1
|
Bolideei M, Barzigar R, Gahrouei RB, Mohebbi E, Haider KH, Paul S, Paul MK, Mehran MJ. Applications of Gene Editing and Nanotechnology in Stem Cell-Based Therapies for Human Diseases. Stem Cell Rev Rep 2025; 21:905-934. [PMID: 40014250 DOI: 10.1007/s12015-025-10857-0] [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] [Accepted: 02/23/2025] [Indexed: 02/28/2025]
Abstract
Stem cell research is a dynamic and fast-advancing discipline with great promise for the treatment of diverse human disorders. The incorporation of gene editing technologies, including ZFNs, TALENs, and the CRISPR/Cas system, in conjunction with progress in nanotechnology, is fundamentally transforming stem cell therapy and research. These innovations not only provide a glimmer of optimism for patients and healthcare practitioners but also possess the capacity to radically reshape medical treatment paradigms. Gene editing and nanotechnology synergistically enhance stem cell-based therapies' precision, efficiency, and applicability, offering transformative potential for treating complex diseases and advancing regenerative medicine. Nevertheless, it is important to acknowledge that these technologies also give rise to ethical considerations and possible hazards, such as inadvertent genetic modifications and the development of genetically modified organisms, therefore creating a new age of designer infants. This review emphasizes the crucial significance of gene editing technologies and nanotechnology in the progress of stem cell treatments, particularly for degenerative pathologies and injuries. It emphasizes their capacity to restructure and comprehensively revolutionize medical treatment paradigms, providing fresh hope and optimism for patients and healthcare practitioners.
Collapse
Affiliation(s)
- Mansoor Bolideei
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China
| | - Rambod Barzigar
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India
| | - Razieh Bahrami Gahrouei
- Department of Pharmacy PES College, Rajiv Gandhi University of Health Sciences, Bangalore, Karnataka, India
| | - Elham Mohebbi
- Department of Medical Microbiology, Immunology, and Cell Biology, Southern Illinois School of Medicine, Springfield, IL, USA
| | - Khawaja Husnain Haider
- Sulaiman AlRajhi Medical School, Al Bukayriyah, AlQaseem, 52726, Kingdom of Saudi Arabia
| | - Sayan Paul
- Department of Biochemistry & Molecular Biology, University of Texas Medical Branch at Galveston, Galveston, TX, 77555, USA.
| | - Manash K Paul
- Department of Radiation Biology and Toxicology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, 576104, India.
| | - Mohammad Javad Mehran
- Department of Biotechnology, SJCE Technical Campus, JSS Research Foundation, University of Mysore, Mysore, 570006, Karnataka, India.
| |
Collapse
|
2
|
Kupke J, Loizou S, Bengtson CP, Sticht C, Oliveira AMM. Hippocampal DNA Methylation Promotes Contextual Fear Memory Persistence by Facilitating Systems Consolidation and Cortical Engram Stabilization. Biol Psychiatry 2025:S0006-3223(25)00058-7. [PMID: 39880069 DOI: 10.1016/j.biopsych.2025.01.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 12/27/2024] [Accepted: 01/22/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Long-term fear memory storage involves gradual reorganization of supporting brain regions over time, a process termed systems consolidation. Memories initially rely on the hippocampus but gradually shift dependence to the neocortex. Although hippocampal activity drives this transfer, the molecular basis of systems consolidation is largely unknown. DNA methylation changes accompany persistent fear memory formation in the hippocampus and cortex, but its causal role in memory storage and systems consolidation remains unclear. METHODS We investigated the role of hippocampal DNA methylation in fear memory persistence through multiple approaches. Using recombinant adeno-associated virus (rAAV)-mediated gene transfer, we overexpressed or knocked down a DNA methyltransferase (DNMT3A2) in the dorsal hippocampus of mice and assessed its impact on fear memory duration. Engram tagging and manipulation tools were applied to study cortical fear engram stabilization. Finally, RNA sequencing analysis was used to identify transcriptional changes driven by DNMT3A2 overexpression. RESULTS Overexpression of hippocampal DNMT3A2 induced a persistent fear memory, while its knockdown impaired remote memory recall. RNA sequencing revealed that DNMT3A2 overexpression modified the expression of synaptic transmission regulatory genes. Furthermore, genetic engram tagging and manipulation revealed that hippocampal DNA methylation promoted the transfer of the fear memory trace from the hippocampus to the cortex and the stabilization of cortical fear memory traces. CONCLUSIONS Our findings demonstrate that hippocampal DNA methylation regulates the long-term storage of persistent fear memories by facilitating the transfer of memory traces from the hippocampus to the cortex and cortical stabilization. These results highlight DNA methylation as a key molecular mechanism underlying systems consolidation and long-term fear memory storage.
Collapse
Affiliation(s)
- Janina Kupke
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Stefanos Loizou
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany; Department of Molecular and Cellular Cognition Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - C Peter Bengtson
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany
| | - Carsten Sticht
- Next Generation Sequencing Core Facility, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ana M M Oliveira
- Department of Neurobiology, Interdisciplinary Center for Neurosciences, Heidelberg University, Heidelberg, Germany; Department of Molecular and Cellular Cognition Research, Central Institute of Mental Health, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| |
Collapse
|
3
|
Lopez-Gordo E, Chamberlain K, Riyad JM, Kohlbrenner E, Weber T. Natural Adeno-Associated Virus Serotypes and Engineered Adeno-Associated Virus Capsid Variants: Tropism Differences and Mechanistic Insights. Viruses 2024; 16:442. [PMID: 38543807 PMCID: PMC10975205 DOI: 10.3390/v16030442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 03/02/2024] [Accepted: 03/06/2024] [Indexed: 05/23/2024] Open
Abstract
Today, adeno-associated virus (AAV)-based vectors are arguably the most promising in vivo gene delivery vehicles for durable therapeutic gene expression. Advances in molecular engineering, high-throughput screening platforms, and computational techniques have resulted in a toolbox of capsid variants with enhanced performance over parental serotypes. Despite their considerable promise and emerging clinical success, there are still obstacles hindering their broader use, including limited transduction capabilities, tissue/cell type-specific tropism and penetration into tissues through anatomical barriers, off-target tissue biodistribution, intracellular degradation, immune recognition, and a lack of translatability from preclinical models to clinical settings. Here, we first describe the transduction mechanisms of natural AAV serotypes and explore the current understanding of the systemic and cellular hurdles to efficient transduction. We then outline progress in developing designer AAV capsid variants, highlighting the seminal discoveries of variants which can transduce the central nervous system upon systemic administration, and, to a lesser extent, discuss the targeting of the peripheral nervous system, eye, ear, lung, liver, heart, and skeletal muscle, emphasizing their tissue and cell specificity and translational promise. In particular, we dive deeper into the molecular mechanisms behind their enhanced properties, with a focus on their engagement with host cell receptors previously inaccessible to natural AAV serotypes. Finally, we summarize the main findings of our review and discuss future directions.
