1
|
Yang L, Fang LZ, Lynch MR, Xu CS, Hahm HJ, Zhang Y, Heitmeier MR, Costa VD, Samineni VK, Creed MC. Transcriptomic landscape of mammalian ventral pallidum at single-cell resolution. SCIENCE ADVANCES 2024; 10:eadq6017. [PMID: 39661664 PMCID: PMC11633743 DOI: 10.1126/sciadv.adq6017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 11/04/2024] [Indexed: 12/13/2024]
Abstract
The ventral pallidum (VP) is critical for motivated behaviors. While contemporary work has begun to elucidate the functional diversity of VP neurons, the molecular heterogeneity underlying this functional diversity remains incompletely understood. We used single-nucleus RNA sequencing and in situ hybridization to define the transcriptional taxonomy of VP cell types in mice, macaques, and baboons. We found transcriptional conservation between all three species, within the broader neurochemical cell types. Unique dopaminoceptive and cholinergic subclusters were identified and conserved across both primate species but had no homolog in mice. This harmonized consensus VP cellular atlas will pave the way for understanding the structure and function of the VP and identified key neuropeptides, neurotransmitters, and neurotransmitter receptors that could be targeted within specific VP cell types for functional investigations.
Collapse
Affiliation(s)
- Lite Yang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Lisa Z. Fang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Michelle R. Lynch
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- NINDS Neuroscience Postbaccalaureate Program, Washington University School of Medicine, St. Louis, MO, USA
| | - Chang S. Xu
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Hannah J. Hahm
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Yufen Zhang
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Monique R. Heitmeier
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
| | - Vincent D. Costa
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA, USA
- Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Atlanta, GA, USA
| | - Vijay K. Samineni
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
| | - Meaghan C. Creed
- Department of Anesthesiology, Washington University Pain Center, Washington University School of Medicine, St. Louis, MO, USA
- Neuroscience Graduate Program, Division of Biology & Biomedical Sciences, Washington University School of Medicine, St. Louis, MO, USA
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
2
|
Wang JH, Zhan W, Gallagher TL, Gao G. Recombinant adeno-associated virus as a delivery platform for ocular gene therapy: A comprehensive review. Mol Ther 2024; 32:4185-4207. [PMID: 39489915 PMCID: PMC11638839 DOI: 10.1016/j.ymthe.2024.10.017] [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: 05/10/2024] [Revised: 09/18/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Adeno-associated virus (AAV) has emerged as a leading platform for in vivo gene therapy, particularly in ocular diseases. AAV-based therapies are characterized by low pathogenicity and broad tissue tropism and have demonstrated clinical success, as exemplified by voretigene neparvovec-rzyl (Luxturna) being the first gene therapy to be approved by the U.S. Food and Drug Administration to treat RPE65-associated Leber congenital amaurosis (LCA). However, several challenges remain in the development of AAV-based gene therapies, including immune responses, limited cargo capacity, and the need for enhanced transduction efficiency, especially for intravitreal delivery to photoreceptors and retinal pigment epithelium cells. This review explores the biology of AAVs in the context of gene therapy, innovations in capsid engineering, and clinical advancements in AAV-based ocular gene therapy. We highlight ongoing clinical trials targeting inherited retinal diseases and acquired conditions, discuss immune-related limitations, and examine novel strategies for enhancing AAV vector performance to address current barriers.
Collapse
Affiliation(s)
- Jiang-Hui Wang
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC 3002, Australia; Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC 3002, Australia
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Thomas L Gallagher
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA 01605, USA; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA 01605, USA; Li Weibo Institute for Rare Diseases Research, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
3
|
Totty MS, Juanes RC, Bach SV, Ameur LB, Valentine MR, Simons E, Romac M, Trinh H, Henderson K, Del Rosario I, Tippani M, Miller RA, Kleinman JE, Page SC, Saunders A, Hyde TM, Martinowich K, Hicks SC, Costa VD. Transcriptomic diversity of amygdalar subdivisions across humans and nonhuman primates. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.18.618721. [PMID: 39463931 PMCID: PMC11507838 DOI: 10.1101/2024.10.18.618721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/29/2024]
Abstract
The amygdaloid complex mediates learning, memory, and emotions. Understanding the cellular and anatomical features that are specialized in the amygdala of primates versus other vertebrates requires a systematic, anatomically-resolved molecular analysis of constituent cell populations. We analyzed five nuclear subdivisions of the primate amygdala with single-nucleus RNA sequencing in macaques, baboons, and humans to examine gene expression profiles for excitatory and inhibitory neurons and confirmed our results with single-molecule FISH analysis. We identified distinct subtypes of FOXP2 + interneurons in the intercalated cell masses and protein-kinase C-δ interneurons in the central nucleus. We also establish that glutamatergic, pyramidal-like neurons are transcriptionally specialized within the basal, lateral, or accessory basal nuclei. Understanding the molecular heterogeneity of anatomically-resolved amygdalar neuron types provides a cellular framework for improving existing models of how amygdalar neural circuits contribute to cognition and mental health in humans by using nonhuman primates as a translational bridge.
Collapse
Affiliation(s)
- Michael S. Totty
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Rita Cervera Juanes
- Department of Translational Neuroscience, Center for Precision Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Svitlana V. Bach
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Lamya Ben Ameur
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Madeline R. Valentine
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Evan Simons
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - McKenna Romac
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
- Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Atlanta, GA, USA
| | - Hoa Trinh
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Krystal Henderson
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
| | - Ishbel Del Rosario
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Madhavi Tippani
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Ryan A. Miller
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Joel E. Kleinman
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Stephanie Cerceo Page
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
| | - Arpiar Saunders
- Vollum Institute, Oregon Health and Science University, Portland, OR, USA
| | - Thomas M. Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Neurology, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Keri Martinowich
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD, USA
- Department of Psychiatry and Behavioral Sciences, Johns Hopkins School of Medicine, Baltimore, MD, USA
- The Solomon H. Snyder Department of Neuroscience, Johns Hopkins School of Medicine, Baltimore, MD, USA
- Johns Hopkins Kavli Neuroscience Discovery Institute, Baltimore, MD, USA
| | - Stephanie C. Hicks
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
- Center for Computational Biology, Johns Hopkins University, Baltimore, MD, USA
- Malone Center for Engineering in Healthcare, Johns Hopkins University, Baltimore, MD, USA
| | - Vincent D. Costa
- Division of Neuroscience, Oregon National Primate Research Center, Beaverton, OR, USA
- Division of Developmental and Cognitive Neuroscience, Emory National Primate Research Center, Atlanta, GA, USA
- Department of Psychiatry and Behavioral Sciences, Emory University, Atlanta, GA 30329, USA
| |
Collapse
|
4
|
Voronin G, Narasimhan J, Gittens J, Sheedy J, Lipari P, Peters M, DeMarco S, Cao L, Varganov Y, Kim MJ, Pear L, Fotouh E, Sinha S, Ray B, Wu MC, Yalamanchili P, Southgate C, Pick J, Saadipour K, Jung S, Lee J, Mollin A, Welch EM, Wu Z, Weetall M. Preclinical studies of gene replacement therapy for CDKL5 deficiency disorder. Mol Ther 2024; 32:3331-3345. [PMID: 39033321 PMCID: PMC11489525 DOI: 10.1016/j.ymthe.2024.07.012] [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: 01/06/2024] [Revised: 04/13/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024] Open
Abstract
Cyclin-dependent kinase-like 5 (CDKL5) deficiency disorder (CDD) is a rare neurodevelopmental disorder caused by a mutation in the X-linked CDKL5 gene. CDKL5 is a serine/threonine kinase that is critical for axon outgrowth and dendritic morphogenesis as well as synapse formation, maturation, and maintenance. This disorder is characterized by early-onset epilepsy, hypotonia, and failure to reach cognitive and motor developmental milestones. Because the disease is monogenic, delivery of the CDKL5 gene to the brain of patients should provide clinical benefit. To this end, we designed a gene therapy vector, adeno-associated virus (AAV)9.Syn.hCDKL5, in which human CDKL5 gene expression is driven by the synapsin promoter. In biodistribution studies conducted in mice, intracerebroventricular (i.c.v.) injection resulted in broader, more optimal biodistribution than did intra-cisterna magna (i.c.m.) delivery. AAV9.Syn.hCDKL5 treatment increased phosphorylation of EB2, a bona fide CDKL5 substrate, demonstrating biological activity in vivo. Our data provide proof of concept that i.c.v. delivery of AAV9.Syn.hCDKL5 to neonatal male Cdkl5 knockout mice reduces pathology and reduces aberrant behavior. Functional improvements were seen at doses of 3e11 to 5e11 vector genomes/g brain, which resulted in transfection of ≥50% of the neurons. Functional improvements were not seen at lower doses, suggesting a requirement for broad distribution for efficacy.
Collapse
Affiliation(s)
- Gregory Voronin
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Jana Narasimhan
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Jamila Gittens
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Josephine Sheedy
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Philip Lipari
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Melinda Peters
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Steven DeMarco
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Liangxian Cao
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Yakov Varganov
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Min Jung Kim
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Lisset Pear
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Eman Fotouh
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Supriya Sinha
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Balmiki Ray
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Michael C Wu
- NeuroDigitech, 8400 Miramar Road, Suite 243C, San Diego, CA 092126, USA
| | | | | | - Joseph Pick
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Khalil Saadipour
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Stephen Jung
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Jeanee Lee
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Anna Mollin
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Ellen M Welch
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Zhijian Wu
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA
| | - Marla Weetall
- PTC Therapeutics, Inc, 500 Warren Corporate Center Drive, Warren, NJ 07059, USA.
| |
Collapse
|
5
|
Yang L, Fang LZ, Lynch MR, Xu CS, Hahm H, Zhang Y, Heitmeier MR, Costa V, Samineni VK, Creed MC. Transcriptomic landscape of mammalian ventral pallidum at single-cell resolution. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.24.595793. [PMID: 38826431 PMCID: PMC11142225 DOI: 10.1101/2024.05.24.595793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
The ventral pallidum (VP) is critical for motivated behaviors. While contemporary work has begun to elucidate the functional diversity of VP neurons, the molecular heterogeneity underlying this functional diversity remains incompletely understood. We used snRNA-seq and in situ hybridization to define the transcriptional taxonomy of VP cell types in mice, macaques, and baboons. We found transcriptional conservation between all three species, within the broader neurochemical cell types. Unique dopaminoceptive and cholinergic subclusters were identified and conserved across both primate species but had no homolog in mice. This harmonized consensus VP cellular atlas will pave the way for understanding the structure and function of the VP and identified key neuropeptides, neurotransmitters, and neuro receptors that could be targeted within specific VP cell types for functional investigations. Teaser Genetic identity of ventral pallidum cell types is conserved across rodents and primates at the transcriptional level.
Collapse
|
6
|
Wang H, Li R, Sadekar S, Kamath AV, Shen BQ. A novel approach to quantitate biodistribution and transduction of adeno-associated virus gene therapy using radiolabeled AAV vectors in mice. Mol Ther Methods Clin Dev 2024; 32:101326. [PMID: 39286334 PMCID: PMC11404148 DOI: 10.1016/j.omtm.2024.101326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Accepted: 08/15/2024] [Indexed: 09/19/2024]
Abstract
An understanding of recombinant adeno-associated virus (AAV) biodistribution profiles is an important element of a preclinical development program. Here, we have developed a radiolabeling strategy utilizing the co-delivery of 125I (non-residualizing) and 111In (residualizing) radionuclide-conjugated AAVs to provide a detailed distribution quantification at tissue level delineating between the cellular internalized AAV (degraded, 111In-125I) and AAV remaining in the extracellular matrix (intact, 125I). This labeling method has been successfully applied to AAV9 and AAV-PHP.eB as tool molecules without altering the physical properties and biological activities of the AAVs. Upon labeling with either of the radioactive probes, these molecules were systemically injected into C57BL/6 mice. The biodistribution results indicate that AAVs, with a fast distribution profile, were mainly located in the extracellular matrix of highly perfused organs such as liver and spleen at early time points, leading to a difference between capsid quantification and vector genome quantification. The results suggest that the 125I-AAV/111In-AAV co-delivery approach offers a robust and efficient analytical strategy to investigate the detailed tissue distribution of AAV vectors, including both vector genome and protein capsids. This novel method has the potential to be applied to capsid optimization, selection, and lead candidate development.
Collapse
Affiliation(s)
- Hongzhi Wang
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ran Li
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Shraddha Sadekar
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Amrita V Kamath
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| | - Ben-Quan Shen
- Preclinical and Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, CA 94080, USA
| |
Collapse
|
7
|
Mandalawatta HP, Rajendra K, Fairfax K, Hewitt AW. Emerging trends in virus and virus-like particle gene therapy delivery to the brain. MOLECULAR THERAPY. NUCLEIC ACIDS 2024; 35:102280. [PMID: 39206077 PMCID: PMC11350507 DOI: 10.1016/j.omtn.2024.102280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Recent advances in gene therapy and gene-editing techniques offer the very real potential for successful treatment of neurological diseases. However, drug delivery constraints continue to impede viable therapeutic interventions targeting the brain due to its anatomical complexity and highly restrictive microvasculature that is impervious to many molecules. Realizing the therapeutic potential of gene-based therapies requires robust encapsulation and safe and efficient delivery to the target cells. Although viral vectors have been widely used for targeted delivery of gene-based therapies, drawbacks such as host genome integration, prolonged expression, undesired off-target mutations, and immunogenicity have led to the development of alternative strategies. Engineered virus-like particles (eVLPs) are an emerging, promising platform that can be engineered to achieve neurotropism through pseudotyping. This review outlines strategies to improve eVLP neurotropism for therapeutic brain delivery of gene-editing agents.
