for Gi/o-biased agonists and suggesting that β-arrestin recruitment is required for SNC80-increased alcohol intake. Overall, these results suggest that activation Gi/o-coupled GPCRs expressed in the dorsal striatum, such as the DOR, by G-protein biased agonists may be a potential strategy to decrease voluntary alcohol consumption and β-arrestin recruitment is to be avoided.
Collapse
Affiliation(s)
- Meridith T Robins
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue University Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States
| | - Terrance Chiang
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue University Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States
| | - Kendall L Mores
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States
| | - Doungkamol Alongkronrusmee
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue University Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States
| | - Richard M van Rijn
- Department of Medicinal Chemistry and Molecular Pharmacology, Purdue University, West Lafayette, IN, United States.,Purdue University Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
26
|
Lathuilière A, Valdés P, Papin S, Cacquevel M, Maclachlan C, Knott GW, Muhs A, Paganetti P, Schneider BL. Motifs in the tau protein that control binding to microtubules and aggregation determine pathological effects. Sci Rep 2017; 7:13556. [PMID: 29051562 PMCID: PMC5648870 DOI: 10.1038/s41598-017-13786-2] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 10/02/2017] [Indexed: 02/04/2023] Open
Abstract
Tau pathology is associated with cognitive decline in Alzheimer’s disease, and missense tau mutations cause frontotemporal dementia. Hyperphosphorylation and misfolding of tau are considered critical steps leading to tauopathies. Here, we determine how motifs controlling conformational changes in the microtubule-binding domain determine tau pathology in vivo. Human tau was overexpressed in the adult mouse forebrain to compare variants carrying residues that modulate tau propensity to acquire a β-sheet conformation. The P301S mutation linked to frontotemporal dementia causes tau aggregation and rapidly progressing motor deficits. By comparison, wild-type tau becomes heavily hyperphosphorylated, and induces behavioral impairments that do not progress over time. However, the behavioral defects caused by wild-type tau can be suppressed when β-sheet breaking proline residues are introduced in the microtubule-binding domain of tau. This modification facilitates tau interaction with microtubules, as shown by lower levels of phosphorylation, and by the enhanced protective effects of mutated tau against the severing of the cytoskeleton in neurons exposed to vinblastine. Altogether, motifs that are critical for tau conformation determine interaction with microtubules and subsequent pathological modifications, including phosphorylation and aggregation.
Collapse
Affiliation(s)
- Aurélien Lathuilière
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Pamela Valdés
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Stéphanie Papin
- AC Immune SA, Lausanne, Switzerland.,Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Via Tesserete 46, CH-6900, Lugano, Switzerland
| | - Matthias Cacquevel
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | | - Graham W Knott
- Centre of Interdisciplinary Electron Microscopy, EPFL, Switzerland
| | | | - Paolo Paganetti
- AC Immune SA, Lausanne, Switzerland.,Laboratory for Biomedical Neurosciences, Neurocenter of Southern Switzerland, Via Tesserete 46, CH-6900, Lugano, Switzerland
| | - Bernard L Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
27
|
Kobayashi K, Inoue KI, Tanabe S, Kato S, Takada M, Kobayashi K. Pseudotyped Lentiviral Vectors for Retrograde Gene Delivery into Target Brain Regions. Front Neuroanat 2017; 11:65. [PMID: 28824385 PMCID: PMC5539090 DOI: 10.3389/fnana.2017.00065] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 07/17/2017] [Indexed: 01/09/2023] Open
Abstract
Gene transfer through retrograde axonal transport of viral vectors offers a substantial advantage for analyzing roles of specific neuronal pathways or cell types forming complex neural networks. This genetic approach may also be useful in gene therapy trials by enabling delivery of transgenes into a target brain region distant from the injection site of the vectors. Pseudotyping of a lentiviral vector based on human immunodeficiency virus type 1 (HIV-1) with various fusion envelope glycoproteins composed of different combinations of rabies virus glycoprotein (RV-G) and vesicular stomatitis virus glycoprotein (VSV-G) enhances the efficiency of retrograde gene transfer in both rodent and nonhuman primate brains. The most recently developed lentiviral vector is a pseudotype with fusion glycoprotein type E (FuG-E), which demonstrates highly efficient retrograde gene transfer in the brain. The FuG-E–pseudotyped vector permits powerful experimental strategies for more precisely investigating the mechanisms underlying various brain functions. It also contributes to the development of new gene therapy approaches for neurodegenerative disorders, such as Parkinson’s disease, by delivering genes required for survival and protection into specific neuronal populations. In this review article, we report the properties of the FuG-E–pseudotyped vector, and we describe the application of the vector to neural circuit analysis and the potential use of the FuG-E vector in gene therapy for Parkinson’s disease.
