1
|
Iyer S, Montmayeur JP, Zolotukhin S, Dotson CD. Exogenous oral application of PYY and exendin-4 impacts upon taste-related behavior and taste perception in wild-type mice. Neuropharmacology 2025; 272:110408. [PMID: 40086622 PMCID: PMC12042652 DOI: 10.1016/j.neuropharm.2025.110408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 03/16/2025]
Abstract
Several gut peptides have been implicated in feeding and body mass accumulation. Glucagon-like peptide 1 (GLP-1) and peptide tyrosine-tyrosine (PYY) have been shown to mediate satiety and reduce food intake. While systemic administration of such peptides has been explored as a therapy for metabolic disease, the effects of these hormones on taste signaling should also be considered given the importance of taste to feeding decisions and considering the fact that components of these signaling systems are expressed in cells of the peripheral gustatory system. We previously demonstrated that genetic disruption of PYY signaling in mice can impact on taste responsiveness and feeding and that viral expression of PYY in the salivary glands of PYY knockout mice can rescue responsiveness. The present work uses adeno-associated virus-mediated salivary gland treatment with both GLP-1 receptor agonist exendin-4 and/or PYY encoding vectors to explore the effect of stimulating these orally present signaling systems on taste-related behavioral responsiveness in male wild-type mice with intact peptide signaling systems. Results showed a significant effect of salivary gland treatment on responsiveness to multiple taste qualities. Data gathered from taste bud cells in vitro suggest that these peptides directly influence the responsiveness of these primary sensory cells. Collectively, these findings show that taste perception can be modulated by the exogenous application of satiety peptides in wild-type mice and suggest that the taste bud is a promising substrate for food intake modulation.
Collapse
Affiliation(s)
- Satya Iyer
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA
| | | | - Sergei Zolotukhin
- Department of Pediatrics, University of Florida College of Medicine, Gainesville, FL, 32610, USA
| | - Cedrick D Dotson
- Neuroscience Institute, Georgia State University, Atlanta, GA, 30303, USA.
| |
Collapse
|
2
|
Kozoriz A, Mora S, Damiano MA, Carballo-Carbajal I, Parent A, Kumarasinghe L, Vila M, Lassot I, Desagher S. ZSCAN21 mediates the pathogenic transcriptional induction of α-synuclein in cellular and animal models of Parkinson's disease. Cell Death Dis 2025; 16:394. [PMID: 40379611 DOI: 10.1038/s41419-025-07722-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 04/25/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
The expression level of α-synuclein is thought to play a crucial role in the pathogenesis of Parkinson's disease. However, little is known about the molecular mechanisms regulating the transcription of its gene, SNCA, particularly in the context of the disease. The transcription factor ZSCAN21 has been shown to act on SNCA, but whether ZSCAN21 is actually involved in the induction of SNCA transcription in Parkinson's disease is unknown. To address this question, we used the MPTP mouse model and LUHMES-derived dopaminergic neuronal spheroids, subjected to Parkinson's disease-related neurotoxins and mutations. We show that MPP+-treated spheroids recapitulate the main features of α-synuclein pathology and that MPP+-triggered transcriptional induction of SNCA is associated with ZSCAN21 stabilisation. Importantly, knock-down of ZSCAN21 prevents both the MPP+-triggered increase in α-synuclein mRNA and pre-mRNA levels in LUHMES-derived spheroids and the death of dopaminergic neurons in the substantia nigra of MPTP-treated mice. These effects are recapitulated by knockdown of TRIM17, a ZSCAN21 stabiliser which prevents its ubiquitination and degradation mediated by TRIM41. Moreover, reducing the interaction between ZSCAN21 and TRIM41, either by inserting Parkinson's disease-associated mutations into the TRIM41 gene or by preventing SUMOylation of ZSCAN21, results in both stabilisation of ZSCAN21 and induction of SNCA. Taken together, our data strongly suggest that ZSCAN21 is a crucial transcription factor for pathogenic α-synuclein expression and neurodegeneration in Parkinson's disease, pointing to its regulators, TRIM17 and TRIM41, as original therapeutic targets for a neuroprotective treatment of Parkinson's disease.
Collapse
Affiliation(s)
- Alina Kozoriz
- IGMM, University of Montpellier, CNRS, Montpellier, France
| | - Stéphan Mora
- IGMM, University of Montpellier, CNRS, Montpellier, France
| | | | - Iria Carballo-Carbajal
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | - Annabelle Parent
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
| | | | - Miquel Vila
- Neurodegenerative Diseases Research Group, Vall d'Hebron Research Institute (VHIR)-Center for Networked Biomedical Research on Neurodegenerative Diseases (CIBERNED), Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), Barcelona, Spain
- Institut de Neurociències (INc-UAB), Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Iréna Lassot
- IGMM, University of Montpellier, CNRS, Montpellier, France
| | - Solange Desagher
- IGMM, University of Montpellier, CNRS, Montpellier, France.