Collapse
|
4
|
Lee MH, Um KH, Lee SW, Sun YJ, Gu DH, Jo YO, Kim SH, Seol W, Hwang H, Baek K, Choi JW. Bi-directional regulation of AIMP2 and its splice variant on PARP-1-dependent neuronal cell death; Therapeutic implication for Parkinson's disease. Acta Neuropathol Commun 2024; 12:5. [PMID: 38172953 PMCID: PMC10765824 DOI: 10.1186/s40478-023-01697-5] [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: 09/07/2023] [Accepted: 11/28/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Parthanatos represents a critical molecular aspect of Parkinson's disease, wherein AIMP2 aberrantly activates PARP-1 through direct physical interaction. Although AIMP2 ought to be a therapeutic target for the disease, regrettably, it is deemed undruggable due to its non-enzymatic nature and predominant localization within the tRNA synthetase multi-complex. Instead, AIMP2 possesses an antagonistic splice variant, designated DX2, which counteracts AIMP2-induced apoptosis in the p53 or inflammatory pathway. Consequently, we examined whether DX2 competes with AIMP2 for PARP-1 activation and is therapeutically effective in Parkinson's disease. METHODS The binding affinity of AIMP2 and DX2 to PARP-1 was contrasted through immunoprecipitation. The efficacy of DX2 in neuronal cell death was assessed under 6-OHDA and H2O2 in vitro conditions. Additionally, endosomal and exosomal activity of synaptic vesicles was gauged in AIMP2 or DX2 overexpressed hippocampal primary neurons utilizing optical live imaging with VAMP-vGlut1 probes. To ascertain the role of DX2 in vivo, rotenone-induced behavioral alterations were compared between wild-type and DX2 transgenic animals. A DX2-encoding self-complementary adeno-associated virus (scAAV) was intracranially injected into 6-OHDA induced in vivo animal models, and their mobility was examined. Subsequently, the isolated brain tissues were analyzed. RESULTS DX2 translocates into the nucleus upon ROS stress more rapidly than AIMP2. The binding affinity of DX2 to PARP-1 appeared to be more robust compared to that of AIMP2, resulting in the inhibition of PARP-1 induced neuronal cell death. DX2 transgenic animals exhibited neuroprotective behavior in rotenone-induced neuronal damage conditions. Following a single intracranial injection of AAV-DX2, both behavior and mobility were consistently ameliorated in neurodegenerative animal models induced by 6-OHDA. CONCLUSION AIMP2 and DX2 are proposed to engage in bidirectional regulation of parthanatos. They physically interact with PARP-1. Notably, DX2's cell survival properties manifest exclusively in the context of abnormal AIMP2 accumulation, devoid of any tumorigenic effects. This suggests that DX2 could represent a distinctive therapeutic target for addressing Parkinson's disease in patients.
Collapse
Affiliation(s)
- Min Hak Lee
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ki-Hwan Um
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Seok Won Lee
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Ye Ji Sun
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Da-Hye Gu
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Young Ok Jo
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sung Hyun Kim
- Department of Neuroscience, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of Physiology, School of Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Wongi Seol
- InAm Neuroscience Research Center, Sanbon Medical Center, College of Medicine, Wonkwang University, Sanbonro 321, Gunposi, Gyeonggido, 15865, Republic of Korea
| | - Hyorin Hwang
- Generoath Ltd., Seoul, 04168, Republic of Korea
- Department of Pharmacology, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangneung, Gangwon-Do, 25457, Republic of Korea
| | - Kyunghwa Baek
- Department of Pharmacology, College of Dentistry and Research Institute of Oral Science, Gangneung-Wonju National University, Gangneung, Gangwon-Do, 25457, Republic of Korea
| | - Jin Woo Choi
- Department of Pharmacology, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Biomedical and Pharmaceutical Science, Graduate School, Kyung Hee University, Seoul, 02447, Republic of Korea.
- Department of Regulatory Science, College of Pharmacy, Kyung Hee University, Seoul, 02447, Republic of Korea.
| |
Collapse
|
5
|
Tian G, Cao C, Li S, Wang W, Zhang Y, Lv Y. rAAV2-Mediated Restoration of GALC in Neural Stem Cells from Krabbe Patient-Derived iPSCs. Pharmaceuticals (Basel) 2023; 16:ph16040624. [PMID: 37111381 PMCID: PMC10143348 DOI: 10.3390/ph16040624] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Revised: 03/28/2023] [Accepted: 04/12/2023] [Indexed: 04/29/2023] Open
Abstract
Krabbe disease is a rare neurodegenerative fatal disease. It is caused by deficiency of the lysosomal enzyme galactocerebrosidase (GALC), which results in progressive accumulation of galactolipid substrates in myelin-forming cells. However, there is still a lack of appropriate neural models and effective approaches for Krabbe disease. We generated induced pluripotent stem cells (iPSCs) from a Krabbe patient previously. Here, Krabbe patient-derived neural stem cells (K-NSCs) were induced from these iPSCs. By using nine kinds of recombinant adeno-associated virus (rAAV) vectors to infect K-NSCs, we found that the rAAV2 vector has high transduction efficiency for K-NSCs. Most importantly, rAAV2-GALC rescued GALC enzymatic activity in K-NSCs. Our findings not only establish a novel patient NSC model for Krabbe disease, but also firstly indicate the potential of rAAV2-mediated gene therapy for this devastating disease.