Collapse
Affiliation(s)
- Kenta Kobayashi
- Section of Viral Vector Development, National Institute for Physiological SciencesOkazaki, Japan.,SOKENDAI (The Graduate University for Advanced Studies)Hayama, Japan
| | - Ken-Ichi Inoue
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto UniversityInuyama, Japan
| | - Soshi Tanabe
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto UniversityInuyama, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of MedicineFukushima, Japan
| | - Masahiko Takada
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, Kyoto UniversityInuyama, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of MedicineFukushima, Japan
| |
Collapse
|
28
|
Kramer PR, Stinson C, Umorin M, Deng M, Rao M, Bellinger LL, Yee MB, Kinchington PR. Lateral thalamic control of nociceptive response after whisker pad injection of varicella zoster virus. Neuroscience 2017; 356:207-216. [PMID: 28549561 DOI: 10.1016/j.neuroscience.2017.05.030] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2017] [Revised: 05/17/2017] [Accepted: 05/18/2017] [Indexed: 12/12/2022]
Abstract
Pain is a common complication of herpes zoster (HZ) infection which results from reactivation of a latent varicella zoster virus (VZV). A third of HZ patients' progress to a chronic pain state known as post herpetic neuralgia (PHN), and about a quarter of these patients' have orofacial pain. The mechanisms controlling the pain responses are not understood. Studies suggest central pathways involving the thalamus could control pain related to HZ, and studies in our lab suggest (VGAT) in the lateral thalamus influences orofacial pain. We hypothesized that thalamic VGAT functions, in part, to reduce pain, particularly orofacial pain, associated with VZV. To address this hypothesis VZV was injected into the whisker pad. Affective and motivational aspects of pain were measured using the Place Escape/Avoidance Paradigm. Thalamic neuronal activity was modulated after injecting an adeno-associated virus (AAV) expressing an engineered acetylcholine Gi-protein-coupled receptor. This receptor inhibits neuronal firing when bound by clozapine-n-oxide (CNO). VGAT expression was attenuated in the thalamus by injecting an AAV construct that expressed a VGAT silencing shRNA. VZV-induced nociception was significantly decreased after administering CNO in male rats. Nociception significantly increased concomitant with increased thalamic c-fos expression after attenuating thalamic VGAT expression. These data establish that the lateral thalamus (posterior, ventral posteromedial, ventral posterolateral and/or reticular thalamic nucleus) controls VZV-induced nociception in the orofacial region, and that GABA in this region appears to reduce the response to VZV-induced nociception possibly by gating facial pain input.
Collapse
Affiliation(s)
- Phillip R Kramer
- Texas A&M University College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, United States.
| | - Crystal Stinson
- Texas A&M University College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, United States
| | - Mikhail Umorin
- Texas A&M University College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, United States
| | - Mohong Deng
- Department of Oral and Maxillofacial Surgery, The State Key Laboratory Breeding Base of Basic Science of Stomatology & Key Laboratory of Oral Biomedicine Ministry of Education, School & Hospital of Stomatology, Wuhan University, Luoyu Road 237, Wuhan 430079, China
| | - Mahesh Rao
- Texas A&M University College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, United States
| | - Larry L Bellinger
- Texas A&M University College of Dentistry, 3302 Gaston Avenue, Dallas, TX 75246, United States
| | - Michael B Yee
- Department of Ophthalmology and of Microbiology and Molecular Genetics, University of Pittsburgh, 203 Lothrop Street, Pittsburgh, PA 15213, United States
| | - Paul R Kinchington
- Department of Ophthalmology and of Microbiology and Molecular Genetics, University of Pittsburgh, 203 Lothrop Street, Pittsburgh, PA 15213, United States.