Collapse
Affiliation(s)
| | - K.C. Rajendra
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
| | - Kirsten Fairfax
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| | - Alex W. Hewitt
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia
- School of Medicine, University of Tasmania, Hobart, TAS, Australia
| |
Collapse
|
8
|
Shay TF, Jang S, Brittain TJ, Chen X, Walker B, Tebbutt C, Fan Y, Wolfe DA, Arokiaraj CM, Sullivan EE, Ding X, Wang TY, Lei Y, Chuapoco MR, Chou TF, Gradinaru V. Human cell surface-AAV interactomes identify LRP6 as blood-brain barrier transcytosis receptor and immune cytokine IL3 as AAV9 binder. Nat Commun 2024; 15:7853. [PMID: 39245720 PMCID: PMC11381518 DOI: 10.1038/s41467-024-52149-0] [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: 06/06/2024] [Accepted: 08/27/2024] [Indexed: 09/10/2024] Open
Abstract
Adeno-associated viruses (AAVs) are foundational gene delivery tools for basic science and clinical therapeutics. However, lack of mechanistic insight, especially for engineered vectors created by directed evolution, can hamper their application. Here, we adapt an unbiased human cell microarray platform to determine the extracellular and cell surface interactomes of natural and engineered AAVs. We identify a naturally-evolved and serotype-specific interaction between the AAV9 capsid and human interleukin 3 (IL3), with possible roles in host immune modulation, as well as lab-evolved low-density lipoprotein receptor-related protein 6 (LRP6) interactions specific to engineered capsids with enhanced blood-brain barrier crossing in non-human primates after intravenous administration. The unbiased cell microarray screening approach also allows us to identify off-target tissue binding interactions of engineered brain-enriched AAV capsids that may inform vectors' peripheral organ tropism and side effects. Our cryo-electron tomography and AlphaFold modeling of capsid-interactor complexes reveal LRP6 and IL3 binding sites. These results allow confident application of engineered AAVs in diverse organisms and unlock future target-informed engineering of improved viral and non-viral vectors for non-invasive therapeutic delivery to the brain.
Collapse
Affiliation(s)
- Timothy F Shay
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| | - Seongmin Jang
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Tyler J Brittain
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Xinhong Chen
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Beth Walker
- Charles River Laboratories, High Peak Business Park, Buxton Road, Chinley, SK23 6FJ, UK
| | - Claire Tebbutt
- Charles River Laboratories, High Peak Business Park, Buxton Road, Chinley, SK23 6FJ, UK
| | - Yujie Fan
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Damien A Wolfe
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Cynthia M Arokiaraj
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Erin E Sullivan
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Xiaozhe Ding
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Ting-Yu Wang
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Yaping Lei
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Miguel R Chuapoco
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Tsui-Fen Chou
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Viviana Gradinaru
- Division of Biology & Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA.
| |
Collapse
|
9
|
Chauhan M, Daugherty AL, Khadir FE, Duzenli OF, Hoffman A, Tinklenberg JA, Kang PB, Aslanidi G, Pacak CA. AAV-DJ is superior to AAV9 for targeting brain and spinal cord, and de-targeting liver across multiple delivery routes in mice. J Transl Med 2024; 22:824. [PMID: 39237935 PMCID: PMC11375878 DOI: 10.1186/s12967-024-05599-5] [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: 05/17/2024] [Accepted: 08/12/2024] [Indexed: 09/07/2024] Open
Abstract
Highly efficient adeno associated viruses (AAVs) targeting the central nervous system (CNS) are needed to deliver safe and effective therapies for inherited neurological disorders. The goal of this study was to compare the organ-specific transduction efficiencies of two AAV capsids across three different delivery routes. We compared AAV9-CBA-fLucYFP to AAV-DJ-CBA-fLucYFP using the following delivery routes in mice: intracerebroventricular (ICV) 1 × 1012 vg/kg, intrathecal (IT) 1 × 1012 vg/kg, and intravenous (IV) 1 × 1013 vg/kg body weight. Our evaluations revealed that following ICV and IT administrations, AAV-DJ demonstrated significantly increased vector genome (vg) uptake throughout the CNS as compared to AAV9. Through the IV route, AAV9 demonstrated significantly increased vg uptake in the CNS. However, significantly fewer vgs were detected in the off-target organs (kidney and liver) following administration of AAV-DJ using the IT and IV delivery routes as compared to AAV9. Distributions of vgs correlate well with transgene transcript levels, luciferase enzyme activities, and immunofluorescence detection of YFP. Overall, between the two vectors, AAV-DJ resulted in better targeting and expression in CNS tissues paired with de-targeting and reduced expression in liver and kidneys. Our findings support further examination of AAV-DJ as a gene therapy capsid for the treatment of neurological disorders.
Collapse
Affiliation(s)
- Monika Chauhan
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - Audrey L Daugherty
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - Fatemeh Ellie Khadir
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - Ozgun F Duzenli
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | | | - Jennifer A Tinklenberg
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - Peter B Kang
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA
| | - George Aslanidi
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Christina A Pacak
- Department of Neurology, Greg Marzolf Jr. Muscular Dystrophy Center, University of Minnesota Medical School, 420 Delaware Street SE, MMC 295, Minneapolis, Minnesota, MN, 55455, USA.
| |
Collapse
|
10
|
Bunuales M, Garduno A, Chillon M, Bosch A, Gonzalez-Aparicio M, Espelosin M, Garcia-Gomara M, Rico AJ, Garcia-Osta A, Cuadrado-Tejedor M, Lanciego JL, Hernandez-Alcoceba R. Characterization of brain transduction capability of a BBB-penetrant AAV vector in mice, rats and macaques reveals differences in expression profiles. Gene Ther 2024; 31:455-466. [PMID: 39039204 PMCID: PMC11399087 DOI: 10.1038/s41434-024-00466-w] [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: 05/16/2024] [Revised: 07/03/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024]
Abstract
Different screening methods are being developed to generate adeno-associated viral vectors (AAV) with the ability to bypass the blood-brain barrier (BBB) upon intravenous administration. Recently, the AAV9P31 stood out as the most efficient version among a library of peptide-displaying capsids selected in C57BL/6 mice using RNA-driven biopanning. In this work we have characterized in detail its biodistribution in different mouse strains (C57BL/6 and Balb/c), as well as in Sprague Dawley rats and non-human primates (Macaca fascicularis). Using GFP and NanoLuc reporter genes, we confirmed homogeneous infection and transgene expression across the CNS of mice injected intravenously with AAV9P31. A more restricted pattern was observed upon either intracerebroventricular or intraparenchymal injection. Following intravenous delivery, region- and cell-specific differential patterns of transduction were observed in the mouse brain, including a preferential transduction of astrocytes and neurons in the cerebral cortex and striatum, whereas neurons were the only transduced cell type in subcortical locations across the hippocampus, thalamus, hypothalamus, mesencephalon, brainstem and cerebellum. Furthermore, transduced microglial cells were never found in any CNS location. Peripheral organs transduced upon intravenous administration included lung, liver, peritoneum, heart and skeletal muscle. However, a comparable performance of AAV9P31 to bypass the BBB in rats and macaques was not observed, although a more limited neuronal transduction was found in the brainstem of rats upon intravenous delivery. Finally, intracerebroventricular delivery in macaques resulted in neuronal transduction in cortical, subcortical structures and cerebellum following a patchy pattern. In conclusion, the widespread CNS transduction obtained in mice upon intravenous delivery of AAV9P31 represents a powerful tool for modeling a wide variety of neurological disorders as well as an appealing choice for the evaluation of gene therapy-based therapeutics.
Collapse
Affiliation(s)
- María Bunuales
- Division of DNA and RNA medicine, CIMA. University of Navarra, Pamplona, Spain
| | - Angeles Garduno
- Division of DNA and RNA medicine, CIMA. University of Navarra, Pamplona, Spain
| | - Miguel Chillon
- Institut de Neurociències, Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona; and Vall d'Hebron Research Institute, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Assumpció Bosch
- Institut de Neurociències, Biochemistry and Molecular Biology Department, Universitat Autònoma de Barcelona; and Vall d'Hebron Research Institute, Barcelona, Spain
| | | | - Maria Espelosin
- Division of DNA and RNA medicine, CIMA. University of Navarra, Pamplona, Spain
| | - Marta Garcia-Gomara
- Division of DNA and RNA medicine, CIMA. University of Navarra, Pamplona, Spain
- Pathology, Anatomy and Physiology Department, School of Medicine, University of Navarra, IdiSNA, 31008, Pamplona, Spain
| | - Alberto J Rico
- Division of DNA and RNA medicine, CIMA. University of Navarra, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), Madrid, Spain
| | - Ana Garcia-Osta
- Division of DNA and RNA medicine, CIMA. University of Navarra, Pamplona, Spain
| | - Mar Cuadrado-Tejedor
- Division of DNA and RNA medicine, CIMA. University of Navarra, Pamplona, Spain
- Pathology, Anatomy and Physiology Department, School of Medicine, University of Navarra, IdiSNA, 31008, Pamplona, Spain
| | - Jose L Lanciego
- Division of DNA and RNA medicine, CIMA. University of Navarra, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CiberNed), Madrid, Spain.
| | | |
Collapse
|
11
|
Fortier M, Cauhapé M, Buono S, Becker J, Menuet A, Branchu J, Ricca I, Mero S, Dorgham K, El Hachimi KH, Dobrenis K, Colsch B, Samaroo D, Devaux M, Durr A, Stevanin G, Santorelli FM, Colombo S, Cowling B, Darios F. Decreasing ganglioside synthesis delays motor and cognitive symptom onset in Spg11 knockout mice. Neurobiol Dis 2024; 199:106564. [PMID: 38876323 DOI: 10.1016/j.nbd.2024.106564] [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: 01/30/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 06/16/2024] Open
Abstract
Biallelic variants in the SPG11 gene account for the most common form of autosomal recessive hereditary spastic paraplegia characterized by motor and cognitive impairment, with currently no therapeutic option. We previously observed in a Spg11 knockout mouse that neurodegeneration is associated with accumulation of gangliosides in lysosomes. To test whether a substrate reduction therapy could be a therapeutic option, we downregulated the key enzyme involved in ganglioside biosynthesis using an AAV-PHP.eB viral vector expressing a miRNA targeting St3gal5. Downregulation of St3gal5 in Spg11 knockout mice prevented the accumulation of gangliosides, delayed the onset of motor and cognitive symptoms, and prevented the upregulation of serum levels of neurofilament light chain, a biomarker widely used in neurodegenerative diseases. Importantly, similar results were observed when Spg11 knockout mice were administrated venglustat, a pharmacological inhibitor of glucosylceramide synthase expected to decrease ganglioside synthesis. Downregulation of St3gal5 or venglustat administration in Spg11 knockout mice strongly decreased the formation of axonal spheroids, previously associated with impaired trafficking. Venglustat had similar effect on cultured human SPG11 neurons. In conclusion, this work identifies the first disease-modifying therapeutic strategy in SPG11, and provides data supporting its relevance for therapeutic testing in SPG11 patients.
Collapse
Affiliation(s)
- Manon Fortier
- Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), INSERM U1127, CNRS UMR 7225, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Margaux Cauhapé
- Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), INSERM U1127, CNRS UMR 7225, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Suzie Buono
- Dynacure SA (now Flamingo Therapeutics NV), Illkirch, France
| | - Julien Becker
- Dynacure SA (now Flamingo Therapeutics NV), Illkirch, France
| | - Alexia Menuet
- Dynacure SA (now Flamingo Therapeutics NV), Illkirch, France
| | - Julien Branchu
- Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), INSERM U1127, CNRS UMR 7225, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Ivana Ricca
- Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Serena Mero
- Molecular Medicine, IRCCS Fondazione Stella Maris, 56128 Pisa, Italy
| | - Karim Dorgham
- Sorbonne Université, INSERM, Centre d'Immunologie et des Maladies Infectieuses-Paris (CIMI-Paris), Paris, France
| | - Khalid-Hamid El Hachimi
- Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), INSERM U1127, CNRS UMR 7225, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; EPHE, PSL Research University, Paris, France
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Benoit Colsch
- Université Paris-Saclay, CEA, INRAE, Département Médicaments et Technologies pour la Santé, MetaboHUB, Gif sur Yvette, France
| | - Dominic Samaroo
- Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), INSERM U1127, CNRS UMR 7225, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Morgan Devaux
- Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), INSERM U1127, CNRS UMR 7225, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Alexandra Durr
- Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), INSERM U1127, CNRS UMR 7225, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France
| | - Giovanni Stevanin
- Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), INSERM U1127, CNRS UMR 7225, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France; EPHE, PSL Research University, Paris, France; University of Bordeaux, CNRS, INCIA, UMR 5287, NRGen Team, Bordeaux, France
| | | | - Sophie Colombo
- Dynacure SA (now Flamingo Therapeutics NV), Illkirch, France
| | - Belinda Cowling
- Dynacure SA (now Flamingo Therapeutics NV), Illkirch, France
| | - Frédéric Darios
- Sorbonne Université, Paris Brain Institute (ICM Institut du Cerveau), INSERM U1127, CNRS UMR 7225, Assistance Publique-Hôpitaux de Paris (AP-HP), Paris, France.
| |
Collapse
|
12
|
Słyk Ż, Stachowiak N, Małecki M. Recombinant Adeno-Associated Virus Vectors for Gene Therapy of the Central Nervous System: Delivery Routes and Clinical Aspects. Biomedicines 2024; 12:1523. [PMID: 39062095 PMCID: PMC11274884 DOI: 10.3390/biomedicines12071523] [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: 04/19/2024] [Revised: 06/23/2024] [Accepted: 07/03/2024] [Indexed: 07/28/2024] Open
Abstract
The Central Nervous System (CNS) is vulnerable to a range of diseases, including neurodegenerative and oncological conditions, which present significant treatment challenges. The blood-brain barrier (BBB) restricts molecule penetration, complicating the achievement of therapeutic concentrations in the CNS following systemic administration. Gene therapy using recombinant adeno-associated virus (rAAV) vectors emerges as a promising strategy for treating CNS diseases, demonstrated by the registration of six gene therapy products in the past six years and 87 ongoing clinical trials. This review explores the implementation of rAAV vectors in CNS disease treatment, emphasizing AAV biology and vector engineering. Various administration methods-such as intravenous, intrathecal, and intraparenchymal routes-and experimental approaches like intranasal and intramuscular administration are evaluated, discussing their advantages and limitations in different CNS contexts. Additionally, the review underscores the importance of optimizing therapeutic efficacy through the pharmacokinetics (PK) and pharmacodynamics (PD) of rAAV vectors. A comprehensive analysis of clinical trials reveals successes and challenges, including barriers to commercialization. This review provides insights into therapeutic strategies using rAAV vectors in neurological diseases and identifies areas requiring further research, particularly in optimizing rAAV PK/PD.
