1
|
Mueller SA, Oler JA, Roseboom PH, Aggarwal N, Kenwood MM, Riedel MK, Elam VR, Olsen ME, DiFilippo AH, Christian BT, Hu X, Galvan A, Boehm MA, Michaelides M, Kalin NH. DREADD-mediated amygdala activation is sufficient to induce anxiety-like responses in young nonhuman primates. CURRENT RESEARCH IN NEUROBIOLOGY 2023; 5:100111. [PMID: 38020807 PMCID: PMC10663133 DOI: 10.1016/j.crneur.2023.100111] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 09/11/2023] [Accepted: 09/25/2023] [Indexed: 12/01/2023] Open
Abstract
Anxiety disorders are among the most prevalent psychiatric disorders, with symptoms often beginning early in life. To model the pathophysiology of human pathological anxiety, we utilized Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) in a nonhuman primate model of anxious temperament to selectively increase neuronal activity of the amygdala. Subjects included 10 young rhesus macaques; 5 received bilateral infusions of AAV5-hSyn-HA-hM3Dq into the dorsal amygdala, and 5 served as controls. Subjects underwent behavioral testing in the human intruder paradigm following clozapine or vehicle administration, prior to and following surgery. Behavioral results indicated that clozapine treatment post-surgery increased freezing across different threat-related contexts in hM3Dq subjects. This effect was again observed approximately 1.9 years following surgery, indicating the long-term functional capacity of DREADD-induced neuronal activation. [11C]deschloroclozapine PET imaging demonstrated amygdala hM3Dq-HA specific binding, and immunohistochemistry revealed that hM3Dq-HA expression was most prominent in basolateral nuclei. Electron microscopy confirmed expression was predominantly on neuronal membranes. Together, these data demonstrate that activation of primate amygdala neurons is sufficient to induce increased anxiety-related behaviors, which could serve as a model to investigate pathological anxiety in humans.
Collapse
Affiliation(s)
- Sascha A.L. Mueller
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Jonathan A. Oler
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Patrick H. Roseboom
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Nakul Aggarwal
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Margaux M. Kenwood
- Department of Psychiatry, Weill Cornell Medical College, New York, NY, 10065, USA
| | - Marissa K. Riedel
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Victoria R. Elam
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Miles E. Olsen
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| | - Alexandra H. DiFilippo
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Bradley T. Christian
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53705, USA
| | - Xing Hu
- Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Adriana Galvan
- Emory National Primate Research Center, Emory University, Atlanta, GA, 30329, USA
| | - Matthew A. Boehm
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Ned H. Kalin
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI, 53719, USA
| |
Collapse
|
2
|
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
|
3
|
Mueller SAL, Oler JA, Roseboom PH, Aggarwal N, Kenwood MM, Riedel MK, Elam VR, Olsen ME, DiFilippo AH, Christian BT, Hu X, Galvan A, Boehm MA, Michaelides M, Kalin NH. DREADD-mediated amygdala activation is sufficient to induce anxiety-like responses in young nonhuman primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.06.06.543911. [PMID: 37333300 PMCID: PMC10274719 DOI: 10.1101/2023.06.06.543911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Anxiety disorders are among the most prevalent psychiatric disorders, with symptoms often beginning early in life. To model the pathophysiology of human pathological anxiety, we utilized Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) in a nonhuman primate model of anxious temperament to selectively increase neuronal activity of the amygdala. Subjects included 10 young rhesus macaques; 5 received bilateral infusions of AAV5-hSyn-HA-hM3Dq into the dorsal amygdala, and 5 served as controls. Subjects underwent behavioral testing in the human intruder paradigm following clozapine or vehicle administration, prior to and following surgery. Behavioral results indicated that clozapine treatment post-surgery increased freezing across different threat-related contexts in hM3Dq subjects. This effect was again observed approximately 1.9 years following surgery, indicating the long-term functional capacity of DREADD-induced neuronal activation. [11C]deschloroclozapine PET imaging demonstrated amygdala hM3Dq-HA specific binding, and immunohistochemistry revealed that hM3Dq-HA expression was most prominent in basolateral nuclei. Electron microscopy confirmed expression was predominantly on neuronal membranes. Together, these data demonstrate that activation of primate amygdala neurons is sufficient to induce increased anxiety-related behaviors, which could serve as a model to investigate pathological anxiety in humans.
