1
|
Enbar T, Hickmott JW, Siu R, Gao D, Garcia-Flores E, Smart J, Casabuenas DL, Faiz M, Morshead CM. Regionally distinct GFAP promoter expression plays a role in off-target neuron expression following AAV5 transduction. Sci Rep 2024; 14:31583. [PMID: 39738170 DOI: 10.1038/s41598-024-79124-5] [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: 08/28/2024] [Accepted: 11/06/2024] [Indexed: 01/01/2025] Open
Abstract
Astrocyte to neuron reprogramming has been performed using viral delivery of neurogenic transcription factors in GFAP expressing cells. Recent reports of off-target expression in cortical neurons following adeno-associated virus (AAV) transduction to deliver the neurogenic factors have confounded our understanding of the efficacy of direct cellular reprogramming. To shed light on potential mechanisms that may underlie the neuronal off-target expression of GFAP promoter driven expression of neurogenic factors in neurons, two regionally distinct cortices were compared-the motor cortex (MC) and medial prefrontal cortex (mPFC)-and investigated: (1) the regional tropism and astrocyte transduction with an AAV5-GFAP vector, (2) the expression of Gfap in MC and mPFC neurons; and (3) material transfer between astrocytes and neurons. Using a Cre-based system (AAV5-hGFAP-Cre; Rosa26R-tdTomato reporter mice), regional differences were observed in tdTomato expression between the MC and mPFC. Interestingly, this correlated with the presence of a greater expression of Gfap mRNA in neurons in the mPFC. Additionally, intercellular material transfer of Cre and tdTomato was observed between astrocytes and neurons in both regions, albeit at very low frequencies. Our study highlights regionally distinct variation in neurons that warrants consideration when designing genetic constructs for gene therapies targeting astrocytes including astrocyte to neuron reprogramming.
Collapse
Affiliation(s)
- T Enbar
- Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - J W Hickmott
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - R Siu
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - D Gao
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada
| | - E Garcia-Flores
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, ON, M5S 3G9, Canada
| | - J Smart
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
| | - D L Casabuenas
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
| | - M Faiz
- Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, 1 King's College Cir, Toronto, ON, M5S 1A8, Canada
| | - C M Morshead
- Institute of Medical Sciences, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Department of Surgery, Division of Anatomy, University of Toronto, 1 King's College Circle, Toronto, ON, M5S 1A8, Canada.
- Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, 160 College Street, Toronto, ON, M5S 3E1, Canada.
- Institute of Biomedical Engineering, University of Toronto, 164 College St., Toronto, ON, M5S 3G9, Canada.
| |
Collapse
|
2
|
Matthews EA, Russ JB, Qian Y, Zhao S, Thompson P, Methani M, Vestal ML, Josh Huang Z, Southwell DG. RNA-programmable cell type monitoring and manipulation in the human cortex with CellREADR. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.03.626590. [PMID: 39677799 PMCID: PMC11642864 DOI: 10.1101/2024.12.03.626590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Reliable and systematic experimental access to diverse cell types is necessary for understanding the neural circuit organization, function, and pathophysiology of the human brain. Methods for targeting human neural populations are scarce and currently center around identifying and engineering transcriptional enhancers and viral capsids. Here we demonstrate the utility of CellREADR, a programmable RNA sensor-effector technology that couples cellular RNA sensing to effector protein translation, for accessing, monitoring, and manipulating specific neuron types in ex vivo human cortical tissues. We designed CellREADR constructs to target two distinct human neuron types, CALB2+ (calretinin) GABAergic interneurons and FOXP2+ (forkhead box protein P2) glutamatergic projection neurons, and validated cell targeting using histological, electrophysiological, and transcriptomic methods. CellREADR-mediated expression of optogenetic effectors and genetically-encoded calcium indicators allowed us to manipulate and monitor these neuronal populations in cortical microcircuits. We further demonstrate that AAV-based CellREADR and enhancer vectors can be jointly used to target different subpopulations in the same preparation. By demonstrating specific, reliable, and programmable experimental access to targeted cell types, our results highlight CellREADR's potential for studying human neural circuits and treating brain disorders with cell type resolution.
Collapse
Affiliation(s)
- Elizabeth A. Matthews
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
| | - Jeffrey B. Russ
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Pediatrics, Division of Neurology, Duke University School of Medicine, Durham, NC USA
| | - Yongjun Qian
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
- Current affiliation: College of Future technology, Peking-Tsinghua Center for Life Sciences, IDG/McGovern Institute for Brain Research, Beijing Advanced Center of RNA Biology (BEACON), Peking University, China
| | - Shengli Zhao
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
| | - Peyton Thompson
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
| | - Muhib Methani
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
| | - Matthew L. Vestal
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Current affiliation: Department of Neurosurgery, Dartmouth University, Dartmouth, MA USA
| | - Z. Josh Huang
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC USA
| | - Derek G. Southwell
- Department of Neurosurgery, Duke University School of Medicine, Durham, NC USA
- Department of Neurobiology, Duke University School of Medicine, Durham, NC USA
- Department of Biomedical Engineering, Duke University Pratt School of Engineering, Durham, NC USA
| |
Collapse
|
3
|
Carneiro AD, Schaffer DV. Engineering novel adeno-associated viruses (AAVs) for improved delivery in the nervous system. Curr Opin Chem Biol 2024; 83:102532. [PMID: 39342684 DOI: 10.1016/j.cbpa.2024.102532] [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: 05/02/2024] [Revised: 08/27/2024] [Accepted: 09/03/2024] [Indexed: 10/01/2024]
Abstract
Harnessing adeno-associated virus (AAV) vectors for therapeutic gene delivery has emerged as a progressively promising strategy to treat disorders of both the central nervous system (CNS) and peripheral nervous system (PNS), and there are many ongoing clinical trials. However, unique physiological and molecular characteristics of the CNS and PNS pose obstacles to efficient vector delivery, ranging from the blood-brain barrier to the diverse nature of nervous system disorders. Engineering novel AAV capsids may help overcome these ongoing challenges and maximize therapeutic transgene delivery. This article discusses strategies for innovative AAV capsid development, highlighting recent advances. Notably, advances in next generation sequencing and machine learning have sparked new approaches for capsid investigation and engineering. Furthermore, we outline future directions and additional challenges in AAV-mediated gene therapy in the CNS and PNS.
Collapse
Affiliation(s)
- Ana D Carneiro
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA 94720, USA; Department of Bioengineering, University of California, Berkeley, CA 94720, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA; California Institute for Quantitative Biosciences, University of California, Berkeley, CA 94720, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, CA 94720, USA.
| |
Collapse
|
4
|
Zappala A, Li H, Inoue K. Focused Ultrasounds as an Adeno-Associated Virus Gene Therapy-Empowering Tool in Juvenile Mice via Intracerebroventricular Administration. Hum Gene Ther 2024; 35:989-999. [PMID: 39585211 DOI: 10.1089/hum.2024.108] [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] [Indexed: 11/26/2024] Open
Abstract
Systemic delivery of adeno-associated virus (AAV) vectors targeting the central nervous system has the potential to solve many neurodevelopmental disorders, yet it is made difficult by the filtering effect of the blood-brain barrier and systemic complications. To overcome this limitation, we attempted to inject a Venus-expressing, oligodendrocyte-selective AAV9 viral vector in the ventricles together with lipid microbubbles and subjected them to focused ultrasound (FUS); the resulting mechanical stimulation on the brain ventricles is able to open small, temporary gaps from which vector particles can leak and spread. Our findings indicate that FUS can increase viral vector diffusion across both the anteroposterior and left-right axes without influencing cell tropism; significant effects were found with 60 and 90 s exposure time, but no effects were observed with longer intervals. Taken together, these results highlight a new strategy for the safe and effective delivery of viral vectors and offer new perspectives for the development and application of gene therapies for central nervous system diseases.
Collapse
Affiliation(s)
- Alessandro Zappala
- Department Mental Retardation & Birth Defect Research, National Center of Neurology & Psychiatry, National Institute of Neuroscience, Tokyo, Japan
| | - Heng Li
- Department Mental Retardation & Birth Defect Research, National Center of Neurology & Psychiatry, National Institute of Neuroscience, Tokyo, Japan
| | - Ken Inoue
- Department Mental Retardation & Birth Defect Research, National Center of Neurology & Psychiatry, National Institute of Neuroscience, Tokyo, Japan
| |
Collapse
|
5
|
Sehara Y, Hashimotodani Y, Watano R, Ohba K, Uchibori R, Shimazaki K, Kawai K, Mizukami H. Adeno-associated Virus-mediated Ezh2 Knockdown Reduced the Increment of Newborn Neurons Induced by Forebrain Ischemia in Gerbil Dentate Gyrus. Mol Neurobiol 2024; 61:9623-9632. [PMID: 38676810 PMCID: PMC11496322 DOI: 10.1007/s12035-024-04200-w] [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: 01/26/2024] [Accepted: 04/23/2024] [Indexed: 04/29/2024]
Abstract
It is established that neurogenesis of dentate gyrus is increased after ischemic insult, although the regulatory mechanisms have not yet been elucidated. In this study, we focused on Ezh2 which suppresses gene expression through catalyzing trimethylation of lysine 27 of histone 3. Male gerbils were injected with adeno-associated virus (AAV) carrying shRNA targeting to Ezh2 into right dentate gyrus 2 weeks prior to forebrain ischemia. One week after ischemia, animals were injected with thymidine analogue to label proliferating cells. Three weeks after ischemia, animals were killed for histological analysis. AAV-mediated knockdown of Ezh2 significantly decreased the ischemia-induced increment of proliferating cells, and the proliferated cells after ischemia showed significantly longer migration from subgranular zone (SGZ), compared to the control group. Furthermore, the number of neural stem cells in SGZ significantly decreased after ischemia with Ezh2 knockdown group. Of note, Ezh2 knockdown did not affect the number of proliferating cells or the migration from SGZ in the non-ischemic condition. Our data showed that, specifically after ischemia, Ezh2 knockdown shifted the balance between self-renewal and differentiation toward differentiation in adult dentate gyrus.
Collapse
Affiliation(s)
- Yoshihide Sehara
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan.
| | | | - Ryota Watano
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Kenji Ohba
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Ryosuke Uchibori
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| | - Kuniko Shimazaki
- Department of Neurosurgery, Jichi Medical University, Shimotsuke, Japan
| | - Kensuke Kawai
- Department of Neurosurgery, Jichi Medical University, Shimotsuke, Japan
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi, 329-0498, Japan
| |
Collapse
|
6
|
Gonzalez-Ramos A, Berglind F, Kudláček J, Rocha ER, Melin E, Sebastião AM, Valente CA, Ledri M, Andersson M, Kokaia M. Chemogenetics with PSAM 4-GlyR decreases excitability and epileptiform activity in epileptic hippocampus. Gene Ther 2024:10.1038/s41434-024-00493-7. [PMID: 39455855 DOI: 10.1038/s41434-024-00493-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 09/28/2024] [Accepted: 10/10/2024] [Indexed: 10/28/2024]
Abstract
Despite the availability of new drugs on the clinics in recent years, drug-resistant epilepsy remains an unresolved challenge for healthcare, and one-third of epilepsy patients remain refractory to anti-seizure medications. Gene therapy in experimental models has emerged as effective treatment targeting specific neuronal populations in the epileptogenic focus. When combined with an external chemical activator using chemogenetics, it also becomes an "on-demand" treatment. Here, we evaluate a targeted and specific chemogenetic therapy, the PSAM/PSEM system, which holds promise as a potential candidate for clinical application in treating drug-resistant epilepsy. We show that the inert ligand uPSEM817, which selectively activates the chloride-permeable channel PSAM4-GlyR, effectively reduces the number of depolarization-induced action potentials in vitro. This effect is likely due to the shunting of depolarizing currents, as evidenced by decreased membrane resistance in these cells. In organotypic slices, uPSEM817 decreased the number of bursts and peak amplitude of events of spontaneous epileptiform activity. Although administration of uPSEM817 in vivo did not significantly alter electrographic seizures in a male mouse model of temporal lobe epilepsy, it did demonstrate a strong trend toward reducing the frequency of interictal epileptiform discharges. These findings indicate that PSAM4-GlyR-based chemogenetics holds potential as an anti-seizure strategy, although further refinement is necessary to enhance its efficacy.
Collapse
Affiliation(s)
- Ana Gonzalez-Ramos
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden.
- Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA.
| | - Fredrik Berglind
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Jan Kudláček
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
- Department of Physiology, Second Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Elza R Rocha
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Esbjörn Melin
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Ana M Sebastião
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Cláudia A Valente
- Instituto de Farmacologia e Neurociências, Faculdade de Medicina, Universidade de Lisboa, Lisboa, Portugal
- Instituto de Medicina Molecular João Lobo Antunes, Universidade de Lisboa, Lisboa, Portugal
| | - Marco Ledri
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - My Andersson
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden
| | - Merab Kokaia
- Epilepsy Center, Department of Clinical Sciences, Lund University Hospital, Lund, Sweden.
| |
Collapse
|
7
|
Choules MP, Bonate PL, Heo N, Weddell J. Prospective approaches to gene therapy computational modeling - spotlight on viral gene therapy. J Pharmacokinet Pharmacodyn 2024; 51:399-416. [PMID: 37848637 DOI: 10.1007/s10928-023-09889-1] [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: 08/29/2022] [Accepted: 09/25/2023] [Indexed: 10/19/2023]
Abstract
Clinical studies have found there still exists a lack of gene therapy dose-toxicity and dose-efficacy data that causes gene therapy dose selection to remain elusive. Model informed drug development (MIDD) has become a standard tool implemented throughout the discovery, development, and approval of pharmaceutical therapies, and has the potential to inform dose-toxicity and dose-efficacy relationships to support gene therapy dose selection. Despite this potential, MIDD approaches for gene therapy remain immature and require standardization to be useful for gene therapy clinical programs. With the goal to advance MIDD approaches for gene therapy, in this review we first provide an overview of gene therapy types and how they differ from a bioanalytical, formulation, route of administration, and regulatory standpoint. With this biological and regulatory background, we propose how MIDD can be advanced for AAV-based gene therapies by utilizing physiological based pharmacokinetic modeling and quantitative systems pharmacology to holistically inform AAV and target protein dynamics following dosing. We discuss how this proposed model, allowing for in-depth exploration of AAV pharmacology, could be the key the field needs to treat these unmet disease populations.
Collapse
Affiliation(s)
- Mary P Choules
- Early Development, New Technologies Group, Astellas, Northbrook, IL, USA
| | - Peter L Bonate
- Early Development, New Technologies Group, Astellas, Northbrook, IL, USA.
| | - Nakyo Heo
- Early Development, New Technologies Group, Astellas, Northbrook, IL, USA
| | - Jared Weddell
- Early Development, New Technologies Group, Astellas, Northbrook, IL, USA
| |
Collapse
|
8
|
Zhou L, Wang Y, Xu Y, Zhang Y, Zhu C. A comprehensive review of AAV-mediated strategies targeting microglia for therapeutic intervention of neurodegenerative diseases. J Neuroinflammation 2024; 21:232. [PMID: 39300451 DOI: 10.1186/s12974-024-03232-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Accepted: 09/10/2024] [Indexed: 09/22/2024] Open
Abstract
Neurodegenerative diseases pose a significant health burden globally, with limited treatment options available. Among the various cell types involved in the pathogenesis of these disorders, microglia, the resident immune cells of the central nervous system, play a pivotal role. Dysregulated microglial activation contributes to neuroinflammation and neuronal damage, making them an attractive target for therapeutic intervention. Adeno-associated virus (AAV) vectors have emerged as powerful tools for delivering therapeutic genes to specific cell types in the central nervous system with remarkable precision and safety. In the current review, we discuss the strategies employed to achieve selective transduction of microglia, including the use of cell-specific promoters, engineered capsids, and microRNA (miRNA) strategies. Additionally, we address the challenges and future directions in the development of AAV-based therapies targeting microglia. Overall, AAV-mediated targeting of microglia holds promise as a novel therapeutic approach for neurodegenerative diseases, offering the potential to modify disease progression and improve patient outcomes.
Collapse
Affiliation(s)
- Livia Zhou
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital Zhengzhou Children's Hospital, Zhengzhou, 450018, China.
| | - Yafeng Wang
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China
| | - Yaodong Zhang
- Henan Neurodevelopment Engineering Research Center for Children, Children's Hospital Affiliated to Zhengzhou University, Henan Children's Hospital Zhengzhou Children's Hospital, Zhengzhou, 450018, China
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury and Henan Pediatric Clinical Research Center, Institute of Neuroscience and The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan Province, China.
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
9
|
Matsuzaka Y, Yashiro R. Therapeutic Application and Structural Features of Adeno-Associated Virus Vector. Curr Issues Mol Biol 2024; 46:8464-8498. [PMID: 39194716 DOI: 10.3390/cimb46080499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 07/02/2024] [Accepted: 07/12/2024] [Indexed: 08/29/2024] Open
Abstract
Adeno-associated virus (AAV) is characterized by non-pathogenicity, long-term infection, and broad tropism and is actively developed as a vector virus for gene therapy products. AAV is classified into more than 100 serotypes based on differences in the amino acid sequence of the capsid protein. Endocytosis involves the uptake of viral particles by AAV and accessory receptors during AAV infection. After entry into the cell, they are transported to the nucleus through the nuclear pore complex. AAVs mainly use proteoglycans as receptors to enter cells, but the types of sugar chains in proteoglycans that have binding ability are different. Therefore, it is necessary to properly evaluate the primary structure of receptor proteins, such as amino acid sequences and post-translational modifications, including glycosylation, and the higher-order structure of proteins, such as the folding of the entire capsid structure and the three-dimensional (3D) structure of functional domains, to ensure the efficacy and safety of biopharmaceuticals. To further enhance safety, it is necessary to further improve the efficiency of gene transfer into target cells, reduce the amount of vector administered, and prevent infection of non-target cells.
Collapse
Affiliation(s)
- Yasunari Matsuzaka
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Minato-ku, Tokyo 108-8639, Japan
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Japan
| | - Ryu Yashiro
- Administrative Section of Radiation Protection, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira 187-8551, Japan
- Department of Mycobacteriology, Leprosy Research Center, National Institute of Infectious Diseases, Tokyo 162-8640, Japan
| |
Collapse
|
10
|
Lu WH, Chang TT, Chang YM, Liu YH, Lin CH, Suen CS, Hwang MJ, Huang YS. CPEB2-activated axonal translation of VGLUT2 mRNA promotes glutamatergic transmission and presynaptic plasticity. J Biomed Sci 2024; 31:69. [PMID: 38992696 PMCID: PMC11241979 DOI: 10.1186/s12929-024-01061-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 07/02/2024] [Indexed: 07/13/2024] Open
Abstract
BACKGROUND Local translation at synapses is important for rapidly remodeling the synaptic proteome to sustain long-term plasticity and memory. While the regulatory mechanisms underlying memory-associated local translation have been widely elucidated in the postsynaptic/dendritic region, there is no direct evidence for which RNA-binding protein (RBP) in axons controls target-specific mRNA translation to promote long-term potentiation (LTP) and memory. We previously reported that translation controlled by cytoplasmic polyadenylation element binding protein 2 (CPEB2) is important for postsynaptic plasticity and memory. Here, we investigated whether CPEB2 regulates axonal translation to support presynaptic plasticity. METHODS Behavioral and electrophysiological assessments were conducted in mice with pan neuron/glia- or glutamatergic neuron-specific knockout of CPEB2. Hippocampal Schaffer collateral (SC)-CA1 and temporoammonic (TA)-CA1 pathways were electro-recorded to monitor synaptic transmission and LTP evoked by 4 trains of high-frequency stimulation. RNA immunoprecipitation, coupled with bioinformatics analysis, were used to unveil CPEB2-binding axonal RNA candidates associated with learning, which were further validated by Western blotting and luciferase reporter assays. Adeno-associated viruses expressing Cre recombinase were stereotaxically delivered to the pre- or post-synaptic region of the TA circuit to ablate Cpeb2 for further electrophysiological investigation. Biochemically isolated synaptosomes and axotomized neurons cultured on a microfluidic platform were applied to measure axonal protein synthesis and FM4-64FX-loaded synaptic vesicles. RESULTS Electrophysiological analysis of hippocampal CA1 neurons detected abnormal excitability and vesicle release probability in CPEB2-depleted SC and TA afferents, so we cross-compared the CPEB2-immunoprecipitated transcriptome with a learning-induced axonal translatome in the adult cortex to identify axonal targets possibly regulated by CPEB2. We validated that Slc17a6, encoding vesicular glutamate transporter 2 (VGLUT2), is translationally upregulated by CPEB2. Conditional knockout of CPEB2 in VGLUT2-expressing glutamatergic neurons impaired consolidation of hippocampus-dependent memory in mice. Presynaptic-specific ablation of Cpeb2 in VGLUT2-dominated TA afferents was sufficient to attenuate protein synthesis-dependent LTP. Moreover, blocking activity-induced axonal Slc17a6 translation by CPEB2 deficiency or cycloheximide diminished the releasable pool of VGLUT2-containing synaptic vesicles. CONCLUSIONS We identified 272 CPEB2-binding transcripts with altered axonal translation post-learning and established a causal link between CPEB2-driven axonal synthesis of VGLUT2 and presynaptic translation-dependent LTP. These findings extend our understanding of memory-related translational control mechanisms in the presynaptic compartment.
Collapse
Affiliation(s)
- Wen-Hsin Lu
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Tzu-Tung Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Yao-Ming Chang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Yi-Hsiang Liu
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Chia-Hsuan Lin
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming Chao-Tung University and Academia Sinica, Taipei, 11529, Taiwan
| | - Ching-Shu Suen
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Ming-Jing Hwang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan
| | - Yi-Shuian Huang
- Institute of Biomedical Sciences, Academia Sinica, 128 Sec. 2, Academia Rd., Taipei, 11529, Taiwan.
- Taiwan International Graduate Program in Interdisciplinary Neuroscience, National Yang-Ming Chao-Tung University and Academia Sinica, Taipei, 11529, Taiwan.
- Neuroscience Program of Academia Sinica, Academia Sinica, Taipei, 11529, Taiwan.
| |
Collapse
|
11
|
Battista M, Carelli V, Bottazzi L, Bandello F, Cascavilla ML, Barboni P. Gene therapy for Leber hereditary optic neuropathy. Expert Opin Biol Ther 2024; 24:521-528. [PMID: 38939999 DOI: 10.1080/14712598.2024.2359015] [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: 12/02/2023] [Accepted: 05/20/2024] [Indexed: 06/29/2024]
Abstract
INTRODUCTION Leber hereditary optic neuropathy (LHON) is among the most frequent inherited mitochondrial disease, causing a severe visual impairment, mostly in young-adult males. The causative mtDNA variants (the three common are m.11778 G>A/MT-ND4, m.3460 G>A/MT-ND1, and m.14484T>C/MT-ND6) by affecting complex I impair oxidative phosphorylation in retinal ganglion cells, ultimately leading to irreversible cell death and consequent functional loss. The gene therapy based on allotopic expression of a wild-type transgene carried by adeno-associated viral vectors (AVV-based) appears a promising approach in mitochondrial disease and its efficacy has been explored in several large clinical trials. AREAS COVERED The review work employed basic concepts in mitochondrial diseases, LHON, and gene therapy procedures. Reports from completed trials in LHON (i.e. RESCUE) were reviewed and critically compared. EXPERT OPINION New challenges, as the improvement of the contralateral untreated eye or the apparently better outcome in patients treated in later stages (6-12 months), were highlighted by the latest gene therapy trials. A better understanding of the pathogenetic mechanisms of the disease together with combined therapy (medical and gene therapy) and optimization in genetic correction approaches could improve the visual outcome of treated eyes.
Collapse
Affiliation(s)
- Marco Battista
- Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Valerio Carelli
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
- Programma di Neurogenetica, IRCCS Istituto di Scienze Neurologiche di Bologna, Bologna, Italy
| | - Leonardo Bottazzi
- Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Francesco Bandello
- Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Maria Lucia Cascavilla
- Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
| | - Piero Barboni
- Department of Ophthalmology, University Vita-Salute, IRCCS Ospedale San Raffaele, Milan, Italy
- Studio Oculistico d'Azeglio, Bologna, Italy
| |
Collapse
|
12
|
Bragg-Gonzalo L, Aguilera A, González-Arias C, De León Reyes NS, Sánchez-Cruz A, Carballeira P, Leroy F, Perea G, Nieto M. Early cortical GABAergic interneurons determine the projection patterns of L4 excitatory neurons. SCIENCE ADVANCES 2024; 10:eadj9911. [PMID: 38728406 PMCID: PMC11086621 DOI: 10.1126/sciadv.adj9911] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Accepted: 04/09/2024] [Indexed: 05/12/2024]
Abstract
During cerebral cortex development, excitatory pyramidal neurons (PNs) establish specific projection patterns while receiving inputs from GABAergic inhibitory interneurons (INs). Whether these inhibitory inputs can shape PNs' projection patterns is, however, unknown. While layer 4 (L4) PNs of the primary somatosensory (S1) cortex are all born as long-range callosal projection neurons (CPNs), most of them acquire local connectivity upon activity-dependent elimination of their interhemispheric axons during postnatal development. Here, we demonstrate that precise developmental regulation of inhibition is key for the retraction of S1L4 PNs' callosal projections. Ablation of somatostatin INs leads to premature inhibition from parvalbumin INs onto S1L4 PNs and prevents them from acquiring their barrel-restricted local connectivity pattern. As a result, adult S1L4 PNs retain interhemispheric projections responding to tactile stimuli, and the mice lose whisker-based texture discrimination. Overall, we show that temporally ordered IN activity during development is key to shaping local ipsilateral S1L4 PNs' projection pattern, which is required for fine somatosensory processing.