- IRIM, University of Montpellier, CNRS, Montpellier, France.
| |
Collapse
|
3
|
Lorek JK, Isaksson M, Nilsson B. Chromatography in Downstream Processing of Recombinant Adeno-Associated Viruses: A Review of Current and Future Practises. Biotechnol Bioeng 2025; 122:1067-1086. [PMID: 39905691 PMCID: PMC11975191 DOI: 10.1002/bit.28932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/06/2025]
Abstract
Recombinant adeno-associated virus (rAAV) has emerged as an attractive gene delivery vector platform to treat both rare and pervasive diseases. With more and more rAAV-based therapies entering late-stage clinical trials and commercialization, there is an increasing pressure on the rAAV manufacturing process to accelerate drug development, account for larger trials, and commercially provide high doses. Still, many of the pre-clinical and clinical manufacturing processes are tied to outdated technologies, which results in substantial production expenses. Those processes face challenges including low productivity and difficult scalability, which limits its ability to provide for required dosages which in turn influences the accessibility of the drug. And as upstream efforts are expected to increase productivities, the downstream part needs to adapt with more scalable and efficient technologies. In this review, both traditional and novel rAAV downstream technologies are presented and discussed. Traditional rAAV downstream processes are based on density gradient ultracentrifugation and have been shown to effectively purify rAAVs with high yields and purities. However, those processes lack scalability and efficiency, which is why novel rAAV downstream processes based on column-chromatography have emerged as an attractive alternative and show potential for integration in continuous processes, following the principle of next-generation manufacturing.
Collapse
Affiliation(s)
| | - Madelène Isaksson
- Department of Process and Life Science EngineeringLund UniversityLundSweden
| | - Bernt Nilsson
- Department of Process and Life Science EngineeringLund UniversityLundSweden
| |
Collapse
|
4
|
Selten M, Bernard C, Mukherjee D, Hamid F, Hanusz-Godoy A, Oozeer F, Zimmer C, Marín O. Regulation of PV interneuron plasticity by neuropeptide-encoding genes. Nature 2025:10.1038/s41586-025-08933-z. [PMID: 40307547 DOI: 10.1038/s41586-025-08933-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/24/2025] [Indexed: 05/02/2025]
Abstract
Neuronal activity must be regulated in a narrow permissive band for the proper operation of neural networks. Changes in synaptic connectivity and network activity-for example, during learning-might disturb this balance, eliciting compensatory mechanisms to maintain network function1-3. In the neocortex, excitatory pyramidal cells and inhibitory interneurons exhibit robust forms of stabilizing plasticity. However, although neuronal plasticity has been thoroughly studied in pyramidal cells4-8, little is known about how interneurons adapt to persistent changes in their activity. Here we describe a critical cellular process through which cortical parvalbumin-expressing (PV+) interneurons adapt to changes in their activity levels. We found that changes in the activity of individual PV+ interneurons drive bidirectional compensatory adjustments of the number and strength of inhibitory synapses received by these cells, specifically from other PV+ interneurons. High-throughput profiling of ribosome-associated mRNA revealed that increasing the activity of a PV+ interneuron leads to upregulation of two genes encoding multiple secreted neuropeptides: Vgf and Scg2. Functional experiments demonstrated that VGF is critically required for the activity-dependent scaling of inhibitory PV+ synapses onto PV+ interneurons. Our findings reveal an instructive role for neuropeptide-encoding genes in regulating synaptic connections among PV+ interneurons in the adult mouse neocortex.
Collapse
Affiliation(s)
- Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Clémence Bernard
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
- Department of Clinical and Biomedical Sciences, Faculty of Health and Life Sciences, University of Exeter, Exeter, UK
| | - Diptendu Mukherjee
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fursham Hamid
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Alicia Hanusz-Godoy
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Christoph Zimmer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK.
- Medical Research Council Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
5
|
Jiang M, Ding H, Huang Y, Lau CW, Guo Y, Luo J, Shih YT, Xia Y, Yao X, Chiu JJ, Wang L, Chien S, Huang Y. Endothelial Serotonin Receptor 1B Acts as a Mechanosensor to Drive Atherosclerosis. Circ Res 2025; 136:887-901. [PMID: 40071330 DOI: 10.1161/circresaha.124.325453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Atherosclerosis is characterized by the accumulation of fatty and fibrotic plaques, which preferentially develop at curvatures and branches along the arterial trees that are exposed to disturbed flow. However, the mechanisms by which endothelial cells sense disturbed flow are still unclear. METHODS The partial carotid ligation mouse model was used to investigate disturbed flow-induced atherogenesis. In vitro experiments were performed using the ibidi system to generate oscillatory shear stress and laminar shear stress. ApoE-/- mice with endothelium-specific knockout or overexpression of 5-HT1B (serotonin receptor 1B) were used to investigate the role of endothelial 5-HT1B in atherosclerosis. RNA sequencing analysis, immunofluorescence analysis, and molecular biological techniques were used to explore the role of 5-HT1B in mechanotransduction and endothelial activation. RESULTS The data showed that human endothelial cells express a high level of 5-HT1B, which is a serotonin receptor subtype. Endothelial 5-HT1B is upregulated in atherosclerotic areas of both humans and rodents and is increased by disturbed flow both in vivo and in vitro. Endothelium-specific overexpression of 5-HT1B exacerbates, whereas knockout or knockdown of 5-HT1B in endothelium inhibits disturbed flow-induced endothelial inflammation and atherogenesis in both male and female ApoE-/- mice. We reveal a previously unknown role of 5-HT1B as a mechanosensor in endothelial cells in response to mechanical stimuli. Upon activation by oscillatory shear stress, 5-HT1B recruits β-arrestin, orchestrates RhoA (ras homolog family member A), and then activates mechanosensitive YAP (yes-associated protein), thereby enhancing endothelial inflammation and monocyte infiltration. Pharmacological blockade of 5-HT1B suppresses endothelial activation and atherogenesis via inhibition of YAP. CONCLUSIONS Taken together, these results uncover that endothelial 5-HT1B acts as a mechanosensor for disturbed flow and contributes to atherogenesis. Inhibition of 5-HT1B could be a promising therapeutic strategy for atherosclerosis.