Collapse
Affiliation(s)
- Sascha A L Mueller
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Jonathan A Oler
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Patrick H Roseboom
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Nakul Aggarwal
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Margaux M Kenwood
- Department of Psychiatry, Weill Cornell Medical College, New York, NY 10065, USA
| | - Marissa K Riedel
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Victoria R Elam
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Miles E Olsen
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Alexandra H DiFilippo
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Bradley T Christian
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Xing Hu
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Adriana Galvan
- Emory National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Matthew A Boehm
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ned H Kalin
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| |
Collapse
|
4
|
Pandey SK, Singh RK. Recent developments in nucleic acid-based therapies for Parkinson's disease: Current status, clinical potential, and future strategies. Front Pharmacol 2022; 13:986668. [PMID: 36339626 PMCID: PMC9632735 DOI: 10.3389/fphar.2022.986668] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 10/06/2022] [Indexed: 11/23/2022] Open
Abstract
Parkinson's disease is the second most common progressive neurodegenerative disease diagnosed mainly based on clinical symptoms caused by loss of nigrostriatal dopaminergic neurons. Although currently available pharmacological therapies provide symptomatic relief, however, the disease continues to progress eventually leading to severe motor and cognitive decline and reduced quality of life. The hallmark pathology of Parkinson's disease includes intraneuronal inclusions known as Lewy bodies and Lewy neurites, including fibrillar α-synuclein aggregates. These aggregates can progressively spread across synaptically connected brain regions leading to emergence of disease symptoms with time. The α-synuclein level is considered important in its fibrillization and aggregation. Nucleic acid therapeutics have recently been shown to be effective in treating various neurological diseases, raising the possibility of developing innovative molecular therapies for Parkinson's disease. In this review, we have described the advancements in genetic dysregulations in Parkinson's disease along with the disease-modifying strategies involved in genetic regulation with particular focus on downregulation of α-synuclein gene using various novel technologies, notably antisense oligonucleotides, microRNA, short interfering RNA, short hairpin RNAs, DNA aptamers, and gene therapy of vector-assisted delivery system-based therapeutics. In addition, the current status of preclinical and clinical development for nucleic acid-based therapies for Parkinson's disease have also been discussed along with their limitations and opportunities.
Collapse
Affiliation(s)
| | - Rakesh Kumar Singh
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli, Uttar Pradesh, India
| |
Collapse
|
5
|
Morais RDVS, Sogorb-González M, Bar C, Timmer NC, Van der Bent ML, Wartel M, Vallès A. Functional Intercellular Transmission of miHTT via Extracellular Vesicles: An In Vitro Proof-of-Mechanism Study. Cells 2022; 11:2748. [PMID: 36078156 PMCID: PMC9455173 DOI: 10.3390/cells11172748] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 08/11/2022] [Accepted: 08/31/2022] [Indexed: 11/16/2022] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder caused by GAG expansion in exon 1 of the huntingtin (HTT) gene. AAV5-miHTT is an adeno-associated virus serotype 5-based vector expressing an engineered HTT-targeting microRNA (miHTT). Preclinical studies demonstrate the brain-wide spread of AAV5-miHTT following a single intrastriatal injection, which is partly mediated by neuronal transport. miHTT has been previously associated with extracellular vesicles (EVs), but whether EVs mediate the intercellular transmission of miHTT remains unknown. A contactless culture system was used to evaluate the transport of miHTT, either from a donor cell line overexpressing miHTT or AAV5-miHTT transduced neurons. Transfer of miHTT to recipient (HEK-293T, HeLa, and HD patient-derived neurons) cells was observed, which significantly reduced HTT mRNA levels. miHTT was present in EV-enriched fractions isolated from culture media. Immunocytochemical and in situ hybridization experiments showed that the signal for miHTT and EV markers co-localized, confirming the transport of miHTT within EVs. In summary, we provide evidence that an engineered miRNA-miHTT-is loaded into EVs, transported across extracellular space, and taken up by neighboring cells, and importantly, that miHTT is active in recipient cells downregulating HTT expression. This represents an additional mechanism contributing to the widespread biodistribution of AAV5-miHTT.
Collapse
Affiliation(s)
- Roberto D. V. S. Morais
- Department of Research and Development, uniQure Biopharma B.V., 1105 BP Amsterdam, The Netherlands
| | - Marina Sogorb-González
- Department of Gastroenterology and Hepatology, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Citlali Bar
- Department of Research and Development, uniQure Biopharma B.V., 1105 BP Amsterdam, The Netherlands
| | - Nikki C. Timmer
- Department of Research and Development, uniQure Biopharma B.V., 1105 BP Amsterdam, The Netherlands
| | - M. Leontien Van der Bent
- Department of Research and Development, uniQure Biopharma B.V., 1105 BP Amsterdam, The Netherlands
| | - Morgane Wartel
- Department of Research and Development, uniQure Biopharma B.V., 1105 BP Amsterdam, The Netherlands
| | - Astrid Vallès
- Department of Research and Development, uniQure Biopharma B.V., 1105 BP Amsterdam, The Netherlands
| |
Collapse
|
6
|
Salegio EA, Cukrov M, Lortz R, Green A, Lambert E, Copeland S, Gonzalez M, Stockinger DE, Yeung JM, Hwa GGC. Feasibility of Targeted Delivery of AAV5-GFP into the Cerebellum of Nonhuman Primates Following a Single Convection-Enhanced Delivery Infusion. Hum Gene Ther 2022; 33:86-93. [PMID: 34779239 DOI: 10.1089/hum.2021.163] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
In this study, we built upon our previous work to demonstrate the distribution and transport of AAV5-green fluorescent protein (GFP) following a single convection-enhanced delivery infusion into the nonhuman primate cerebellum, with no untoward side effects noted. Dosing under magnetic resonance imaging guidance revealed a sixfold larger volume of distribution compared with the volume of infusion, with no evidence of reflux underscoring the convective properties of the cerebellum and step design of the cannula. Postmortem tissue analysis, 4 weeks post-adeno-associated viral (AAV) delivery, revealed the robust presence of the transgene in situ, with GFP detection in secondary regions not directly targeted by the infusion, denoting distal transport of the vector. Irrespective of tropism, a twofold larger area of transgene expression was found and was corroborated against the presence of contrast on T1-weighted images. Different levels of transduction were detected between animals, which were negatively correlated with the level of antibody titer against the GFP construct, whereby the higher the antibody titer, the lower the level of transgene expression. These findings support the use of the posterior fossa as a potential target site for direct delivery of gene-based therapeutics for cerebellar diseases.