Collapse
Affiliation(s)
- Lorena Bragg-Gonzalo
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid 28049, Spain
| | - Alfonso Aguilera
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid 28049, Spain
| | - Candela González-Arias
- Functional and Systems Neurobiology Department, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain
| | - Noelia S. De León Reyes
- Instituto de Neurociencias (CSIC-UMH), Av. Ramón y Cajal s/n, San Juan de Alicante, Alicante, Spain
| | - Alonso Sánchez-Cruz
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid 28049, Spain
| | - Paula Carballeira
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid 28049, Spain
| | - Félix Leroy
- Instituto de Neurociencias (CSIC-UMH), Av. Ramón y Cajal s/n, San Juan de Alicante, Alicante, Spain
| | - Gertrudis Perea
- Functional and Systems Neurobiology Department, Instituto Cajal, Consejo Superior de Investigaciones Científicas, Madrid 28002, Spain
| | - Marta Nieto
- Department of Molecular and Cellular Biology, Centro Nacional de Biotecnología, Consejo Superior de Investigaciones Científicas (CNB-CSIC), Madrid 28049, Spain
| |
Collapse
|
13
|
Berkowitz ST, Finn AP. Gene therapy for age-related macular degeneration: potential, feasibility, and pitfalls. Curr Opin Ophthalmol 2024; 35:170-177. [PMID: 38441066 DOI: 10.1097/icu.0000000000001043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
PURPOSE OF REVIEW The landscape for age-related macular degeneration (AMD) is rapidly changing with addition of biosimilars and now United States Food and Drug Administration (FDA) approved nonneovascular AMD (nnAMD) treatment options. These developments have inspired a burgeoning pipeline of gene therapy approaches focused on similar antivascular endothelial growth factors (VEGF) and complement related pathways. Historic and more recent setbacks in the gene therapy pipeline, including intraocular inflammatory reactions, have raised important concerns for adverse events related to AMD therapeutics both for gene and nongene approaches. The specific clinical profile of these therapeutics approaching later stage clinical trials are complex and under active investigation; however, these options hold promise to disrupt the current landscape and change management paradigms for one of the leading causes of vision loss worldwide. RECENT FINDINGS This review covers current gene therapy approaches for neovascular AMD (nAMD) and nnAMD. Intravitreal, suprachoroidal, and subretinal delivery routes are discussed with attention to technical procedure, capabilities for transgene delivery to target tissue, immunogenicity, and collateral effects. Suprachoroidal delivery is an emerging approach which may bridge some of the practical drawbacks for intravitreal and subretinal methods, though with less elaborated immunologic profile. In parallel to delivery modification, viral vectors have been cultivated to target specific cells, with promising enhancements in adeno-associated viral (AAV) vectors and persistent interest in alternate viral and nonviral delivery vectors. Ongoing questions such as steroid or immunosuppressive regimen and economic considerations from a payer and societal perspective are discussed. SUMMARY The present review discusses emerging gene therapy options which could foster new, more durable nAMD and nnAMD therapeutics. These options will need refinement with regards to route, vector, and dosage, and specialists must decipher the specific clinical risk benefit profile for individual patients. Ongoing concerns for immunogenicity or dosage related adverse events could stifle progress, while further vector development and refined delivery techniques have the potential to change the safety and efficacy of currently options in the pipeline.
Collapse
Affiliation(s)
- Sean T Berkowitz
- Vanderbilt University Medical Center, Department of Ophthalmology, Nashville, Tennessee, USA
| | | |
Collapse
|
14
|
Kulkarni A, Chen T, Sidransky E, Han TU. Advancements in Viral Gene Therapy for Gaucher Disease. Genes (Basel) 2024; 15:364. [PMID: 38540423 PMCID: PMC10970163 DOI: 10.3390/genes15030364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 06/14/2024] Open
Abstract
Gaucher disease, an autosomal recessively inherited lysosomal storage disorder, results from biallelic mutations in the GBA1 gene resulting in deficient activity of the enzyme glucocerebrosidase. In Gaucher disease, the reduced levels and activity of glucocerebrosidase lead to a disparity in the rates of formation and breakdown of glucocerebroside and glucosylsphingosine, resulting in the accumulation of these lipid substrates in the lysosome. This gives rise to the development of Gaucher cells, engorged macrophages with a characteristic wrinkled tissue paper appearance. There are both non-neuronopathic (type 1) and neuronopathic (types 2 and 3) forms of Gaucher disease, associated with varying degrees of severity. The visceral and hematologic manifestations of Gaucher disease respond well to both enzyme replacement therapy and substrate reduction therapy. However, these therapies do not improve the neuronopathic manifestations, as they cannot cross the blood-brain barrier. There is now an established precedent for treating lysosomal storage disorders with gene therapy strategies, as many have the potential to cross into the brain. The range of the gene therapies being employed is broad, but this review aimed to discuss the progress, advances, and challenges in developing viral gene therapy as a treatment for Gaucher disease.
Collapse
Affiliation(s)
| | | | - Ellen Sidransky
- Section on Molecular Neurogenetics, Medical Genetics Branch, National Human Genome Research Institute, Building 35A, Room 1E623, 35A Convent Drive, MSC 3708, Bethesda, MD 20892-3708, USA; (A.K.); (T.C.); (T.-U.H.)
| | | |
Collapse
|
15
|
Li W, Feng SL, Herrschaft L, Samulski RJ, Li C. Rationally engineered novel AAV capsids for intra-articular gene delivery. Mol Ther Methods Clin Dev 2024; 32:101211. [PMID: 38435130 PMCID: PMC10907215 DOI: 10.1016/j.omtm.2024.101211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 02/12/2024] [Indexed: 03/05/2024]
Abstract
Intra-articular adeno-associated virus (AAV) gene therapy has been explored as a potential strategy for joint diseases. However, concerns of low transduction efficacy, off-target expression, and neutralizing antibodies (Nabs) still need to be addressed. In this study, we demonstrated that AAV6 was the best serotype to transduce joints after screening serotypes 1 to 9. To develop a more effective AAV vector, a set of novel AAV capsids were rationally engineered. The mutant AAV62 created by swapping variable region I (VRI) of AAV2 into AAV6 induced a higher transduction efficiency per AAV genome copy number. To further investigate the roles of specific amino acids in the transduction of AAV62 and AAV6, we found out that AAV6D with the deletion of threonine at residue 265 induced a 2-fold higher transduction than AAV6, while the transduction efficiency from AAV6M with the mutation of alanine to glutamine at residue 263 was 10-fold lower. AAV6D efficiently transduced both synoviocytes and chondrocytes with low AAV genome copy numbers in other tissues and less Nab formation. This study demonstrates that novel AAV mutants with rational engineering may enhance joint transduction after intra-articular administration in mice, with the potential to evade AAV Nabs and minimize off-target effects in the liver.
Collapse
Affiliation(s)
- Wenjun Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Division of Oral and Craniofacial Biomedicine, University of North Carolina Adams School of Dentistry, Chapel Hill, NC, USA
| | - Susi Liu Feng
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Lizette Herrschaft
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - R. Jude Samulski
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
| | - Chengwen Li
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA
- Carolina Institute for Developmental Disabilities, University of North Carolina at Chapel Hill, Chapel Hill, NC 27510, USA
| |
Collapse
|
16
|
Hayashi Y, Sehara Y, Watano R, Ohba K, Takayanagi Y, Sakiyama Y, Muramatsu K, Mizukami H. Therapeutic Strategy for Fabry Disease by Intravenous Administration of Adeno-Associated Virus 9 in a Symptomatic Mouse Model. Hum Gene Ther 2024; 35:192-201. [PMID: 38386497 DOI: 10.1089/hum.2023.106] [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] [Indexed: 02/24/2024] Open
Abstract
Fabry disease (FD) is an inherited lysosomal storage disease caused by deficiency of α-galactosidase A (α-Gal A), an enzyme that hydrolyzes glycosphingolipids in lysosome. Accumulation of glycosphingolipids, mainly globotriaosylceramide (Gb3) in tissues, induces cellular dysfunction leading to multi-organ disorder. Gene therapy is a promising strategy that can overcome these problems, and virus vectors such as adeno-associated virus (AAV) have been used for study on gene therapy. We used human Gb3 synthetase-transgenic (TgG3S)/α-Gal A knockout (GLAko) mice. TgG3S/GLAko mice have elevated Gb3 accumulation in the major organs compared with GLAko mice, which have been widely used as a model for FD. At the age of 6 weeks, male TgG3S/GLAko were injected with 2 × 1012 vector genome AAV9 vectors containing human α-Gal A cDNA. Eight weeks after intravenous injection of AAV, α-Gal A enzymatic activity was elevated in the plasma, heart, and liver of TgG3S/GLAko mice to levels corresponding to 224%, 293%, and 105% of wild-type, respectively. Gb3 amount 8 weeks after AAV injection in the heart and liver of this group was successfully reduced to levels corresponding to 16% and 3% of untreated TgG3S/GLAko mice. Although the brain and kidney of AAV9-treated TgG3S/GLAko mice showed no significant increases in α-Gal A activity, Gb3 amount was smaller than untreated littermates (48% and 44%, respectively). In this study, systemic AAV administration did not show significant extension of the lifespan of TgG3S/GLAko mice compared with the untreated littermates. The timing of AAV injection, capsid choice, administration route, and injection volume may be important to achieve sufficient expression of α-Gal A in the whole body for the amelioration of lifespan.
Collapse
Affiliation(s)
- Yuka Hayashi
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
- Department of Neurology, Jichi Medical University Saitama Medical Center, Omiya, Japan
| | - Yoshihide Sehara
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Ryota Watano
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Kenji Ohba
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| | - Yuki Takayanagi
- Division of Brain and Neurophysiology, Department of Physiology, Jichi Medical University, Shimotsuke, Japan
| | - Yoshio Sakiyama
- Department of Neurology, Jichi Medical University Saitama Medical Center, Omiya, Japan
| | | | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Japan
| |
Collapse
|
17
|
Soto JS, Jami-Alahmadi Y, Wohlschlegel JA, Khakh BS. In vivo identification of astrocyte and neuron subproteomes by proximity-dependent biotinylation. Nat Protoc 2024; 19:896-927. [PMID: 38062165 DOI: 10.1038/s41596-023-00923-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 10/03/2023] [Indexed: 02/08/2024]
Abstract
The central nervous system (CNS) comprises diverse and morphologically complex cells. To understand the molecular basis of their physiology, it is crucial to assess proteins expressed within intact cells. Commonly used methods utilize cell dissociation and sorting to isolate specific cell types such as neurons and astrocytes, the major CNS cells. Proteins purified from isolated cells are identified by mass spectrometry-based proteomics. However, dissociation and cell-sorting methods lead to near total loss of cellular morphology, thereby losing proteins from key relevant subcompartments such as processes, end feet, dendrites and axons. Here we provide a systematic protocol for cell- and subcompartment-specific labeling and identification of proteins found within intact astrocytes and neurons in vivo. This protocol utilizes the proximity-dependent biotinylation system BioID2, selectively expressed in either astrocytes or neurons, to label proximal proteins in a cell-specific manner. BioID2 is targeted genetically to assess the subproteomes of subcellular compartments such as the plasma membrane and sites of cell-cell contacts. We describe in detail the expression methods (variable timing), stereotaxic surgeries for expression (1-2 d and then 3 weeks), in vivo protein labeling (7 d), protein isolation (2-3 d), protein identification methods (2-3 d) and data analysis (1 week). The protocol can be applied to any area of the CNS in mouse models of physiological processes and for disease-related research.