Collapse
Affiliation(s)
- Żaneta Słyk
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Natalia Stachowiak
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
13
|
Stamataki M, Rissiek B, Magnus T, Körbelin J. Microglia targeting by adeno-associated viral vectors. Front Immunol 2024; 15:1425892. [PMID: 39035004 PMCID: PMC11257843 DOI: 10.3389/fimmu.2024.1425892] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024] Open
Abstract
Microglia play a crucial role in maintaining homeostasis of the central nervous system and they are actively involved in shaping the brain's inflammatory response to stress. Among the multitude of involved molecules, purinergic receptors and enzymes are of special importance due to their ability to regulate microglia activation. By investigating the mechanisms underlying microglial responses and dysregulation, researchers can develop more precise interventions to modulate microglial behavior and alleviate neuroinflammatory processes. Studying gene function selectively in microglia, however, remains technically challenging. This review article provides an overview of adeno-associated virus (AAV)-based microglia targeting approaches, discussing potential prospects for refining these approaches to improve both specificity and effectiveness and encouraging future investigations aimed at connecting the potential of AAV-mediated microglial targeting for therapeutic benefit in neurological disorders.
Collapse
Affiliation(s)
- Maria Stamataki
- ENDomics Lab, Department of Oncology, Hematology & Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Björn Rissiek
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Tim Magnus
- Department of Neurology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jakob Körbelin
- ENDomics Lab, Department of Oncology, Hematology & Bone Marrow Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
14
|
Reich M, Simon MJ, Polke B, Paris I, Werner G, Schrader C, Spieth L, Davis SS, Robinson S, de Melo GL, Schlaphoff L, Buschmann K, Berghoff S, Logan T, Nuscher B, de Weerd L, Edbauer D, Simons M, Suh JH, Sandmann T, Kariolis MS, DeVos SL, Lewcock JW, Paquet D, Capell A, Di Paolo G, Haass C. Peripheral expression of brain-penetrant progranulin rescues pathologies in mouse models of frontotemporal lobar degeneration. Sci Transl Med 2024; 16:eadj7308. [PMID: 38838131 DOI: 10.1126/scitranslmed.adj7308] [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: 07/13/2023] [Accepted: 05/10/2024] [Indexed: 06/07/2024]
Abstract
Progranulin (PGRN) haploinsufficiency is a major risk factor for frontotemporal lobar degeneration with TAR DNA-binding protein 43 (TDP-43) pathology (FTLD-GRN). Multiple therapeutic strategies are in clinical development to restore PGRN in the CNS, including gene therapy. However, a limitation of current gene therapy approaches aimed to alleviate FTLD-associated pathologies may be their inefficient brain exposure and biodistribution. We therefore developed an adeno-associated virus (AAV) targeting the liver (L) to achieve sustained peripheral expression of a transferrin receptor (TfR) binding, brain-penetrant (b) PGRN variant [AAV(L):bPGRN] in two mouse models of FTLD-GRN, namely, Grn knockout and GrnxTmem106b double knockout mice. This therapeutic strategy avoids potential safety and biodistribution issues of CNS-administered AAVs and maintains sustained concentrations of PGRN in the brain after a single dose. AAV(L):bPGRN treatment reduced several FTLD-GRN-associated pathologies including severe motor function deficits, aberrant TDP-43 phosphorylation, dysfunctional protein degradation, lipid metabolism, gliosis, and neurodegeneration in the brain. The potential translatability of our findings was tested in an in vitro model using cocultured human induced pluripotent stem cell (hiPSC)-derived microglia lacking PGRN and TMEM106B and wild-type hiPSC-derived neurons. As in mice, aberrant TDP-43, lysosomal dysfunction, and neuronal loss were ameliorated after treatment with exogenous TfR-binding protein transport vehicle fused to PGRN (PTV:PGRN). Together, our studies suggest that peripherally administered brain-penetrant PGRN replacement strategies ameliorate FTLD-GRN relevant phenotypes including TDP-43 pathology, neurodegeneration, and behavioral deficits. Our data provide preclinical proof of concept for the use of this AAV platform for treatment of FTLD-GRN and potentially other CNS disorders.
Collapse
Affiliation(s)
- Marvin Reich
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), LMU Munich, 82152 Planegg, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Matthew J Simon
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Beate Polke
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Iñaki Paris
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Georg Werner
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Christian Schrader
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Lena Spieth
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Sonnet S Davis
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Sophie Robinson
- Graduate School of Systemic Neurosciences (GSN), LMU Munich, 82152 Planegg, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | | | - Lennart Schlaphoff
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), LMU Munich, 82152 Planegg, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany
| | - Katrin Buschmann
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Stefan Berghoff
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Todd Logan
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Brigitte Nuscher
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Lis de Weerd
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), 81377 Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), 81377 Munich, Germany
| | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Thomas Sandmann
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | | | - Sarah L DeVos
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | | | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), 81377 Munich, Germany
| | - Anja Capell
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | | | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), 81377 Munich, Germany
| |
Collapse
|
15
|
Wang JH, Gessler DJ, Zhan W, Gallagher TL, Gao G. Adeno-associated virus as a delivery vector for gene therapy of human diseases. Signal Transduct Target Ther 2024; 9:78. [PMID: 38565561 PMCID: PMC10987683 DOI: 10.1038/s41392-024-01780-w] [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: 07/05/2023] [Revised: 02/08/2024] [Accepted: 02/19/2024] [Indexed: 04/04/2024] Open
Abstract
Adeno-associated virus (AAV) has emerged as a pivotal delivery tool in clinical gene therapy owing to its minimal pathogenicity and ability to establish long-term gene expression in different tissues. Recombinant AAV (rAAV) has been engineered for enhanced specificity and developed as a tool for treating various diseases. However, as rAAV is being more widely used as a therapy, the increased demand has created challenges for the existing manufacturing methods. Seven rAAV-based gene therapy products have received regulatory approval, but there continue to be concerns about safely using high-dose viral therapies in humans, including immune responses and adverse effects such as genotoxicity, hepatotoxicity, thrombotic microangiopathy, and neurotoxicity. In this review, we explore AAV biology with an emphasis on current vector engineering strategies and manufacturing technologies. We discuss how rAAVs are being employed in ongoing clinical trials for ocular, neurological, metabolic, hematological, neuromuscular, and cardiovascular diseases as well as cancers. We outline immune responses triggered by rAAV, address associated side effects, and discuss strategies to mitigate these reactions. We hope that discussing recent advancements and current challenges in the field will be a helpful guide for researchers and clinicians navigating the ever-evolving landscape of rAAV-based gene therapy.
Collapse
Affiliation(s)
- Jiang-Hui Wang
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC, 3002, Australia
- Ophthalmology, Department of Surgery, University of Melbourne, East Melbourne, VIC, 3002, Australia
| | - Dominic J Gessler
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurological Surgery, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Neurosurgery, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Wei Zhan
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Thomas L Gallagher
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
- Li Weibo Institute for Rare Diseases Research, University of Massachusetts Chan Medical School, Worcester, MA, 01605, USA.
| |
Collapse
|
16
|
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] [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
|
17
|
Keshavan N, Minczuk M, Viscomi C, Rahman S. Gene therapy for mitochondrial disorders. J Inherit Metab Dis 2024; 47:145-175. [PMID: 38171948 DOI: 10.1002/jimd.12699] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 10/30/2023] [Accepted: 11/30/2023] [Indexed: 01/05/2024]
Abstract
In this review, we detail the current state of application of gene therapy to primary mitochondrial disorders (PMDs). Recombinant adeno-associated virus-based (rAAV) gene replacement approaches for nuclear gene disorders have been undertaken successfully in more than ten preclinical mouse models of PMDs which has been made possible by the development of novel rAAV technologies that achieve more efficient organ targeting. So far, however, the greatest progress has been made for Leber Hereditary Optic Neuropathy, for which phase 3 clinical trials of lenadogene nolparvovec demonstrated efficacy and good tolerability. Other methods of treating mitochondrial DNA (mtDNA) disorders have also had traction, including refinements to nucleases that degrade mtDNA molecules with pathogenic variants, including transcription activator-like effector nucleases, zinc-finger nucleases, and meganucleases (mitoARCUS). rAAV-based approaches have been used successfully to deliver these nucleases in vivo in mice. Exciting developments in CRISPR-Cas9 gene editing technology have achieved in vivo gene editing in mouse models of PMDs due to nuclear gene defects and new CRISPR-free gene editing approaches have shown great potential for therapeutic application in mtDNA disorders. We conclude the review by discussing the challenges of translating gene therapy in patients both from the point of view of achieving adequate organ transduction as well as clinical trial design.
Collapse
Affiliation(s)
- Nandaki Keshavan
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital, London, UK
| | - Michal Minczuk
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Carlo Viscomi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Veneto Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Shamima Rahman
- UCL Great Ormond Street Institute of Child Health, London, UK
- Great Ormond Street Hospital, London, UK
| |
Collapse
|
18
|
Słyk Ż, Wrzesień R, Barszcz S, Gawrychowski K, Małecki M. Adeno-associated virus vector hydrogel formulations for brain cancer gene therapy applications. Biomed Pharmacother 2024; 170:116061. [PMID: 38154269 DOI: 10.1016/j.biopha.2023.116061] [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: 09/10/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 12/30/2023] Open
Abstract
Gelatin-based formulations are utilized in neurosurgical procedures, with Medisponge® serving as an illustration of a secure and biocompatible hemostatic formulation. Noteworthy are combined hemostatic products that integrate pharmacological agents with gelatin. Gelatin matrices, which host biologically active substances, provide a platform for a variety of molecules. Biopolymers function as carriers for chemicals and genes, a facet particularly pertinent in brain cancer therapy, as gene therapy complement conventional approaches. The registration of Zolgensma underscores the efficacy of rAAV vectors in therapeutic gene delivery to the CNS. rAAVs, renowned for their safety, stability, and neuron-targeting capabilities, predominate in CNS gene therapy studies. The effectiveness of rAAV vector therapy varies based on the serotype and administration route. Local gene therapy employing hydrogel (e.g., post-tumor resection) enables the circumvention of the blood-brain barrier and restricts formulation diffusion. This study formulates gelatin rAAV gene formulations and evaluates vector transduction potential. Transduction efficiency was assessed using ex vivo mouse brains and in vitro cancer cell lines. In vitro, the transduction of rAAV vectors in gelatin matrices was quantified through qPCR, measuring the itr and Gfp expression. rAAVDJ and rAAV2 demonstrated superior transduction in ex vivo and in vitro models. Among the cell lines tested (Hs683, B16-F10, NIH:OVCAR-3), gelatin matrix F1 exhibited selective transduction, particularly with Hs683 human glioma cells, surpassing the performance Medisponge®. This research highlights the exploration of local brain cancer therapy, emphasizing the potential of gelatin as an rAAV vector carrier for gene therapy. The functional transduction activity of gelatin rAAV formulations is demonstrated.
Collapse
Affiliation(s)
- Żaneta Słyk
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland; Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland.
| | - Robert Wrzesień
- Central Laboratory of Experimental Animals, Center for Preclinical Research and Technology, Medical University of Warsaw, Warsaw, Poland
| | - Sławomir Barszcz
- Department of Neurosurgery, Children's Clinical Hospital, University Clinical Centre of the Medical University of Warsaw, Warsaw, Poland
| | - Krzysztof Gawrychowski
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| | - Maciej Małecki
- Department of Applied Pharmacy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland; Laboratory of Gene Therapy, Faculty of Pharmacy, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
19
|
He X, Wang X, Wang H, Wang T, Yang F, Chen Y, Pei Z, Bai Y, Li W, Wu Z, Chen G. NeuroD1 Regulated Endothelial Gene Expression to Modulate Transduction of AAV-PHP.eB and Recovery Progress after Ischemic Stroke. Aging Dis 2023; 15:2632-2649. [PMID: 38270116 PMCID: PMC11567258 DOI: 10.14336/ad.2023.1213] [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: 09/21/2023] [Accepted: 12/13/2023] [Indexed: 01/26/2024] Open
Abstract
AAV-PHP.eB depends on endothelial cells to highly transduce the central nervous system (CNS) and is widely used for intravenous gene therapy. However, the transduction profile and therapeutic efficiency after endothelial cell injury such as ischemic stroke is largely unknown. In this study, we tested the transduction profiles of AAV-PHP.eB and developed intravenous NeuroD1 gene therapy to treat ischemic stroke in mice. We found that AAV-PHP.eB-GFP control virus crossed the BBB and infected brain cells efficiently in normal brain. However, after stroke, AAV-PHP.eB-GFP control virus was highly restricted in the blood vessels. Surprisingly, after switching to therapeutic vector AAV-PHP.eB-NeuroD1-GFP, the viral vector successfully crossed blood vessels and infected brain cells. Using Tie2-cre transgenic mice, we demonstrated that NeuroD1 regulated endothelial gene expression to modulate AAV-PHP.eB transduction. Following the changes of signaling pathways in endothelial cells, NeuroD1 effectively protected BBB integrity, attenuated neuroinflammation, inhibited neuron apoptosis and rescued motor deficits after ischemic stroke. Moreover, NeuroD1 over-expression in brain cells further promoted neural regeneration. These results indicate that intravenous gene therapy using AAV-PHP.eB for ischemic stroke differs from intracranial gene therapy and NeuroD1 intravenous delivery using AAV-PHP.eB efficiently rescue both vascular damage and neuronal loss, providing an advancing therapeutic treatment for stroke.
Collapse
Affiliation(s)
- Xiaosong He
- Emergency Department, Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, China.
- Department of Neurology of the Sixth Affiliated Hospital of Guangzhou Medical University, Department of Human Anatomy in School of Basic Science of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, China.
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Xin Wang
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Hui Wang
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
- Department of Neurology, Affiliated ZhongDa Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Tao Wang
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Fuhan Yang
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Yuchen Chen
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Zifei Pei
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Yuting Bai
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
| | - Wen Li
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Zheng Wu
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| | - Gong Chen
- Department of Biology, Huck Institutes of Life Sciences, Pennsylvania State University, University Park, PA 16802, USA.
- GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China.
| |
Collapse
|
20
|
Li C, Lin Y, Chen Y, Song X, Zheng X, Li J, He J, Chen X, Huang C, Wang W, Wu J, Wu J, Gao J, Tu Z, Li XJ, Yan S, Li S. A Specific Mini-Intrabody Mediates Lysosome Degradation of Mutant Huntingtin. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2301120. [PMID: 37688357 PMCID: PMC10625127 DOI: 10.1002/advs.202301120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 08/01/2023] [Indexed: 09/10/2023]
Abstract
Accumulation of misfolded proteins leads to many neurodegenerative diseases that can be treated by lowering or removing mutant proteins. Huntington's disease (HD) is characterized by the intracellular accumulation of mutant huntingtin (mHTT) that can be soluble and aggregated in the central nervous system and causes neuronal damage and death. Here, an intracellular antibody (intrabody) fragment is generated that can specifically bind mHTT and link to the lysosome for degradation. It is found that delivery of this peptide by either brain injection or intravenous administration can efficiently clear the soluble and aggregated mHTT by activating the lysosomal degradation pathway, resulting in amelioration of gliosis and dyskinesia in HD knock-in (KI-140Q) mice. These findings suggest that the small intrabody peptide linked to lysosomes can effectively lower mutant proteins and provide a new approach for treating neurodegenerative diseases that are caused by the accumulation of mutant proteins.
Collapse
Affiliation(s)
- Caijuan Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yingqi Lin
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Yizhi Chen
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xichen Song
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao Zheng
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiawei Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jun He
- Institute of Laboratory Animal Science, Jinan University, Guangzhou, 510632, China
| | - Xiusheng Chen
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Chunhui Huang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Wei Wang
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jianhao Wu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiaxi Wu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Jiale Gao
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Zhuchi Tu
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| | - Sen Yan
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
- Department of Pathophysiology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Shihua Li
- Guangdong Key Laboratory of Non-human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), GHM Institute of CNS Regeneration, Jinan University, Guangzhou, 510632, China
| |
Collapse
|
21
|
Boespflug-Tanguy O, Sevin C, Piguet F. Gene therapy for neurodegenerative disorders in children: dreams and realities. Arch Pediatr 2023; 30:8S32-8S40. [PMID: 38043981 DOI: 10.1016/s0929-693x(23)00225-7] [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: 12/05/2023]
Abstract
Gene therapy encompasses the administration of biological medicinal products containing recombinant nucleic acids, mainly DNA, with the aim of treating or curing diseases. This represents a unique therapeutic strategy to reach the brain, in order to prevent or halt a neurodegenerative process. During the past decade, active multidisciplinary research has started to solve many issues for gene therapy in neurodegenerative disorders in terms of vectors, modes of administration, and expression of the therapeutic DNA. The engineering of hematopoietic stem cells (HSC) with lentivirus vectors for ex vivo gene therapy has demonstrated efficiency in reaching the brain through their transformation into microglial/macrophages cells with a long-term gene expression of the therapeutic vector as an alternative to autologous HSC transplants. Two drugs based on this strategy have been approved to date. The first is for metachromatic leukodystrophy (MLD), a severe lysosomal storage disease, and provides high levels of the deficient enzyme; the second one is for cerebral forms of X-linked adrenoleukodystrophy (X-ALD), and works by halting the neuroinflammation process. However, due to the long-lasting effect of the procedure, the therapy is applicable only to pre- or pauci/oligo-symptomatic patients. In vivo gene therapy via direct injection into the brain or the cerebrospinal fluid, but also by intravenous injection, represents a more efficient approach; however, many challenges remain to be solved despite the approval of two drugs: one for the early infantile form of spinal muscular atrophy (SMA), in which the gene product injected intravenously is able to prevent spinal motoneuron neurodegeneration. The second one, for aromatic L-amino acid decarboxylase (AADC) deficiency, provides the defective enzyme to the basal ganglia via intraparenchymal injection. The production of vectors able to reach the brain target cells with a sufficiently high expression remains a major bottleneck. In parallel, efforts must continue in order to better define (i) the natural history and clinical outcomes of many neurodegenerative disorders with childhood onset, and (ii) the mechanisms involved in the neurodegenerative process. © 2023 Published by Elsevier Masson SAS on behalf of French Society of Pediatrics.
Collapse
Affiliation(s)
- Odile Boespflug-Tanguy
- APHP, Service de Neuropediatrie, Hôpital Robert Debré, Paris, France; Université Paris Cité, INSERM UMR 1141, Hôpital Robert Debré, Paris France.
| | - Caroline Sevin
- APHP, Service de Neuropediatrie, Hôpital du Kremlin Bicêtre, Paris, France; GENOV, Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013, Paris, France
| | - Francoise Piguet
- GENOV, Institut du Cerveau, ICM, Inserm U 1127, CNRS UMR 7225, Sorbonne Université, F-75013, Paris, France
| |
Collapse
|
22
|
Gardin A, Ronzitti G. Current limitations of gene therapy for rare pediatric diseases: Lessons learned from clinical experience with AAV vectors. Arch Pediatr 2023; 30:8S46-8S52. [PMID: 38043983 DOI: 10.1016/s0929-693x(23)00227-0] [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] [Indexed: 12/05/2023]
Abstract
Gene therapy using adeno-associated viral (AAV) vectors is a promising therapeutic strategy for multiple inherited diseases. Following intravenous injection, AAV vectors carrying a copy of the missing gene or the genome-editing machinery reach their target cells and deliver the genetic material. Several clinical trials are currently ongoing and significant success has already been achieved with at least six AAV gene therapy products with market approval in Europe and the United States. Nonetheless, clinical trials and preclinical studies have uncovered several limitations of AAV gene transfer, which need to be addressed in order to improve the safety and enable the treatment of the largest patient population. Limitations include the occurrence of immune-mediated toxicities, the potential loss of correction in the long run, and the development of neutralizing antibodies against AAV vectors preventing re-administration. In this review, we summarize these limitations and discuss the potential technological developments to overcome them. © 2023 Published by Elsevier Masson SAS on behalf of French Society of Pediatrics.
Collapse
Affiliation(s)
- Antoine Gardin
- Genethon, 91000 Evry, France; Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951, 91000 Evry, France; Hépatologie et Transplantation Hépatique Pédiatriques, Centre de référence de l'atrésie des voies biliaires et des cholestases génétiques, FSMR FILFOIE, Health Care Provider of the European Reference Network on Rare Liver Disorders (ERN RARE LIVER), Hôpital Bicêtre, AP-HP, Université Paris-Saclay, Le Kremlin-Bicêtre, France
| | - Giuseppe Ronzitti
- Genethon, 91000 Evry, France; Université Paris-Saclay, Univ Evry, Inserm, Genethon, Integrare research unit UMR_S951, 91000 Evry, France.
| |
Collapse
|
23
|
Chuapoco MR, Flytzanis NC, Goeden N, Christopher Octeau J, Roxas KM, Chan KY, Scherrer J, Winchester J, Blackburn RJ, Campos LJ, Man KNM, Sun J, Chen X, Lefevre A, Singh VP, Arokiaraj CM, Shay TF, Vendemiatti J, Jang MJ, Mich JK, Bishaw Y, Gore BB, Omstead V, Taskin N, Weed N, Levi BP, Ting JT, Miller CT, Deverman BE, Pickel J, Tian L, Fox AS, Gradinaru V. Adeno-associated viral vectors for functional intravenous gene transfer throughout the non-human primate brain. NATURE NANOTECHNOLOGY 2023; 18:1241-1251. [PMID: 37430038 PMCID: PMC10575780 DOI: 10.1038/s41565-023-01419-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 05/15/2023] [Indexed: 07/12/2023]
Abstract
Crossing the blood-brain barrier in primates is a major obstacle for gene delivery to the brain. Adeno-associated viruses (AAVs) promise robust, non-invasive gene delivery from the bloodstream to the brain. However, unlike in rodents, few neurotropic AAVs efficiently cross the blood-brain barrier in non-human primates. Here we report on AAV.CAP-Mac, an engineered variant identified by screening in adult marmosets and newborn macaques, which has improved delivery efficiency in the brains of multiple non-human primate species: marmoset, rhesus macaque and green monkey. CAP-Mac is neuron biased in infant Old World primates, exhibits broad tropism in adult rhesus macaques and is vasculature biased in adult marmosets. We demonstrate applications of a single, intravenous dose of CAP-Mac to deliver functional GCaMP for ex vivo calcium imaging across multiple brain areas, or a cocktail of fluorescent reporters for Brainbow-like labelling throughout the macaque brain, circumventing the need for germline manipulations in Old World primates. As such, CAP-Mac is shown to have potential for non-invasive systemic gene transfer in the brains of non-human primates.
Collapse
Affiliation(s)
- Miguel R Chuapoco
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Nicholas C Flytzanis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Capsida Biotherapeutics, Thousand Oaks, CA, USA.
| | - Nick Goeden
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Capsida Biotherapeutics, Thousand Oaks, CA, USA
| | | | | | - Ken Y Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Stanley Center for Psychiatric Research at Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | | | | | - Lillian J Campos
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Psychology and the California National Primate Research Center, University of California Davis, Davis, CA, USA
| | - Kwun Nok Mimi Man
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - Junqing Sun
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Arthur Lefevre
- Cortical Systems and Behavior Laboratory, University of California San Diego, San Diego, CA, USA
| | - Vikram Pal Singh
- Cortical Systems and Behavior Laboratory, University of California San Diego, San Diego, CA, USA
| | - Cynthia M Arokiaraj
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - Timothy F Shay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Julia Vendemiatti
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Min J Jang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
| | - John K Mich
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Bryan B Gore
- Allen Institute for Brain Science, Seattle, WA, USA
| | | | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Boaz P Levi
- Allen Institute for Brain Science, Seattle, WA, USA
| | - Jonathan T Ting
- Allen Institute for Brain Science, Seattle, WA, USA
- Washington National Primate Research Center, University of Washington, Seattle, WA, USA
| | - Cory T Miller
- Cortical Systems and Behavior Laboratory, University of California San Diego, San Diego, CA, USA
| | - Benjamin E Deverman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Stanley Center for Psychiatric Research at Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - James Pickel
- National Institute of Mental Health, National Institutes of Health, Bethesda, MD, USA
| | - Lin Tian
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Biochemistry and Molecular Medicine, University of California Davis, Davis, CA, USA
| | - Andrew S Fox
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA
- Department of Psychology and the California National Primate Research Center, University of California Davis, Davis, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA.
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
24
|
Asher D, Dai D, Klimchak AC, Sedita LE, Gooch KL, Rodino-Klapac L. Paving the way for future gene therapies: A case study of scientific spillover from delandistrogene moxeparvovec. Mol Ther Methods Clin Dev 2023; 30:474-483. [PMID: 37674905 PMCID: PMC10477757 DOI: 10.1016/j.omtm.2023.08.002] [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: 09/08/2023]
Abstract
Gene therapies have potential to improve outcomes of severe diseases after only a single administration. Novel therapies are continually being developed using knowledge gained from prior successes, a concept known as scientific spillover. Gene therapy advancement requires extensive development at each stage: preclinical work to create and evaluate vehicles for delivery of the therapy, design of clinical development programs, and establishment of a large-scale manufacturing process. Pioneering gene therapies are generating spillover as investigators confront myriad issues specific to this treatment modality. These include frameworks for construct engineering, dose evaluation, patient selection, outcome assessment, and safety monitoring. Consequently, the benefits of these therapies extend beyond offering knowledge for treating any one disease to establishing new platforms and paradigms that will accelerate advancement of future gene therapies. This impact is even more profound in rare diseases, where developing therapies in isolation may not be possible. This review describes some instances of scientific spillover in healthcare, and specifically gene therapy, using delandistrogene moxeparvovec (SRP-9001), a gene therapy recently approved by the US Food and Drug Administration for the treatment of ambulatory pediatric patients aged 4-5 years with Duchenne muscular dystrophy with a confirmed mutation in the DMD gene, as a case study.
Collapse
Affiliation(s)
- Damon Asher
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Daisy Dai
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Alexa C. Klimchak
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | - Lauren E. Sedita
- Sarepta Therapeutics, Inc., 215 First Street, Cambridge, MA 02142, USA
| | | | | |
Collapse
|
25
|
Mirjalili Mohanna SZ, Korecki AJ, Simpson EM. rAAV-PHP.B escapes the mouse eye and causes lethality whereas rAAV9 can transduce aniridic corneal limbal stem cells without lethality. Gene Ther 2023; 30:670-684. [PMID: 37072572 PMCID: PMC10506911 DOI: 10.1038/s41434-023-00400-6] [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: 01/06/2023] [Revised: 03/28/2023] [Accepted: 04/05/2023] [Indexed: 04/20/2023]
Abstract
Recently safety concerns have been raised in connection with high doses of recombinant adeno-associated viruses (rAAV). Therefore, we undertook a series of experiments to test viral capsid (rAAV9 and rAAV-PHP.B), dose, and route of administration (intrastromal, intravitreal, and intravenous) focused on aniridia, a congenital blindness that currently has no cure. The success of gene therapy for aniridia may depend on the presence of functional limbal stem cells (LSCs) in the damaged aniridic corneas and whether rAAV can transduce them. Both these concerns were unknown, and thus were also addressed by our studies. For the first time, we report ataxia and lethality after intravitreal or intrastromal rAAV-PHP.B virus injections. We demonstrated virus escape from the eye and transduction of non-ocular tissues by rAAV9 and rAAV-PHP.B capsids. We have also shown that intrastromal and intravitreal delivery of rAAV9 can transduce functional LSCs, as well as all four PAX6-expressing retinal cell types in aniridic eye, respectively. Overall, lack of adverse events and successful transduction of LSCs and retinal cells makes it clear that rAAV9 is the capsid of choice for future aniridia gene therapy. Our finding of rAAV lethality after intraocular injections will be impactful for other researchers developing rAAV-based gene therapies.
Collapse
Affiliation(s)
- Seyedeh Zeinab Mirjalili Mohanna
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC, Canada
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada
| | - Andrea J Korecki
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC, Canada
| | - Elizabeth M Simpson
- Centre for Molecular Medicine and Therapeutics at British Columbia Children's Hospital, The University of British Columbia, Vancouver, BC, Canada.