Collapse
Affiliation(s)
| | - Mira Cukrov
- Valley Biosystems, Inc., West Sacramento, California, USA
| | - Rachel Lortz
- Valley Biosystems, Inc., West Sacramento, California, USA
| | - Abigail Green
- Valley Biosystems, Inc., West Sacramento, California, USA
| | - Emily Lambert
- Valley Biosystems, Inc., West Sacramento, California, USA
| | | | - Marc Gonzalez
- Valley Biosystems, Inc., West Sacramento, California, USA
| | | | - Jeremy M Yeung
- Valley Biosystems, Inc., West Sacramento, California, USA
| | | |
Collapse
|
7
|
Roseboom PH, Mueller SAL, Oler JA, Fox AS, Riedel MK, Elam VR, Olsen ME, Gomez JL, Boehm MA, DiFilippo AH, Christian BT, Michaelides M, Kalin NH. Evidence in primates supporting the use of chemogenetics for the treatment of human refractory neuropsychiatric disorders. Mol Ther 2021; 29:3484-3497. [PMID: 33895327 PMCID: PMC8636156 DOI: 10.1016/j.ymthe.2021.04.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Revised: 04/01/2021] [Accepted: 04/16/2021] [Indexed: 10/21/2022] Open
Abstract
Non-human primate (NHP) models are essential for developing and translating new treatments that target neural circuit dysfunction underlying human psychopathology. As a proof-of-concept for treating neuropsychiatric disorders, we used a NHP model of pathological anxiety to investigate the feasibility of decreasing anxiety by chemogenetically (DREADDs [designer receptors exclusively activated by designer drugs]) reducing amygdala neuronal activity. Intraoperative MRI surgery was used to infect dorsal amygdala neurons with AAV5-hSyn-HA-hM4Di in young rhesus monkeys. In vivo microPET studies with [11C]-deschloroclozapine and postmortem autoradiography with [3H]-clozapine demonstrated selective hM4Di binding in the amygdala, and neuronal expression of hM4Di was confirmed with immunohistochemistry. Additionally, because of its high affinity for DREADDs, and its approved use in humans, we developed an individualized, low-dose clozapine administration strategy to induce DREADD-mediated amygdala inhibition. Compared to controls, clozapine selectively decreased anxiety-related freezing behavior in the human intruder paradigm in hM4Di-expressing monkeys, while coo vocalizations and locomotion were unaffected. These results are an important step in establishing chemogenetic strategies for patients with refractory neuropsychiatric disorders in which amygdala alterations are central to disease pathophysiology.
Collapse
Affiliation(s)
- Patrick H Roseboom
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA.
| | - Sascha A L Mueller
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Jonathan A Oler
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Andrew S Fox
- Department of Psychology and the California National Primate Research Center, University of California-Davis, Davis, CA 95616, USA
| | - Marissa K Riedel
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Victoria R Elam
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Miles E Olsen
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| | - Juan L Gomez
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Matthew A Boehm
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Alexandra H DiFilippo
- Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Bradley T Christian
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA; Department of Medical Physics, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705, USA
| | - Michael Michaelides
- Biobehavioral Imaging and Molecular Neuropsychopharmacology Unit, National Institute on Drug Abuse Intramural Research Program, National Institutes of Health, Baltimore, MD 21224, USA
| | - Ned H Kalin
- Department of Psychiatry and the HealthEmotions Research Institute, University of Wisconsin School of Medicine and Public Health, Madison, WI 53719, USA
| |
Collapse
|
8
|
Pietersz KL, Plessis FD, Pouw SM, Liefhebber JM, van Deventer SJ, Martens GJM, Konstantinova PS, Blits B. PhP.B Enhanced Adeno-Associated Virus Mediated-Expression Following Systemic Delivery or Direct Brain Administration. Front Bioeng Biotechnol 2021; 9:679483. [PMID: 34414171 PMCID: PMC8370029 DOI: 10.3389/fbioe.2021.679483] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 06/24/2021] [Indexed: 01/14/2023] Open
Abstract