Collapse
Affiliation(s)
- Joselyn S Soto
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| | - Yasaman Jami-Alahmadi
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - James A Wohlschlegel
- Department of Biological Chemistry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Baljit S Khakh
- Department of Physiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|
18
|
Daci R, Flotte TR. Delivery of Adeno-Associated Virus Vectors to the Central Nervous System for Correction of Single Gene Disorders. Int J Mol Sci 2024; 25:1050. [PMID: 38256124 PMCID: PMC10816966 DOI: 10.3390/ijms25021050] [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: 11/30/2023] [Revised: 12/26/2023] [Accepted: 01/05/2024] [Indexed: 01/24/2024] Open
Abstract
Genetic disorders of the central nervous system (CNS) comprise a significant portion of disability in both children and adults. Several preclinical animal models have shown effective adeno-associated virus (AAV) mediated gene transfer for either treatment or prevention of autosomal recessive genetic disorders. Owing to the intricacy of the human CNS and the blood-brain barrier, it is difficult to deliver genes, particularly since the expression of any given gene may be required in a particular CNS structure or cell type at a specific time during development. In this review, we analyzed delivery methods for AAV-mediated gene therapy in past and current clinical trials. The delivery routes analyzed were direct intraparenchymal (IP), intracerebroventricular (ICV), intra-cisterna magna (CM), lumbar intrathecal (IT), and intravenous (IV). The results demonstrated that the dose used in these routes varies dramatically. The average total doses used were calculated and were 1.03 × 1013 for IP, 5.00 × 1013 for ICV, 1.26 × 1014 for CM, and 3.14 × 1014 for IT delivery. The dose for IV delivery varies by patient weight and is 1.13 × 1015 IV for a 10 kg infant. Ultimately, the choice of intervention must weigh the risk of an invasive surgical procedure to the toxicity and immune response associated with a high dose vector.
Collapse
Affiliation(s)
- Rrita Daci
- Department of Neurosurgery, University of Massachusetts Chan Medical School, 55 N Lake Ave, Worcester, MA 01655, USA;
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
| | - Terence R. Flotte
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, 368 Plantation Street, Worcester, MA 01605, USA
- Department of Pediatrics, University of Massachusetts Chan Medical School, 55 N Lake Ave, Worcester, MA 01655, USA
| |
Collapse
|
19
|
Xu F, Liu Q. Virus-Based Neural Circuit Tracing. ADVANCES IN NEUROBIOLOGY 2024; 41:113-131. [PMID: 39589712 DOI: 10.1007/978-3-031-69188-1_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/27/2024]
Abstract
Neural circuits provide an anatomical basis for functional networks. Therefore, dissecting the structure of neural circuits is an indispensable prerequisite to understanding how the brain functions. Knowing how the neural circuits organize and function under physiological conditions and their progressive alterations under pathophysiological conditions are key to understanding the underlying circuit mechanism of diseases, thus finding cures for the diseases. Recombinant neurotropic viruses are important tools for neural circuit tracing with many advantages over non-viral tracers: they allow for anterograde, retrograde, and trans-synaptic delivery of tracers in a cell-type-specific, circuit-selective manner. We herein summarize the recent developments in the viral tools for neural circuit tracing, discuss the key principles of using viral tools in neuroscience research, and highlight innovations for developing and optimizing viral tools for neural circuit tracing across diverse animal species, including nonhuman primates.
Collapse
Affiliation(s)
- Fuqiang Xu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, China.
| | - Qing Liu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen, Guangdong, China
| |
Collapse
|
20
|
Day JW, Mendell JR, Burghes AH, van Olden RW, Adhikary RR, Dilly KW. Adeno-associated virus serotype 9 antibody seroprevalence for patients in the United States with spinal muscular atrophy. Mol Ther Methods Clin Dev 2023; 31:101117. [PMID: 37822718 PMCID: PMC10562739 DOI: 10.1016/j.omtm.2023.101117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2023] [Accepted: 09/14/2023] [Indexed: 10/13/2023]
Abstract
Onasemnogene abeparvovec is a recombinant adeno-associated virus serotype 9 (AAV9) vector-based gene therapy for spinal muscular atrophy (SMA). Patients with elevated titers of anti-AAV9 antibodies (AAV9-Ab) should not receive onasemnogene abeparvovec because of potential safety and efficacy implications. We conducted a retrospective study to describe the seroprevalence of anti-AAV9 binding antibodies for pediatric patients with SMA in the United States. At initial testing, 13.0% (115 of 882) of patients (mean [SD] age, 26.29 [33.66] weeks) had elevated AAV9-Ab titers. The prevalence of elevated titers decreased as age increased, with 18.2% (92 of 507) of patients ≤3 months old but only 1.1% (1 of 92) of patients ≥21 months old having elevated titers. This suggests transplacental maternal transfer of antibodies. No patterns of geographic variations in AAV9-Ab prevalence were confirmed. Elevated AAV9-Ab titers in children <6 weeks old decreased in all circumstances. Lower magnitudes of elevated titers declined more rapidly than greater magnitudes. Retesting was completed at the discretion of the treating clinician, so age at testing and time between tests varied. AAV9-Ab retesting should be considered when patients have elevated titers, and elevations at a young age are not a deterrent to eventual onasemnogene abeparvovec administration. Early disease-modifying treatment for SMA leads to optimal outcomes.
Collapse
Affiliation(s)
- John W. Day
- Department of Neurology, Stanford University Medical Center, Stanford, CA, USA
| | - Jerry R. Mendell
- Center for Gene Therapy, Nationwide Children’s Hospital, Columbus, OH, USA
- Department of Pediatrics and Department of Neurology, The Ohio State University, Columbus, OH, USA
| | - Arthur H.M. Burghes
- Department of Neurology and Department of Biological Chemistry and Pharmacology, The Ohio State University, Columbus, OH, USA
| | | | - Rishi R. Adhikary
- CONEXTS-Real World Evidence, Novartis Healthcare Private Limited, Hyderabad, India
| | | |
Collapse
|
21
|
Aweidah H, Xi Z, Sahel JA, Byrne LC. PRPF31-retinitis pigmentosa: Challenges and opportunities for clinical translation. Vision Res 2023; 213:108315. [PMID: 37714045 PMCID: PMC10872823 DOI: 10.1016/j.visres.2023.108315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/17/2023]
Abstract
Mutations in pre-mRNA processing factor 31 cause autosomal dominant retinitis pigmentosa (PRPF31-RP), for which there is currently no efficient treatment, making this disease a prime target for the development of novel therapeutic strategies. PRPF31-RP exhibits incomplete penetrance due to haploinsufficiency, in which reduced levels of gene expression from the mutated allele result in disease. A variety of model systems have been used in the investigation of disease etiology and therapy development. In this review, we discuss recent advances in both in vivo and in vitro model systems, evaluating their advantages and limitations in the context of therapy development for PRPF31-RP. Additionally, we describe the latest approaches for treatment, including AAV-mediated gene augmentation, genome editing, and late-stage therapies such as optogenetics, cell transplantation, and retinal prostheses.
Collapse
Affiliation(s)
- Hamzah Aweidah
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Zhouhuan Xi
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Ophthalmology, Eye Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Leah C Byrne
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Neurobiology, University of Pittsburgh, Pittsburgh, PA, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
22
|
Boylan MA, Pincetic A, Romano G, Tatton N, Kenkare-Mitra S, Rosenthal A. Targeting Progranulin as an Immuno-Neurology Therapeutic Approach. Int J Mol Sci 2023; 24:15946. [PMID: 37958929 PMCID: PMC10647331 DOI: 10.3390/ijms242115946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/25/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Immuno-neurology is an emerging therapeutic strategy for dementia and neurodegeneration designed to address immune surveillance failure in the brain. Microglia, as central nervous system (CNS)-resident myeloid cells, routinely perform surveillance of the brain and support neuronal function. Loss-of-function (LOF) mutations causing decreased levels of progranulin (PGRN), an immune regulatory protein, lead to dysfunctional microglia and are associated with multiple neurodegenerative diseases, including frontotemporal dementia caused by the progranulin gene (GRN) mutation (FTD-GRN), Alzheimer's disease (AD), Parkinson's disease (PD), limbic-predominant age-related transactivation response deoxyribonucleic acid binding protein 43 (TDP-43) encephalopathy (LATE), and amyotrophic lateral sclerosis (ALS). Immuno-neurology targets immune checkpoint-like proteins, offering the potential to convert aging and dysfunctional microglia into disease-fighting cells that counteract multiple disease pathologies, clear misfolded proteins and debris, promote myelin and synapse repair, optimize neuronal function, support astrocytes and oligodendrocytes, and maintain brain vasculature. Several clinical trials are underway to elevate PGRN levels as one strategy to modulate the function of microglia and counteract neurodegenerative changes associated with various disease states. If successful, these and other immuno-neurology drugs have the potential to revolutionize the treatment of neurodegenerative disorders by harnessing the brain's immune system and shifting it from an inflammatory/pathological state to an enhanced physiological/homeostatic state.
Collapse
Affiliation(s)
| | | | | | | | | | - Arnon Rosenthal
- Alector, Inc., 131 Oyster Point Blvd, Suite 600, South San Francisco, CA 94080, USA
| |
Collapse
|
23
|
Yang X, Zhang Y, Liu Y, Wang Y, Zhou N. Fluorescence imaging of peripheral nerve function and structure. J Mater Chem B 2023; 11:10052-10071. [PMID: 37846619 DOI: 10.1039/d3tb01927f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2023]
Abstract
Peripheral nerve injuries are common and can cause catastrophic consequences. Although peripheral nerves have notable regenerative capacity, full functional recovery is often challenging due to a number of factors, including age, the type of injury, and delayed healing, resulting in chronic disorders that cause lifelong miseries and significant financial burdens. Fluorescence imaging, among the various techniques, may be the key to overcome these restrictions and improve the prognosis because of its feasibility and dynamic real-time imaging. Intraoperative dynamic fluorescence imaging allows the visualization of the morphological structure of the nerve so that surgeons can reduce the incidence of medically induced injury. Axoplasmic transport-based neuroimaging allows the visualization of the internal transport function of the nerve, facilitating early, objective, and accurate assessment of the degree of regenerative repair, allowing early intervention in patients with poor recovery, thereby improving prognosis. This review briefly discusses peripheral nerve fluorescent dyes that have been reported or could potentially be employed, with a focus on their role in visualizing the nerve's function and anatomy.
Collapse
Affiliation(s)
- Xiaoqi Yang
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China.
| | - Yumin Zhang
- Department of Geriatric Endocrinology, The First Hospital Affiliated to Nanjing Medical University, Nanjing, Jiangsu 210029, P. R. China
| | - Yadong Liu
- Department of Spinal Surgery, The First Hospital of Jilin University, Jilin Engineering Research Center For Spine and Spinal Cord Injury, 1 Xinmin St, Changchun, 130021, China.
| | - Yuanyi Wang
- Department of Spinal Surgery, The First Hospital of Jilin University, Jilin Engineering Research Center For Spine and Spinal Cord Injury, 1 Xinmin St, Changchun, 130021, China.
| | - Nan Zhou
- Department of Orthopedics, The First Affiliated Hospital of Zhengzhou University, No. 1 Jianshe East Road, Zhengzhou 450052, Henan, China.
| |
Collapse
|
24
|
Abstract
The neural retina of mammals, like most of the rest of the central nervous system, does not regenerate new neurons after they are lost through damage or disease. The ability of nonmammalian vertebrates, like fish and amphibians, is remarkable, and lessons learned over the last 20 years have revealed some of the mechanisms underlying this potential. This knowledge has recently been applied to mammals to develop methods that can stimulate regeneration in mice. In this review, we highlight the progress in this area, and propose a "wish list" of how the clinical implementation of regenerative strategies could be applicable to various human retinal diseases.
Collapse
Affiliation(s)
- Marina Pavlou
- Department of Biological Structure, University of Washington School of Medicine, Institute of Stem Cells and Regenerative Medicine, Seattle, Washington 98195, USA
| | - Thomas A Reh
- Department of Biological Structure, University of Washington School of Medicine, Institute of Stem Cells and Regenerative Medicine, Seattle, Washington 98195, USA
| |
Collapse
|
25
|
Liu B, Li Y, Ren M, Li X. Targeted approaches to delineate neuronal morphology during early development. Front Cell Neurosci 2023; 17:1259360. [PMID: 37854514 PMCID: PMC10579594 DOI: 10.3389/fncel.2023.1259360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2023] [Accepted: 09/18/2023] [Indexed: 10/20/2023] Open
Abstract
Understanding the developmental changes that affect neurons is a key step in exploring the assembly and maturation of neural circuits in the brain. For decades, researchers have used a number of labeling techniques to visualize neuronal morphology at different stages of development. However, the efficiency and accuracy of neuronal labeling technologies are limited by the complexity and fragility of neonatal brains. In this review, we illustrate the various labeling techniques utilized for examining the neurogenesis and morphological changes occurring during the early stages of development. We compare the advantages and limitations of each technique from different aspects. Then, we highlight the gaps remaining in our understanding of the structure of neurons in the neonatal mouse brain.