| |
Collapse
|
29
|
Brys I, Nunes J, Fuentes R. Motor deficits and beta oscillations are dissociable in an alpha-synuclein model of Parkinson's disease. Eur J Neurosci 2017; 46:1906-1917. [DOI: 10.1111/ejn.13568] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Revised: 02/27/2017] [Accepted: 03/24/2017] [Indexed: 12/22/2022]
Affiliation(s)
- Ivani Brys
- Psychobiology Post Graduation Program; Federal University of Rio Grande do Norte; Natal Brazil
- Federal University of Vale do São Francisco; Petrolina Brazil
| | - Jessica Nunes
- Edmond e Lily Safra Institute of Neuroscience; Natal Brazil
- Physical Education Department; Potiguar University; Natal Brazil
| | - Romulo Fuentes
- Edmond e Lily Safra Institute of Neuroscience; Natal Brazil
- Biomedical Sciences Institute; Universidad de Chile; Santiago Chile
| |
Collapse
|
30
|
Pignataro D, Sucunza D, Vanrell L, Lopez-Franco E, Dopeso-Reyes IG, Vales A, Hommel M, Rico AJ, Lanciego JL, Gonzalez-Aseguinolaza G. Adeno-Associated Viral Vectors Serotype 8 for Cell-Specific Delivery of Therapeutic Genes in the Central Nervous System. Front Neuroanat 2017; 11:2. [PMID: 28239341 PMCID: PMC5301009 DOI: 10.3389/fnana.2017.00002] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2016] [Accepted: 01/13/2017] [Indexed: 12/19/2022] Open
Abstract
Adeno-associated viruses (AAVs) have become highly promising tools for research and clinical applications in the central nervous system (CNS). However, specific delivery of genes to the cell type of interest is essential for the success of gene therapy and therefore a correct selection of the promoter plays a very important role. Here, AAV8 vectors carrying enhanced green fluorescent protein (eGFP) as reporter gene under the transcriptional control of different CNS-specific promoters were used and compared with a strong ubiquitous promoter. Since one of the main limitations of AAV-mediated gene delivery lies in its restricted cloning capacity, we focused our work on small-sized promoters. We tested the transduction efficacy and specificity of each vector after stereotactic injection into the mouse striatum. Three glia-specific AAV vectors were generated using two truncated forms of the human promoter for glial fibrillar acidic protein (GFAP) as well as a truncated form of the murine GFAP promoter. All three vectors resulted in predominantly glial expression; however we also observed eGFP expression in other cell-types such as oligodendrocytes, but never in neurons. In addition, robust and neuron-specific eGFP expression was observed using the minimal promoters for the neural protein BM88 and the neuronal nicotinic receptor β2 (CHRNB2). In summary, we developed a set of AAV vectors designed for specific expression in cells of the CNS using minimal promoters to drive gene expression when the size of the therapeutic gene matters.
Collapse
Affiliation(s)
- Diego Pignataro
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical ResearchPamplona, Spain; Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain
| | - Diego Sucunza
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical ResearchPamplona, Spain; Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain
| | - Lucia Vanrell
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research Pamplona, Spain
| | | | - Iria G Dopeso-Reyes
- Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| | - Africa Vales
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical Research Pamplona, Spain
| | - Mirja Hommel
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical ResearchPamplona, Spain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| | - Alberto J Rico
- Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| | - Jose L Lanciego
- Department of Neurosciences, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| | - Gloria Gonzalez-Aseguinolaza
- Department of Gene Therapy and Regulation of Gene Expression, Center for Applied Medical ResearchPamplona, Spain; Centro de Investigación Biomédica en Red sobre Enfermedades NeurodegenerativasSpain; Instituto de Investigación Sanitaria de NavarraPamplona, Spain
| |
Collapse
|
31
|
Albert K, Voutilainen MH, Domanskyi A, Airavaara M. AAV Vector-Mediated Gene Delivery to Substantia Nigra Dopamine Neurons: Implications for Gene Therapy and Disease Models. Genes (Basel) 2017; 8:genes8020063. [PMID: 28208742 PMCID: PMC5333052 DOI: 10.3390/genes8020063] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2016] [Accepted: 02/03/2017] [Indexed: 12/21/2022] Open
Abstract
Gene delivery using adeno-associated virus (AAV) vectors is a widely used method to transduce neurons in the brain, especially due to its safety, efficacy, and long-lasting expression. In addition, by varying AAV serotype, promotor, and titer, it is possible to affect the cell specificity of expression or the expression levels of the protein of interest. Dopamine neurons in the substantia nigra projecting to the striatum, comprising the nigrostriatal pathway, are involved in movement control and degenerate in Parkinson’s disease. AAV-based gene targeting to the projection area of these neurons in the striatum has been studied extensively to induce the production of neurotrophic factors for disease-modifying therapies for Parkinson’s disease. Much less emphasis has been put on AAV-based gene therapy targeting dopamine neurons in substantia nigra. We will review the literature related to targeting striatum and/or substantia nigra dopamine neurons using AAVs in order to express neuroprotective and neurorestorative molecules, as well as produce animal disease models of Parkinson’s disease. We discuss difficulties in targeting substantia nigra dopamine neurons and their vulnerability to stress in general. Therefore, choosing a proper control for experimental work is not trivial. Since the axons along the nigrostriatal tract are the first to degenerate in Parkinson’s disease, the location to deliver the therapy must be carefully considered. We also review studies using AAV-α-synuclein (α-syn) to target substantia nigra dopamine neurons to produce an α-syn overexpression disease model in rats. Though these studies are able to produce mild dopamine system degeneration in the striatum and substantia nigra and some behavioural effects, there are studies pointing to the toxicity of AAV-carrying green fluorescent protein (GFP), which is often used as a control. Therefore, we discuss the potential difficulties in overexpressing proteins in general in the substantia nigra.