Of the adeno-associated viruses (AAVs), AAV9 is known for its capability to cross the blood–brain barrier (BBB) and can, therefore, be used as a noninvasive method to target the central nervous system. Furthermore, the addition of the peptide PhP.B to AAV9 increases its transduction across the BBB by 40-fold. Another neurotropic serotype, AAV5, has been shown as a gene therapeutic delivery vehicle to ameliorate several neurodegenerative diseases in preclinical models, but its administration requires invasive surgery. In this study, AAV9-PhP.B and AAV5-PhP.B were designed and produced in an insect cell–based system. To AAV9, the PhP.B peptide TLAVPFK was added, whereas in AAV5-PhP.B (AQTLAVPFKAQAQ), with AQ-AQAQ sequences used to swap with the corresponding sequence of AAV5. The addition of PhP.B to AAV5 did not affect its capacity to cross the mouse BBB, while increased transduction of liver tissue was observed. Then, intravenous (IV) and intrastriatal (IStr) delivery of AAV9-PhP.B and AAV5 were compared. For AAV9-PhP.B, similar transduction and expression levels were achieved in the striatum and cortex, irrespective of the delivery method used. IStr administration of AAV5 resulted in significantly higher amounts of vector DNA and therapeutic miRNA in the target regions such as striatum and cortex when compared with an IV administration of AAV9-PhP.B. These results illustrate the challenge in developing a vector that can be delivered noninvasively while achieving a transduction level similar to that of direct administration of AAV5. Thus, for therapeutic miRNA delivery with high local expression requirements, intraparenchymal delivery of AAV5 is preferred, whereas a humanized AAV9-PhP.B may be useful when widespread brain (and peripheral) transduction is needed.
Collapse
Affiliation(s)
- Kimberly L Pietersz
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, Netherlands.,Department of Molecular Animal Physiology, Faculty of Science, Centre for Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
| | - Francois Du Plessis
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, Netherlands
| | - Stephan M Pouw
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, Netherlands
| | - Jolanda M Liefhebber
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, Netherlands
| | - Sander J van Deventer
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, Netherlands.,Department of Gastroenterology and Hepatology, Leiden University Medical Center, Leiden, Netherlands
| | - Gerard J M Martens
- Department of Molecular Animal Physiology, Faculty of Science, Centre for Neuroscience, Donders Institute for Brain, Cognition and Behavior, Radboud University, Nijmegen, Netherlands
| | | | - Bas Blits
- Department of Research & Development, uniQure Biopharma B.V., Amsterdam, Netherlands
| |
Collapse
|
9
|
Vallès A, Evers MM, Stam A, Sogorb-Gonzalez M, Brouwers C, Vendrell-Tornero C, Acar-Broekmans S, Paerels L, Klima J, Bohuslavova B, Pintauro R, Fodale V, Bresciani A, Liscak R, Urgosik D, Starek Z, Crha M, Blits B, Petry H, Ellederova Z, Motlik J, van Deventer S, Konstantinova P. Widespread and sustained target engagement in Huntington's disease minipigs upon intrastriatal microRNA-based gene therapy. Sci Transl Med 2021; 13:13/588/eabb8920. [PMID: 33827977 DOI: 10.1126/scitranslmed.abb8920] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Accepted: 01/09/2021] [Indexed: 12/12/2022]
Abstract
Huntingtin (HTT)-lowering therapies hold promise to slow down neurodegeneration in Huntington's disease (HD). Here, we assessed the translatability and long-term durability of recombinant adeno-associated viral vector serotype 5 expressing a microRNA targeting human HTT (rAAV5-miHTT) administered by magnetic resonance imaging-guided convention-enhanced delivery in transgenic HD minipigs. rAAV5-miHTT (1.2 × 1013 vector genome (VG) copies per brain) was successfully administered into the striatum (bilaterally in caudate and putamen), using age-matched untreated animals as controls. Widespread brain biodistribution of vector DNA was observed, with the highest concentration in target (striatal) regions, thalamus, and cortical regions. Vector DNA presence and transgene expression were similar at 6 and 12 months after administration. Expression of miHTT strongly correlated with vector DNA, with a corresponding reduction of mutant HTT (mHTT) protein of more than 75% in injected areas, and 30 to 50% lowering in distal regions. Translational pharmacokinetic and pharmacodynamic measures in cerebrospinal fluid (CSF) were largely in line with the effects observed in the brain. CSF miHTT expression was detected up to 12 months, with CSF mHTT protein lowering of 25 to 30% at 6 and 12 months after dosing. This study demonstrates widespread biodistribution, strong and durable efficiency of rAAV5-miHTT in disease-relevant regions in a large brain, and the potential of using CSF analysis to determine vector expression and efficacy in the clinic.