Collapse
Affiliation(s)
- Bimin Liu
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Yuxiao Li
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Miao Ren
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Xiangning Li
- State Key Laboratory of Digital Medical Engineering, School of Biomedical Engineering, Hainan University, Haikou, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
- Research Unit of Multimodal Cross Scale Neural Signal Detection and Imaging, Chinese Academy of Medical Sciences, HUST-Suzhou Institute for Brainsmatics, JITRI, Suzhou, China
| |
Collapse
|
26
|
Amato A, Wongchaisuwat N, Lamborn A, Schmidt R, Everett L, Yang P, Pennesi ME. Gene therapy in bestrophinopathies: Insights from preclinical studies in preparation for clinical trials. Saudi J Ophthalmol 2023; 37:287-295. [PMID: 38155675 PMCID: PMC10752275 DOI: 10.4103/sjopt.sjopt_175_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 12/30/2023] Open
Abstract
The BEST1 gene encodes bestrophin-1, a homopentameric ion channel expressed in the retinal pigment epithelium (RPE), where it localizes to the basolateral plasma membrane. Pathogenic variants in this gene can cause different autosomal dominant and recessive inherited retinal diseases (IRDs), collectively named "bestrophinopathies." These disorders share a number of clinical and molecular features that make them an appealing target for gene therapy. Clinically, bestrophinopathies are often slowly progressive with a wide window of opportunity, and the presence of subretinal material (vitelliform deposits and/or fluid) as a hallmark of these conditions provides an easily quantifiable endpoint in view of future clinical trials. From a molecular standpoint, most BEST1 pathogenic variants have been shown to cause either loss of function (LOF) of the protein or a dominant-negative (DN) effect, with a smaller subset causing a toxic gain of function (GOF). Both LOF and DN mutations may be amenable to gene augmentation alone. On the other hand, individuals harboring GOF variants would require a combination of gene silencing and gene augmentation, which has been shown to be effective in RPE cells derived from patients with Best disease. In this article, we review the current knowledge of BEST1-related IRDs and we discuss how their molecular and clinical features are being used to design novel and promising therapeutic strategies.
Collapse
Affiliation(s)
- Alessia Amato
- Department of Ophthalmic Genetics, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Nida Wongchaisuwat
- Department of Ophthalmic Genetics, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Andrew Lamborn
- Department of Ophthalmic Genetics, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Ryan Schmidt
- Department of Ophthalmic Genetics, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Lesley Everett
- Department of Ophthalmic Genetics, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Paul Yang
- Department of Ophthalmic Genetics, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| | - Mark E. Pennesi
- Department of Ophthalmic Genetics, Casey Eye Institute, Oregon Health and Science University, Portland, Oregon, United States of America
| |
Collapse
|
27
|
Basurco L, Abellanas MA, Ayerra L, Conde E, Vinueza-Gavilanes R, Luquin E, Vales A, Vilas A, Martin-Uriz PS, Tamayo I, Alonso MM, Hernaez M, Gonzalez-Aseguinolaza G, Clavero P, Mengual E, Arrasate M, Hervás-Stubbs S, Aymerich MS. Microglia and astrocyte activation is region-dependent in the α-synuclein mouse model of Parkinson's disease. Glia 2023; 71:571-587. [PMID: 36353934 PMCID: PMC10100513 DOI: 10.1002/glia.24295] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 10/16/2022] [Accepted: 10/21/2022] [Indexed: 11/11/2022]
Abstract
Inflammation is a common feature in neurodegenerative diseases that contributes to neuronal loss. Previously, we demonstrated that the basal inflammatory tone differed between brain regions and, consequently, the reaction generated to a pro-inflammatory stimulus was different. In this study, we assessed the innate immune reaction in the midbrain and in the striatum using an experimental model of Parkinson's disease. An adeno-associated virus serotype 9 expressing the α-synuclein and mCherry genes or the mCherry gene was administered into the substantia nigra. Myeloid cells (CD11b+ ) and astrocytes (ACSA2+ ) were purified from the midbrain and striatum for bulk RNA sequencing. In the parkinsonian midbrain, CD11b+ cells presented a unique anti-inflammatory transcriptomic profile that differed from degenerative microglia signatures described in experimental models for other neurodegenerative conditions. By contrast, striatal CD11b+ cells showed a pro-inflammatory state and were similar to disease-associated microglia. In the midbrain, a prominent increase of infiltrated monocytes/macrophages was observed and, together with microglia, participated actively in the phagocytosis of dopaminergic neuronal bodies. Although striatal microglia presented a phagocytic transcriptomic profile, morphology and cell density was preserved and no active phagocytosis was detected. Interestingly, astrocytes presented a pro-inflammatory fingerprint in the midbrain and a low number of differentially displayed transcripts in the striatum. During α-synuclein-dependent degeneration, microglia and astrocytes experience context-dependent activation states with a different contribution to the inflammatory reaction. Our results point towards the relevance of selecting appropriate cell targets to design neuroprotective strategies aimed to modulate the innate immune system during the active phase of dopaminergic degeneration.
Collapse
Affiliation(s)
- Leyre Basurco
- Departamentode Bioquímica y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain.,Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Miguel Angel Abellanas
- Departamentode Bioquímica y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain.,Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Leyre Ayerra
- Departamentode Bioquímica y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain.,Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Enrique Conde
- Programa de Inmunología, CIMA-Universidad de Navarra, Pamplona, Spain
| | | | - Esther Luquin
- Departamento de Patología, Anatomía y Fisiología, Facultad de Medicina, Universidad de Navarra, Pamplona, Spain
| | - Africa Vales
- Programa de Terapia Génica, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Amaya Vilas
- Programa de Oncohematología, CIMA-Universidad de Navarra, Pamplona, Spain
| | | | - Ibon Tamayo
- Programa de Biología Computacional, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Marta M Alonso
- Programa de Tumores Sólidos, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| | - Mikel Hernaez
- Programa de Biología Computacional, CIMA-Universidad de Navarra, Pamplona, Spain
| | - Gloria Gonzalez-Aseguinolaza
- Programa de Terapia Génica, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| | - Pedro Clavero
- Servicio de Neurología, Complejo Hospitalario de Navarra, Pamplona, Spain
| | - Elisa Mengual
- Departamento de Patología, Anatomía y Fisiología, Facultad de Medicina, Universidad de Navarra, Pamplona, Spain
| | - Montserrat Arrasate
- Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| | - Sandra Hervás-Stubbs
- Programa de Inmunología, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| | - Maria S Aymerich
- Departamentode Bioquímica y Genética, Facultad de Ciencias, Universidad de Navarra, Pamplona, Spain.,Programa de Neurociencias, CIMA-Universidad de Navarra, Pamplona, Spain.,Programa de Tumores Sólidos, CIMA-Universidad de Navarra, Pamplona, Spain.,Neurociencias y Salud Mental, Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain
| |
Collapse
|
28
|
Calvo-López T, Grueso E, Sánchez-Martínez C, Almendral JM. Intracellular virion traffic to the endosome driven by cell type specific sialic acid receptors determines parvovirus tropism. Front Microbiol 2023; 13:1063706. [PMID: 36756201 PMCID: PMC9899843 DOI: 10.3389/fmicb.2022.1063706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/28/2022] [Indexed: 01/24/2023] Open
Abstract
Parvoviruses are promising anticancer and gene therapy agents, but a deep knowledge of the entry process is crucial to exploit their therapeutic potential. We addressed this issue while attempting to retarget the oncolytic parvovirus minute virus of mice (MVMp) to the tumor vasculature. Residues at three functional domains of the icosahedral capsid were substituted by rational design with peptides competing with the vascular endothelial growth factor. Most substitutions impaired virus maturation, though some yielded infectious chimeric virions, and substitutions in a dimple at the twofold axis that allocates sialic acid (SIA) receptors altered viral tropism. One dimple-modified chimeric virion was efficiently attached as MVMp to α2-linked SIA moieties, but the infection was impaired by the binding to some inhibitory α2-3,-6,-8 SIA pseudoreceptors, which hampers intracellular virus traffic to the endosome in a cell type-dependent manner. Infectious from nonproductive traffic could be mechanistically discriminated by an endosomal drastic capsid structural transition comprising the cleavage of some VP2-Nt sequences and its associated VP1-Nt exposure. Correspondingly, neuraminidase removal of inhibitory SIA moieties enhanced the infection quantitatively, correlating to the restored virus traffic to the endosome and the extent of VP2-Nt cleavage/VP1-Nt exposure. This study illustrates (i) structural constraints to retarget parvoviruses with evolutionary adopted narrow grooves allocating small SIA receptors, (ii) the possibility to enhance parvovirus oncolysis by relaxing the glycan network on the cancer cell surface, and (iii) the major role played by the attachment to cell type-specific SIAs in the intracellular virus traffic to the endosome, which may determine parvovirus tropism and host range.
Collapse
Affiliation(s)
- Tania Calvo-López
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain,Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Esther Grueso
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain,Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Sánchez-Martínez
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain,Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M. Almendral
- Centro de Biología Molecular Severo Ochoa (CSIC-UAM), Madrid, Spain,Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain,*Correspondence: José M. Almendral ✉
| |
Collapse
|
29
|
Wu WH, Tso A, Breazzano MP, Jenny LA, Levi SR, Tsang SH, Quinn PMJ. Culture of Human Retinal Explants for Ex Vivo Assessment of AAV Gene Delivery. Methods Mol Biol 2022; 2560:303-311. [PMID: 36481906 DOI: 10.1007/978-1-0716-2651-1_28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Due to the clinically established safety and efficacy profile of recombinant adeno-associated viral (rAAV) vectors, they are considered the "go to" vector for retinal gene therapy. Design of a rAAV-mediated gene therapy focuses on cell tropism, high transduction efficiency, and high transgene expression levels to achieve the lowest therapeutic treatment dosage and avoid toxicity. Human retinal explants are a clinically relevant model system for exploring these aspects of rAAV-mediated gene delivery. In this chapter, we describe an ex vivo human retinal explant culture protocol to evaluate transgene expression in order to determine the selectivity and efficacy of rAAV vectors for human retinal gene therapy.
Collapse
Affiliation(s)
- Wen-Hsuan Wu
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA
| | - Amy Tso
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA
| | - Mark P Breazzano
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA.,Department of Ophthalmology, New York University School of Medicine, New York University Langone Health, New York, NY, USA.,Manhattan Eye, Ear and Throat Hospital, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Laura A Jenny
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA.,Department of Ophthalmology, New York University School of Medicine, New York University Langone Health, New York, NY, USA.,Manhattan Eye, Ear and Throat Hospital, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Sarah R Levi
- Edward S. Harkness Eye Institute, Department of Ophthalmology, Columbia University Irving Medical Center/New York-Presbyterian Hospital, New York, NY, USA.,Department of Ophthalmology, New York University School of Medicine, New York University Langone Health, New York, NY, USA.,Manhattan Eye, Ear and Throat Hospital, Lenox Hill Hospital, Northwell Health, New York, NY, USA
| | - Stephen H Tsang
- Departments of Ophthalmology, Pathology & Cell Biology, Graduate Programs in Nutritional & Metabolic Biology and Neurobiology & Behavior, Columbia Stem Cell Initiative, New York, NY, USA
| | - Peter M J Quinn
- Department of Opthalmology, Columbia University Medical Center, New York, NY, USA.
| |
Collapse
|
30
|
Liu Q, Wu Y, Wang H, Jia F, Xu F. Viral Tools for Neural Circuit Tracing. Neurosci Bull 2022; 38:1508-1518. [PMID: 36136267 PMCID: PMC9723069 DOI: 10.1007/s12264-022-00949-z] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 06/09/2022] [Indexed: 10/14/2022] Open
Abstract
Neural circuits provide an anatomical basis for functional networks. Therefore, dissecting the structure of neural circuits is essential to understanding how the brain works. Recombinant neurotropic viruses are important tools for neural circuit tracing with many advantages over non-viral tracers: they allow for anterograde, retrograde, and trans-synaptic delivery of tracers in a cell type-specific, circuit-selective manner. In this review, we summarize the recent developments in the viral tools for neural circuit tracing, discuss the key principles of using viral tools in neuroscience research, and highlight innovations for developing and optimizing viral tools for neural circuit tracing across diverse animal species, including nonhuman primates.
Collapse
Affiliation(s)
- Qing Liu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yang Wu
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Huadong Wang
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fan Jia
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fuqiang Xu
- The Brain Cognition and Brain Disease Institute (BCBDI), Shenzhen Key Laboratory of Viral Vectors for Biomedicine, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, NMPA Key Laboratory for Research and Evaluation of Viral Vector Technology in Cell and Gene Therapy Medicinal Products, Shenzhen Key Laboratory of Quality Control Technology for Virus-Based Therapeutics, Guangdong Provincial Medical Products Administration, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- State Key Laboratory of Magnetic Resonance and Atomic and Molecular Physics, Key Laboratory of Magnetic Resonance in Biological Systems, Wuhan Center for Magnetic Resonance, Innovation Academy for Precision Measurement Science and Technology, Chinese Academy of Sciences, Wuhan, 430071, China.