- Department of Medical Genetics, The University of British Columbia, Vancouver, BC, Canada.
| |
Collapse
|
26
|
Casy W, Garza IT, Chen X, Dong T, Hu Y, Kanchwala M, Trygg CB, Shyng C, Xing C, Bunnell BA, Braun SE, Gray SJ. SMRT Sequencing Enables High-Throughput Identification of Novel AAVs from Capsid Shuffling and Directed Evolution. Genes (Basel) 2023; 14:1660. [PMID: 37628711 PMCID: PMC10454592 DOI: 10.3390/genes14081660] [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: 07/21/2023] [Revised: 08/15/2023] [Accepted: 08/17/2023] [Indexed: 08/27/2023] Open
Abstract
The use of AAV capsid libraries coupled with various selection strategies has proven to be a remarkable approach for generating novel AAVs with enhanced and desired features. The inability to reliably sequence the complete capsid gene in a high-throughput manner has been the bottleneck of capsid engineering. As a result, many library strategies are confined to localized and modest alterations in the capsid, such as peptide insertions or single variable region (VR) alterations. The caveat of short reads by means of next-generation sequencing (NGS) hinders the diversity of capsid library construction, shifting the field away from whole-capsid modifications. We generated AAV capsid shuffled libraries of naturally occurring AAVs and applied directed evolution in both mice and non-human primates (NHPs), with the goal of yielding AAVs that are compatible across both species for translational applications. We recovered DNA from the tissues of injected animal and used single molecule real-time (SMRT) sequencing to identify variants enriched in the central nervous system (CNS). We provide insights and considerations for variant identification by comparing bulk tissue sequencing to that of isolated nuclei. Our work highlights the potential advantages of whole-capsid engineering, as well as indispensable methodological improvements for the analysis of recovered capsids, including the nuclei-enrichment step and SMRT sequencing.
Collapse
Affiliation(s)
- Widler Casy
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (I.T.G.); (X.C.); (Y.H.)
| | - Irvin T. Garza
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (I.T.G.); (X.C.); (Y.H.)
- Graduate School of Basic Biomedical Sciences, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Xin Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (I.T.G.); (X.C.); (Y.H.)
| | - Thomas Dong
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (I.T.G.); (X.C.); (Y.H.)
| | - Yuhui Hu
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (I.T.G.); (X.C.); (Y.H.)
| | - Mohammed Kanchwala
- Eugene McDermott Center for Human Growth & Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.K.)
| | - Cynthia B. Trygg
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA (B.A.B.); (S.E.B.)
| | - Charles Shyng
- Gene Therapy Center, University of North Carolina, Chapel Hill, NC 27599, USA;
| | - Chao Xing
- Eugene McDermott Center for Human Growth & Development, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; (M.K.)
- Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bruce A. Bunnell
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA (B.A.B.); (S.E.B.)
| | - Stephen E. Braun
- Tulane National Primate Research Center, Tulane University School of Medicine, Covington, LA 70433, USA (B.A.B.); (S.E.B.)
| | - Steven J. Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA (I.T.G.); (X.C.); (Y.H.)
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| |
Collapse
|
27
|
Ajeeb R, Clegg JR. Intrathecal delivery of Macromolecules: Clinical status and emerging technologies. Adv Drug Deliv Rev 2023; 199:114949. [PMID: 37286086 DOI: 10.1016/j.addr.2023.114949] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 05/31/2023] [Accepted: 06/03/2023] [Indexed: 06/09/2023]
Abstract
The proximity and association of cerebrospinal fluid (CSF) and the intrathecal (IT) space with deep targets in the central nervous system (CNS) parenchyma makes IT injection an attractive route of administration for brain drug delivery. However, the extent to which intrathecally administered macromolecules are effective in treating neurological diseases is a question of both clinical debate and technological interest. We present the biological, chemical, and physical properties of the intrathecal space that are relevant to drug absorption, distribution, metabolism, and elimination from CSF. We then analyze the evolution of IT drug delivery in clinical trials over the last 20 years. Our analysis revealed that the percentage of clinical trials assessing IT delivery for the delivery of biologics (i.e., macromolecules, cells) for treatment of chronic conditions (e.g., neurodegeneration, cancer, and metabolic diseases) has steadily increased. Clinical trials exploring cell or macromolecular delivery within the IT space have not evaluated engineering technologies, such as depots, particles, or other delivery systems. Recent pre-clinical studies have evaluated IT macromolecule delivery in small animals, postulating that delivery efficacy can be assisted by external medical devices, micro- or nanoparticles, bulk biomaterials, and viral vectors. Further studies are necessary to evaluate the extent to which engineering technologies and IT administration improve CNS targeting and therapeutic outcome.
Collapse
Affiliation(s)
- Rana Ajeeb
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK, United States
| | - John R Clegg
- Stephenson School of Biomedical Engineering, The University of Oklahoma, Norman, OK, United States; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Harold Hamm Diabetes Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States; Institute for Biomedical Engineering, Science, and Technology, University of Oklahoma, Norman, OK, United States.
| |
Collapse
|
28
|
Blesa J, Pineda-Pardo JA, Inoue KI, Gasca-Salas C, Balzano T, Del Rey NLG, Reinares-Sebastián A, Esteban-García N, Rodríguez-Rojas R, Márquez R, Ciorraga M, del Álamo M, García-Cañamaque L, Ruiz de Aguiar S, Rachmilevitch I, Trigo-Damas I, Takada M, Obeso JA. BBB opening with focused ultrasound in nonhuman primates and Parkinson's disease patients: Targeted AAV vector delivery and PET imaging. SCIENCE ADVANCES 2023; 9:eadf4888. [PMID: 37075119 PMCID: PMC10115413 DOI: 10.1126/sciadv.adf4888] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
Intracerebral vector delivery in nonhuman primates has been a major challenge. We report successful blood-brain barrier opening and focal delivery of adeno-associated virus serotype 9 vectors into brain regions involved in Parkinson's disease using low-intensity focus ultrasound in adult macaque monkeys. Openings were well tolerated with generally no associated abnormal magnetic resonance imaging signals. Neuronal green fluorescent protein expression was observed specifically in regions with confirmed blood-brain barrier opening. Similar blood-brain barrier openings were safely demonstrated in three patients with Parkinson's disease. In these patients and in one monkey, blood-brain barrier opening was followed by 18F-Choline uptake in the putamen and midbrain regions based on positron emission tomography. This indicates focal and cellular binding of molecules that otherwise would not enter the brain parenchyma. The less-invasive nature of this methodology could facilitate focal viral vector delivery for gene therapy and might allow early and repeated interventions to treat neurodegenerative disorders.
Collapse
Affiliation(s)
- Javier Blesa
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - José A. Pineda-Pardo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Ken-ichi Inoue
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University; Inuyama, Aichi 484-8506, Japan
| | - Carmen Gasca-Salas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- University CEU-San Pablo, Madrid, Spain
| | - Tiziano Balzano
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Natalia López-González Del Rey
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Alejandro Reinares-Sebastián
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Noelia Esteban-García
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- PhD Program in Neuroscience Autónoma de Madrid University-Cajal Institute, Madrid, Spain
| | - Rafael Rodríguez-Rojas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Raquel Márquez
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - María Ciorraga
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Marta del Álamo
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | - Lina García-Cañamaque
- University CEU-San Pablo, Madrid, Spain
- Department of Nuclear Medicine, Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
| | | | | | - Inés Trigo-Damas
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
| | - Masahiko Takada
- Systems Neuroscience Section, Department of Neuroscience, Primate Research Institute, and Center for the Evolutionary Origins of Human Behavior, Kyoto University; Inuyama, Aichi 484-8506, Japan
- Department of Neurology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
| | - José A. Obeso
- HM CINAC (Centro Integral de Neurociencias Abarca Campal), Hospital Universitario HM Puerta del Sur, HM Hospitales, Madrid, Spain
- Network Center for Biomedical Research on Neurodegenerative Diseases (CIBERNED), Instituto Carlos III, Madrid, Spain
- University CEU-San Pablo, Madrid, Spain
- Corresponding author.
| |
Collapse
|
29
|
Shay TF, Sullivan EE, Ding X, Chen X, Ravindra Kumar S, Goertsen D, Brown D, Crosby A, Vielmetter J, Borsos M, Wolfe DA, Lam AW, Gradinaru V. Primate-conserved carbonic anhydrase IV and murine-restricted LY6C1 enable blood-brain barrier crossing by engineered viral vectors. SCIENCE ADVANCES 2023; 9:eadg6618. [PMID: 37075114 PMCID: PMC10115422 DOI: 10.1126/sciadv.adg6618] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
The blood-brain barrier (BBB) presents a major challenge for delivering large molecules to study and treat the central nervous system. This is due in part to the scarcity of targets known to mediate BBB crossing. To identify novel targets, we leverage a panel of adeno-associated viruses (AAVs) previously identified through mechanism-agnostic directed evolution for improved BBB transcytosis. Screening potential cognate receptors for enhanced BBB crossing, we identify two targets: murine-restricted LY6C1 and widely conserved carbonic anhydrase IV (CA-IV). We apply AlphaFold-based in silico methods to generate capsid-receptor binding models to predict the affinity of AAVs for these identified receptors. Demonstrating how these tools can unlock target-focused engineering strategies, we create an enhanced LY6C1-binding vector, AAV-PHP.eC, that, unlike our prior PHP.eB, also works in Ly6a-deficient mouse strains such as BALB/cJ. Combined with structural insights from computational modeling, the identification of primate-conserved CA-IV enables the design of more specific and potent human brain-penetrant chemicals and biologicals, including gene delivery vectors.
Collapse
Affiliation(s)
- Timothy F. Shay
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Corresponding author. (T.F.S.); (V.G.)
| | - Erin E. Sullivan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Xiaozhe Ding
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - David Goertsen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - David Brown
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Anaya Crosby
- California State Polytechnic University, Pomona, Pomona, CA, USA
| | - Jost Vielmetter
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Máté Borsos
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Damien A. Wolfe
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Annie W. Lam
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, USA
- Corresponding author. (T.F.S.); (V.G.)
| |
Collapse
|
30
|
Simon MJ, Logan T, DeVos SL, Di Paolo G. Lysosomal functions of progranulin and implications for treatment of frontotemporal dementia. Trends Cell Biol 2023; 33:324-339. [PMID: 36244875 DOI: 10.1016/j.tcb.2022.09.006] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 12/12/2022]
Abstract
Loss-of-function heterozygous mutations in GRN, the gene encoding progranulin (PGRN), were identified in patients with frontotemporal lobar degeneration (FTLD) almost two decades ago and are generally linked to reduced PGRN protein expression levels. Although initial characterization of PGRN function primarily focused on its role in extracellular signaling as a secreted protein, more recent studies revealed critical roles of PGRN in regulating lysosome function, including proteolysis and lipid degradation, consistent with its lysosomal localization. Emerging from these studies is the notion that PGRN regulates glucocerebrosidase activity via direct chaperone activities and via interaction with prosaposin (i.e., a key regulator of lysosomal sphingolipid-metabolizing enzymes), as well as with the anionic phospholipid bis(monoacylglycero)phosphate. This emerging lysosomal biology of PGRN identified novel and promising opportunities in therapeutic discovery as well as biomarker development.
Collapse
Affiliation(s)
| | - Todd Logan
- Denali Therapeutics, South San Francisco, CA, USA
| | | | | |
Collapse
|
31
|
Daw TB, El-Nahal HG, Basso MA, Jun EJ, Bautista AR, Samulski RJ, Sommer MA, Bohlen MO. Direct Comparison of Epifluorescence and Immunostaining for Assessing Viral Mediated Gene Expression in the Primate Brain. Hum Gene Ther 2023; 34:228-246. [PMID: 36719771 PMCID: PMC10031143 DOI: 10.1089/hum.2022.194] [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: 10/08/2022] [Accepted: 01/03/2023] [Indexed: 02/01/2023] Open
Abstract
Viral vector technologies are commonly used in neuroscience research to understand and manipulate neural circuits, but successful applications of these technologies in non-human primate models have been inconsistent. An essential component to improve these technologies is an impartial and accurate assessment of the effectiveness of different viral constructs in the primate brain. We tested a diverse array of viral vectors delivered to the brain and extraocular muscles of macaques and compared three methods for histological assessment of viral-mediated fluorescent transgene expression: epifluorescence (Epi), immunofluorescence (IF), and immunohistochemistry (IHC). Importantly, IF and IHC identified a greater number of transduced neurons compared to Epi. Furthermore, IF and IHC reliably provided enhanced visualization of transgene in most cellular compartments (i.e., dendritic, axonal, and terminal fields), whereas the degree of labeling provided by Epi was inconsistent and predominantly restricted to somas and apical dendrites. Because Epi signals are unamplified (in contrast to IF and IHC), Epi may provide a more veridical assessment for the amount of accumulated transgene and, thus, the potential to chemogenetically or optogenetically manipulate neuronal activity. The comparatively weak Epi signals suggest that the current generations of viral constructs, regardless of delivered transgene, are not optimized for primates. This reinforces an emerging viewpoint that viral vectors tailored for the primate brain are necessary for basic research and human gene therapy.
Collapse
Affiliation(s)
- Tierney B. Daw
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
| | - Hala G. El-Nahal
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| | - Michele A. Basso
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
- Department of Biological Structure, Washington National Primate Research Center, University of Washington, Seattle, Seattle, Washington, USA
- Department of Physiology and Biophysics, Washington National Primate Research Center, University of Washington, Seattle, Seattle, Washington, USA
| | - Elizabeth J. Jun
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
| | - Alex R. Bautista
- Fuster Laboratory of Cognitive Neuroscience, Department of Psychiatry and Biobehavioral Sciences, Jane and Terry Semel Institute for Neuroscience and Human Behavior, University of California, Los Angeles, Los Angeles, California, USA
| | - R. Jude Samulski
- Gene Therapy Center and Department of Pharmacology, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, USA
- R&D Department, Asklepios BioPharmaceutical, Inc. (AskBio), Research Triangle, North Carolina, USA
| | - Marc A. Sommer
- Department of Neurobiology, Duke University School of Medicine, Durham, North Carolina, USA
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
- Department of Psychology and Neuroscience, Duke University, Durham, North Carolina, USA
- Center for Cognitive Neuroscience, Duke Institute for Brain Sciences, Duke University, Durham, North Carolina, USA
| | - Martin O. Bohlen
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, USA
| |
Collapse
|
32
|
Chen X, Lim DA, Lawlor MW, Dimmock D, Vite CH, Lester T, Tavakkoli F, Sadhu C, Prasad S, Gray SJ. Biodistribution of Adeno-Associated Virus Gene Therapy Following Cerebrospinal Fluid-Directed Administration. Hum Gene Ther 2023; 34:94-111. [PMID: 36606687 DOI: 10.1089/hum.2022.163] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Adeno-associated virus (AAV)-based gene therapies, exemplified by the approved therapy for spinal muscular atrophy, have the potential to deliver disease-course-altering treatments for central nervous system (CNS) indications. However, several clinical trials have reported severe adverse events, including patient deaths following high-dose systemic administration for muscle-directed gene transfer, highlighting the need to explore approaches utilizing lower doses when targeting the CNS. Animal models of disease provide insight into the response to new AAV therapies. However, translation from small to larger animals and eventually to humans is hampered by anatomical and biological differences across the species and their impact on AAV delivery. We performed a literature review of preclinical studies of AAV gene therapy biodistribution following cerebrospinal fluid (CSF) delivery (intracerebroventricular, intra-cisterna magna, and intrathecal lumbar). The reviewed literature varies greatly in the reported biodistribution of AAV following administration into the CSF. Differences between studies, including animal model, vector serotype used, method used to assess biodistribution, and route of administration, among other variables, contribute to differing outcomes and difficulties in translating these preclinical results. For example, only half of the published AAV-based gene therapy studies report vector copy number, the most direct readout following administration of a vector; none of these studies reported details such as the empty:full capsid ratio and quality of encapsidated genome. Analysis of the last decade's literature focusing on AAV-based gene therapies targeting the CNS underscores limitations of the body of knowledge and room for continued research. In particular, there is a need to understand the biodistribution achieved by different CSF-directed routes of administration and determining if specific cell types/structures of interest will be transduced. Our findings point to a clear need for a more systematic approach across the field to align the assessments and elements reported in preclinical research to enable more reliable translation across animal models and into human studies.