Collapse
Affiliation(s)
- Katrina Albert
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
| | - Merja H Voutilainen
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
| | - Andrii Domanskyi
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
| | - Mikko Airavaara
- Institute of Biotechnology, University of Helsinki, Helsinki 00014, Finland.
| |
Collapse
|
32
|
Kobayashi K, Kato S, Kobayashi K. Genetic manipulation of specific neural circuits by use of a viral vector system. J Neural Transm (Vienna) 2017; 125:67-75. [DOI: 10.1007/s00702-016-1674-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2016] [Accepted: 12/30/2016] [Indexed: 01/05/2023]
|
33
|
Valdés P, Schneider BL. Gene Therapy: A Promising Approach for Neuroprotection in Parkinson's Disease? Front Neuroanat 2016; 10:123. [PMID: 28066194 PMCID: PMC5168434 DOI: 10.3389/fnana.2016.00123] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/06/2016] [Indexed: 01/22/2023] Open
Abstract
With the development of effective systems for gene delivery to the central nervous system (CNS), gene therapy has become a therapeutic option for the treatment of Parkinson’s disease (PD). Gene therapies that are the most advanced in the clinic have been designed to more effectively compensate for the lack of dopamine signaling in the basal ganglia and rescue the cardinal motor symptoms of PD. However, it remains essential to devise novel therapies to prevent neurodegeneration and disease progression. Since gene therapy has been initially proposed for the delivery of neurotrophins to support the survival and function of dopaminergic neurons, our understanding of PD etiology has changed dramatically. Genes implicated in familial forms of the disease and genetic risk factors associated with sporadic PD have been identified. The spreading of the α-synuclein pathology, as well as perturbations of the lysosomal and mitochondrial activities, appear to play critical roles in the pathogenesis. These findings provide novel targets for gene therapy against PD, but at the same time underline the complexity of this chronic disease. Here we review and discuss the successes and limitations of gene therapy approaches, which have been proposed to provide neuroprotection in PD.
Collapse
Affiliation(s)
- Pamela Valdés
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne, Switzerland
| | - Bernard L Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) Lausanne, Switzerland
| |
Collapse
|
34
|
Murlidharan G, Crowther A, Reardon RA, Song J, Asokan A. Glymphatic fluid transport controls paravascular clearance of AAV vectors from the brain. JCI Insight 2016; 1:e88034. [PMID: 27699236 DOI: 10.1172/jci.insight.88034] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Adeno-associated viruses (AAV) are currently being evaluated in clinical trials for gene therapy of CNS disorders. However, host factors that influence the spread, clearance, and transduction efficiency of AAV vectors in the brain are not well understood. Recent studies have demonstrated that fluid flow mediated by aquaporin-4 (AQP4) channels located on astroglial end feet is essential for exchange of solutes between interstitial and cerebrospinal fluid. This phenomenon, which is essential for interstitial clearance of solutes from the CNS, has been termed glial-associated lymphatic transport or glymphatic transport. In the current study, we demonstrate that glymphatic transport profoundly affects various aspects of AAV gene transfer in the CNS. Altered localization of AQP4 in aged mouse brains correlated with significantly increased retention of AAV vectors in the parenchyma and reduced systemic leakage following ventricular administration. We observed a similar increase in AAV retention and transgene expression upon i.c.v. administration in AQP4-/- mice. Consistent with this observation, fluorophore-labeled AAV vectors showed markedly reduced flux from the ventricles of AQP4-/- mice compared with WT mice. These results were further corroborated by reduced AAV clearance from the AQP4-null brain, as demonstrated by reduced transgene expression and vector genome accumulation in systemic organs. We postulate that deregulation of glymphatic transport in aged and diseased brains could markedly affect the parenchymal spread, clearance, and gene transfer efficiency of AAV vectors. Assessment of biomarkers that report the kinetics of CSF flux in prospective gene therapy patients might inform variable treatment outcomes and guide future clinical trial design.