Collapse
MESH Headings
- Animals
- Humans
- Mechanotransduction, Cellular
- Atherosclerosis/metabolism
- Atherosclerosis/pathology
- Atherosclerosis/genetics
- Receptor, Serotonin, 5-HT1B/metabolism
- Receptor, Serotonin, 5-HT1B/genetics
- Mice
- Male
- Mice, Knockout, ApoE
- Mice, Inbred C57BL
- Endothelial Cells/metabolism
- Stress, Mechanical
- Cells, Cultured
- Female
- Human Umbilical Vein Endothelial Cells/metabolism
- Apolipoproteins E/genetics
- Mice, Knockout
- Disease Models, Animal
Collapse
Affiliation(s)
- Minchun Jiang
- Department of Endocrinology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China (M.J.)
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Huanyu Ding
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Yuhong Huang
- Department of Biomedical Sciences (Yuhong Huang, L.W., Yu Huang), City University of Hong Kong, Hong Kong, China
| | - Chi Wai Lau
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Ying Guo
- Department of Endocrinology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China (Y.G.)
| | - Jianfang Luo
- Department of Cardiology, Guangdong Provincial Key Laboratory of Coronary Heart Disease Prevention, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China (J.L.)
| | - Yu-Tsung Shih
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan (Y.-T.S., J.-J.C.)
| | - Yin Xia
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Xiaoqiang Yao
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
| | - Jeng-Jiann Chiu
- College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan (J.-J.C.)
- Institute of Cellular and System Medicine, National Health Research Institutes, Miaoli, Taiwan (Y.-T.S., J.-J.C.)
| | - Li Wang
- Department of Biomedical Sciences (Yuhong Huang, L.W., Yu Huang), City University of Hong Kong, Hong Kong, China
| | - Shu Chien
- Departments of Bioengineering and Medicine, and Institute of Engineering in Medicine, University of California, San Diego, CA (S.C.)
| | - Yu Huang
- School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China (M.J., H.D., C.W.L., Y.X., X.Y., Yu Huang)
- Department of Biomedical Sciences (Yuhong Huang, L.W., Yu Huang), City University of Hong Kong, Hong Kong, China
- Tung Biomedical Sciences Centre (Yu Huang), City University of Hong Kong, Hong Kong, China
| |
Collapse
|
6
|
Zhu X, Yang K, Xie J, Feng X, Wu T, Hu M, Wang H, Yu C, Yu X, Hemmatzadeh F, Zhu L, Zhang L. An SDS-NaOH-based method to isolate genome of recombinant adeno-associated virus vectors for physical titer measurement. PLoS One 2025; 20:e0315921. [PMID: 40179116 PMCID: PMC11967980 DOI: 10.1371/journal.pone.0315921] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Accepted: 12/03/2024] [Indexed: 04/05/2025] Open
Abstract
Recombinant adeno-associated viruses (rAAVs) vectors are promising for their safety and sustained expression of genetic payloads across various tissues. These vectors consist of a protein capsid enclosing a 4.7 kb single-stranded DNA genome. Rapid and accurate determination of the physical titers of rAAV vector is crucial for quality control in rAAV manufacturing and precise drug dosage in clinical trials. To prepare vector DNA for genome titer assessment, it is essential to completely degrade unencapsulated DNA and dissociate the capsid. Conventional methods typically involve co-incubation with DNase I to degrade unencapsidated DNA, followed by co-incubation with Proteinase K to cleave protein shells. Here, we present a "Benzonase & SDS-NaOH" pretreatment as an effective alkaline lysis for releasing the vector DNA. In the presence of producer cell crude extract, Benzonase demonstrated superior efficacy in degrading unencapsidated DNA compared to DNase I. Additionally, the use of SDS-NaOH, effective at 65 °C for 30 min, significantly reduces the time required compared to that of Proteinase K at 56 °C for 2 hours. We also showed that the "Benzonase & SDS-NaOH" pretreatment is applicable for vector genome titration in rAAV production, harvest, and purified stock. Moreover, our method is effective for both scAAV and ssAAV forms and across all serotypes, including the thermally stable rAAV5. Overall, this method offers a rapid and straightforward solution to determine rAAV vector genome titers in both purified preparations and during the manufacturing process.
Collapse
Affiliation(s)
- Xiangying Zhu
- Zhejiang Hengyu Biological Technology Co., Ltd., Jiaxing, Zhejiang, China
| | - Keying Yang
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Fudan University, Shanghai, China
| | - Jinyan Xie
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Fudan University, Shanghai, China
| | - Xilin Feng
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Fudan University, Shanghai, China
| | - Tao Wu
- Zhejiang Hengyu Biological Technology Co., Ltd., Jiaxing, Zhejiang, China
| | - Mengjun Hu
- Zhejiang Hengyu Biological Technology Co., Ltd., Jiaxing, Zhejiang, China
| | - Haijian Wang
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Chenghui Yu
- State Key Laboratory of Genetic Engineering and Engineering Research Center of Gene Technology (Ministry of Education), School of Life Sciences, Fudan University, Shanghai, China
| | - Xiaomin Yu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Farhid Hemmatzadeh
- School of Animal and Veterinary Sciences, The University of Adelaide, Adelaide, South Australia, Australia
| | - Liqing Zhu
- Department of Clinical Laboratory, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- Department of Clinical Laboratory, Peking University Cancer Hospital and Institute, Beijing, China
| | - Linhua Zhang
- Department of Clinical Laboratory, The People’s Hospital of Yuhuan, Taizhou, Zhejiang, China
| |
Collapse
|
7
|
Hoffmann M, Sorensen RJ, Extross A, He Y, Schmidt D. Protein Carrier Adeno-Associated Virus. ACS NANO 2025; 19:12308-12322. [PMID: 40117458 PMCID: PMC11966780 DOI: 10.1021/acsnano.5c01498] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/10/2025] [Accepted: 03/11/2025] [Indexed: 03/23/2025]
Abstract
Adeno-associated virus (AAV) has emerged as a leading platform for gene therapy, enabling the delivery of therapeutic DNA to target cells. However, the potential of AAV to deliver protein payloads has been unexplored. In this study, we engineered a protein carrier AAV (pcAAV) to package and deliver proteins by inserting binding domains on the interior capsid surface. These binding domains mediate the packaging of specific target proteins through interaction with cognate peptides or protein tags during the capsid assembly process. We demonstrate the packaging of multiple proteins, including green fluorescent protein, Streptococcus pyogenes Cas9, Cre recombinase, and the engineered peroxidase APEX2. Packaging efficiency is modulated by the binding domain insertion site, the viral protein isoform containing the binding domain, and the subcellular localization of the target protein. We show that pcAAV can enter cells and deliver the protein payload and that enzymes retain their activity after packaging. Importantly, this protein packaging capability can be translated to multiple AAV serotypes. Our work establishes AAV as a protein delivery vehicle, significantly expanding the utility of this viral vector for biomedical applications.