Collapse
Affiliation(s)
- Xin Chen
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Daniel A Lim
- Department of Neurological Surgery, Eli and Edythe Broad Center for Regeneration Medicine, and the Weill Institute for Neurosciences, University of California San Francisco School of Medicine, San Francisco, California, USA
| | - Michael W Lawlor
- Medical College of Wisconsin and Diverge Translational Science Laboratory, Milwaukee, Wisconsin, USA
| | - David Dimmock
- Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Charles H Vite
- School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA; and
| | | | | | | | | | - Steven J Gray
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
33
|
Chuapoco MR, Flytzanis NC, Goeden N, Octeau JC, Roxas KM, Chan KY, Scherrer J, Winchester J, Blackburn RJ, Campos LJ, Man KNM, Sun J, Chen X, Lefevre A, Singh VP, Arokiaraj CM, Shaya TF, Vendemiatti J, Jang MJ, Mich J, Bishaw Y, Gore B, Omstead V, Taskin N, Weed N, Ting J, Miller CT, Deverman BE, Pickel J, Tian L, Fox AS, Gradinaru V. Intravenous functional gene transfer throughout the brain of non-human primates using AAV. RESEARCH SQUARE 2023:rs.3.rs-1370972. [PMID: 36789432 PMCID: PMC9928057 DOI: 10.21203/rs.3.rs-1370972/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Adeno-associated viruses (AAVs) promise robust gene delivery to the brain through non-invasive, intravenous delivery. However, unlike in rodents, few neurotropic AAVs efficiently cross the blood-brain barrier in non-human primates (NHPs). Here we describe AAV.CAP-Mac, an engineered variant identified by screening in adult marmosets and newborn macaques with improved efficiency in the brain of multiple NHP species: marmoset, rhesus macaque, and green monkey. CAP-Mac is neuron-biased in infant Old World primates, exhibits broad tropism in adult rhesus macaques, and is vasculature-biased in adult marmosets. We demonstrate applications of a single, intravenous dose of CAP-Mac to deliver (1) functional GCaMP for ex vivo calcium imaging across multiple brain areas, and (2) a cocktail of fluorescent reporters for Brainbow-like labeling throughout the macaque brain, circumventing the need for germline manipulations in Old World primates. Given its capabilities for systemic gene transfer in NHPs, CAP-Mac promises to help unlock non-invasive access to the brain.
Collapse
Affiliation(s)
- Miguel R. Chuapoco
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Nicholas C. Flytzanis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Capsida Biotherapeutics, Thousand Oaks, CA 91320, USA
- Present address: Capsida Biotherapeutics, Thousand Oaks, CA 91320, USA
| | - Nick Goeden
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Capsida Biotherapeutics, Thousand Oaks, CA 91320, USA
- Present address: Capsida Biotherapeutics, Thousand Oaks, CA 91320, USA
| | | | | | - Ken Y. Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Present address: Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jon Scherrer
- Capsida Biotherapeutics, Thousand Oaks, CA 91320, USA
| | | | | | - Lillian J. Campos
- Department of Psychology and the California National Primate Research Center, University of California-Davis, Davis, CA 95616, USA
| | - Kwun Nok Mimi Man
- Department of Psychology and the California National Primate Research Center, University of California-Davis, Davis, CA 95616, USA
| | - Junqing Sun
- Department of Psychology and the California National Primate Research Center, University of California-Davis, Davis, CA 95616, USA
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Arthur Lefevre
- Cortical Systems and Behavior Laboratory, University of California-San Diego, La Jolla, CA 92039, USA
| | - Vikram Pal Singh
- Cortical Systems and Behavior Laboratory, University of California-San Diego, La Jolla, CA 92039, USA
| | - Cynthia M. Arokiaraj
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Timothy F. Shaya
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Julia Vendemiatti
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - Min J. Jang
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
| | - John Mich
- Allen Institute for Brain Science, Seattle, WA, 98109, USA
| | - Yeme Bishaw
- Allen Institute for Brain Science, Seattle, WA, 98109, USA
| | - Bryan Gore
- Allen Institute for Brain Science, Seattle, WA, 98109, USA
| | | | - Naz Taskin
- Allen Institute for Brain Science, Seattle, WA, 98109, USA
| | - Natalie Weed
- Allen Institute for Brain Science, Seattle, WA, 98109, USA
| | - Jonathan Ting
- Allen Institute for Brain Science, Seattle, WA, 98109, USA
| | - Cory T. Miller
- Cortical Systems and Behavior Laboratory, University of California-San Diego, La Jolla, CA 92039, USA
| | - Benjamin E. Deverman
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Present address: Stanley Center for Psychiatric Research, Broad Institute of MIT and Harvard, Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - James Pickel
- Cortical Systems and Behavior Laboratory, University of California-San Diego, La Jolla, CA 92039, USA
| | - Lin Tian
- Department of Psychology and the California National Primate Research Center, University of California-Davis, Davis, CA 95616, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Andrew S. Fox
- Department of Psychology and the California National Primate Research Center, University of California-Davis, Davis, CA 95616, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA 91125, USA
- Aligning Science Across Parkinson’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815
| |
Collapse
|
34
|
Park J, Won J, Jeon CY, Lim KS, Choi WS, Park SH, Seo J, Cho J, Seong JB, Yeo HG, Kim K, Kim YG, Kim M, Yi KS, Lee Y. XperCT-guided Intra-cisterna Magna Injection of Streptozotocin for Establishing an Alzheimer's Disease Model Using the Cynomolgus Monkey ( Macaca fascicularis). Exp Neurobiol 2022; 31:409-418. [PMID: 36631849 PMCID: PMC9841743 DOI: 10.5607/en22027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/14/2022] [Accepted: 11/14/2022] [Indexed: 01/13/2023] Open
Abstract
Till date, researchers have been developing animal models of Alzheimer's disease (AD) in various species to understand the pathological characterization and molecular mechanistic pathways associated with this condition in humans to identify potential therapeutic treatments. A widely recognized AD model that mimics the pathology of human AD involves the intracerebroventricular (ICV) injection with streptozotocin (STZ). However, ICV injection as an invasive approach has several limitations related to complicated surgical procedures. Therefore, in the present study, we created a customized stereotaxic frame using the XperCT-guided system for injecting STZ in cynomolgus monkeys, aiming to establish an AD model. The anatomical structures surrounding the cisterna magna (CM) were confirmed using CT/MRI fusion images of monkey brain with XperCT, the c-arm cone beam computed tomography. XperCT was used to determine the appropriate direction in which the needle tip should be inserted within the CM region. Cerebrospinal fluid (CSF) was collected to confirm the accurate target site when STZ was injected into the CM. Cynomolgus monkeys were administered STZ dissolved in artificial CSF once every week for 4 weeks via intracisterna magna (ICM) injection using XperCT-guided stereotactic system. The molecular mechanisms underlying the progression of STZ-induced AD pathology were analyzed two weeks after the final injection. The monkeys subjected to XperCT-based STZ injection via the ICM route showed features of AD pathology, including markedly enhanced neuronal loss, synaptic impairment, and tau phosphorylation in the hippocampus. These findings suggest a new approach for the construction of neurodegenerative disease models and development of therapeutic strategies.
Collapse
Affiliation(s)
- Junghyung Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jinyoung Won
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Chang-Yeop Jeon
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Kyung Seob Lim
- Futuristic Animal Resource and Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Won Seok Choi
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Sung-hyun Park
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jincheol Seo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jiyeon Cho
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Jung Bae Seong
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea
| | - Hyeon-Gu Yeo
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Keonwoo Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,School of Life Sciences, BK21 Plus KNU Creative BioResearch Group, Kyungpook National University, Daegu 41566, Korea
| | - Yu Gyeong Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea
| | - Minji Kim
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
| | - Kyung Sik Yi
- Department of Radiology, Chungbuk National University Hospital, Cheongju 28644, Korea
| | - Youngjeon Lee
- National Primate Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Cheongju 28116, Korea,KRIBB School of Bioscience, University of Science and Technology (UST), Daejeon 34113, Korea,To whom correspondence should be addressed. TEL: 82-43-240-6316, FAX: 82-43-240-6309, e-mail:
| |
Collapse
|
35
|
Guo L, Li Z, Li Y, Qu B, Jiao G, Liang C, Lu Z, Wang XG, Huang C, Du H, Liang J, Zhou Q, Li W. Treatment of glutaric aciduria type I (GA-I) via intracerebroventricular delivery of GCDH. FUNDAMENTAL RESEARCH 2022; 2:836-842. [PMID: 38933374 PMCID: PMC11197790 DOI: 10.1016/j.fmre.2022.08.013] [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: 05/31/2022] [Revised: 07/24/2022] [Accepted: 08/23/2022] [Indexed: 10/14/2022] Open
Abstract
Glutaric aciduria type I (GA-I) is an autosomal recessive genetic disorder caused by a deficiency in glutaryl-CoA dehydrogenase (GCDH). Patients who do not receive proper treatment may die from acute encephalopathic crisis. Current treatments for GA-I include a low-lysine diet combined with oral supplementation of L-carnitine. A mouse model of Gcdh c.422_428del/c.422_428del (Gcdh -/-) was generated in our laboratory using CRISPR/Cas9. Gcdh -/- mice had significantly higher levels of glutaric acid (GA) in the plasma, liver, and brain than those in wild-type C57BL/6 mice. When given a high-protein diet (HPD) for two days, approximately 60% of Gcdh -/- mice did not survive the metabolic stress. To evaluate whether GCDH gene replacement therapy could be used to provide sustained treatment for patients with GA-1, we prepared a recombinant adeno-associated virus (rAAV) carrying a human GCDH expression cassette and injected it into Gcdh -/- neonates for a proof-of-concept (PoC) study. Our study demonstrated that delivering rAAV to the central nervous system (CNS), but not the peripheral system, significantly increased the survival rate under HPD exposure. Our study also demonstrated that rAAVPHP.eB mediated a higher efficiency than that of rAAV9 in increasing the survival rate. Surviving mice showed dose-dependent GCDH protein expression in the CNS and downregulation of GA levels. Our study demonstrated that AAV-based gene replacement therapy was effective for GA-I treatment and provided a feasible solution for this unmet medical need.
Collapse
Affiliation(s)
- Lu Guo
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhikun Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Yuhuan Li
- The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Bin Qu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guanyi Jiao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Chen Liang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zongbao Lu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Xin-Ge Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Cheng Huang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Hongwei Du
- The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Jianmin Liang
- The First Hospital of Jilin University, Changchun, Jilin 130021, China
| | - Qi Zhou
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| | - Wei Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing 100101, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
36
|
Tone D, Ode KL, Zhang Q, Fujishima H, Yamada RG, Nagashima Y, Matsumoto K, Wen Z, Yoshida SY, Mitani TT, Arisato Y, Ohno RI, Ukai-Tadenuma M, Yoshida Garçon J, Kaneko M, Shi S, Ukai H, Miyamichi K, Okada T, Sumiyama K, Kiyonari H, Ueda HR. Distinct phosphorylation states of mammalian CaMKIIβ control the induction and maintenance of sleep. PLoS Biol 2022; 20:e3001813. [PMID: 36194579 PMCID: PMC9531794 DOI: 10.1371/journal.pbio.3001813] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
The reduced sleep duration previously observed in Camk2b knockout mice revealed a role for Ca2+/calmodulin-dependent protein kinase II (CaMKII)β as a sleep-promoting kinase. However, the underlying mechanism by which CaMKIIβ supports sleep regulation is largely unknown. Here, we demonstrate that activation or inhibition of CaMKIIβ can increase or decrease sleep duration in mice by almost 2-fold, supporting the role of CaMKIIβ as a core sleep regulator in mammals. Importantly, we show that this sleep regulation depends on the kinase activity of CaMKIIβ. A CaMKIIβ mutant mimicking the constitutive-active (auto)phosphorylation state promotes the transition from awake state to sleep state, while mutants mimicking subsequent multisite (auto)phosphorylation states suppress the transition from sleep state to awake state. These results suggest that the phosphorylation states of CaMKIIβ differently control sleep induction and maintenance processes, leading us to propose a "phosphorylation hypothesis of sleep" for the molecular control of sleep in mammals.