Collapse
Affiliation(s)
- Guoshuai Tian
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Chunyu Cao
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443000, China
| | - Shuyue Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443000, China
| | - Wei Wang
- Department of Neurology, China-Japan Friendship Hospital, Beijing 100029, China
| | - Ye Zhang
- State Key Laboratory of Medical Molecular Biology, Department of Biochemistry and Molecular Biology, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences, School of Basic Medicine Peking Union Medical College, Beijing 100005, China
| | - Yafeng Lv
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Sciences, China Three Gorges University, Yichang 443000, China
| |
Collapse
|
6
|
Ghauri MS, Ou L. AAV Engineering for Improving Tropism to the Central Nervous System. BIOLOGY 2023; 12:186. [PMID: 36829465 PMCID: PMC9953251 DOI: 10.3390/biology12020186] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/01/2023] [Revised: 01/21/2023] [Accepted: 01/24/2023] [Indexed: 01/28/2023]
Abstract
Adeno-associated virus (AAV) is a non-pathogenic virus that mainly infects primates with the help of adenoviruses. AAV is being widely used as a delivery vector for in vivo gene therapy, as evidenced by five currently approved drugs and more than 255 clinical trials across the world. Due to its relatively low immunogenicity and toxicity, sustained efficacy, and broad tropism, AAV holds great promise for treating many indications, including central nervous system (CNS), ocular, muscular, and liver diseases. However, low delivery efficiency, especially for the CNS due to the blood-brain barrier (BBB), remains a significant challenge for more clinical application of AAV gene therapy. Thus, there is an urgent need for utilizing AAV engineering to discover next-generation capsids with improved properties, e.g., enhanced BBB penetrance, lower immunogenicity, and higher packaging efficiency. AAV engineering methods, including directed evolution, rational design, and in silico design, have been developed, resulting in the discovery of novel capsids (e.g., PhP.B, B10, PAL1A/B/C). In this review, we discuss key studies that identified engineered CNS capsids and/or established methodological improvements. Further, we also discussed important issues that need to be addressed, including cross-species translatability, cell specificity, and modular engineering to improve multiple properties simultaneously.
Collapse
Affiliation(s)
- Muhammad S. Ghauri
- School of Medicine, California University of Science and Medicine, Colton, CA 92324, USA
| | - Li Ou
- Genemagic Biosciences, Media, PA 19086, USA
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55454, USA
| |
Collapse
|
7
|
Bharti S, Anant PS, Kumar A. Nanotechnology in stem cell research and therapy. JOURNAL OF NANOPARTICLE RESEARCH 2023; 25:6. [DOI: 10.1007/s11051-022-05654-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2025]
|
8
|
Kremer LP, Cerrizuela S, Dehler S, Stiehl T, Weinmann J, Abendroth H, Kleber S, Laure A, El Andari J, Anders S, Marciniak-Czochra A, Grimm D, Martin-Villalba A. High throughput screening of novel AAV capsids identifies variants for transduction of adult NSCs within the subventricular zone. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 23:33-50. [PMID: 34553001 PMCID: PMC8427210 DOI: 10.1016/j.omtm.2021.07.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 07/09/2021] [Indexed: 12/19/2022]
Abstract
The adult mammalian brain entails a reservoir of neural stem cells (NSCs) generating glial cells and neurons. However, NSCs become increasingly quiescent with age, which hampers their regenerative capacity. New means are therefore required to genetically modify adult NSCs for re-enabling endogenous brain repair. Recombinant adeno-associated viruses (AAVs) are ideal gene-therapy vectors due to an excellent safety profile and high transduction efficiency. We thus conducted a high-throughput screening of 177 intraventricularly injected barcoded AAV variants profiled by RNA sequencing. Quantification of barcoded AAV mRNAs identified two synthetic capsids, peptide-modified derivative of wild-type AAV9 (AAV9_A2) and peptide-modified derivative of wild-type AAV1 (AAV1_P5), both of which transduce active and quiescent NSCs. Further optimization of AAV1_P5 by judicious selection of the promoter and dose of injected viral genomes enabled labeling of 30%–60% of the NSC compartment, which was validated by fluorescence-activated cell sorting (FACS) analyses and single-cell RNA sequencing. Importantly, transduced NSCs readily produced neurons. The present study identifies AAV variants with a high regional tropism toward the ventricular-subventricular zone (v-SVZ) with high efficiency in targeting adult NSCs, thereby paving the way for preclinical testing of regenerative gene therapy.
Collapse
Affiliation(s)
- Lukas P.M. Kremer
- Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Center for Molecular Biology of Heidelberg University (ZMBH), 69120 Heidelberg, Germany
| | - Santiago Cerrizuela
- Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Sascha Dehler
- Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Thomas Stiehl
- Institute of Applied Mathematics, Interdisciplinary Center for Scientific Computing and BioQuant, Heidelberg University, 69120 Heidelberg, Germany
| | - Jonas Weinmann
- Virus-Host Interaction Group, Department of Infectious Diseases/Virology, Heidelberg University Hospital, Cluster of Excellence Cell Networks, BioQuant, 69120 Heidelberg, Germany
| | - Heike Abendroth
- Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Susanne Kleber
- Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Alexander Laure
- Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Jihad El Andari
- Virus-Host Interaction Group, Department of Infectious Diseases/Virology, Heidelberg University Hospital, Cluster of Excellence Cell Networks, BioQuant, 69120 Heidelberg, Germany
| | - Simon Anders
- Center for Molecular Biology of Heidelberg University (ZMBH), 69120 Heidelberg, Germany
| | - Anna Marciniak-Czochra
- Institute of Applied Mathematics, Interdisciplinary Center for Scientific Computing and BioQuant, Heidelberg University, 69120 Heidelberg, Germany
| | - Dirk Grimm
- Virus-Host Interaction Group, Department of Infectious Diseases/Virology, Heidelberg University Hospital, Cluster of Excellence Cell Networks, BioQuant, 69120 Heidelberg, Germany
- German Center for Infection Research (DZIF) and German Center for Cardiovascular Research (DZHK), partner site Heidelberg, 69120 Heidelberg, Germany
| | - Ana Martin-Villalba
- Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Corresponding author: Ana Martin-Villalba, Molecular Neurobiology, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany.
| |
Collapse
|
9
|
Novel Approaches Used to Examine and Control Neurogenesis in Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22179608. [PMID: 34502516 PMCID: PMC8431772 DOI: 10.3390/ijms22179608] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/31/2021] [Accepted: 09/02/2021] [Indexed: 12/16/2022] Open
Abstract
Neurogenesis is a key mechanism of brain development and plasticity, which is impaired in chronic neurodegeneration, including Parkinson’s disease. The accumulation of aberrant α-synuclein is one of the features of PD. Being secreted, this protein produces a prominent neurotoxic effect, alters synaptic plasticity, deregulates intercellular communication, and supports the development of neuroinflammation, thereby providing propagation of pathological events leading to the establishment of a PD-specific phenotype. Multidirectional and ambiguous effects of α-synuclein on adult neurogenesis suggest that impaired neurogenesis should be considered as a target for the prevention of cell loss and restoration of neurological functions. Thus, stimulation of endogenous neurogenesis or cell-replacement therapy with stem cell-derived differentiated neurons raises new hopes for the development of effective and safe technologies for treating PD neurodegeneration. Given the rapid development of optogenetics, it is not surprising that this method has already been repeatedly tested in manipulating neurogenesis in vivo and in vitro via targeting stem or progenitor cells. However, niche astrocytes could also serve as promising candidates for controlling neuronal differentiation and improving the functional integration of newly formed neurons within the brain tissue. In this review, we mainly focus on current approaches to assess neurogenesis and prospects in the application of optogenetic protocols to restore the neurogenesis in Parkinson’s disease.