Collapse
Affiliation(s)
- Mareike
Daniela Hoffmann
- Department
of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ryan James Sorensen
- Department
of Biochemistry, Molecular Biology and Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ajay Extross
- Department
of Molecular, Cellular, Developmental Biology, and Genetics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Yungui He
- Department
of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Daniel Schmidt
- Department
of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota 55455, United States
| |
Collapse
|
8
|
Gliwa K, Hull J, Kansol A, Zembruski V, Lakshmanan R, Mietzsch M, Chipman P, Bennett A, McKenna R. Biophysical and structural insights into AAV genome ejection. J Virol 2025; 99:e0089924. [PMID: 39907279 PMCID: PMC11915859 DOI: 10.1128/jvi.00899-24] [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: 05/22/2024] [Accepted: 10/09/2024] [Indexed: 02/06/2025] Open
Abstract
Recombinant adeno-associated virus (rAAV) is comprised of non-enveloped capsids that can package a therapeutic transgene and are currently being developed and utilized as gene therapy vectors. The therapeutic efficiency of rAAV is dependent on successful cytoplasmic trafficking and transgene delivery to the nucleus. It is hypothesized that an increased understanding of the effects of the cellular environment and biophysical properties of the capsid as it traffics to the nucleus could provide insight to improve vector efficiency. The AAV capsid is exposed to increasing [H+] during endo-lysosomal trafficking. Exposure to low pH facilitates the externalization of the viral protein 1 unique region (VP1u). This VP1u contains a phospholipase A2 domain required for endosomal escape and nuclear localization signals that facilitate nuclear targeting and entry. The viral genome is released either after total capsid disassembly or via a concerted DNA ejection mechanism in the nucleus. This study presents the characterization of genome ejection (GE) for two diverse serotypes, AAV2 and AAV5, using temperature. The temperature required to disassemble the virus capsid (TM) is significantly higher than the temperature required to expose the transgene (TE) for both serotypes. This was verified by quantitative PCR (qPCR) and transmission electron microscopy. Additionally, the absence of VP1/VP2 in the capsids and a decrease in pH increase the temperature of GE. Furthermore, cryo-electron microscopy structures of the AAV5 capsid pre- and post-GE reveal dynamics at the twofold, threefold, and fivefold regions of the capsid interior consistent with a concerted egress of the viral genome.IMPORTANCEThe development of recombinant adeno-associated virus (rAAV) capsids has grown rapidly in recent years, with five of the eight established therapeutics gaining approval in the past 2 years alone. Clinical progression with AAV2 and AAV5 represents a growing need to further characterize the molecular biology of these viruses. The goal of AAV-based gene therapy is to treat monogenic disorders with a vector-delivered transgene to provide wild-type protein function. A better understanding of the dynamics and conditions enabling transgene release may improve therapeutic efficiency. In addition to their clinical importance, AAV2 and 5 were chosen in this study for their diverse antigenic and biophysical properties compared to more closely related serotypes. Characterization of a shared genome ejection process may imply a conserved mechanism for all rAAV therapies.
Collapse
Affiliation(s)
- Keely Gliwa
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Joshua Hull
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Austin Kansol
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Victoria Zembruski
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Renuk Lakshmanan
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Mario Mietzsch
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Paul Chipman
- ICBR Electron Microscopy Core Facility, University of Florida, Gainesville, Florida, USA
| | - Antonette Bennett
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| | - Robert McKenna
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
9
|
Inagaki T, Espera J, Wang KH, Komaki S, Nair S, Davis RR, Kumar A, Nakajima KI, Izumiya Y. Design, development, and evaluation of gene therapeutics specific to KSHV-associated diseases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639178. [PMID: 40027700 PMCID: PMC11870588 DOI: 10.1101/2025.02.19.639178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the causative agent of Kaposi's sarcoma (KS) and two human lymphoproliferative diseases: primary effusion lymphoma and AIDS-related multicentric Castleman's disease. KSHV-encoded latency-associated nuclear antigen (LANA) is expressed in KSHV-infected cancer cells and is responsible for maintaining viral genomes in infected cells. Thus, LANA is an attractive target for therapeutic intervention for KSHV-associated diseases. Here, we devised a cancer gene therapy vector using the adeno-associated virus (AAV), which capitalizes the LANA's function to maintain terminal repeat (TR) containing circular genome in latently infected cells and the TR's enhancer function for KSHV inducible gene promoters. By including two TR copies with a lytic inducible gene promoter (TR2 -OriP ), we prepared an AAV vector, which expresses an engineered thymidine kinase (TK) selectively in KSHV-infected cells. Ganciclovir (GCV), an anti-herpesvirus drug, effectively eradicated multiple KSHV-infected cells that include iPSC-derived epithelial colony-forming cells, but not non-KSHV-infected counterparts in the presence of AAV8-TR2 -OriP -TK. In addition, AAV8-TR2 -OriP -TK prevents KSHV virion production from reactivated cells, spreading KSHV infections from reactivated cells. Anti-cancer drugs, known to reactivate KSHV, stimulated TK expression from the vector and, therefore, synergized with AAV8 TR2 -OriP -TK to induce KSHV-infected cancer cell death. Finally, the AAV8-TR2 -OriP -TK with GCV completely diminished KSHV-infected cancer cells in the xenograft tumor model. The new cancer gene therapeutics should augment the current clinical protocol for KS.