Collapse
Affiliation(s)
- Astrid Vallès
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands.
| | - Melvin M Evers
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands.
| | - Anouk Stam
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| | - Marina Sogorb-Gonzalez
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| | - Cynthia Brouwers
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| | - Carlos Vendrell-Tornero
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| | - Seyda Acar-Broekmans
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| | - Lieke Paerels
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| | - Jiri Klima
- Institute of Animal Physiology and Genetics, Rumburská 89, 277 21 Libechov, Czech Republic
| | - Bozena Bohuslavova
- Institute of Animal Physiology and Genetics, Rumburská 89, 277 21 Libechov, Czech Republic
| | - Roberta Pintauro
- Department of Translational Biology, IRBM Science Park S.p.A., Via Pontina km 30,600, 00071 Pomezia, Italy
| | - Valentina Fodale
- Department of Translational Biology, IRBM Science Park S.p.A., Via Pontina km 30,600, 00071 Pomezia, Italy
| | - Alberto Bresciani
- Department of Translational Biology, IRBM Science Park S.p.A., Via Pontina km 30,600, 00071 Pomezia, Italy
| | - Roman Liscak
- Department of Stereotactic Radioneurosurgery, Na Homolce Hospital, Roentgenova 37/2, 150 30, Prague 5, Czech Republic
| | - Dusan Urgosik
- Department of Stereotactic Radioneurosurgery, Na Homolce Hospital, Roentgenova 37/2, 150 30, Prague 5, Czech Republic
| | - Zdenek Starek
- Interventional Cardiac Electrophysiology, St. Anne's University Hospital, Pekařská 53, 656 91 Brno, Czech Republic
| | - Michal Crha
- Small Animal Clinic, Veterinary and Pharmaceutical University, Palackého třída 1946/1, 612 42 Brno, Czech Republic
| | - Bas Blits
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| | - Harald Petry
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| | - Zdenka Ellederova
- Institute of Animal Physiology and Genetics, Rumburská 89, 277 21 Libechov, Czech Republic
| | - Jan Motlik
- Institute of Animal Physiology and Genetics, Rumburská 89, 277 21 Libechov, Czech Republic
| | - Sander van Deventer
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| | - Pavlina Konstantinova
- Department of Research and Development, uniQure biopharma B.V., Paasheuvelweg 25a, 1105 BP Amsterdam, Netherlands
| |
Collapse
|
10
|
Pietersz KL, Martier RM, Baatje MS, Liefhebber JM, Brouwers CC, Pouw SM, Fokkert L, Lubelski J, Petry H, Martens GJM, van Deventer SJ, Konstantinova P, Blits B. Transduction patterns in the CNS following various routes of AAV-5-mediated gene delivery. Gene Ther 2020; 28:435-446. [PMID: 32801344 DOI: 10.1038/s41434-020-0178-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 05/12/2020] [Accepted: 07/08/2020] [Indexed: 02/06/2023]
Abstract
Various administration routes of adeno-associated virus (AAV)-based gene therapy have been examined to target the central nervous system to answer the question what the most optimal delivery route is for treatment of the brain with certain indications. In this study, we evaluated AAV5 vector system for its capability to target the central nervous system via intrastriatal, intrathalamic or intracerebroventricular delivery routes in rats. AAV5 is an ideal candidate for gene therapy because of its relatively low level of existing neutralizing antibodies compared to other serotypes, and its broad tissue and cell tropism. Intrastriatal administration of AAV5-GFP resulted in centralized localized vector distribution and expression in the frontal part of the brain. Intrathalamic injection showed transduction and gradient expression from the rostral brain into lumbar spinal cord, while intracerebroventricular administration led to a more evenly, albeit relatively superficially distributed, transduction and expression throughout the central nervous system. To visualize the differences between localized and intra-cerebral spinal fluid administration routes, we compared intrastriatal to intracerebroventricular and intrathecal administration of AAV5-GFP. Together, our results demonstrate that for efficient transgene expression, various administration routes can be applied.
Collapse
|
11
|
Cerebral Organoids: A Human Model for AAV Capsid Selection and Therapeutic Transgene Efficacy in the Brain. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2020; 18:167-175. [PMID: 32637448 PMCID: PMC7327852 DOI: 10.1016/j.omtm.2020.05.028] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/24/2020] [Accepted: 05/27/2020] [Indexed: 12/15/2022]
Abstract
The development of gene therapies for central nervous system disorders is challenging because it is difficult to translate preclinical data from current in vitro and in vivo models to the clinic. Therefore, we developed induced pluripotent stem cell (iPSC)-derived cerebral organoids as a model for recombinant adeno-associated virus (rAAV) capsid selection and for testing efficacy of AAV-based gene therapy in a human context. Cerebral organoids are physiological 3D structures that better recapitulate the human brain compared with 2D cell lines. To validate the model, we compared the transduction efficiency and distribution of two commonly used AAV serotypes (rAAV5 and rAAV9). In cerebral organoids, transduction with rAAV5 led to higher levels of vector DNA, transgenic mRNA, and protein expression as compared with rAAV9. The superior transduction of rAAV5 was replicated in iPSC-derived neuronal cells. Furthermore, rAAV5-mediated delivery of a human sequence-specific engineered microRNA to cerebral organoids led to a lower expression of its target ataxin-3. Our studies provide a new tool for selecting and deselecting AAV serotypes, and for demonstrating therapeutic efficacy of transgenes in a human context. Implementing cerebral organoids during gene therapy development could reduce the usage of animal models and improve translation to the clinic.