Collapse
Affiliation(s)
- Daisuke Tone
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Koji L. Ode
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Qianhui Zhang
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hiroshi Fujishima
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Rikuhiro G. Yamada
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Yoshiki Nagashima
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Thermo Fisher Scientific K.K., Yokohama, Kanagawa, Japan
| | - Katsuhiko Matsumoto
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Zhiqing Wen
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shota Y. Yoshida
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Graduate school of Medicine, Osaka University, Suita, Osaka, Japan
| | - Tomoki T. Mitani
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Graduate school of Medicine, Osaka University, Suita, Osaka, Japan
| | - Yuki Arisato
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- Faculty of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Rei-ichiro Ohno
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Maki Ukai-Tadenuma
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Junko Yoshida Garçon
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan
| | - Shoi Shi
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Hideki Ukai
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Kazunari Miyamichi
- Laboratory for Comparative Connections, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, the University of Tokyo, Minato-city, Tokyo, Japan
| | - Kenta Sumiyama
- Laboratory for Mouse Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
| | - Hiroshi Kiyonari
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Chuo-ku, Kobe, Hyogo, Japan
| | - Hiroki R. Ueda
- Laboratory for Synthetic Biology, RIKEN Center for Biosystems Dynamics Research, Suita, Osaka, Japan
- Department of Systems Pharmacology, Graduate School of Medicine, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
- * E-mail:
| |
Collapse
|
37
|
Ye D, Yuan J, Yang Y, Yue Y, Hu Z, Fadera S, Chen H. Incisionless targeted adeno-associated viral vector delivery to the brain by focused ultrasound-mediated intranasal administration. EBioMedicine 2022; 84:104277. [PMID: 36152518 PMCID: PMC9508404 DOI: 10.1016/j.ebiom.2022.104277] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2022] [Revised: 09/05/2022] [Accepted: 09/06/2022] [Indexed: 12/02/2022] Open
Abstract
Background Adeno-associated viral (AAV) vectors are currently the leading platform for gene therapy with the potential to treat a variety of central nervous system (CNS) diseases. There are numerous methods for delivering AAVs to the CNS, such as direct intracranial injection (DI), intranasal delivery (IN), and intravenous injection with focused ultrasound-induced blood–brain barrier disruption (FUS-BBBD). However, non-invasive and efficient delivery of AAVs to the brain with minimal systemic toxicity remain the major challenge. This study aims to investigate the potential of focused ultrasound-mediated intranasal delivery (FUSIN) in AAV delivery to brain. Methods Mice were intranasally administered with AAV5 encoding enhanced green fluorescence protein (AAV5-EGFP) followed by FUS sonication in the presence of systemically injected microbubbles. Mouse brains and other major organs were harvested for immunohistological staining, PCR quantification, and in situ hybridization. The AAV delivery outcomes were compared with those of DI, FUS-BBBD, and IN delivery. Findings FUSIN achieved safe and efficient delivery of AAV5-EGFP to spatially targeted brain locations, including a superficial brain site (cortex) and a deep brain region (brainstem). FUSIN achieved comparable delivery outcomes as the established DI, and displayed 414.9-fold and 2073.7-fold higher delivery efficiency than FUS-BBBD and IN. FUSIN was associated with minimal biodistribution in peripheral organs, which was comparable to that of DI. Interpretation Our results suggest that FUSIN is a promising technique for non-invasive, efficient, safe, and spatially targeted AAV delivery to the brain. Funding National Institutes of Health (NIH) grants R01EB027223, R01EB030102, R01MH116981, and UG3MH126861.
Collapse
|
38
|
Georgieva JV, Katt M, Ye Z, Umlauf BJ, Wenthur CJ, Shusta EV. The 46.1 Antibody Mediates Neurotensin Uptake into the CNS and the Effects Depend on the Route of Intravenous Administration. Pharmaceutics 2022; 14:pharmaceutics14081706. [PMID: 36015332 PMCID: PMC9414616 DOI: 10.3390/pharmaceutics14081706] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 08/10/2022] [Accepted: 08/11/2022] [Indexed: 11/16/2022] Open
Abstract
Central nervous system (CNS) exposure to blood-borne biotherapeutics is limited by the restrictive nature of the brain vasculature. In particular, tightly sealed endothelial cells of the blood-brain barrier (BBB) prevent the uptake of protein and gene medicines. An approach to increase the bioavailability of such therapeutics is harnessing the BBB endothelial cells' own receptor-mediated transcytosis (RMT) mechanisms. Key to this process is a targeting ligand that can engage a BBB-resident RMT receptor. We recently identified an antibody, named 46.1, that accumulates in the mouse brain after intravenous injection. To further characterize the brain targeting and penetrating properties of clone 46.1, we conjugated neurotensin (NT) to an scFv-Fc form of the antibody (46.1-scFv-Fc-LongLinker-NT). While centrally administered NT decreases the core body temperature and locomotor activity, effects attributed to two spatially segregated brain areas, systemically administered NT has limited effects. Hence, NT can be used as a model therapeutic payload to evaluate the brain penetration of BBB-targeting antibodies and their capability to accumulate in discrete brain areas. We demonstrate that intravenously administered 46.1-scFv-Fc-LL-NT can elicit transient hypothermia and reduce drug-induced hyperlocomotion, confirming that 46.1 can deliver drug cargo to the CNS at pharmacologically relevant doses. Interestingly, when two intravenous administration routes in mice, retro-orbital and tail vein, were compared, only retro-orbital administration led to transient hypothermia. We further explored the retro-orbital route and demonstrated that the 46.1-scFv-Fc-LL-NT could enter the brain arterial blood supply directly from the retro-orbital/cavernous sinus. Taken together, the 46.1 antibody is capable of transporting drug cargo into the CNS, and at least of a portion of its CNS accumulation occurs via the cavernous sinus-arterial route.
Collapse
Affiliation(s)
- Julia V. Georgieva
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Moriah Katt
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Zhou Ye
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Benjamin J. Umlauf
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
| | - Cody J. Wenthur
- Divisions of Pharmaceutical Sciences and Pharmacy Practice, School of Pharmacy, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Eric V. Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI 53792, USA
- Correspondence:
| |
Collapse
|
39
|
Hunter JE, Molony CM, Bagel JH, O’Donnell PA, Kaler SG, Wolfe JH. Transduction characteristics of alternative adeno-associated virus serotypes in the cat brain by intracisternal delivery. Mol Ther Methods Clin Dev 2022; 26:384-393. [PMID: 36034772 PMCID: PMC9391516 DOI: 10.1016/j.omtm.2022.07.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 07/12/2022] [Indexed: 11/18/2022]
Abstract
Multiple studies have examined the transduction characteristics of different AAV serotypes in the mouse brain, where they can exhibit significantly different patterns of transduction. The pattern of transduction also varies with the route of administration. Much less information exists for the transduction characteristics in large-brained animals. Large animal models have brains that are closer in size and organization to the human brain, such as being gyrencephalic compared to the lissencephalic rodent brains, pathway organization, and certain electrophysiologic properties. Large animal models are used as translational intermediates to develop gene therapies to treat human diseases. Various AAV serotypes and routes of delivery have been used to study the correction of pathology in the brain in lysosomal storage diseases. In this study, we evaluated the ability of selected AAV serotypes to transduce cells in the cat brain when delivered into the cerebrospinal fluid via the cisterna magna. We previously showed that AAV1 transduced significantly greater numbers of cells than AAV9 in the cat brain by this route. In the present study, we evaluated serotypes closely related to AAVs 1 and 9 (AAVs 6, AS, hu32) that may mediate more extensive transduction, as well as AAVs 4 and 5, which primarily transduce choroid plexus epithelial (CPE) and ependymal lining cells in the rodent brain. The related serotypes tended to have similar patterns of transduction but were divergent in some specific brain structures.
Collapse
Affiliation(s)
- Jacqueline E. Hunter
- Research Institute of Children’s Hospital of Philadelphia, 502-G Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA
| | - Caitlyn M. Molony
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jessica H. Bagel
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patricia A. O’Donnell
- W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Stephen G. Kaler
- Section on Translational Neuroscience, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD 20892, USA
| | - John H. Wolfe
- Research Institute of Children’s Hospital of Philadelphia, 502-G Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104, USA,W.F. Goodman Center for Comparative Medical Genetics, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA,Corresponding author John H. Wolfe, Children’s Hospital of Philadelphia, 502-G Abramson Research Center, 3615 Civic Center Boulevard, Philadelphia, PA 19104-4399, USA.
| |
Collapse
|
40
|
Pouliopoulos AN, Murillo MF, Noel RL, Batts AJ, Ji R, Kwon N, Yu H, Tong CK, Gelinas JN, Araghy DK, Hussaini SA, Konofagou EE. Non-invasive optogenetics with ultrasound-mediated gene delivery and red-light excitation. Brain Stimul 2022; 15:927-941. [PMID: 35718324 PMCID: PMC9379392 DOI: 10.1016/j.brs.2022.06.007] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/30/2022] [Accepted: 06/11/2022] [Indexed: 12/03/2022] Open
Abstract
Optogenetics has revolutionized the capability of controlling genetically modified neurons in vitro and in vivo and has become an indispensable neuroscience tool. Using light as a probe for selective neuronal activation or inhibition and as a means to read out neural activity has dramatically enhanced our understanding of complex neural circuits. However, a common limitation of optogenetic studies to date is their invasiveness and spatiotemporal range. Direct viral injections into the brain tissue along with implantation of optical fibers and recording electrodes can disrupt the neuronal circuitry and cause significant damage. Conventional approaches are spatially limited around the site of the direct injection and insufficient in examining large networks throughout the brain. Lastly, optogenetics is currently not easily scalable to large animals or humans. Here, we demonstrate that optogenetic excitation can be achieved entirely non-invasively through the intact skull in mice. Using a needle-free combination of focused ultrasound-mediated viral delivery and extracorporeal illumination with red light, we achieved selective neuronal activation at depths up to 4 mm in the murine brain, confirmed through cFos expression and electrophysiology measurements within the treated areas. Ultrasound treatment significantly reduced freezing time during recall in fear conditioning experiments, but remote light exposure had a moderate effect on the freezing behavior of mice treated with viral vectors. The proposed method has the potential to open new avenues of studying, but also stimulating, neuronal networks, in an effort to elucidate normal or dysfunctional brain activity and treat neurological diseases. Finally, the same non-invasive methodology could be combined with gene therapy and applied to other organs, such as the eye and the heart.
Collapse
Affiliation(s)
| | - Maria F Murillo
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Rebecca Lynn Noel
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Alec J Batts
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Robin Ji
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Nancy Kwon
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA
| | - Han Yu
- Department of Electrical Engineering, Columbia University, New York City, NY, USA
| | - Chi-Kun Tong
- Department of Physiology and Cellular Biophysics, Columbia University, New York City, NY, USA
| | | | | | - S Abid Hussaini
- Department of Pathology and Cell Biology, Columbia University, New York City, NY, USA
| | - Elisa E Konofagou
- Department of Biomedical Engineering, Columbia University, New York City, NY, USA; Department of Radiology, Columbia University, New York City, NY, USA.
| |
Collapse
|
41
|
Palfi A, Chadderton N, Millington-Ward S, Post I, Humphries P, Kenna PF, Farrar GJ. AAV-PHP.eB transduces both the inner and outer retina with high efficacy in mice. Mol Ther Methods Clin Dev 2022; 25:236-249. [PMID: 35474956 PMCID: PMC9018541 DOI: 10.1016/j.omtm.2022.03.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2021] [Accepted: 03/27/2022] [Indexed: 12/18/2022]
Abstract
Recombinant adeno-associated virus (AAV) vectors are one of the main gene delivery vehicles used in retinal gene therapy approaches; however, there is a need to further improve the efficacy, tropism, and safety of these vectors. In this study, using a CMV-EGFP expression cassette, we characterize the retinal utility of AAV-PHP.eB, a serotype recently developed by in vivo directed evolution, which can cross the blood-brain barrier and target neurons with high efficacy in mice. Systemic and intravitreal delivery of AAV-PHP.eB resulted in the high transduction efficacy of retinal ganglion and horizontal cells, with systemic delivery providing pan-retinal coverage of the mouse retina. Subretinal delivery transduced photoreceptors and retinal pigment epithelium cells robustly. EGFP expression (number of transduced cells and mRNA levels) were similar when the retinas were transduced systemically or intravitreally with AAV-PHP.eB or intravitreally with AAV2/2. Notably, in photoreceptors, EGFP fluorescence intensities and mRNA levels were 50–70 times higher, when subretinal injections with AAV-PHP.eB were compared to AAV2/8. Our results demonstrate the pan-retinal transduction of ganglion cells and extremely efficient transduction of photoreceptor and retinal pigment epithelium cells as the most valuable features of AAV-PHP.eB in the mouse retina.
Collapse
Affiliation(s)
- Arpad Palfi
- Department of Genetics, School of Genetics and Microbiology, Trinity College Dublin, D02 VF25, Dublin, Ireland
| | - Naomi Chadderton
- Department of Genetics, School of Genetics and Microbiology, Trinity College Dublin, D02 VF25, Dublin, Ireland
| | - Sophia Millington-Ward
- Department of Genetics, School of Genetics and Microbiology, Trinity College Dublin, D02 VF25, Dublin, Ireland
| | - Iris Post
- Department of Genetics, School of Genetics and Microbiology, Trinity College Dublin, D02 VF25, Dublin, Ireland
| | - Pete Humphries
- Department of Genetics, School of Genetics and Microbiology, Trinity College Dublin, D02 VF25, Dublin, Ireland
| | - Paul F Kenna
- Department of Genetics, School of Genetics and Microbiology, Trinity College Dublin, D02 VF25, Dublin, Ireland.,The Research Foundation, Royal Victoria Eye and Ear Hospital, D02 XK51, Dublin, Ireland
| | - G Jane Farrar
- Department of Genetics, School of Genetics and Microbiology, Trinity College Dublin, D02 VF25, Dublin, Ireland
| |
Collapse
|
42
|
Islam A, Tom VJ. The use of viral vectors to promote repair after spinal cord injury. Exp Neurol 2022; 354:114102. [PMID: 35513025 DOI: 10.1016/j.expneurol.2022.114102] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Revised: 04/21/2022] [Accepted: 04/27/2022] [Indexed: 11/16/2022]
Abstract
Spinal cord injury (SCI) is a devastating event that can permanently disrupt multiple modalities. Unfortunately, the combination of the inhibitory environment at a central nervous system (CNS) injury site and the diminished intrinsic capacity of adult axons for growth results in the failure for robust axonal regeneration, limiting the ability for repair. Delivering genetic material that can either positively or negatively modulate gene expression has the potential to counter the obstacles that hinder axon growth within the spinal cord after injury. A popular gene therapy method is to deliver the genetic material using viral vectors. There are considerations when deciding on a viral vector approach for a particular application, including the type of vector, as well as serotypes, and promoters. In this review, we will discuss some of the aspects to consider when utilizing a viral vector approach to as a therapy for SCI. Additionally, we will discuss some recent applications of gene therapy to target extrinsic and/or intrinsic barriers to promote axon regeneration after SCI in preclinical models. While still in early stages, this approach has potential to treat those living with SCI.