Collapse
Affiliation(s)
- Tomoki Inagaki
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, USA
| | - Jonna Espera
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, USA
| | - Kang-Hsin Wang
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, USA
| | - Somayeh Komaki
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, USA
| | - Sonali Nair
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, USA
- Department of Medical Health Science, Touro University California, Vallejo, California. USA
| | - Ryan R. Davis
- Department of Pathology and Laboratory Medicine, School of Medicine, UC Davis, Sacramento, California, USA
| | - Ashish Kumar
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, USA
| | - Ken-ichi Nakajima
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, USA
| | - Yoshihiro Izumiya
- Department of Dermatology, School of Medicine, the University of California Davis (UC Davis), Sacramento, California, USA
- Department of Biochemistry and Molecular Medicine, School of Medicine, UC Davis, Sacramento, California, USA
- UC Davis Comprehensive Cancer Center, Sacramento, California, USA
| |
Collapse
|
10
|
Kirk RW, Sun L, Xiao R, Clark EA, Nelson S. Multiplexed CRISPRi Reveals a Transcriptional Switch Between KLF Activators and Repressors in the Maturing Neocortex. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.07.636951. [PMID: 39975013 PMCID: PMC11839100 DOI: 10.1101/2025.02.07.636951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
A critical phase of mammalian brain development takes place after birth. Neurons of the mouse neocortex undergo dramatic changes in their morphology, physiology, and synaptic connections during the first postnatal month, while properties of immature neurons, such as the capacity for robust axon outgrowth, are lost. The genetic and epigenetic programs controlling prenatal development are well studied, but our understanding of the transcriptional mechanisms that regulate postnatal neuronal maturation is comparatively lacking. By integrating chromatin accessibility and gene expression data from two subtypes of neocortical pyramidal neurons in the neonatal and maturing brain, we predicted a role for the Krüppel-Like Factor (KLF) family of Transcription Factors in the developmental regulation of neonatally expressed genes. Using a multiplexed CRISPR Interference (CRISPRi) knockdown strategy, we found that a shift in expression from KLF activators (Klf6, Klf7) to repressors (Klf9, Klf13) during early postnatal development functions as a transcriptional 'switch' to first activate, then repress a set of shared targets with cytoskeletal functions including Tubb2b and Dpysl3. We demonstrate that this switch is buffered by redundancy between KLF paralogs, which our multiplexed CRISPRi strategy is equipped to overcome and study. Our results indicate that competition between activators and repressors within the KLF family regulates a conserved component of the postnatal maturation program that may underlie the loss of intrinsic axon growth in maturing neurons. This could facilitate the transition from axon growth to synaptic refinement required to stabilize mature circuits.
Collapse
Affiliation(s)
- Ryan W Kirk
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Liwei Sun
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Ruixuan Xiao
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Erin A Clark
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| | - Sacha Nelson
- Department of Biology, Brandeis University, Waltham, MA 02453, USA
| |
Collapse
|
11
|
Zwi-Dantsis L, Mohamed S, Massaro G, Moeendarbary E. Adeno-Associated Virus Vectors: Principles, Practices, and Prospects in Gene Therapy. Viruses 2025; 17:239. [PMID: 40006994 PMCID: PMC11861813 DOI: 10.3390/v17020239] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 01/27/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Gene therapy offers promising potential as an efficacious and long-lasting therapeutic option for genetic conditions, by correcting defective mutations using engineered vectors to deliver genetic material to host cells. Among these vectors, adeno-associated viruses (AAVs) stand out for their efficiency, versatility, and safety, making them one of the leading platforms in gene therapy. The enormous potential of AAVs has been demonstrated through their use in over 225 clinical trials and the FDA's approval of six AAV-based gene therapy products, positioning these vectors at the forefront of the field. This review highlights the evolution and current applications of AAVs in gene therapy, focusing on their clinical successes, ongoing developments, and the manufacturing processes required for the rapid commercial growth anticipated in the AAV therapy market. It also discusses the broader implications of these advancements for future therapeutic strategies targeting more complex and multi-systemic conditions and biological processes such as aging. Finally, we explore some of the major challenges currently confronting the field.