Collapse
|
10
|
Carrier-Ruiz A, Sugaya Y, Kumar D, Vergara P, Koyanagi I, Srinivasan S, Naoi T, Kano M, Sakaguchi M. Calcium imaging of adult-born neurons in freely moving mice. STAR Protoc 2021; 2:100238. [PMID: 33458703 PMCID: PMC7797924 DOI: 10.1016/j.xpro.2020.100238] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Adult-born neurons (ABNs) in the dentate gyrus bestow unique cellular plasticity to the mammalian brain. We recently found that the activity of ABNs during sleep is necessary for memory consolidation. Here, we describe our method for Ca2+ imaging of ABN activity using a miniaturized fluorescent microscope and sleep recordings. As preparatory surgery and post-recording data processing can be major obstacles, we provide detailed descriptions and problem-solving tips. For complete details on the use and execution of this protocol, please refer to Kumar et al. (2020).
Collapse
Affiliation(s)
- Alvaro Carrier-Ruiz
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo 113-0033, Japan
- Department of Neuroscience, Karolinska Institute, Stockholm 17165, Sweden
| | - Yuki Sugaya
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo 113-0033, Japan
| | - Deependra Kumar
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Pablo Vergara
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Iyo Koyanagi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Sakthivel Srinivasan
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Toshie Naoi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| | - Masanobu Kano
- Department of Neurophysiology, Graduate School of Medicine, The University of Tokyo, Tokyo 113-0033, Japan
- International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), Tokyo 113-0033, Japan
| | - Masanori Sakaguchi
- International Institute for Integrative Sleep Medicine (WPI-IIIS), University of Tsukuba, Tsukuba, Ibaraki 305-0006, Japan
| |
Collapse
|
11
|
Hu S, Yang T, Wang Y. Widespread labeling and genomic editing of the fetal central nervous system by in utero CRISPR AAV9-PHP.eB administration. Development 2021; 148:dev.195586. [PMID: 33334860 PMCID: PMC7847274 DOI: 10.1242/dev.195586] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2020] [Accepted: 12/07/2020] [Indexed: 01/20/2023]
Abstract
Efficient genetic manipulation in the developing central nervous system is crucial for investigating mechanisms of neurodevelopmental disorders and the development of promising therapeutics. Common approaches including transgenic mice and in utero electroporation, although powerful in many aspects, have their own limitations. In this study, we delivered vectors based on the AAV9.PHP.eB pseudo-type to the fetal mouse brain, and achieved widespread and extensive transduction of neural cells. When AAV9.PHP.eB-coding gRNA targeting PogZ or Depdc5 was delivered to Cas9 transgenic mice, widespread gene knockout was also achieved at the whole brain level. Our studies provide a useful platform for studying brain development and devising genetic intervention for severe developmental diseases. Summary:In utero CRISPR AAV9-PHP.eB provides a powerful platform to efficiently manipulate gene expression in the developing CNS to investigate mechanisms of neurodevelopmental disorders.
Collapse
Affiliation(s)
- Shuntong Hu
- Department of Neurology, The Third Xiangya Hospital, Central South University, Changsha 410013, China.,Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Tao Yang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Yu Wang
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| |
Collapse
|
12
|
Wang Y, Bruggeman KF, Franks S, Gautam V, Hodgetts SI, Harvey AR, Williams RJ, Nisbet DR. Is Viral Vector Gene Delivery More Effective Using Biomaterials? Adv Healthc Mater 2021; 10:e2001238. [PMID: 33191667 DOI: 10.1002/adhm.202001238] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/03/2020] [Indexed: 12/16/2022]
Abstract
Gene delivery has been extensively investigated for introducing foreign genetic material into cells to promote expression of therapeutic proteins or to silence relevant genes. This approach can regulate genetic or epigenetic disorders, offering an attractive alternative to pharmacological therapy or invasive protein delivery options. However, the exciting potential of viral gene therapy has yet to be fully realized, with a number of clinical trials failing to deliver optimal therapeutic outcomes. Reasons for this include difficulty in achieving localized delivery, and subsequently lower efficacy at the target site, as well as poor or inconsistent transduction efficiency. Thus, ongoing efforts are focused on improving local viral delivery and enhancing its efficiency. Recently, biomaterials have been exploited as an option for more controlled, targeted and programmable gene delivery. There is a growing body of literature demonstrating the efficacy of biomaterials and their potential advantages over other delivery strategies. This review explores current limitations of gene delivery and the progress of biomaterial-mediated gene delivery. The combination of biomaterials and gene vectors holds the potential to surmount major challenges, including the uncontrolled release of viral vectors with random delivery duration, poorly localized viral delivery with associated off-target effects, limited viral tropism, and immune safety concerns.