Collapse
|
12
|
Aron Badin R. Nonhuman Primate Models of Huntington's Disease and Their Application in Translational Research. Methods Mol Biol 2018; 1780:267-284. [PMID: 29856024 DOI: 10.1007/978-1-4939-7825-0_14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Huntington's disease (HD) is a monogenic, autosomal dominant inherited fatal disease that affects 1 in 10,000 people worldwide. Given its unique genetic characteristics, HD would appear as one of the most straightforward neurodegenerative diseases to replicate in animal models. Indeed, mutations in the HTT gene have been used to generate a variety of animal models that display differential pathologies and have significantly increased our understanding of the pathological mechanisms of HD. However, decades of efforts have also shown the complexity of recapitulating the human condition in other species. Here we describe the three different types of models that have been generated in nonhuman primate species, stating their advantages and limitations and attempt to give a critical perspective of their translational value to test the efficacy of novel therapeutic strategies. Obtaining construct, phenotypic, and predictive validity has proven to be challenging in most animal models of human diseases. In HD in particular, it is hard to assess the predictive validity of a new therapeutic strategy when no effective "benchmark" treatment is available in the clinic. In this light, only phenotypic/face validity and construct validity are discussed.
Collapse
Affiliation(s)
- Romina Aron Badin
- Commissariat à l'Energie Atomique (CEA), Molecular Imaging Research Center (MIRCen), Fontenay-aux-Roses, France.
- Centre National de la Recherche Scientifique (CNRS), Université Paris-Sud, Université Paris-Saclay, UMR 9199, Neurodegenerative Diseases Laboratory, Fontenay-aux-Roses, France.
| |
Collapse
|
13
|
Axelsen TM, Woldbye DP. Gene Therapy for Parkinson's Disease, An Update. JOURNAL OF PARKINSON'S DISEASE 2018; 8:195-215. [PMID: 29710735 PMCID: PMC6027861 DOI: 10.3233/jpd-181331] [Citation(s) in RCA: 105] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 03/25/2018] [Indexed: 12/19/2022]
Abstract
The current mainstay treatment of Parkinson's disease (PD) consists of dopamine replacement therapy which, in addition to causing several side effects, does not delay disease progression. The field of gene therapy offers a potential means to improve current therapy. The present review gives an update of the present status of gene therapy for PD. Both non-disease and disease modifying transgenes have been tested for PD gene therapy in animal and human studies. Non-disease modifying treatments targeting dopamine or GABA synthesis have been successful and promising at improving PD symptomatology in randomized clinical studies, but substantial testing remains before these can be implemented in the standard clinical treatment repertoire. As for disease modifying targets that theoretically offer the possibility of slowing the progression of disease, several neurotrophic factors show encouraging results in preclinical models (e.g., neurturin, GDNF, BDNF, CDNF, VEGF-A). However, so far, clinical trials have only tested neurturin, and, unfortunately, no trial has been able to meet its primary endpoint. Future clinical trials with neurotrophic factors clearly deserve to be conducted, considering the still enticing goal of actually slowing the disease process of PD. As alternative types of gene therapy, opto- and chemogenetics might also find future use in PD treatment and novel genome-editing technology could also potentially be applied as individualized gene therapy for genetic types of PD.
Collapse
Affiliation(s)
- Tobias M. Axelsen
- Department of Neurology, Herlev University Hospital, Herlev, Denmark
| | - David P.D. Woldbye
- Department of Neuroscience, Panum Institute, Mærsk Tower, University of Copenhagen, Copenhagen N, Denmark
| |
Collapse
|
14
|
Torres N, Molet J, Moro C, Mitrofanis J, Benabid AL. Neuroprotective Surgical Strategies in Parkinson's Disease: Role of Preclinical Data. Int J Mol Sci 2017; 18:ijms18102190. [PMID: 29053638 PMCID: PMC5666871 DOI: 10.3390/ijms18102190] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2017] [Revised: 10/10/2017] [Accepted: 10/13/2017] [Indexed: 12/18/2022] Open
Abstract
Although there have been many pharmacological agents considered to be neuroprotective therapy in Parkinson's disease (PD) patients, neurosurgical approaches aimed to neuroprotect or restore the degenerative nigrostriatal system have rarely been the focus of in depth reviews. Here, we explore the neuroprotective strategies involving invasive surgical approaches (NSI) using neurotoxic models 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) and 6-hydroxydopamine (6-OHDA), which have led to clinical trials. We focus on several NSI approaches, namely deep brain stimulation of the subthalamic nucleus, glial neurotrophic derived factor (GDNF) administration and cell grafting methods. Although most of these interventions have produced positive results in preclinical animal models, either from behavioral or histological studies, they have generally failed to pass randomized clinical trials to validate each approach. We argue that NSI are promising approaches for neurorestoration in PD, but preclinical studies should be planned carefully in order not only to detect benefits but also to detect potential adverse effects. Further, clinical trials should be designed to be able to detect and disentangle neuroprotection from symptomatic effects. In summary, our review study evaluates the pertinence of preclinical models to study NSI for PD and how this affects their efficacy when translated into clinical trials.