Collapse
Affiliation(s)
- Limor Zwi-Dantsis
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Saira Mohamed
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| | - Giulia Massaro
- UCL School of Pharmacy, University College London, London WC1N 1AX, UK
| | - Emad Moeendarbary
- Department of Mechanical Engineering, Roberts Building, University College London, London WC1E 6BT, UK
| |
Collapse
|
12
|
Sedorovitz M, Byrne LC, Betegon M. Chromatographic Purification and Polishing of AAV Particles. Methods Mol Biol 2025; 2848:249-257. [PMID: 39240527 DOI: 10.1007/978-1-0716-4087-6_15] [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: 09/07/2024]
Abstract
The production of Adeno-associated virus (AAV) vectors in the lab setting has typically involved expression in adherent cells followed by purification through ultracentrifugation in density gradients. This production method is, however, not easily scalable, presents high levels of cellular impurities that co-purify with the virus, and results in a mixture of empty and full capsids. Here we describe a detailed AAV production protocol that overcomes these limitations through AAV expression in suspension cells followed by AAV affinity purification and AAV polishing to separate empty and full capsids, resulting in high yields of ultra-pure AAV that is highly enriched in full capsids.
Collapse
Affiliation(s)
- Morgan Sedorovitz
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
- Avista Therapeutics, Pittsburgh, PA, USA
| | - Leah C Byrne
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Miguel Betegon
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA, USA.
| |
Collapse
|
13
|
Oestreicher D, Chepurwar S, Kusch K, Rankovic V, Jung S, Strenzke N, Pangrsic T. CaBP1 and 2 enable sustained Ca V1.3 calcium currents and synaptic transmission in inner hair cells. eLife 2024; 13:RP93646. [PMID: 39718549 DOI: 10.7554/elife.93646] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2024] Open
Abstract
To encode continuous sound stimuli, the inner hair cell (IHC) ribbon synapses utilize calcium-binding proteins (CaBPs), which reduce the inactivation of their CaV1.3 calcium channels. Mutations in the CABP2 gene underlie non-syndromic autosomal recessive hearing loss DFNB93. Besides CaBP2, the structurally related CaBP1 is highly abundant in the IHCs. Here, we investigated how the two CaBPs cooperatively regulate IHC synaptic function. In Cabp1/2 double-knockout mice, we find strongly enhanced CaV1.3 inactivation, slowed recovery from inactivation and impaired sustained exocytosis. Already mild IHC activation further reduces the availability of channels to trigger synaptic transmission and may effectively silence synapses. Spontaneous and sound-evoked responses of spiral ganglion neurons in vivo are strikingly reduced and strongly depend on stimulation rates. Transgenic expression of CaBP2 leads to substantial recovery of IHC synaptic function and hearing sensitivity. We conclude that CaBP1 and 2 act together to suppress voltage- and calcium-dependent inactivation of IHC CaV1.3 channels in order to support sufficient rate of exocytosis and enable fast, temporally precise and indefatigable sound encoding.
Collapse
Affiliation(s)
- David Oestreicher
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
| | - Shashank Chepurwar
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Kathrin Kusch
- Functional Auditory Genomics, Institute for Auditory Neuroscience, University Medical Center Göttingen, Göttingen, Germany
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Vladan Rankovic
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Restorative Cochlear Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Cente, Göttingen, Germany
| | - Sangyong Jung
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
| | - Nicola Strenzke
- Auditory Systems Physiology Group, Institute for Auditory Neuroscience, InnerEarLab, University Medical Center Göttingen, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
| | - Tina Pangrsic
- Experimental Otology Group, InnerEarLab, Department of Otolaryngology, University Medical Center Göttingen, Göttingen, Germany
- Auditory Neuroscience Group, Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Collaborative Research Center 889, University of Göttingen, Göttingen, Germany
- Multiscale Bioimaging Cluster of Excellence (MBExC), University of Göttingen, Göttingen, Germany
| |
Collapse
|
14
|
Lam P, Zygmunt DA, Ashbrook A, Yan C, Du H, Martin PT. Liver-directed AAV gene therapy normalizes disease symptoms and provides cross-correction in a model of lysosomal acid lipase deficiency. Mol Ther 2024; 32:4272-4284. [PMID: 39489913 PMCID: PMC11638878 DOI: 10.1016/j.ymthe.2024.10.022] [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: 06/20/2024] [Revised: 09/11/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Lysosomal acid lipase deficiency (LAL-D) is caused by mutations in the LIPA gene, which encodes the lysosomal enzyme that hydrolyzes triglycerides and cholesteryl esters to free fatty acids and free cholesterol. The objective of this study was to develop a curative single-treatment therapy for LAL-D using adeno-associated virus (AAV). Treatment at both early (1-2 days) and late (8-week) timepoints with rscAAVrh74.LP1.LIPA, a liver-directed AAV gene therapy, normalized many disease measures in Lipa-/- mice when measured at 24 weeks of age, including hepatosplenomegaly, serum transaminase activity, organ triglyceride and cholesterol levels, and biomarkers of liver inflammation and fibrosis. For most measures, liver-directed therapy was superior to therapy utilizing a constitutive tissue expression approach. rscAAVrh74.LP1.LIPA treatment elevated LAL enzyme activity above wild-type levels in all tissues tested, including liver, spleen, intestine, muscle, and brain, and treatment elicited minimal serum antibody responses to transgenic protein. AAV treatment at 8 weeks of age with 1 × 1013 vg/kg extended survival significantly, with all AAV-treated mice surviving beyond the maximal lifespan of untreated Lipa-/- mice. These results show that this liver-directed LIPA gene therapy has the potential to be a transformative treatment for LAL-D.