Collapse
Affiliation(s)
| | - Andrew Crowther
- Neurobiology Curriculum.,University of North Carolina Neuroscience Center
| | | | - Juan Song
- Department of Pharmacology.,University of North Carolina Neuroscience Center
| | - Aravind Asokan
- Gene Therapy Center.,Department of Genetics, and.,Department of Biochemistry and Biophysics, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
| |
Collapse
|
35
|
Blessing D, Déglon N. Adeno-associated virus and lentivirus vectors: a refined toolkit for the central nervous system. Curr Opin Virol 2016; 21:61-66. [PMID: 27559630 DOI: 10.1016/j.coviro.2016.08.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 08/05/2016] [Accepted: 08/08/2016] [Indexed: 12/20/2022]
Abstract
The last two decades have witnessed the increasing instrumentalization of viruses, which have progressively evolved into highly potent gene transfer vehicles for a wide spectrum of applications. In the context of the central nervous system (CNS), their unique gene delivery features and targeting specificities have been exploited not only to improve our understanding of basic neurobiology, but also to investigate diseases or deliver therapeutic candidates. As a result, we have started moving away from the opportunistic use of recombinant vectors that are derived from naturally existing viruses toward the rational engineering of tailored lentivirus (LV) and adeno-associated virus (AAV) vectors for specific use in the CNS.
Collapse
Affiliation(s)
- Daniel Blessing
- Laboratory of Cellular and Molecular Neurotherapies (LCMN), Department of Clinical Neurosciences (DNC), Lausanne University Hospital (CHUV), Lausanne, Switzerland; Laboratory of Cellular and Molecular Neurotherapies (LCMN), Neuroscience Research Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland
| | - Nicole Déglon
- Laboratory of Cellular and Molecular Neurotherapies (LCMN), Department of Clinical Neurosciences (DNC), Lausanne University Hospital (CHUV), Lausanne, Switzerland; Laboratory of Cellular and Molecular Neurotherapies (LCMN), Neuroscience Research Center, Lausanne University Hospital (CHUV), Lausanne, Switzerland.
| |
Collapse
|
36
|
Abstract
Unraveling the complex network of neural circuits that form the nervous system demands tools that can manipulate specific circuits. The recent evolution of genetic tools to target neural circuits allows an unprecedented precision in elucidating their function. Here we describe two general approaches for achieving circuit specificity. The first uses the genetic identity of a cell, such as a transcription factor unique to a circuit, to drive expression of a molecule that can manipulate cell function. The second uses the spatial connectivity of a circuit to achieve specificity: one genetic element is introduced at the origin of a circuit and the other at its termination. When the two genetic elements combine within a neuron, they can alter its function. These two general approaches can be combined to allow manipulation of neurons with a specific genetic identity by introducing a regulatory gene into the origin or termination of the circuit. We consider the advantages and disadvantages of both these general approaches with regard to specificity and efficacy of the manipulations. We also review the genetic techniques that allow gain- and loss-of-function within specific neural circuits. These approaches introduce light-sensitive channels (optogenetic) or drug sensitive channels (chemogenetic) into neurons that form specific circuits. We compare these tools with others developed for circuit-specific manipulation and describe the advantages of each. Finally, we discuss how these tools might be applied for identification of the neural circuits that mediate behavior and for repair of neural connections.
Collapse
Affiliation(s)
- Hong Geun Park
- Burke Medical Research Institute, White Plains, NY, USA.
| | - Jason B Carmel
- Burke Medical Research Institute, White Plains, NY, USA
- Brain and Mind Research Institute and Departments of Neurology and Pediatrics, Weill Cornell Medical College, New York, NY, USA
| |
Collapse
|
37
|
Brys I, Bobela W, Schneider BL, Aebischer P, Fuentes R. Spinal cord stimulation improves forelimb use in an alpha-synuclein animal model of Parkinson's disease. Int J Neurosci 2016; 127:28-36. [PMID: 26856727 DOI: 10.3109/00207454.2016.1138296] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Neuromodulation by spinal cord stimulation has been proposed as a symptomatic treatment for Parkinson's disease. We tested the chronic effects of spinal cord stimulation in a progressive model of Parkinson's based on overexpression of alpha-synuclein in the substantia nigra. Adult Sprague Dawley rats received unilateral injections of adeno-associated virus serotype 6 (AAV6) in the substantia nigra to express alpha-synuclein. Locomotion and forepaw use of the rats were evaluated during the next 10 weeks. Starting on week 6, a group of AAV6-injected rats received spinal cord stimulation once a week. At the end of the experiment, tyrosine hydroxylase and alpha-synuclein immunostaining were performed. Rats with unilateral alpha-synuclein expression showed a significant decrease in the use of the contralateral forepaw, which was mildly but significantly reverted by spinal cord stimulation applied once a week from the 6th to the 10th week after the AAV6 injection. Long-term spinal cord stimulation proved to be effective to suppress or delay motor symptoms in a sustained and progressive model of Parkinson's and might become an alternative, less invasive neuromodulation option to treat this disease.