Collapse
Affiliation(s)
- Yi Wang
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Kiara F. Bruggeman
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Stephanie Franks
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
| | - Vini Gautam
- Department of Biomedical Engineering The University of Melbourne Melbourne Victoria 3010 Australia
| | - Stuart I. Hodgetts
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Alan R. Harvey
- School of Human Sciences The University of Western Australia Perth WA 6009 Australia
- Perron Institute for Neurological and Translational Science Perth WA 6009 Australia
| | - Richard J. Williams
- The Institute for Mental and Physical Health and Clinical Translation (IMPACT) School of Medicine Deakin University Waurn Ponds VIC 3216 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| | - David R. Nisbet
- Laboratory of Advanced Biomaterials Research School of Engineering The Australian National University Canberra ACT 2601 Australia
- Biofab3D St. Vincent's Hospital Fitzroy 3065 Australia
| |
Collapse
|
13
|
Chen SH, Papaneri A, Walker M, Scappini E, Keys RD, Martin NP. A Simple, Two-Step, Small-Scale Purification of Recombinant Adeno-Associated Viruses. J Virol Methods 2020; 281:113863. [PMID: 32371233 DOI: 10.1016/j.jviromet.2020.113863] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2019] [Revised: 02/22/2020] [Accepted: 04/03/2020] [Indexed: 10/24/2022]
Abstract
Recombinant adeno-associated viruses (rAAVs) are robust and versatile tools for in vivo gene delivery. Natural and designer capsid variations in rAAVs allow for targeted gene delivery to specific cell types. Low immunogenicity and lack of pathogenesis also add to the popularity of this virus as an innocuous gene delivery vector for gene therapy. rAAVs are routinely used to express recombinases, sensors, detectors, CRISPR-Cas9 components, or to simply overexpress a gene of interest for functional studies. High production demand has given rise to multiple platforms for the production and purification of rAAVs. However, most platforms rely heavily on large amounts of starting material and multiple purification steps to produce highly purified viral particles. Often, researchers require several small-scale purified rAAVs. Here, we describe a simple and efficient technique for purification of recombinant rAAVs from small amounts of starting material in a two-step purification method. In this method, rAAVs are released into the packaging cell medium using high salt concentration, pelleted by ultracentrifugation to remove soluble impurities. Then, the resuspended pellet is purified using a protein spin-concentrator. In this protocol, we modify the conventional rAAV purification methods to eliminate the need for fraction collection and the labor-intensive steps for evaluating the titer and purity of individual fractions. The resulting rAAV preparations are comparable in titer and purity to commercially available samples. This simplified process can be used to generate highly purified rAAV particles on a small scale, thereby saving resources, generating less waste, and reducing a laboratory's environmental footprint.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Neurobiology Laboratory, U.S.A; Viral Vector Core, U.S.A
| | | | - Mitzie Walker
- Neurobiology Laboratory, U.S.A; Viral Vector Core, U.S.A
| | | | - Robert D Keys
- Cellular and Molecular Pathology Branch, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C. 27709, U.S.A
| | - Negin P Martin
- Neurobiology Laboratory, U.S.A; Viral Vector Core, U.S.A.
| |
Collapse
|
14
|
Nemirovich-Danchenko NM, Khodanovich MY. Telomerase Gene Editing in the Neural Stem Cells in vivo as a Possible New Approach against Brain Aging. RUSS J GENET+ 2020. [DOI: 10.1134/s1022795420040092] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
15
|
Romeo C, Chen SH, Goulding E, Van Gorder L, Schwartz M, Walker M, Scott G, Scappini E, Ray M, Martin NP. AAV diffuses across zona pellucida for effortless gene delivery to fertilized eggs. Biochem Biophys Res Commun 2020; 526:85-90. [PMID: 32197836 DOI: 10.1016/j.bbrc.2020.03.026] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Accepted: 03/05/2020] [Indexed: 12/14/2022]
Abstract
Gene delivery to fertilized eggs is often the first step in creation of transgenic animals, CRISPR knock-out, or early developmental studies. The zona pellucida, a hardened glycoprotein matrix surrounding the mammalian fertilized eggs, often complicates gene delivery by forming a barrier against transfection reagents and viruses. High efficiency techniques to perforate or penetrate the zona allow for access and gene delivery to fertilized eggs. However, these techniques often rely on highly skilled technologists, are costly, and require specialized equipment for micromanipulation, laser perforation, or electroporation. Here, we report that adenoassociated viruses (AAVs) with serotypes 1 or DJ can efficiently diffuse across the zona to deliver genes without any manipulations to fertilized eggs. We observe lowered rates of embryo development after treatment of embryos with all AAV serotypes. However, we were able to reduce adverse effects on embryo development by exposing embryos to AAVs at later stages of in vitro development. AAVs have low immune response and do not incorporate into their host chromosomes to cause insertional mutations. Hence, AAVs can serve as a highly effective tool for transient delivery of genes to fertilized mammalian eggs.
Collapse
Affiliation(s)
- Charles Romeo
- Neurobiology Laboratory, Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA
| | - Shih-Heng Chen
- Neurobiology Laboratory, Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA
| | - Eugenia Goulding
- Neurobiology Laboratory, Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA
| | - Lucas Van Gorder
- Neurobiology Laboratory, Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA
| | - Maura Schwartz
- Neurobiology Laboratory, Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA
| | - Mitzie Walker
- Neurobiology Laboratory, Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA
| | - Gregory Scott
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA
| | - Erica Scappini
- Signal Transduction Laboratory, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA
| | - Manas Ray
- Reproductive and Developmental Biology Laboratory, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA
| | - Negin P Martin
- Neurobiology Laboratory, Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, 111 T.W. Alexander Drive, Research Triangle Park, N.C, 27709, USA.
| |
Collapse
|
16
|
Kim SH, Lee S, Lee H, Cho M, Schaffer DV, Jang JH. AAVR-Displaying Interfaces: Serotype-Independent Adeno-Associated Virus Capture and Local Delivery Systems. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 18:432-443. [PMID: 31670142 PMCID: PMC6831863 DOI: 10.1016/j.omtn.2019.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 09/17/2019] [Accepted: 09/17/2019] [Indexed: 12/12/2022]
Abstract
Interfacing gene delivery vehicles with biomaterials has the potential to play a key role in diversifying gene transfer capabilities, including localized, patterned, and controlled delivery. However, strategies for modifying biomaterials to interact with delivery vectors must be redesigned whenever new delivery vehicles and applications are explored. We have developed a vector-independent biomaterial platform capable of interacting with various adeno-associated viral (AAV) serotypes. A water-soluble, cysteine-tagged, recombinant protein version of the recently discovered multi-AAV serotype receptor (AAVR), referred to as cys-AAVR, was conjugated to maleimide-displaying polycaprolactone (PCL) materials using click chemistry. The resulting cys-AAVR-PCL system bound to a broad range of therapeutically relevant AAV serotypes, thereby providing a platform capable of modulating the delivery of all AAV serotypes. Intramuscular injection of cys-AAVR-PCL microspheres with bound AAV vectors resulted in localized and sustained gene delivery as well as reduced spread to off-target organs compared to a vector solution. This cys-AAVR-PCL system is thus an effective approach for biomaterial-based AAV gene delivery for a broad range of therapeutic applications.