Collapse
Affiliation(s)
- Patricia Lam
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Deborah A Zygmunt
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Anna Ashbrook
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA
| | - Cong Yan
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Hong Du
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Paul T Martin
- Center for Gene Therapy, Abigail Wexner Research Institute, Nationwide Children's Hospital, Columbus, OH 43215, USA; Department of Pediatrics, The Ohio State University School of Medicine, Columbus, OH 43210, USA.
| |
Collapse
|
15
|
Hussein MN. Labeling of the serotonergic neuronal circuits emerging from the raphe nuclei via some retrograde tracers. Microsc Res Tech 2024; 87:2894-2914. [PMID: 39041701 DOI: 10.1002/jemt.24662] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 06/20/2024] [Accepted: 07/11/2024] [Indexed: 07/24/2024]
Abstract
Serotonin (5-hydroxytryptamine, 5-HT) is a very important neurotransmitter emerging from the raphe nuclei to several brain regions. Serotonergic neuronal connectivity has multiple functions in the brain. In this study, several techniques were used to trace serotonergic neurons in the dorsal raphe (DR) and median raphe (MnR) that project toward the arcuate nucleus of the hypothalamus (Arc), dorsomedial hypothalamic nucleus (DM), lateral hypothalamic area (LH), paraventricular hypothalamic nucleus (PVH), ventromedial hypothalamic nucleus (VMH), fasciola cinereum (FC), and medial habenular nucleus (MHb). Cholera toxin subunit B (CTB), retro-adeno-associated virus (rAAV-CMV-mCherry), glycoprotein-deleted rabies virus (RV-ΔG), and simultaneous microinjection of rAAV2-retro-Cre-tagBFP with AAV-dio-mCherry in C57BL/6 mice were used in this study. In addition, rAAV2-retro-Cre-tagBFP was microinjected into Ai9 mice. Serotonin immunohistochemistry was used for the detection of retrogradely traced serotonergic neurons in the raphe nuclei. The results indicated that rAAV2-retro-Cre-tagBFP microinjection in Ai9 mice was the best method for tracing serotonergic neuron circuits. All of the previously listed nuclei exhibited serotonergic neuronal projections from the DR and MnR, with the exception of the FC, which had very few projections from the DR. The serotonergic neuronal projections were directed toward the Arc by the subpeduncular tegmental (SPTg) nuclei. Moreover, the RV-ΔG tracer revealed monosynaptic non-serotonergic neuronal projections from the DR that were directed toward the Arc. Furthermore, rAAV tracers revealed monosynaptic serotonergic neuronal connections from the raphe nuclei toward Arc. These findings validate the variations in neurotropism among several retrograde tracers. The continued discovery of several novel serotonergic neural circuits is crucial for the future discovery of the functions of these circuits. RESEARCH HIGHLIGHTS: Various kinds of retrograde tracers were microinjected into C57BL/6 and Ai9 mice. The optimum method for characterizing serotonergic neuronal circuits is rAAV2-retro-Cre-tagBFP microinjection in Ai9 mice. The DR, MnR, and SPTg nuclei send monosynaptic serotonergic neuronal projections toward the arcuate nucleus of the hypothalamus. Whole-brain quantification analysis of retrograde-labeled neurons in different brain nuclei following rAAV2-retro-Cre-tagBFP microinjection in the Arc, DM, LH, and VMH is shown. Differential quantitative analysis of median and dorsal raphe serotonergic neurons emerging toward the PVH, DM, LH, Arc, VMH, MHb, and FC is shown.
Collapse
Affiliation(s)
- Mona N Hussein
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan, China
- College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China
- Histology and Cytology Department, Faculty of Veterinary Medicine, Benha University, Benha, Egypt
| |
Collapse
|
16
|
Shih FH, Chang HH, Wang YC. Utilizing adeno-associated virus as a vector in treating genetic disorders or human cancers. IUBMB Life 2024; 76:1000-1010. [PMID: 38970351 DOI: 10.1002/iub.2896] [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: 06/01/2024] [Accepted: 06/17/2024] [Indexed: 07/08/2024]
Abstract
Clinical data from over two decades, involving more than 3000 treated patients, demonstrate that adeno-associated virus (AAV) gene therapy is a safe, effective, and well-tolerated therapeutic method. Clinical trials using AAV-mediated gene delivery to accessible tissues have led to successful treatments for numerous monogenic disorders and advancements in tissue engineering. Although the US Food and Drug Administration (FDA) has approved AAV for clinical use, systemic administration remains a significant challenge. In this review, we delve into AAV biology, focusing on current manufacturing technologies and transgene engineering strategies. We examine the use of AAVs in ongoing clinical trials for ocular, neurological, and hematological disorders, as well as cancers. By discussing recent advancements and current challenges in the field, we aim to provide valuable insights for researchers and clinicians navigating the evolving landscape of AAV-based gene therapy.