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, 200031, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
31
|
Qiu L, Zhang B, Gao Z. Lighting Up Neural Circuits by Viral Tracing. Neurosci Bull 2022; 38:1383-1396. [PMID: 35578093 PMCID: PMC9672192 DOI: 10.1007/s12264-022-00860-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/23/2022] [Indexed: 12/03/2022] Open
Abstract
Neurons are highly interwoven to form intricate neural circuits that underlie the diverse functions of the brain. Dissecting the anatomical organization of neural circuits is key to deciphering how the brain processes information, produces thoughts, and instructs behaviors. Over the past decades, recombinant viral vectors have become the most commonly used tracing tools to define circuit architecture. In this review, we introduce the current categories of viral tools and their proper application in circuit tracing. We further discuss some advances in viral tracing strategy and prospective innovations of viral tools for future study.
Collapse
Affiliation(s)
- Liyao Qiu
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Bin Zhang
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China
| | - Zhihua Gao
- Department of Neurobiology and Department of Neurology of the Second Affiliated Hospital, NHC and CAMS Key Laboratory of Medical Neurobiology, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- The MOE Frontier Science Center for Brain Research and Brain-Machine Integration, School of Brain Science and Brain Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
32
|
Allen JK, Sutherland TC, Prater AR, Geoffroy CG, Pellois JP. In vivo peptide-based delivery of a gene-modifying enzyme into cells of the central nervous system. SCIENCE ADVANCES 2022; 8:eabo2954. [PMID: 36170360 PMCID: PMC9519033 DOI: 10.1126/sciadv.abo2954] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/10/2022] [Indexed: 06/16/2023]
Abstract
We report on the successful delivery of the Cre recombinase enzyme in the neural cells of mice in vivo by simple coinjection with peptides derived from HIV-TAT. Cre delivery activates the expression of a reporter gene in both neurons and astrocytes of the cortex without tissue damage and with a transduction efficiency that parallels or exceeds that of a commonly used adeno-associated virus. Our data indicate that the delivery peptides mediate efficient endosomal leakage and cytosolic escape in cells that have endocytosed Cre. The peptides, therefore, act in trans and do not require conjugation to the payload, greatly simplifying sample preparation. Moreover, the delivery peptides are exclusively composed of natural amino acids and are consequently readily degradable and processed by cells. We envision that this approach will be beneficial to applications that require the transient introduction of proteins into cells in vivo.
Collapse
Affiliation(s)
- Jason K. Allen
- Department of Biochemistry and Biophysics, and Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Theresa C. Sutherland
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Austin R. Prater
- Department of Biochemistry and Biophysics, and Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| | - Cédric G. Geoffroy
- Department of Neuroscience and Experimental Therapeutics, School of Medicine, Texas A&M University, Bryan, TX 77807, USA
| | - Jean-Philippe Pellois
- Department of Biochemistry and Biophysics, and Department of Chemistry, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
33
|
Abstract
In 2001, the first large animal was successfully treated with a gene therapy that restored its vision. Lancelot, the Briard dog that was treated, suffered from a human childhood blindness called Leber's congenital amaurosis type 2. Sixteen years later, the gene therapy was approved by the U.S. Food and Drug Administration. The success of this gene therapy in dogs led to a fast expansion of the ocular gene therapy field. By now every class of inherited retinal dystrophy has been treated in at least one animal model and many clinical trials have been initiated in humans. In this study, we review the status of viral gene therapies for the retina, with a focus on ongoing human clinical trials. It is likely that in the next decade we will see several new viral gene therapies approved.
Collapse
Affiliation(s)
- Shun-Yun Cheng
- University of Massachusetts Medical School, Ophthalmology, Worcester, Massachusetts, United States;
| | - Claudio Punzo
- University of Massachusetts Medical School, Ophthalmology, 368 Plantation Street, Albert Sherman Center, AS6-2041, Worcester, Massachusetts, United States, 01605;
| |
Collapse
|
34
|
Skukan L, Brezak M, Ister R, Klimaschewski L, Vojta A, Zoldoš V, Gajović S. Lentivirus- or AAV-mediated gene therapy interventions in ischemic stroke: A systematic review of preclinical in vivo studies. J Cereb Blood Flow Metab 2022; 42:219-236. [PMID: 34427147 PMCID: PMC8795232 DOI: 10.1177/0271678x211039997] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Due to the limited therapeutic options after ischemic stroke, gene therapy has emerged as a promising choice, especially with recent advances in viral vector delivery systems. Therefore, we aimed to provide the current state of the art of lentivirus (LV) and adeno-associated virus (AAV) mediated gene interventions in preclinical ischemic stroke models. A systematic analysis including qualitative and quantitative syntheses of studies published until December 2020 was performed. Most of the 87 selected publications used adult male rodents and the preferred stroke model was transient middle cerebral artery occlusion. LV and AAV vectors were equally used for transgene delivery, however loads of AAVs were higher than LVs. Serotypes having broad cell tropism, the use of constitutive promoters, and virus delivery before the stroke induction via stereotaxic injection in the cortex and striatum were preferred in the analyzed studies. The meta-analysis based on infarct volume as the primary outcome confirmed the efficacy of the preclinical interventions. The quality assessment exposed publication bias and setbacks in regard to risks of bias and study relevance. The translational potential could increase by using specific cell targeting, post-stroke interventions, non-invasive systematic delivery, and use of large animals.
Collapse
Affiliation(s)
- Laura Skukan
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Matea Brezak
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Rok Ister
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| | - Lars Klimaschewski
- Institute of Neuroanatomy, Medical University of Innsbruck, Innsbruck, Austria
| | - Aleksandar Vojta
- Department for Molecular Biology, University of Zagreb Faculty of Science, Zagreb, Croatia
| | - Vlatka Zoldoš
- Department for Molecular Biology, University of Zagreb Faculty of Science, Zagreb, Croatia
| | - Srećko Gajović
- Croatian Institute for Brain Research, University of Zagreb School of Medicine, Zagreb, Croatia
| |
Collapse
|
35
|
Qureshi YH, Berman DE, Marsh SE, Klein RL, Patel VM, Simoes S, Kannan S, Petsko GA, Stevens B, Small SA. The neuronal retromer can regulate both neuronal and microglial phenotypes of Alzheimer's disease. Cell Rep 2022; 38:110262. [PMID: 35045281 PMCID: PMC8830374 DOI: 10.1016/j.celrep.2021.110262] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 09/14/2021] [Accepted: 12/22/2021] [Indexed: 02/03/2023] Open
Abstract
Disruption of retromer-dependent endosomal trafficking is considered pathogenic in late-onset Alzheimer's disease (AD). Here, to investigate this disruption in the intact brain, we turn to a genetic mouse model where the retromer core protein VPS35 is depleted in hippocampal neurons, and then we replete VPS35 using an optimized viral vector protocol. The VPS35 depletion-repletion studies strengthen the causal link between the neuronal retromer and AD-associated neuronal phenotypes, including the acceleration of amyloid precursor protein cleavage and the loss of synaptic glutamate receptors. Moreover, the studies show that the neuronal retromer can regulate a distinct, dystrophic, microglia morphology, phenotypic of hippocampal microglia in AD. Finally, the neuronal and, in part, the microglia responses to VPS35 depletion were found to occur independent of tau. Showing that the neuronal retromer can regulate AD-associated pathologies in two of AD's principal cell types strengthens the link, and clarifies the mechanism, between endosomal trafficking and late-onset sporadic AD.
Collapse
Affiliation(s)
- Yasir H Qureshi
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Diego E Berman
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Samuel E Marsh
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ronald L Klein
- Department of Pharmacology, Toxicology and Neuroscience, Louisiana State University Health Sciences Center, Shreveport, LA, USA
| | - Vivek M Patel
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Sabrina Simoes
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Suvarnambiga Kannan
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA
| | - Gregory A Petsko
- Ann Romney Center for Neurologic Diseases, Department of Neurology, Harvard Medical School and Brigham and Women's Hospital, Boston, MA, USA
| | - Beth Stevens
- Boston Children's Hospital, F.M. Kirby Neurobiology Center, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Cambridge, MA, USA; Howard Hughes Medical Institute, Boston Children's Hospital, Boston, MA, USA.
| | - Scott A Small
- Departments of Neurology and the Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Columbia University, New York, NY, USA.
| |
Collapse
|
36
|
Iseppon F, Linley JE, Wood JN. Calcium imaging for analgesic drug discovery. NEUROBIOLOGY OF PAIN (CAMBRIDGE, MASS.) 2022; 11:100083. [PMID: 35079661 PMCID: PMC8777277 DOI: 10.1016/j.ynpai.2021.100083] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 12/17/2021] [Accepted: 12/27/2021] [Indexed: 11/24/2022]
Abstract
Somatosensation and pain are complex phenomena involving a rangeofspecialised cell types forming different circuits within the peripheral and central nervous systems. In recent decades, advances in the investigation of these networks, as well as their function in sensation, resulted from the constant evolution of electrophysiology and imaging techniques to allow the observation of cellular activity at the population level both in vitro and in vivo. Genetically encoded indicators of neuronal activity, combined with recent advances in DNA engineering and modern microscopy, offer powerful tools to dissect and visualise the activity of specific neuronal subpopulations with high spatial and temporal resolution. In recent years various groups developed in vivo imaging techniques to image calcium transients in the dorsal root ganglia, the spinal cord and the brain of anesthetised and awake, behaving animals to address fundamental questions in both the physiology and pathophysiology of somatosensation and pain. This approach, besides giving unprecedented details on the circuitry of innocuous and painful sensation, can be a very powerful tool for pharmacological research, from the characterisation of new potential drugs to the discovery of new, druggable targets within specific neuronal subpopulations. Here we summarise recent developments in calcium imaging for pain research, discuss technical challenges and advances, and examine the potential positive impact of this technique in early preclinical phases of the analgesic drug discovery process.
Collapse
Affiliation(s)
- Federico Iseppon
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, WC1E 6BT London, UK
- Discovery UK, Neuroscience, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - John E. Linley
- Discovery UK, Neuroscience, Biopharmaceuticals R&D, AstraZeneca, Cambridge, UK
| | - John N. Wood
- Molecular Nociception Group, Wolfson Institute for Biomedical Research, University College London, Gower Street, WC1E 6BT London, UK
| |
Collapse
|
37
|
Hamann MV, Beschorner N, Vu XK, Hauber I, Lange UC, Traenkle B, Kaiser PD, Foth D, Schneider C, Büning H, Rothbauer U, Hauber J. Improved targeting of human CD4+ T cells by nanobody-modified AAV2 gene therapy vectors. PLoS One 2021; 16:e0261269. [PMID: 34928979 PMCID: PMC8687595 DOI: 10.1371/journal.pone.0261269] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 11/26/2021] [Indexed: 12/12/2022] Open
Abstract
Adeno-associated viruses (AAV) are considered non-pathogenic in humans, and thus have been developed into powerful vector platforms for in vivo gene therapy. Although the various AAV serotypes display broad tropism, frequently infecting multiple tissues and cell types, vectors for specific and efficient targeting of human CD4+ T lymphocytes are largely missing. In fact, a substantial translational bottleneck exists in the field of therapeutic gene transfer that would require in vivo delivery into peripheral disease-related lymphocytes for subsequent genome editing. To solve this issue, capsid modification for retargeting AAV tropism, and in turn improving vector potency, is considered a promising strategy. Here, we genetically modified the minor AAV2 capsid proteins, VP1 and VP2, with a set of novel nanobodies with high-affinity for the human CD4 receptor. These novel vector variants demonstrated improved targeting of human CD4+ cells, including primary human peripheral blood mononuclear cells (PBMC) and purified human CD4+ T lymphocytes. Thus, the technical approach presented here provides a promising strategy for developing specific gene therapy vectors, particularly targeting disease-related peripheral blood CD4+ leukocytes.