Collapse
Affiliation(s)
- Ashraful Islam
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA
| | - Veronica J Tom
- Drexel University College of Medicine, Department of Neurobiology and Anatomy, Marion Murray Spinal Cord Research Center, Philadelphia, PA, USA.
| |
Collapse
|
43
|
Challis RC, Ravindra Kumar S, Chen X, Goertsen D, Coughlin GM, Hori AM, Chuapoco MR, Otis TS, Miles TF, Gradinaru V. Adeno-Associated Virus Toolkit to Target Diverse Brain Cells. Annu Rev Neurosci 2022; 45:447-469. [PMID: 35440143 DOI: 10.1146/annurev-neuro-111020-100834] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recombinant adeno-associated viruses (AAVs) are commonly used gene delivery vehicles for neuroscience research. They have two engineerable features: the capsid (outer protein shell) and cargo (encapsulated genome). These features can be modified to enhance cell type or tissue tropism and control transgene expression, respectively. Several engineered AAV capsids with unique tropisms have been identified, including variants with enhanced central nervous system transduction, cell type specificity, and retrograde transport in neurons. Pairing these AAVs with modern gene regulatory elements and state-of-the-art reporter, sensor, and effector cargo enables highly specific transgene expression for anatomical and functional analyses of brain cells and circuits. Here, we discuss recent advances that provide a comprehensive (capsid and cargo) AAV toolkit for genetic access to molecularly defined brain cell types. Expected final online publication date for the Annual Review of Neuroscience, Volume 45 is July 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Rosemary C Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - David Goertsen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Gerard M Coughlin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Acacia M Hori
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Miguel R Chuapoco
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Thomas S Otis
- Sainsbury Wellcome Centre, University College London, London, United Kingdom
| | - Timothy F Miles
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| |
Collapse
|
44
|
Marino M, Holt MG. AAV Vector-Mediated Antibody Delivery (A-MAD) in the Central Nervous System. Front Neurol 2022; 13:870799. [PMID: 35493843 PMCID: PMC9039256 DOI: 10.3389/fneur.2022.870799] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 02/21/2022] [Indexed: 11/13/2022] Open
Abstract
In the last four decades, monoclonal antibodies and their derivatives have emerged as a powerful class of therapeutics, largely due to their exquisite targeting specificity. Several clinical areas, most notably oncology and autoimmune disorders, have seen the successful introduction of monoclonal-based therapeutics. However, their adoption for treatment of Central Nervous System diseases has been comparatively slow, largely due to issues of efficient delivery resulting from limited permeability of the Blood Brain Barrier. Nevertheless, CNS diseases are becoming increasingly prevalent as societies age, accounting for ~6.5 million fatalities worldwide per year. Therefore, harnessing the full therapeutic potential of monoclonal antibodies (and their derivatives) in this clinical area has become a priority. Adeno-associated virus-based vectors (AAVs) are a potential solution to this problem. Preclinical studies have shown that AAV vector-mediated antibody delivery provides protection against a broad range of peripheral diseases, such as the human immunodeficiency virus (HIV), influenza and malaria. The parallel identification and optimization of AAV vector platforms which cross the Blood Brain Barrier with high efficiency, widely transducing the Central Nervous System and allowing high levels of local transgene production, has now opened a number of interesting scenarios for the development of AAV vector-mediated antibody delivery strategies to target Central Nervous System proteinopathies.
Collapse
Affiliation(s)
- Marika Marino
- Laboratory of Glia Biology, VIB-KU Leuven, Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
| | - Matthew G. Holt
- Laboratory of Glia Biology, VIB-KU Leuven, Center for Brain & Disease Research, Leuven, Belgium
- Department of Neurosciences, KU Leuven, Leuven, Belgium
- Leuven Brain Institute, Leuven, Belgium
- Synapse Biology Group, Instituto de Investigação e Inovação em Saúde (i3S), University of Porto, Porto, Portugal
- *Correspondence: Matthew G. Holt
| |
Collapse
|
45
|
Applications of chemogenetics in non-human primates. Curr Opin Pharmacol 2022; 64:102204. [DOI: 10.1016/j.coph.2022.102204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/10/2022] [Accepted: 02/11/2022] [Indexed: 11/23/2022]
|
46
|
Björklund T, Davidsson M. Next-Generation Gene Therapy for Parkinson's Disease Using Engineered Viral Vectors. JOURNAL OF PARKINSON'S DISEASE 2022; 11:S209-S217. [PMID: 34366370 PMCID: PMC8543274 DOI: 10.3233/jpd-212674] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 07/14/2021] [Indexed: 11/23/2022]
Abstract
Recent technological and conceptual advances have resulted in a plethora of exciting novel engineered adeno associated viral (AAV) vector variants. They all have unique characteristics and abilities. This review summarizes the development and their potential in treating Parkinson's disease (PD). Clinical trials in PD have shown over the last decade that AAV is a safe and suitable vector for gene therapy but that it also is a vehicle that can benefit significantly from improvement in specificity and potency. This review provides a concise collection of the state-of-the-art for synthetic capsids and their utility in PD. We also summarize what therapeutical strategies may become feasible with novel engineered vectors, including genome editing and neuronal rejuvenation.
Collapse
Affiliation(s)
- Tomas Björklund
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| | - Marcus Davidsson
- Molecular Neuromodulation, Wallenberg Neuroscience Center, Lund University, Lund, Sweden
| |
Collapse
|
47
|
Arotcarena ML, Dovero S, Biendon N, Dutheil N, Planche V, Bezard E, Dehay B. Pilot Study Assessing the Impact of Intrathecal Administration of Variants AAV-PHP.B and AAV-PHP.eB on Brain Transduction in Adult Rhesus Macaques. Front Bioeng Biotechnol 2021; 9:762209. [PMID: 34869273 PMCID: PMC8634843 DOI: 10.3389/fbioe.2021.762209] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Accepted: 10/26/2021] [Indexed: 01/15/2023] Open
Abstract
Adeno-associated virus (AAV) vectors are increasingly used as an effective and safe approach to deliver genetic material to the central nervous system (CNS). The AAV9-derived variants, AAV-PHP. B and AAV-PHP.eB, reportedly broadly transduce cells throughout the CNS compared to the original serotype 9, AAV9. As non-human primate data are scarce, we here evaluated the CNS transduction efficiencies after lumbar intrathecal bolus delivery of identical doses of either AAV-PHP. B:CAG-EGFP or AAV-PHP. eB:CAG-EGFP in rhesus macaque monkeys. AAV-PHP.eB achieved a more efficient and widespread CNS transduction compared to AAV-PHP.B. We report a strong neuronal and oligodendroglial tropism for both variants in the putamen and in the hippocampus. This proof-of-concept experiment highlights the potential value of intrathecal infusions of AAV-PHP.eB to distribute genetic material in the CNS with cell-type specificity and introduces a new opportunity to model brain diseases in rhesus macaque monkeys and further develop gene therapies targeting the CNS in humans.
Collapse
Affiliation(s)
| | - Sandra Dovero
- CNRS, IMN, UMR 5293, Univ. Bordeaux, Bordeaux, France
| | | | | | - Vincent Planche
- CNRS, IMN, UMR 5293, Univ. Bordeaux, Bordeaux, France.,Centre Memoire de Ressources et de Recherches, Pôle de Neurosciences Cliniques, CHU de Bordeaux, Bordeaux, France
| | - Erwan Bezard
- CNRS, IMN, UMR 5293, Univ. Bordeaux, Bordeaux, France
| | | |
Collapse
|
48
|
Ding Y, Shusta EV, Palecek SP. Integrating in vitro disease models of the neurovascular unit into discovery and development of neurotherapeutics. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2021; 20:100341. [PMID: 34693102 PMCID: PMC8530278 DOI: 10.1016/j.cobme.2021.100341] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The blood-brain barrier (BBB) regulates the transport of small molecules, proteins, and cells between the bloodstream and the central nervous system (CNS). Brain microvascular endothelial cells work with other resident brain cell types, including pericytes, astrocytes, neurons, and microglia, to form the neurovascular unit (NVU) and maintain BBB integrity. The restrictive barrier influences the pathogenesis of many CNS diseases, and impedes the delivery of neurotherapeutics into the CNS. In vitro NVU models enable the discovery of complex cell-cell interactions involved in human BBB pathophysiology in diseases including Alzheimer's Disease (AD), Parkinson's Disease (PD) and viral infections of the brain. In vitro NVU models have also been deployed to study the delivery of neurotherapeutics across the BBB, including small molecule drugs, monoclonal antibodies, gene therapy vectors and immune cells. The high scalability, accessibility, and phenotype fidelity of in vitro NVU models can facilitate the discovery and development of effective neurotherapeutics.
Collapse
Affiliation(s)
- Yunfeng Ding
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eric V Shusta
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
- Department of Neurological Surgery, University of Wisconsin-Madison, Madison, WI, USA
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
49
|
Hana S, Peterson M, McLaughlin H, Marshall E, Fabian AJ, McKissick O, Koszka K, Marsh G, Craft M, Xu S, Sorets A, Torregrosa T, Sun C, Henderson CE, Lo SC. Highly efficient neuronal gene knockout in vivo by CRISPR-Cas9 via neonatal intracerebroventricular injection of AAV in mice. Gene Ther 2021; 28:646-658. [PMID: 33558692 PMCID: PMC8599009 DOI: 10.1038/s41434-021-00224-2] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 11/29/2020] [Accepted: 01/15/2021] [Indexed: 02/08/2023]
Abstract
CRISPR-Cas systems have emerged as a powerful tool to generate genetic models for studying normal and diseased central nervous system (CNS). Targeted gene disruption at specific loci has been demonstrated successfully in non-dividing neurons. Despite its simplicity, high specificity and low cost, the efficiency of CRISPR-mediated knockout in vivo can be substantially impacted by many parameters. Here, we used CRISPR-Cas9 to disrupt the neuronal-specific gene, NeuN, and optimized key parameters to achieve effective gene knockout broadly in the CNS in postnatal mice. Three cell lines and two primary neuron cultures were used to validate the disruption of NeuN by single-guide RNAs (sgRNA) harboring distinct spacers and scaffold sequences. This triage identified an optimal sgRNA design with the highest NeuN disruption in in vitro and in vivo systems. To enhance CRISPR efficiency, AAV-PHP.B, a vector with superior neuronal transduction, was used to deliver this sgRNA in Cas9 mice via neonatal intracerebroventricular (ICV) injection. This approach resulted in 99.4% biallelic indels rate in the transduced cells, leading to greater than 70% reduction of total NeuN proteins in the cortex, hippocampus and spinal cord. This work contributes to the optimization of CRISPR-mediated knockout and will be beneficial for fundamental and preclinical research.
Collapse
|
50
|
Judson MC, Shyng C, Simon JM, Davis CR, Punt AM, Salmon MT, Miller NW, Ritola KD, Elgersma Y, Amaral DG, Gray SJ, Philpot BD. Dual-isoform hUBE3A gene transfer improves behavioral and seizure outcomes in Angelman syndrome model mice. JCI Insight 2021; 6:144712. [PMID: 34676830 PMCID: PMC8564914 DOI: 10.1172/jci.insight.144712] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Accepted: 09/02/2021] [Indexed: 12/23/2022] Open
Abstract
Loss of the maternal UBE3A allele causes Angelman syndrome (AS), a debilitating neurodevelopmental disorder. Here, we devised an AS treatment strategy based on reinstating dual-isoform expression of human UBE3A (hUBE3A) in the developing brain. Kozak sequence engineering of our codon-optimized vector (hUBE3Aopt) enabled translation of both short and long hUBE3A protein isoforms at a near-endogenous 3:1 (short/long) ratio, a feature that could help to support optimal therapeutic outcomes. To model widespread brain delivery and early postnatal onset of hUBE3A expression, we packaged the hUBE3Aopt vector into PHP.B capsids and performed intracerebroventricular injections in neonates. This treatment significantly improved motor learning and innate behaviors in AS mice, and it rendered them resilient to epileptogenesis and associated hippocampal neuropathologies induced by seizure kindling. hUBE3A overexpression occurred frequently in the hippocampus but was uncommon in the neocortex and other major brain structures; furthermore, it did not correlate with behavioral performance. Our results demonstrate the feasibility, tolerability, and therapeutic potential for dual-isoform hUBE3A gene transfer in the treatment of AS.
Collapse
Affiliation(s)
- Matthew C Judson
- Neuroscience Center.,Department of Cell Biology and Physiology.,Carolina Institute for Developmental Disabilities
| | - Charles Shyng
- Carolina Institute for Developmental Disabilities.,Gene Therapy Center, and
| | - Jeremy M Simon
- Neuroscience Center.,Carolina Institute for Developmental Disabilities.,Department of Genetics, University of North Carolina (UNC), Chapel Hill, North Carolina, USA
| | | | - A Mattijs Punt
- Department of Clinical Genetics and.,Department of Neuroscience, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | | | - Noah W Miller
- Neuroscience Center.,Department of Cell Biology and Physiology
| | - Kimberly D Ritola
- Neuroscience Center.,Department of Pharmacology, UNC, Chapel Hill, North Carolina, USA.,Scientific Operations Manager-Viral Tools, Janelia Research Campus, Howard Hughes Medical Institute, Ashburn, Virginia, USA
| | - Ype Elgersma
- Department of Clinical Genetics and.,Department of Neuroscience, Erasmus MC University Medical Center, Rotterdam, The Netherlands.,ENCORE Expertise Center for Neurodevelopmental Disorders, Erasmus MC University Medical Center, Rotterdam, The Netherlands
| | - David G Amaral
- Department of Psychiatry and Behavioral Sciences, MIND Institute, and.,California National Primate Research Center, University of California, Davis, California, USA
| | - Steven J Gray
- Gene Therapy Center, and.,Department of Pediatrics and.,Eugene McDermott Center for Human Growth and Development, University of Texas (UT) Southwestern Medical Center, Dallas, Texas, USA
| | - Benjamin D Philpot
- Neuroscience Center.,Department of Cell Biology and Physiology.,Carolina Institute for Developmental Disabilities
| |
Collapse
|