Collapse
Affiliation(s)
- Ivani Brys
- a Department of Psychobiology , Federal University of Rio Grande do Norte , Natal , Brazil.,b Edmond and Lily Safra Institute of Neuroscience of Natal , 590660 , Brazil
| | - Wojciech Bobela
- c Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) , Lausanne , Switzerland
| | - Bernard L Schneider
- c Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) , Lausanne , Switzerland
| | - Patrick Aebischer
- c Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL) , Lausanne , Switzerland
| | - Romulo Fuentes
- b Edmond and Lily Safra Institute of Neuroscience of Natal , 590660 , Brazil .,d Facultad de Medicina, Programa de Fisiología y Biofísica, Instituto de Ciencias Biomédicas , Universidad de Chile , Santiago , Chile
| |
Collapse
|
38
|
Cook-Snyder DR, Jones A, Reijmers LG. A retrograde adeno-associated virus for collecting ribosome-bound mRNA from anatomically defined projection neurons. Front Mol Neurosci 2015; 8:56. [PMID: 26557053 PMCID: PMC4617378 DOI: 10.3389/fnmol.2015.00056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Accepted: 09/07/2015] [Indexed: 01/23/2023] Open
Abstract
The brain contains a large variety of projection neurons with different functional properties. The functional properties of projection neurons arise from their connectivity with other neurons and their molecular composition. We describe a novel tool for obtaining the gene expression profiles of projection neurons that are anatomically defined by the location of their soma and axon terminals. Our tool utilizes adeno-associated virus serotype 9 (AAV9), which we found to retrogradely transduce projection neurons after injection at the site of the axon terminals. We used AAV9 to express Enhanced Green Fluorescent Protein (EGFP)-tagged ribosomal protein L10a (EGFP-L10a), which enables the immunoprecipitation of EGFP-tagged ribosomes and associated mRNA with a method known as Translating Ribosome Affinity Purification (TRAP). To achieve high expression of the EGFP-L10a protein in projection neurons, we placed its expression under control of a 1.3 kb alpha-calcium/calmodulin-dependent protein kinase II (Camk2a) promoter. We injected the AAV9-Camk2a-TRAP virus in either the hippocampus or the bed nucleus of the stria terminalis (BNST) of the mouse brain. In both brain regions the 1.3 kb Camk2a promoter did not confer complete cell-type specificity around the site of injection, as EGFP-L10a expression was observed in Camk2a-expressing neurons as well as in neuronal and non-neuronal cells that did not express Camk2a. In contrast, cell-type specific expression was observed in Camk2a-positive projection neurons that were retrogradely transduced by AAV9-Camk2a-TRAP. Injection of AAV9-Camk2a-TRAP into the BNST enabled the use of TRAP to collect ribosome-bound mRNA from basal amygdala projection neurons that innervate the BNST. AAV9-Camk2a-TRAP provides a single-virus system that can be used for the molecular profiling of anatomically defined projection neurons in mice and other mammalian model organisms. In addition, AAV9-Camk2a-TRAP may enable the discovery of protein synthesis events that support information storage in projection neurons.