Collapse
Affiliation(s)
- Seung-Hyun Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea
| | - Slgirim Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea; Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, IL 60611, USA
| | - Heehyung Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea
| | - Mira Cho
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720-3220, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720-3220, USA; Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720-3220, USA.
| | - Jae-Hyung Jang
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 120-749, Korea.
| |
Collapse
|
17
|
Farzamfar S, Nazeri N, Salehi M, Valizadeh A, Marashi SM, Savari Kouzehkonan G, Ghanbari H. Will Nanotechnology Bring New Hope for Stem Cell Therapy? Cells Tissues Organs 2019; 206:229-241. [PMID: 31288229 DOI: 10.1159/000500517] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Accepted: 04/21/2019] [Indexed: 01/05/2025] Open
Abstract
The potential of stem cell therapy has been shown in preclinical trials for the treatment of damage and replacement of organs and degenerative diseases. After many years of research, its clinical application is limited. Currently there is not a single stem cell therapy product or procedure. Nanotechnology is an emerging field in medicine and has huge potential due to its unique characteristics such as its size, surface effects, tunnel effects, and quantum size effect. The importance of application of nanotechnology in stem cell technology and cell-based therapies has been recognized. In particular, the effects of nanotopography on stem cell differentiation, proliferation, and adhesion have become an area of intense research in tissue engineering and regenerative medicine. Despite the many opportunities that nanotechnology can create to change the fate of stem cell technology and cell therapies, it poses several risks since some nanomaterials are cytotoxic and can affect the differentiation program of stem cells and their viability. Here we review some of the advances and the prospects of nanotechnology in stem cell research and cell-based therapies and discuss the issues, obstacles, applications, and approaches with the aim of opening new avenues for further research.
Collapse
Affiliation(s)
- Saeed Farzamfar
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Niloofar Nazeri
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran,
| | - Majid Salehi
- Tissue Engineering and Stem Cell Research Center, Shahroud University of Medical Sciences, Shahroud, Iran
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Alireza Valizadeh
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Sayed Mahdi Marashi
- Virology Department, School of Public Health, Tehran University of Medical Sciences, Tehran, Iran
| | - Gholamreza Savari Kouzehkonan
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hossein Ghanbari
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
18
|
Chen SH, Haam J, Walker M, Scappini E, Naughton J, Martin NP. Recombinant Viral Vectors as Neuroscience Tools. ACTA ACUST UNITED AC 2019; 87:e67. [PMID: 30901512 DOI: 10.1002/cpns.67] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Recombinant viruses are highly efficient vehicles for in vivo gene delivery. Viral vectors expand the neurobiology toolbox to include direct and rapid anterograde, retrograde, and trans-synaptic delivery of tracers, sensors, and actuators to the mammalian brain. Each viral type offers unique advantages and limitations. To establish strategies for selecting a suitable viral type, this article aims to provide readers with an overview of viral recombinant technology, viral structure, tropism, and differences between serotypes and pseudotypes for three of the most commonly used vectors in neurobiology research: adeno-associated viruses, retro/lentiviruses, and glycoprotein-deleted rabies viruses. © 2019 by John Wiley & Sons, Inc.
Collapse
Affiliation(s)
- Shih-Heng Chen
- Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, Research Triangle Park, North Carolina.,Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH/DHHS, Research Triangle Park, North Carolina
| | - Juhee Haam
- Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH/DHHS, Research Triangle Park, North Carolina
| | - Mitzie Walker
- Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, Research Triangle Park, North Carolina.,Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH/DHHS, Research Triangle Park, North Carolina
| | - Erica Scappini
- Signal Transduction Laboratory, Fluorescence Microscopy and Imaging Center, National Institute of Environmental Health Sciences, NIH/DHHS, Research Triangle Park, North Carolina
| | - John Naughton
- Gene Transfer, Targeting and Therapeutics (GT3) Core, Salk Institute for Biological Studies, La Jolla, California
| | - Negin P Martin
- Viral Vector Core, National Institute of Environmental Health Sciences, NIH/DHHS, Research Triangle Park, North Carolina.,Neurobiology Laboratory, National Institute of Environmental Health Sciences, NIH/DHHS, Research Triangle Park, North Carolina
| |
Collapse
|
19
|
Celikkaya H, Cosacak MI, Papadimitriou C, Popova S, Bhattarai P, Biswas SN, Siddiqui T, Wistorf S, Nevado-Alcalde I, Naumann L, Mashkaryan V, Brandt K, Freudenberg U, Werner C, Kizil C. GATA3 Promotes the Neural Progenitor State but Not Neurogenesis in 3D Traumatic Injury Model of Primary Human Cortical Astrocytes. Front Cell Neurosci 2019; 13:23. [PMID: 30809125 PMCID: PMC6380212 DOI: 10.3389/fncel.2019.00023] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/18/2019] [Indexed: 01/05/2023] Open
Abstract
Astrocytes are abundant cell types in the vertebrate central nervous system and can act as neural stem cells in specialized niches where they constitutively generate new neurons. Outside the stem cell niches, however, these glial cells are not neurogenic. Although injuries in the mammalian central nervous system lead to profound proliferation of astrocytes, which cluster at the lesion site to form a gliotic scar, neurogenesis does not take place. Therefore, a plausible regenerative therapeutic option is to coax the endogenous reactive astrocytes to a pre-neurogenic progenitor state and use them as an endogenous reservoir for repair. However, little is known on the mechanisms that promote the neural progenitor state after injuries in humans. Gata3 was previously found to be a mechanism that zebrafish brain uses to injury-dependent induction of neural progenitors. However, the effects of GATA3 in human astrocytes after injury are not known. Therefore, in this report, we investigated how overexpression of GATA3 in primary human astrocytes would affect the neurogenic potential before and after injury in 2D and 3D cultures. We found that primary human astrocytes are unable to induce GATA3 after injury. Lentivirus-mediated overexpression of GATA3 significantly increased the number of GFAP/SOX2 double positive astrocytes and expression of pro-neural factor ASCL1, but failed to induce neurogenesis, suggesting that GATA3 is required for enhancing the neurogenic potential of primary human astrocytes and is not sufficient to induce neurogenesis alone.