Collapse
Affiliation(s)
- Martin V. Hamann
- Heinrich Pette Institute – Leibniz Institute for Experimental Virology (HPI), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg – Lübeck – Borstel – Riems, Hamburg, Germany
| | - Niklas Beschorner
- Heinrich Pette Institute – Leibniz Institute for Experimental Virology (HPI), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg – Lübeck – Borstel – Riems, Hamburg, Germany
| | - Xuan-Khang Vu
- Institute of Experimental Haematology, Hannover Medical School, Hannover, Germany
| | - Ilona Hauber
- Heinrich Pette Institute – Leibniz Institute for Experimental Virology (HPI), Hamburg, Germany
| | - Ulrike C. Lange
- Heinrich Pette Institute – Leibniz Institute for Experimental Virology (HPI), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg – Lübeck – Borstel – Riems, Hamburg, Germany
- Department of Anaesthesiology, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Bjoern Traenkle
- Natural and Medical Science Institute at the University Tübingen (NMI), Reutlingen, Germany
| | - Philipp D. Kaiser
- Natural and Medical Science Institute at the University Tübingen (NMI), Reutlingen, Germany
| | - Daniel Foth
- Heinrich Pette Institute – Leibniz Institute for Experimental Virology (HPI), Hamburg, Germany
| | - Carola Schneider
- Heinrich Pette Institute – Leibniz Institute for Experimental Virology (HPI), Hamburg, Germany
| | - Hildegard Büning
- German Center for Infection Research (DZIF), Partner Site Hamburg – Lübeck – Borstel – Riems, Hamburg, Germany
- Institute of Experimental Haematology, Hannover Medical School, Hannover, Germany
| | - Ulrich Rothbauer
- Natural and Medical Science Institute at the University Tübingen (NMI), Reutlingen, Germany
- Pharmaceutical Biotechnology, Eberhard Karls University Tübingen, Reutlingen, Germany
| | - Joachim Hauber
- Heinrich Pette Institute – Leibniz Institute for Experimental Virology (HPI), Hamburg, Germany
- German Center for Infection Research (DZIF), Partner Site Hamburg – Lübeck – Borstel – Riems, Hamburg, Germany
| |
Collapse
|
38
|
Nasir G, Chopra R, Elwood F, Ahmed SS. Krabbe Disease: Prospects of Finding a Cure Using AAV Gene Therapy. Front Med (Lausanne) 2021; 8:760236. [PMID: 34869463 PMCID: PMC8633897 DOI: 10.3389/fmed.2021.760236] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2021] [Accepted: 10/15/2021] [Indexed: 11/13/2022] Open
Abstract
Krabbe Disease (KD) is an autosomal metabolic disorder that affects both the central and peripheral nervous systems. It is caused by a functional deficiency of the lysosomal enzyme, galactocerebrosidase (GALC), resulting in an accumulation of the toxic metabolite, psychosine. Psychosine accumulation affects many different cellular pathways, leading to severe demyelination. Although there is currently no effective therapy for Krabbe disease, recent gene therapy-based approaches in animal models have indicated a promising outlook for clinical treatment. This review highlights recent findings in the pathogenesis of Krabbe disease, and evaluates AAV-based gene therapy as a promising strategy for treating this devastating pediatric disease.
Collapse
Affiliation(s)
- Gibran Nasir
- Department of Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA, United States
| | - Rajiv Chopra
- AllianThera Biopharma, Boston, MA, United States
| | - Fiona Elwood
- Department of Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA, United States
| | - Seemin S Ahmed
- Department of Neuroscience, Novartis Institutes for BioMedical Research (NIBR), Cambridge, MA, United States
| |
Collapse
|
39
|
Narcisse D, Mustafi SM, Carlson M, Batabyal S, Kim S, Wright W, Kumar Mohanty S. Bioluminescent Multi-Characteristic Opsin for Simultaneous Optical Stimulation and Continuous Monitoring of Cortical Activities. Front Cell Neurosci 2021; 15:750663. [PMID: 34759801 PMCID: PMC8573050 DOI: 10.3389/fncel.2021.750663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Accepted: 09/20/2021] [Indexed: 12/02/2022] Open
Abstract
Stimulation and continuous monitoring of neural activities at cellular resolution are required for the understanding of the sensory processing of stimuli and development of effective neuromodulation therapies. We present bioluminescence multi-characteristic opsin (bMCOII), a hybrid optogenetic actuator, and a bioluminescence Ca2+ sensor for excitation-free, continuous monitoring of neural activities in the visual cortex, with high spatiotemporal resolution. An exceptionally low intensity (10 μW/mm2) of light could elicit neural activation that could be detected by Ca2+ bioluminescence imaging. An uninterrupted (>14 h) recording of visually evoked neural activities in the cortex of mice enabled the determination of strength of sensory activation. Furthermore, an artificial intelligence-based neural activation parameter transformed Ca2+ bioluminescence signals to network activity patterns. During continuous Ca2+-bioluminescence recordings, visual cortical activity peaked at the seventh to eighth hour of anesthesia, coinciding with circadian rhythm. For both direct optogenetic stimulation in cortical slices and visually evoked activities in the visual cortex, we observed secondary delayed Ca2+-bioluminescence responses, suggesting the involvement of neuron-astrocyte-neuron pathway. Our approach will enable the development of a modular and scalable interface system capable of serving a multiplicity of applications to modulate and monitor large-scale activities in the brain.
Collapse
|
40
|
Hutt JA, Assaf BT, Bolon B, Cavagnaro J, Galbreath E, Grubor B, Kattenhorn LM, Romeike A, Whiteley LO. Scientific and Regulatory Policy Committee Points to Consider: Nonclinical Research and Development of In Vivo Gene Therapy Products, Emphasizing Adeno-Associated Virus Vectors. Toxicol Pathol 2021; 50:118-146. [PMID: 34657529 DOI: 10.1177/01926233211041962] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Sequencing of the human genome and numerous advances in molecular techniques have launched the era of genetic medicine. Increasingly precise technologies for genetic modification, manufacturing, and administration of pharmaceutical-grade biologics have proved the viability of in vivo gene therapy (GTx) as a therapeutic modality as shown in several thousand clinical trials and recent approval of several GTx products for treating rare diseases and cancers. In recognition of the rapidly advancing knowledge in this field, the regulatory landscape has evolved considerably to maintain appropriate monitoring of safety concerns associated with this modality. Nonetheless, GTx safety assessment remains complex and is designed on a case-by-case basis that is determined by the disease indication and product attributes. This article describes our current understanding of fundamental biological principles and possible procedures (emphasizing those related to toxicology and toxicologic pathology) needed to support research and development of in vivo GTx products. This article is not intended to provide comprehensive guidance on all GTx modalities but instead provides an overview relevant to in vivo GTx generally by utilizing recombinant adeno-associated virus-based GTx-the most common in vivo GTx platform-to exemplify the main points to be considered in nonclinical research and development of GTx products.
Collapse
Affiliation(s)
- Julie A Hutt
- Greenfield Pathology Services, Inc, Greenfield, IN, USA
| | - Basel T Assaf
- Drug Safety Research and Development, Pfizer Inc, Cambridge, MA, USA
| | | | | | | | - Branka Grubor
- Biogen, Preclinical Safety/Comparative Pathology, Cambridge, MA, USA
| | | | | | | |
Collapse
|
41
|
Davidson CD, Gibson AL, Gu T, Baxter LL, Deverman BE, Beadle K, Incao AA, Rodriguez-Gil JL, Fujiwara H, Jiang X, Chandler RJ, Ory DS, Gradinaru V, Venditti CP, Pavan WJ. Improved systemic AAV gene therapy with a neurotrophic capsid in Niemann-Pick disease type C1 mice. Life Sci Alliance 2021; 4:e202101040. [PMID: 34407999 PMCID: PMC8380657 DOI: 10.26508/lsa.202101040] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 11/24/2022] Open
Abstract
Niemann-Pick C1 disease (NPC1) is a rare, fatal neurodegenerative disease caused by mutations in NPC1, which encodes the lysosomal cholesterol transport protein NPC1. Disease pathology involves lysosomal accumulation of cholesterol and lipids, leading to neurological and visceral complications. Targeting the central nervous system (CNS) from systemic circulation complicates treatment of neurological diseases with gene transfer techniques. Selected and engineered capsids, for example, adeno-associated virus (AAV)-PHP.B facilitate peripheral-to-CNS transfer and hence greater CNS transduction than parental predecessors. We report that systemic delivery to Npc1 m1N/m1N mice using an AAV-PHP.B vector ubiquitously expressing NPC1 led to greater disease amelioration than an otherwise identical AAV9 vector. In addition, viral copy number and biodistribution of GFP-expressing reporters showed that AAV-PHP.B achieved more efficient, albeit variable, CNS transduction than AAV9 in Npc1 m1N/m1N mice. This variability was associated with segregation of two alleles of the putative AAV-PHP.B receptor Ly6a in Npc1 m1N/m1N mice. Our data suggest that robust improvements in NPC1 disease phenotypes occur even with modest CNS transduction and that improved neurotrophic capsids have the potential for superior NPC1 AAV gene therapy vectors.
Collapse
Affiliation(s)
- Cristin D Davidson
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alana L Gibson
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tansy Gu
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Laura L Baxter
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Benjamin E Deverman
- Division of Biology and Biological Engineering, California Institutes of Technology, Pasadena, CA, USA
| | - Keith Beadle
- Division of Biology and Biological Engineering, California Institutes of Technology, Pasadena, CA, USA
| | - Arturo A Incao
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jorge L Rodriguez-Gil
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Hideji Fujiwara
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xuntian Jiang
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Randy J Chandler
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - Daniel S Ory
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institutes of Technology, Pasadena, CA, USA
| | - Charles P Venditti
- Medical Genomics and Metabolic Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
42
|
Arango D, Bittar A, Esmeral NP, Ocasión C, Muñoz-Camargo C, Cruz JC, Reyes LH, Bloch NI. Understanding the Potential of Genome Editing in Parkinson's Disease. Int J Mol Sci 2021; 22:9241. [PMID: 34502143 PMCID: PMC8430539 DOI: 10.3390/ijms22179241] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 01/05/2023] Open
Abstract
CRISPR is a simple and cost-efficient gene-editing technique that has become increasingly popular over the last decades. Various CRISPR/Cas-based applications have been developed to introduce changes in the genome and alter gene expression in diverse systems and tissues. These novel gene-editing techniques are particularly promising for investigating and treating neurodegenerative diseases, including Parkinson's disease, for which we currently lack efficient disease-modifying treatment options. Gene therapy could thus provide treatment alternatives, revolutionizing our ability to treat this disease. Here, we review our current knowledge on the genetic basis of Parkinson's disease to highlight the main biological pathways that become disrupted in Parkinson's disease and their potential as gene therapy targets. Next, we perform a comprehensive review of novel delivery vehicles available for gene-editing applications, critical for their successful application in both innovative research and potential therapies. Finally, we review the latest developments in CRISPR-based applications and gene therapies to understand and treat Parkinson's disease. We carefully examine their advantages and shortcomings for diverse gene-editing applications in the brain, highlighting promising avenues for future research.
Collapse
Affiliation(s)
- David Arango
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Amaury Bittar
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Natalia P. Esmeral
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Camila Ocasión
- Grupo de Diseño de Productos y Procesos, Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (C.O.); (L.H.R.)
| | - Carolina Muñoz-Camargo
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Juan C. Cruz
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| | - Luis H. Reyes
- Grupo de Diseño de Productos y Procesos, Department of Chemical and Food Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (C.O.); (L.H.R.)
| | - Natasha I. Bloch
- Department of Biomedical Engineering, Universidad de los Andes, Bogotá 111711, Colombia; (D.A.); (A.B.); (N.P.E.); (C.M.-C.); (J.C.C.)
| |
Collapse
|
43
|
Diversity of Adult Neural Stem and Progenitor Cells in Physiology and Disease. Cells 2021; 10:cells10082045. [PMID: 34440814 PMCID: PMC8392301 DOI: 10.3390/cells10082045] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 07/23/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023] Open
Abstract
Adult neural stem and progenitor cells (NSPCs) contribute to learning, memory, maintenance of homeostasis, energy metabolism and many other essential processes. They are highly heterogeneous populations that require input from a regionally distinct microenvironment including a mix of neurons, oligodendrocytes, astrocytes, ependymal cells, NG2+ glia, vasculature, cerebrospinal fluid (CSF), and others. The diversity of NSPCs is present in all three major parts of the CNS, i.e., the brain, spinal cord, and retina. Intrinsic and extrinsic signals, e.g., neurotrophic and growth factors, master transcription factors, and mechanical properties of the extracellular matrix (ECM), collectively regulate activities and characteristics of NSPCs: quiescence/survival, proliferation, migration, differentiation, and integration. This review discusses the heterogeneous NSPC populations in the normal physiology and highlights their potentials and roles in injured/diseased states for regenerative medicine.