Collapse
Affiliation(s)
- Napoleon Torres
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| | - Jenny Molet
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| | - Cecile Moro
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| | - John Mitrofanis
- Department of Anatomy, University of Sydney; Sydney Medical School, Sydney NSW 2006, Australia.
| | - Alim Louis Benabid
- University Grenoble Alpes, CEA, LETI, CLINATEC, MINATEC Campus, 38000 Grenoble, France.
| |
Collapse
|
15
|
Vermilyea SC, Emborg ME. The role of nonhuman primate models in the development of cell-based therapies for Parkinson's disease. J Neural Transm (Vienna) 2017; 125:365-384. [PMID: 28326445 PMCID: PMC5847191 DOI: 10.1007/s00702-017-1708-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Accepted: 03/12/2017] [Indexed: 12/23/2022]
Abstract
Through the course of over three decades, nonhuman primate (NHP) studies on cell-based therapies (CBTs) for Parkinson’s disease (PD) have provided insight into the feasibility, safety and efficacy of the approach, methods of cell collection and preparation, cell viability, as well as potential brain targets. Today, NHP research continues to be a vital source of information for improving cell grafts and analyzing how the host affects graft survival, integration and function. Overall, this article aims to discuss the role that NHP models of PD have played in CBT development and highlights specific issues that need to be considered to maximize the value of NHP studies for the successful clinical translation of CBTs.
Collapse
Affiliation(s)
- Scott C Vermilyea
- Neuroscience Training Program, University of Wisconsin, Madison, 1220 Capitol Court, Madison, WI, 53715, USA.,Wisconsin National Primate Research Center, University of Wisconsin, Madison, USA
| | - Marina E Emborg
- Neuroscience Training Program, University of Wisconsin, Madison, 1220 Capitol Court, Madison, WI, 53715, USA. .,Wisconsin National Primate Research Center, University of Wisconsin, Madison, USA. .,Department of Medical Physics, University of Wisconsin, Madison, USA.
| |
Collapse
|
16
|
Samaranch L, Blits B, San Sebastian W, Hadaczek P, Bringas J, Sudhakar V, Macayan M, Pivirotto PJ, Petry H, Bankiewicz KS. MR-guided parenchymal delivery of adeno-associated viral vector serotype 5 in non-human primate brain. Gene Ther 2017; 24:253-261. [PMID: 28300083 PMCID: PMC5404203 DOI: 10.1038/gt.2017.14] [Citation(s) in RCA: 58] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2016] [Revised: 01/19/2017] [Accepted: 02/01/2017] [Indexed: 12/19/2022]
Abstract
The present study was designed to characterize transduction of non-human primate brain and spinal cord with AAV5 viral vector after parenchymal delivery. AAV5-CAG-GFP (1 × 1013 vector genomes per milliliter (vg ml−1)) was bilaterally infused either into putamen, thalamus or with the combination left putamen and right thalamus. Robust expression of GFP was seen throughout infusion sites and also in other distal nuclei. Interestingly, thalamic infusion of AAV5 resulted in the transduction of the entire corticospinal axis, indicating transport of AAV5 over long distances. Regardless of site of injection, AAV5 transduced both neurons and astrocytes equally. Our data demonstrate that AAV5 is a very powerful vector for the central nervous system and has potential for treatment of a wide range of neurological pathologies with cortical, subcortical and/or spinal cord affection.
Collapse
Affiliation(s)
- L Samaranch
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - B Blits
- Neurobiology, Research and Development, UniQure NV, Amsterdam 1105BA, The Netherlands
| | - W San Sebastian
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - P Hadaczek
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - J Bringas
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - V Sudhakar
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - M Macayan
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - P J Pivirotto
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| | - H Petry
- Neurobiology, Research and Development, UniQure NV, Amsterdam 1105BA, The Netherlands
| | - K S Bankiewicz
- Department of Neurological Surgery, University of California, San Francisco, CA, USA
| |
Collapse
|
17
|
Vermilyea SC, Lu J, Olsen M, Guthrie S, Tao Y, Fekete EM, Riedel MK, Brunner K, Boettcher C, Bondarenko V, Brodsky E, Block WF, Alexander A, Zhang SC, Emborg ME. Real-Time Intraoperative MRI Intracerebral Delivery of Induced Pluripotent Stem Cell-Derived Neurons. Cell Transplant 2016; 26:613-624. [PMID: 27633706 DOI: 10.3727/096368916x692979] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Induced pluripotent stem cell (iPSC)-derived neurons represent an opportunity for cell replacement strategies for neurodegenerative disorders such as Parkinson's disease (PD). Improvement in cell graft targeting, distribution, and density can be key for disease modification. We have previously developed a trajectory guide system for real-time intraoperative magnetic resonance imaging (RT-IMRI) delivery of infusates, such as viral vector suspensions for gene therapy strategies. Intracerebral delivery of iPSC-derived neurons presents different challenges than viral vectors, including limited cell survival if cells are kept at room temperature for prolonged periods of time, precipitation and aggregation of cells in the cannula, and obstruction during injection, which must be solved for successful application of this delivery approach. To develop procedures suitable for RT-IMRI cell delivery, we first performed in vitro studies to tailor the delivery hardware (e.g., cannula) and defined a range of parameters to be applied (e.g., maximal time span allowable between cell loading in the system and intracerebral injection) to ensure cell survival. Then we performed an in vivo study to evaluate the feasibility of applying the system to nonhuman primates. Our results demonstrate that the RT-IMRI delivery system provides valuable guidance, monitoring, and visualization during intracerebral cell delivery that are compatible with cell survival.