Collapse
Affiliation(s)
- Hilal Celikkaya
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany.,Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Mehmet Ilyas Cosacak
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany
| | | | - Stanislava Popova
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany.,Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Prabesh Bhattarai
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany
| | - Srijeeta Nag Biswas
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany.,Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Tohid Siddiqui
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany
| | - Sabrina Wistorf
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany
| | - Isabel Nevado-Alcalde
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany
| | - Lisa Naumann
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany
| | - Violeta Mashkaryan
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany.,Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| | - Kerstin Brandt
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany
| | - Uwe Freudenberg
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany.,Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
| | - Carsten Werner
- Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany.,Max Bergmann Center of Biomaterials Dresden, Leibniz Institute of Polymer Research Dresden, Dresden, Germany
| | - Caghan Kizil
- German Center for Neurodegenerative Diseases, Helmholtz Association, Dresden, Germany.,Center for Regenerative Therapies Dresden, TU Dresden, Dresden, Germany
| |
Collapse
|
20
|
Cwetsch AW, Pinto B, Savardi A, Cancedda L. In vivo methods for acute modulation of gene expression in the central nervous system. Prog Neurobiol 2018; 168:69-85. [PMID: 29694844 PMCID: PMC6080705 DOI: 10.1016/j.pneurobio.2018.04.008] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2017] [Revised: 04/17/2018] [Accepted: 04/20/2018] [Indexed: 12/17/2022]
Abstract
Accurate and timely expression of specific genes guarantees the healthy development and function of the brain. Indeed, variations in the correct amount or timing of gene expression lead to improper development and/or pathological conditions. Almost forty years after the first successful gene transfection in in vitro cell cultures, it is currently possible to regulate gene expression in an area-specific manner at any step of central nervous system development and in adulthood in experimental animals in vivo, even overcoming the very poor accessibility of the brain. Here, we will review the diverse approaches for acute gene transfer in vivo, highlighting their advantages and disadvantages with respect to the efficiency and specificity of transfection as well as to brain accessibility. In particular, we will present well-established chemical, physical and virus-based approaches suitable for different animal models, pointing out their current and future possible applications in basic and translational research as well as in gene therapy.
Collapse
Affiliation(s)
- Andrzej W Cwetsch
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Bruno Pinto
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Bio@SNS, Scuola Normale Superiore, Piazza dei Cavalieri 7, 56126, Pisa, Italy
| | - Annalisa Savardi
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; Università degli Studi di Genova, Via Balbi, 5, 16126 Genova, Italy
| | - Laura Cancedda
- Local Micro-environment and Brain Development Laboratory, Istituto Italiano di Tecnologia, via Morego, 30, 16163 Genova, Italy; DulbeccoTelethon Institute, Italy.
| |
Collapse
|
21
|
Sun S, Schaffer DV. Engineered viral vectors for functional interrogation, deconvolution, and manipulation of neural circuits. Curr Opin Neurobiol 2018; 50:163-170. [PMID: 29614429 PMCID: PMC5984719 DOI: 10.1016/j.conb.2017.12.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 11/27/2017] [Accepted: 12/16/2017] [Indexed: 12/19/2022]
Abstract
Optimization of traditional replication-competent viral tracers has granted access to immediate synaptic partners of target neuronal populations, enabling the dissection of complex brain circuits into functional neural pathways. The excessive virulence of most conventional tracers, however, impedes their utility in revealing and genetically perturbing cellular function on long time scales. As a promising alternative, the natural capacity of adeno-associated viral (AAV) vectors to safely mediate persistent and robust gene expression has stimulated strong interest in adapting them for sparse neuronal labeling and physiological studies. Furthermore, increasingly refined engineering strategies have yielded novel AAV variants with enhanced target specificity, transduction, and retrograde trafficking in the CNS. These potent vectors offer new opportunities for characterizing the identity and connectivity of single neurons within immense networks and modulating their activity via robust delivery of functional genetic tools.
Collapse
Affiliation(s)
- Sabrina Sun
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA; Department of Bioengineering, University of California, Berkeley, CA, USA; The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA.
| |
Collapse
|
22
|
Crowther AJ, Lim SA, Asrican B, Albright BH, Wooten J, Yeh CY, Bao H, Cerri DH, Hu J, Ian Shih YY, Asokan A, Song J. An Adeno-Associated Virus-Based Toolkit for Preferential Targeting and Manipulating Quiescent Neural Stem Cells in the Adult Hippocampus. Stem Cell Reports 2018; 10:1146-1159. [PMID: 29478897 PMCID: PMC5918266 DOI: 10.1016/j.stemcr.2018.01.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/17/2018] [Accepted: 01/18/2018] [Indexed: 01/12/2023] Open
Abstract
Quiescent neural stem cells (qNSCs) with radial morphology are the only proven source of new neurons in the adult mammalian brain. Our understanding of the roles of newly generated neurons depends on the ability to target and manipulate adult qNSCs. Although various strategies have been developed to target and manipulate adult hippocampal qNSCs, they often suffer from prolonged breeding, low recombination efficiency, and non-specific labeling. Therefore, developing a readily manufactured viral vector that allows flexible packaging and robust expression of various transgenes in qNSCs is a pressing need. Here, we report a recombinant adeno-associated virus serotype 4 (rAAV4)-based toolkit that preferentially targets hippocampal qNSCs and allows for lineage tracing, functional analyses, and activity manipulation of adult qNSCs. Importantly, targeting qNSCs in a non-Cre-dependent fashion opens the possibility for studying qNSCs in less genetically tractable animal species and may have translational impact in gene therapy by preferentially targeting qNSCs. rAAV4 vectors preferentially target quiescent NSCs in the adult hippocampus rAAV4 vectors with distinct promoters reveal differential selectivity for radial NSCs rAAV4 allows for genetic manipulation and lineage tracing of quiescent NSCs rAAV4 allows for calcium imaging and activity manipulation of quiescent NSCs
Collapse
Affiliation(s)
- Andrew J Crowther
- Department of Pharmacology and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Szu-Aun Lim
- Department of Pharmacology and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Brent Asrican
- Department of Pharmacology and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Blake H Albright
- Department of Genetics and Gene Therapy Center, University of North Carolina, Chapel Hill, NC 27599, USA; Genetics and Molecular Biology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Josh Wooten
- Department of Pharmacology and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Genetics and Molecular Biology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Chia-Yu Yeh
- Department of Pharmacology and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Hechen Bao
- Department of Pharmacology and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Domenic H Cerri
- Department of Neurology and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Jessica Hu
- Department of Pharmacology and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Yen-Yu Ian Shih
- Department of Neurology and Biomedical Research Imaging Center, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Aravind Asokan
- Department of Genetics and Gene Therapy Center, University of North Carolina, Chapel Hill, NC 27599, USA; Genetics and Molecular Biology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA
| | - Juan Song
- Department of Pharmacology and Neuroscience Center, University of North Carolina, Chapel Hill, NC 27599, USA; Neurobiology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA; Genetics and Molecular Biology Curriculum, University of North Carolina, Chapel Hill, NC 27599, USA.