Collapse
|
44
|
Sehara Y, Hayashi Y, Ohba K, Uchibori R, Urabe M, Inutsuka A, Shimazaki K, Kawai K, Mizukami H. Higher Transduction Efficiency of AAV5 to Neural Stem Cells and Immature Neurons in Gerbil Dentate Gyrus Compared to AAV2 and rh10. Hum Gene Ther 2021; 33:76-85. [PMID: 34348481 DOI: 10.1089/hum.2021.106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The safety and high efficiency of adeno-associated virus (AAV) vectors has facilitated their wide scale use to deliver therapeutic genes for experimental and clinical purposes in diseases affecting the central nervous system (CNS). AAV1, 2, 5, 8, 9, and rh10 are the most commonly used serotypes for CNS applications. Most AAVs are known to transduce genes predominantly into neurons. However, the precise tropism of AAVs in the dentate gyrus (DG), the region where persistent neurogenesis occurs in the adult brain, is not fully understood. We stereotaxically injected 1.5 × 1010 viral genomes of AAV2, 5, or rh10 carrying green fluorescent protein (GFP) into the right side of gerbil hippocampus, and performed immunofluorescent analysis using differentiation stage-specific markers one week after injection. We found that AAV5 showed a significantly larger number of double positive cells for GFP and Sox2 in the DG, compared to the AAV2 and rh10 groups. On the other hand, AAVrh10 presented a substantially larger number of double positive cells for GFP and NeuN in the DG, compared to AAV2 and AAV5. Our findings indicated that AAV5 showed high transduction efficiency to neural stem cells and precursor cells, while AAVrh10 showed much higher efficiency to mature neurons in the DG.
Collapse
Affiliation(s)
- Yoshihide Sehara
- Jichi Medical University, Division of Genetic Therapeutics, Center for Molecular Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi, Japan, 329-0498;
| | - Yuka Hayashi
- Jichi Medical University, Division of Genetic Therapeutics, Center for Molecular Medicine, Shimotsuke, Tochigi, Japan;
| | - Kenji Ohba
- Jichi Medical University, Division of Genetic Therapeutics, Center for Molecular Medicine, Shimotsuke, Tochigi, Japan;
| | - Ryosuke Uchibori
- Jichi Medical University, Division of Genetic Therapeutics, Center for Molecular Medicine, Shimotsuke, Tochigi, Japan;
| | - Masashi Urabe
- Jichi Medical University, Division of Genetic Therapeutics, Center for Molecular Medicine, Shimotsuke, Tochigi, Japan;
| | - Ayumu Inutsuka
- Jichi Medical University, 12838, Division of Brain and Neurophysiology, Department of Physiology, Shimotsuke, Tochigi, Japan;
| | - Kuniko Shimazaki
- Jichi Medical University, 12838, Department of Neurosurgery, Shimotsuke, Tochigi, Japan;
| | - Kensuke Kawai
- Jichi Medical University, 12838, Department of Neurosurgery, Shimotsuke, Tochigi, Japan;
| | - Hiroaki Mizukami
- Jichi Medical University, Division of Genetic Therapeutics, Center for Molecular Medicine, Shimotsuke, Tochigi, Japan;
| |
Collapse
|
45
|
Völkner M, Pavlou M, Büning H, Michalakis S, Karl MO. Optimized Adeno-Associated Virus Vectors for Efficient Transduction of Human Retinal Organoids. Hum Gene Ther 2021; 32:694-706. [PMID: 33752467 DOI: 10.1089/hum.2020.321] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The most widely used vectors for gene delivery in the retina are recombinant adeno-associated virus (rAAV) vectors. They have proven to be safe and effective in retinal gene therapy studies aimed to treat inherited retinal dystrophies, although with various limitations in transduction efficiency. Novel variants with modified capsid sequences have been engineered to improve transduction and overcome limitations of naturally occurring variants. Although preclinical evaluation of rAAV vectors based on such novel capsids is mostly done in animal models, the use of human induced pluripotent stem cell (hiPSC)-derived organoids offers an accessible and abundant human testing platform for rAAV evaluation. In this study, we tested the novel capsids, AAV9.GL and AAV9.NN, for their tropism and transduction efficiency in hiPSC-derived human retinal organoids (HROs) with all major neuronal and glial cell types in a laminated structure. These variants are based on the AAV9 capsid and were engineered to display specific surface-exposed peptide sequences, previously shown to improve the retinal transduction properties in the context of AAV2. To this end, HROs were transduced with increasing concentrations of rAAV9, rAAV9.GL, or rAAV9.NN carrying a self-complementary genome with a cytomegalovirus-enhanced green fluorescent protein (eGFP) cassette and were monitored for eGFP expression. The rAAV vectors transduced HROs in a dose-dependent manner, with rAAV9.NN achieving the highest efficiency and fastest onset kinetics, leading to detectable eGFP signals in photoreceptors, some interneurons, and Müller glia already at 2 days post-transduction. The potency-enhancing effect of the NN peptide insert was replicated when using the corresponding AAV2-based version (rAAV2.NN). Taken together, we report the application of an HRO system for screening novel AAV vectors and introduce novel vector candidates with enhanced transduction efficiency for human retinal cells.
Collapse
Affiliation(s)
- Manuela Völkner
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany
| | - Marina Pavlou
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Germany.,Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany.,REBIRTH Center for Translational Regenerative Medicine, Hannover Medical School, Hannover, Germany
| | - Stylianos Michalakis
- Department of Ophthalmology, University Hospital, LMU Munich, Munich, Germany.,Department of Pharmacy-Center for Drug Research, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Mike O Karl
- German Center for Neurodegenerative Diseases (DZNE), Dresden, Germany.,CRTD-Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany.,TU Dresden, Faculty of Medicine Carl Gustav Carus, Dresden, Germany
| |
Collapse
|
46
|
von Jonquieres G, Rae CD, Housley GD. Emerging Concepts in Vector Development for Glial Gene Therapy: Implications for Leukodystrophies. Front Cell Neurosci 2021; 15:661857. [PMID: 34239416 PMCID: PMC8258421 DOI: 10.3389/fncel.2021.661857] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 05/12/2021] [Indexed: 12/12/2022] Open
Abstract
Central Nervous System (CNS) homeostasis and function rely on intercellular synchronization of metabolic pathways. Developmental and neurochemical imbalances arising from mutations are frequently associated with devastating and often intractable neurological dysfunction. In the absence of pharmacological treatment options, but with knowledge of the genetic cause underlying the pathophysiology, gene therapy holds promise for disease control. Consideration of leukodystrophies provide a case in point; we review cell type – specific expression pattern of the disease – causing genes and reflect on genetic and cellular treatment approaches including ex vivo hematopoietic stem cell gene therapies and in vivo approaches using adeno-associated virus (AAV) vectors. We link recent advances in vectorology to glial targeting directed towards gene therapies for specific leukodystrophies and related developmental or neurometabolic disorders affecting the CNS white matter and frame strategies for therapy development in future.
Collapse
Affiliation(s)
- Georg von Jonquieres
- Translational Neuroscience Facility, Department of Physiology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| | - Caroline D Rae
- Neuroscience Research Australia, Randwick, NSW, Australia
| | - Gary D Housley
- Translational Neuroscience Facility, Department of Physiology, School of Medical Sciences, UNSW Sydney, Sydney, NSW, Australia
| |
Collapse
|
47
|
Pickering CA, Mazarakis ND. Viral Vector Delivery of DREADDs for CNS Therapy. Curr Gene Ther 2021; 21:191-206. [PMID: 33573551 DOI: 10.2174/1566523221666210211102435] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/15/2021] [Accepted: 01/25/2021] [Indexed: 11/22/2022]
Abstract
Designer Receptors Exclusively Activated by Designer Drugs (DREADDs) are genetically modified G-protein-coupled receptors (GPCRs), that can be activated by a synthetic ligand which is otherwise inert at endogenous receptors. DREADDs can be expressed in cells in the central nervous system (CNS) and subsequently offer the opportunity for remote and reversible silencing or activation of the target cells when the synthetic ligand is systemically administered. In neuroscience, DREADDs have thus far shown to be useful tools for several areas of research and offer considerable potential for the development of gene therapy strategies for neurological disorders. However, in order to design a DREADD-based gene therapy, it is necessary to first evaluate the viral vector delivery methods utilised in the literature to deliver these chemogenetic tools. This review evaluates each of the prominent strategies currently utilised for DREADD delivery, discussing their respective advantages and limitations. We focus on adeno-associated virus (AAV)-based and lentivirus-based systems, and the manipulation of these through cell-type specific promoters and pseudotyping. Furthermore, we address how virally mediated DREADD delivery could be improved in order to make it a viable gene therapy strategy and thus expand its translational potential.
Collapse
Affiliation(s)
- Ceri A Pickering
- Division of Neuroscience, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| | - Nicholas D Mazarakis
- Division of Neuroscience, Department of Brain Sciences, Faculty of Medicine, Imperial College London, London, United Kingdom
| |
Collapse
|
48
|
Lonser RR, Akhter AS, Zabek M, Elder JB, Bankiewicz KS. Direct convective delivery of adeno-associated virus gene therapy for treatment of neurological disorders. J Neurosurg 2021; 134:1751-1763. [PMID: 32915526 DOI: 10.3171/2020.4.jns20701] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/16/2020] [Indexed: 11/06/2022]
Abstract
Molecular biological insights have led to a fundamental understanding of the underlying genomic mechanisms of nervous system disease. These findings have resulted in the identification of therapeutic genes that can be packaged in viral capsids for the treatment of a variety of neurological conditions, including neurodegenerative, metabolic, and enzyme deficiency disorders. Recent data have demonstrated that gene-carrying viral vectors (most often adeno-associated viruses) can be effectively distributed by convection-enhanced delivery (CED) in a safe, reliable, targeted, and homogeneous manner across the blood-brain barrier. Critically, these vectors can be monitored using real-time MRI of a co-infused surrogate tracer to accurately predict vector distribution and transgene expression at the perfused site. The unique properties of CED of adeno-associated virus vectors allow for cell-specific transgene manipulation of the infused anatomical site and/or widespread interconnected sites via antero- and/or retrograde transport. The authors review the convective properties of viral vectors, associated technology, and clinical applications.
Collapse
Affiliation(s)
- Russell R Lonser
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Asad S Akhter
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Mirosław Zabek
- 2Department of Neurological Surgery, Bródno Hospital, Warsaw, Poland
| | - J Bradley Elder
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| | - Krystof S Bankiewicz
- 1Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Columbus, Ohio; and
| |
Collapse
|
49
|
Ozawa A, Arakawa H. Chemogenetics drives paradigm change in the investigation of behavioral circuits and neural mechanisms underlying drug action. Behav Brain Res 2021; 406:113234. [PMID: 33741409 PMCID: PMC8110310 DOI: 10.1016/j.bbr.2021.113234] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 03/06/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022]
Abstract
Recent developments in chemogenetic approaches to the investigation of brain function have ushered in a paradigm change in the strategy for drug and behavior research and clinical drug-based medications. As the nature of the drug action is based on humoral regulation, it is a challenge to identify the neuronal mechanisms responsible for the expression of certain targeted behavior induced by drug application. The development of chemogenetic approaches has allowed researchers to control neural activities in targeted neurons through a toolbox, including engineered G protein-coupled receptors or ligand-gated ion channels together with exogenously inert synthetic ligands. This review provides a brief overview of the chemogenetics toolbox with an emphasis on the DREADDs (Designer Receptors Exclusively Activated by Designer Drugs) technique used in rodent models, which is applicable to the investigation of how specific neural circuits regulate behavioral processes. The use of chemogenetics has had a significant impact on basic neuroscience for a better understanding of the relationships between brain activity and the expression of behaviors with cell- and circuit-specific orders. Furthermore, chemogenetics is potentially a useful tool to deconstruct the neuropathological mechanisms of mental diseases and its regulation by drug, and provide us with transformative therapeutics with medication. We also review recent findings in the use of chemogenetic techniques to uncover functional circuit connections of serotonergic neurons in rodent models.
Collapse
Affiliation(s)
- Akihiko Ozawa
- Department of Biomedical Science, Florida Atlantic University, Boca Raton, FL, USA
| | - Hiroyuki Arakawa
- Department of Psychology, Tokiwa University, Mito, Ibaraki, Japan; Department of Systems Physiology, University of Ryukyus, Faculty of Medicine, Nakagami District, Okinawa, Japan.
| |
Collapse
|
50
|
Poth KM, Texakalidis P, Boulis NM. Chemogenetics: Beyond Lesions and Electrodes. Neurosurgery 2021; 89:185-195. [PMID: 33913505 PMCID: PMC8279839 DOI: 10.1093/neuros/nyab147] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 02/26/2021] [Indexed: 01/14/2023] Open
Abstract
The field of chemogenetics has rapidly expanded over the last decade, and engineered receptors are currently utilized in the lab to better understand molecular interactions in the nervous system. We propose that chemogenetic receptors can be used for far more than investigational purposes. The potential benefit of adding chemogenetic neuromodulation to the current neurosurgical toolkit is substantial. There are several conditions currently treated surgically, electrically, and pharmacologically in clinic, and this review highlights how chemogenetic neuromodulation could improve patient outcomes over current neurosurgical techniques. We aim to emphasize the need to take these techniques from bench to bedside.
Collapse
Affiliation(s)
- Kelly M Poth
- Department of Neurosurgery, Emory University, Atlanta, Georgia, USA
| | | | | |
Collapse
|