Collapse
Affiliation(s)
- Denise R Cook-Snyder
- Department of Neuroscience, School of Medicine, Tufts University Boston, MA, USA ; Department of Biology and Neuroscience Program, Carthage College Kenosha, WI, USA
| | - Alexander Jones
- Department of Neuroscience, School of Medicine, Tufts University Boston, MA, USA ; Graduate Program in Neuroscience, Sackler School of Graduate Biomedical Sciences, Tufts University Boston, MA, USA
| | - Leon G Reijmers
- Department of Neuroscience, School of Medicine, Tufts University Boston, MA, USA
| |
Collapse
|
39
|
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: 31] [Impact Index Per Article: 3.1] [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
|
40
|
Ciron C, Zheng L, Bobela W, Knott GW, Leone TC, Kelly DP, Schneider BL. PGC-1α activity in nigral dopamine neurons determines vulnerability to α-synuclein. Acta Neuropathol Commun 2015; 3:16. [PMID: 25853296 PMCID: PMC4379693 DOI: 10.1186/s40478-015-0200-8] [Citation(s) in RCA: 74] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2015] [Accepted: 03/06/2015] [Indexed: 12/12/2022] Open
Abstract
Introduction Mitochondrial dysfunction and oxidative stress are critical factors in the pathogenesis of age-dependent neurodegenerative diseases. PGC-1α, a master regulator of mitochondrial biogenesis and cellular antioxidant defense, has emerged as a possible therapeutic target for Parkinson’s disease, with important roles in the function and survival of dopaminergic neurons in the substantia nigra. The objective of this study is to determine if the loss of PGC-1α activity contributes to α-synuclein-induced degeneration. Results We explore the vulnerability of PGC-1α null mice to the accumulation of human α-synuclein in nigral neurons, and assess the neuroprotective effect of AAV-mediated PGC-1α expression in this experimental model. Using neuronal cultures derived from these mice, mitochondrial respiration and production of reactive oxygen species are assessed in conditions of human α-synuclein overexpression. We find ultrastructural evidence for abnormal mitochondria and fragmented endoplasmic reticulum in the nigral dopaminergic neurons of PGC-1α null mice. Furthermore, PGC-1α null nigral neurons are more prone to degenerate following overexpression of human α-synuclein, an effect more apparent in male mice. PGC-1α overexpression restores mitochondrial morphology, oxidative stress detoxification and basal respiration, which is consistent with the observed neuroprotection against α-synuclein toxicity in male PGC-1α null mice. Conclusions Altogether, our results highlight an important role for PGC-1α in controlling the mitochondrial function of nigral neurons accumulating α-synuclein, which may be critical for gender-dependent vulnerability to Parkinson’s disease. Electronic supplementary material The online version of this article (doi:10.1186/s40478-015-0200-8) contains supplementary material, which is available to authorized users.
Collapse
|
41
|
Murlidharan G, Samulski RJ, Asokan A. Biology of adeno-associated viral vectors in the central nervous system. Front Mol Neurosci 2014; 7:76. [PMID: 25285067 PMCID: PMC4168676 DOI: 10.3389/fnmol.2014.00076] [Citation(s) in RCA: 123] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 09/04/2014] [Indexed: 01/11/2023] Open
Abstract
Gene therapy is a promising approach for treating a spectrum of neurological and neurodegenerative disorders by delivering corrective genes to the central nervous system (CNS). In particular, adeno-associated viruses (AAVs) have emerged as promising tools for clinical gene transfer in a broad range of genetic disorders with neurological manifestations. In the current review, we have attempted to bridge our understanding of the biology of different AAV strains with their transduction profiles, cellular tropisms, and transport mechanisms within the CNS. Continued efforts to dissect AAV-host interactions within the brain are likely to aid in the development of improved vectors for CNS-directed gene transfer applications in the clinic.
Collapse
Affiliation(s)
- Giridhar Murlidharan
- Curriculum in Genetics and Molecular Biology, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA ; Gene Therapy Center, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| | - Richard J Samulski
- Gene Therapy Center, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA ; Department of Pharmacology, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill NC, USA
| | - Aravind Asokan
- Gene Therapy Center, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA ; Department of Genetics and Department of Biochemistry and Biophysics, School of Medicine, University of North Carolina at Chapel Hill Chapel Hill, NC, USA
| |
Collapse
|
42
|
Mosser S, Alattia JR, Dimitrov M, Matz A, Pascual J, Schneider BL, Fraering PC. The adipocyte differentiation protein APMAP is an endogenous suppressor of Aβ production in the brain. Hum Mol Genet 2014; 24:371-82. [PMID: 25180020 PMCID: PMC4275069 DOI: 10.1093/hmg/ddu449] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The deposition of amyloid-beta (Aβ) aggregates in the brain is a major pathological hallmark of Alzheimer's disease (AD). Aβ is generated from the cleavage of C-terminal fragments of the amyloid precursor protein (APP-CTFs) by γ-secretase, an intramembrane-cleaving protease with multiple substrates, including the Notch receptors. Endogenous modulation of γ-secretase is pointed to be implicated in the sporadic, age-dependent form of AD. Moreover, specifically modulating Aβ production has become a priority for the safe treatment of AD because the inhibition of γ-secretase results in adverse effects that are related to impaired Notch cleavage. Here, we report the identification of the adipocyte differentiation protein APMAP as a novel endogenous suppressor of Aβ generation. We found that APMAP interacts physically with γ-secretase and its substrate APP. In cells, the partial depletion of APMAP drastically increased the levels of APP-CTFs, as well as uniquely affecting their stability, with the consequence being increased secretion of Aβ. In wild-type and APP/ presenilin 1 transgenic mice, partial adeno-associated virus-mediated APMAP knockdown in the hippocampus increased Aβ production by ∼20 and ∼55%, respectively. Together, our data demonstrate that APMAP is a negative regulator of Aβ production through its interaction with APP and γ-secretase. All observed APMAP phenotypes can be explained by an impaired degradation of APP-CTFs, likely caused by an altered substrate transport capacity to the lysosomal/autophagic system.