| |
Collapse
|
23
|
Encinas JM, Fitzsimons CP. Gene regulation in adult neural stem cells. Current challenges and possible applications. Adv Drug Deliv Rev 2017; 120:118-132. [PMID: 28751200 DOI: 10.1016/j.addr.2017.07.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/17/2017] [Accepted: 07/19/2017] [Indexed: 12/13/2022]
Abstract
Adult neural stem and progenitor cells (NSPCs) offer a unique opportunity for neural regeneration and niche modification in physiopathological conditions, harnessing the capability to modify from neuronal circuits to glial scar. Findings exposing the vast plasticity and potential of NSPCs have accumulated over the past years and we currently know that adult NSPCs can naturally give rise not only to neurons but also to astrocytes and reactive astrocytes, and eventually to oligodendrocytes through genetic manipulation. We can consider NSPCs as endogenous flexible tools to fight against neurodegenerative and neurological disorders and aging. In addition, NSPCs can be considered as active agents contributing to chronic brain alterations and as relevant cell populations to be preserved, so that their main function, neurogenesis, is not lost in damage or disease. Altogether we believe that learning to manipulate NSPC is essential to prevent, ameliorate or restore some of the cognitive deficits associated with brain disease and injury, and therefore should be considered as target for future therapeutic strategies. The first step to accomplish this goal is to target them specifically, by unveiling and understanding their unique markers and signaling pathways.
Collapse
Affiliation(s)
- Juan Manuel Encinas
- Achucarro Basque Center for Neuroscience, Bizkaia Science and Technology Park, 205, 48170 Zamudio, Spain; Ikerbasque, The Basque Science Foundation, María Díaz de Haro 3, 6(th) Floor, 48013 Bilbao, Spain; University of the Basque Country (UPV/EHU), Barrio Sarriena s/n, 48940 Leioa, Spain.
| | - Carlos P Fitzsimons
- Neuroscience Program, Swammerdam Institute for Life Sciences, Faculty of Sciences, University of Amsterdam, SciencePark 904, 1098XH Amsterdam, The Netherlands.
| |
Collapse
|
24
|
In Vivo Selection of a Computationally Designed SCHEMA AAV Library Yields a Novel Variant for Infection of Adult Neural Stem Cells in the SVZ. Mol Ther 2017; 26:304-319. [PMID: 28988711 DOI: 10.1016/j.ymthe.2017.09.006] [Citation(s) in RCA: 71] [Impact Index Per Article: 8.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2017] [Revised: 08/29/2017] [Accepted: 09/03/2017] [Indexed: 01/17/2023] Open
Abstract
Directed evolution continues to expand the capabilities of complex biomolecules for a range of applications, such as adeno-associated virus vectors for gene therapy; however, advances in library design and selection strategies are key to develop variants that overcome barriers to clinical translation. To address this need, we applied structure-guided SCHEMA recombination of the multimeric adeno-associated virus (AAV) capsid to generate a highly diversified chimeric library with minimal structural disruption. A stringent in vivo Cre-dependent selection strategy was implemented to identify variants that transduce adult neural stem cells (NSCs) in the subventricular zone. A novel variant, SCH9, infected 60% of NSCs and mediated 24-fold higher GFP expression and a 12-fold greater transduction volume than AAV9. SCH9 utilizes both galactose and heparan sulfate as cell surface receptors and exhibits increased resistance to neutralizing antibodies. These results establish the SCHEMA library as a valuable tool for directed evolution and SCH9 as an effective gene delivery vector to investigate subventricular NSCs.
Collapse
|
25
|
Combining Engineered Nucleases with Adeno-associated Viral Vectors for Therapeutic Gene Editing. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1016:29-42. [PMID: 29130152 DOI: 10.1007/978-3-319-63904-8_2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
With the recent advent of several generations of targeted DNA nucleases, most recently CRISPR/Cas9, genome editing has become broadly accessible across the biomedical community. Importantly, the capacity of these nucleases to modify specific genomic loci associated with human disease could render new classes of genetic disease, including autosomal dominant or even idiopathic disease, accessible to gene therapy. In parallel, the emergence of adeno-associated virus (AAV) as a clinically important vector raises the possibility of integrating these two technologies towards the development of gene editing therapies. Though clear challenges exist, numerous proof-of-concept studies in preclinical models offer exciting promise for the future of gene therapy.
Collapse
|
26
|
A Kotterman M, Schaffer DV. Engineered AAV vectors for improved central nervous system gene delivery. NEUROGENESIS 2015; 2:e1122700. [PMID: 27606332 PMCID: PMC4973602 DOI: 10.1080/23262133.2015.1122700] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 08/20/2015] [Revised: 11/11/2015] [Accepted: 11/17/2015] [Indexed: 12/02/2022]
Abstract
Adeno-associated viruses (AAV) are non-pathogenic members of the Parvoviridae family that are being harnessed as delivery vehicles for both basic research and increasingly successful clinical gene therapy. To address a number of delivery shortcomings with natural AAV variants, we have developed and implemented directed evolution—a high-throughput molecular engineering approach to generate novel biomolecules with enhanced function—to create novel AAV vectors that are designed to preferentially transduce specific cell types in the central nervous system (CNS), including astrocytes, neural stem cells, and cells within the retina. These novel AAV vectors—which have enhanced infectivity in vitro and enhanced infectivity and selectivity in vivo—can enable more efficient studies to further our understanding of neurogenesis, development, aging, and disease. Furthermore, such engineered vectors may aid gene or cell replacement therapies to treat neurodegenerative disease or injury.
Collapse
Affiliation(s)
| | - David V Schaffer
- 4D Molecular Therapeutics; Emeryville, CA USA; Departments of Chemical and Biomolecular Engineering, Bioengineering, and Molecular and Cell Biology; University of California; Berkeley, CA USA
| |
Collapse
|