Collapse
|
18
|
Kalin NH, Fox AS, Kovner R, Riedel MK, Fekete EM, Roseboom PH, Tromp DPM, Grabow BP, Olsen ME, Brodsky EK, McFarlin DR, Alexander AL, Emborg ME, Block WF, Fudge JL, Oler JA. Overexpressing Corticotropin-Releasing Factor in the Primate Amygdala Increases Anxious Temperament and Alters Its Neural Circuit. Biol Psychiatry 2016; 80:345-55. [PMID: 27016385 PMCID: PMC4967405 DOI: 10.1016/j.biopsych.2016.01.010] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Revised: 12/23/2015] [Accepted: 01/14/2016] [Indexed: 12/28/2022]
Abstract
BACKGROUND Nonhuman primate models are critical for understanding mechanisms underlying human psychopathology. We established a nonhuman primate model of anxious temperament (AT) for studying the early-life risk to develop anxiety and depression. Studies have identified the central nucleus of the amygdala (Ce) as an essential component of AT's neural substrates. Corticotropin-releasing factor (CRF) is expressed in the Ce, has a role in stress, and is linked to psychopathology. Here, in young rhesus monkeys, we combined viral vector technology with assessments of anxiety and multimodal neuroimaging to understand the consequences of chronically increased CRF in the Ce region. METHODS Using real-time intraoperative magnetic resonance imaging-guided convection-enhanced delivery, five monkeys received bilateral dorsal amygdala Ce-region infusions of adeno-associated virus serotype 2 containing the CRF construct. Their cagemates served as unoperated control subjects. AT, regional brain metabolism, resting functional magnetic resonance imaging, and diffusion tensor imaging were assessed before and 2 months after viral infusions. RESULTS Dorsal amygdala CRF overexpression significantly increased AT and metabolism within the dorsal amygdala. Additionally, we observed changes in metabolism in other AT-related regions, as well as in measures of functional and structural connectivity. CONCLUSIONS This study provides a translational roadmap that is important for understanding human psychopathology by combining molecular manipulations used in rodents with behavioral phenotyping and multimodal neuroimaging measures used in humans. The results indicate that chronic CRF overexpression in primates not only increases AT but also affects metabolism and connectivity within components of AT's neural circuitry.
Collapse
Affiliation(s)
- Ned H Kalin
- Department of Psychiatry, University of Wisconsin, Madison, WI,Neuroscience Training Program, University of Wisconsin, Madison, WI,Wisconsin National Primate Research Center, Madison, WI
| | - Andrew S Fox
- Department of Psychiatry, University of Wisconsin, Madison, WI
| | - Rothem Kovner
- Department of Psychiatry, University of Wisconsin, Madison, WI,Neuroscience Training Program, University of Wisconsin, Madison, WI
| | | | - Eva M Fekete
- Department of Psychiatry, University of Wisconsin, Madison, WI
| | - Patrick H Roseboom
- Department of Psychiatry, University of Wisconsin, Madison, WI,Neuroscience Training Program, University of Wisconsin, Madison, WI
| | - Do P M Tromp
- Department of Psychiatry, University of Wisconsin, Madison, WI,Neuroscience Training Program, University of Wisconsin, Madison, WI
| | | | - Miles E Olsen
- Department of Medical Physics, University of Wisconsin, Madison, WI
| | - Ethan K Brodsky
- Department of Medical Physics, University of Wisconsin, Madison, WI,inseRT MRI, Inc
| | | | - Andrew L Alexander
- Department of Psychiatry, University of Wisconsin, Madison, WI,Department of Medical Physics, University of Wisconsin, Madison, WI,inseRT MRI, Inc
| | - Marina E Emborg
- Neuroscience Training Program, University of Wisconsin, Madison, WI,Department of Medical Physics, University of Wisconsin, Madison, WI,Wisconsin National Primate Research Center, Madison, WI
| | - Walter F Block
- Department of Medical Physics, University of Wisconsin, Madison, WI,inseRT MRI, Inc
| | - Julie L Fudge
- Departments of Neurobiology and Anatomy, and Psychiatry, University of Rochester Medical Center
| | - Jonathan A Oler
- Department of Psychiatry, University of Wisconsin, Madison, Wisconsin.
| |
Collapse
|