Collapse
Affiliation(s)
- Sebastien Mosser
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH1015, Switzerland
| | - Jean-René Alattia
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH1015, Switzerland
| | - Mitko Dimitrov
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH1015, Switzerland
| | - Alexandre Matz
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH1015, Switzerland
| | - Justine Pascual
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH1015, Switzerland
| | - Bernard L Schneider
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH1015, Switzerland
| | - Patrick C Fraering
- Brain Mind Institute and School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne CH1015, Switzerland
| |
Collapse
|
43
|
Tsika E, Glauser L, Moser R, Fiser A, Daniel G, Sheerin UM, Lees A, Troncoso JC, Lewis PA, Bandopadhyay R, Schneider BL, Moore DJ. Parkinson's disease-linked mutations in VPS35 induce dopaminergic neurodegeneration. Hum Mol Genet 2014; 23:4621-38. [PMID: 24740878 PMCID: PMC4119414 DOI: 10.1093/hmg/ddu178] [Citation(s) in RCA: 117] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Mutations in the vacuolar protein sorting 35 homolog (VPS35) gene at the PARK17 locus, encoding a key component of the retromer complex, were recently identified as a new cause of late-onset, autosomal dominant Parkinson's disease (PD). Here we explore the pathogenic consequences of PD-associated mutations in VPS35 using a number of model systems. VPS35 exhibits a broad neuronal distribution throughout the rodent brain, including within the nigrostriatal dopaminergic pathway. In the human brain, VPS35 protein levels and distribution are similar in tissues from control and PD subjects, and VPS35 is not associated with Lewy body pathology. The common D620N missense mutation in VPS35 does not compromise its protein stability or localization to endosomal and lysosomal vesicles, or the vesicular sorting of the retromer cargo, sortilin, SorLA and cation-independent mannose 6-phosphate receptor, in rodent primary neurons or patient-derived human fibroblasts. In yeast we show that PD-linked VPS35 mutations are functional and can normally complement VPS35 null phenotypes suggesting that they do not result in a loss-of-function. In rat primary cortical cultures the overexpression of human VPS35 induces neuronal cell death and increases neuronal vulnerability to PD-relevant cellular stress. In a novel viral-mediated gene transfer rat model, the expression of D620N VPS35 induces the marked degeneration of substantia nigra dopaminergic neurons and axonal pathology, a cardinal pathological hallmark of PD. Collectively, these studies establish that dominant VPS35 mutations lead to neurodegeneration in PD consistent with a gain-of-function mechanism, and support a key role for VPS35 in the development of PD.
Collapse
Affiliation(s)
- Elpida Tsika
- Laboratory of Molecular Neurodegenerative Research
| | | | - Roger Moser
- Laboratory of Molecular Neurodegenerative Research
| | - Aris Fiser
- Laboratory of Molecular Neurodegenerative Research
| | | | | | - Andrew Lees
- Queen Square Brain Bank for Neurological Disorders, University College London Institute of Neurology, London WC1N 3BG, UK
| | - Juan C Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Patrick A Lewis
- Department of Molecular Neuroscience School of Pharmacy, University of Reading, Reading RG6 6AP, UK
| | - Rina Bandopadhyay
- Reta Lila Weston Institute of Neurological Studies, University College London Institute of Neurology, London WC1N 1PJ, UK
| | - Bernard L Schneider
- Neurodegenerative Disease Laboratory, Brain Mind Institute, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Darren J Moore
- Laboratory of Molecular Neurodegenerative Research Center for Neurodegenerative Science, Van Andel Institute, Grand Rapids, MI 49503, USA
| |
Collapse
|