Collapse
Affiliation(s)
- Fu-Hsuan Shih
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Hsiung-Hao Chang
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| | - Yi-Ching Wang
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
- Department of Pharmacology, College of Medicine, National Cheng Kung University, Tainan, Taiwan, ROC
| |
Collapse
|
17
|
Lkharrazi A, Tobler K, Marti S, Bratus-Neuenschwander A, Vogt B, Fraefel C. AAV2 can replicate its DNA by a rolling hairpin or rolling circle mechanism, depending on the helper virus. J Virol 2024; 98:e0128224. [PMID: 39382273 PMCID: PMC11575299 DOI: 10.1128/jvi.01282-24] [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: 07/26/2024] [Accepted: 09/13/2024] [Indexed: 10/10/2024] Open
Abstract
Adeno-associated virus type 2 (AAV2) is a small, non-pathogenic, helper virus-dependent parvovirus with a single-stranded (ss) DNA genome of approximately 4.7 kb. AAV2 DNA replication requires the presence of a helper virus such as adenovirus type 5 (AdV5) or herpes simplex virus type 1 (HSV-1) and is generally assumed to occur as a strand-displacement rolling hairpin (RHR) mechanism initiated at the AAV2 3' inverted terminal repeat (ITR). We have recently shown that AAV2 replication supported by HSV-1 leads to the formation of double-stranded head-to-tail concatemers, which provides evidence for a rolling circle replication (RCR) mechanism. We have revisited AAV2 DNA replication and specifically compared the formation of AAV2 replication intermediates in the presence of either HSV-1 or AdV5 as the helper virus. The results confirmed that the AAV2 DNA replication mechanism is helper virus-dependent and follows a strand-displacement RHR mechanism when AdV5 is the helper virus and primarily an RCR mechanism when HSV-1 is the helper virus. We also demonstrate that recombination plays a negligible role in AAV2 genome replication. Interestingly, the formation of high-molecular-weight AAV2 DNA concatemers in the presence of HSV-1 as the helper virus was dependent on an intact HSV-1 DNA polymerase. IMPORTANCE AAV is a small helper virus-dependent, non-pathogenic parvovirus. The AAV genome replication mechanism was extensively studied in the presence of AdV as the helper virus and described to proceed using RHR. Surprisingly, HSV-1 co-infection facilitates RCR of the AAV2 DNA. We directly compared AdV5 and HSV-1 supported AAV2 DNA replication and showed that AAV2 can adapt its replication mechanism to the helper virus. A detailed understanding of the AAV replication mechanism expands our knowledge of virus biology and can contribute to increase gene therapy vector production.
Collapse
Affiliation(s)
- Anouk Lkharrazi
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Kurt Tobler
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Sara Marti
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | | | - Bernd Vogt
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Cornel Fraefel
- Institute of Virology, University of Zurich, Zurich, Switzerland
| |
Collapse
|
18
|
Lee H, Assaraf R, Subramanian S, Goetschius D, Bieri J, DiNunno NM, Leisi R, Bator CM, Hafenstein SL, Ros C. Infectious parvovirus B19 circulates in the blood coated with active host protease inhibitors. Nat Commun 2024; 15:9543. [PMID: 39500886 PMCID: PMC11538491 DOI: 10.1038/s41467-024-53794-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Accepted: 10/20/2024] [Indexed: 11/08/2024] Open
Abstract
The lack of a permissive cell culture system has limited high-resolution structures of parvovirus B19 (B19V) to virus-like particles (VLPs). In this study, we present the atomic resolution structure (2.2 Å) of authentic B19V purified from a patient blood sample. There are significant differences compared to non-infectious VLPs. Most strikingly, two host protease inhibitors (PIs), inter-alpha-trypsin inhibitor heavy chain 4 (ITIH4) and serpinA3, were identified in complex with the capsids in all patient samples tested. The ITIH4 binds specifically to the icosahedral fivefold axis and serpinA3 occupies the twofold axis. The protein-coated virions remain infectious, and the capsid-associated PIs retain activity; however, upon virion interaction with target cells, the PIs dissociate from the capsid prior to viral entry. Our finding of an infectious virion shielded by bound host serum proteins suggests an evolutionarily favored phenomenon to evade immune surveillance and escape host protease activity.
Collapse
Affiliation(s)
- Hyunwook Lee
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Ruben Assaraf
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, Bern, Switzerland
| | | | - Dan Goetschius
- The Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Jan Bieri
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Nadia M DiNunno
- The Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Remo Leisi
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland
| | - Carol M Bator
- The Hormel Institute, University of Minnesota, Austin, MN, USA
| | - Susan L Hafenstein
- The Hormel Institute, University of Minnesota, Austin, MN, USA.
- Department of Biochemistry, Biophysics and Molecular Biology, University of Minnesota, Minneapolis, MN, USA.
- Department of Infectious Diseases, Mayo Clinic, Rochester, MN, USA.
| | - Carlos Ros
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, University of Bern, Bern, Switzerland.
| |
Collapse
|
19
|
Dell'Aquila F, Di Cunto R, Marrocco E, Del Prete E, D'Alessio A, De Stefano L, Notaro S, Nusco E, Auricchio A. Combined intraocular and intravenous gene delivery for therapy of gyrate atrophy of the choroid and retina. Mol Ther 2024:S1525-0016(24)00680-4. [PMID: 39489914 DOI: 10.1016/j.ymthe.2024.10.019] [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: 07/07/2024] [Revised: 09/21/2024] [Accepted: 10/23/2024] [Indexed: 11/05/2024] Open
Abstract
Gyrate atrophy of the choroid and retina (GACR) is due to ornithine aminotransferase (OAT) deficiency, which causes hyperornithinemia, leading to retinal pigment epithelium, followed by choroidal and retinal degeneration. Adeno-associated virus serotype 8 (AAV8) vector-mediated OAT (AAV8-OAT) liver gene transfer reduces ornithinemia in the Oat-/- mouse model of GACR and improves retinal function and structure. Since OAT is expressed in various tissues including the retina, we investigated the efficacy of restoration of OAT expression in either retina or liver or both tissues on the retinal phenotype of Oat-/- mice. Intravenous and subretinal administration of AAV8-OAT resulted in intraocular and liver OAT expression with reduced ornithinemia after intravenous AAV8-OAT administration, while intraocular ornithine levels were significantly reduced only following combined gene delivery. Accordingly, only Oat-/- animals treated with combined intravenous and subretinal AAV8-OAT administrations showed significant improvements in both retinal morphology and function. This work shows the benefits of combined liver and retinal OAT supplementation for the treatment of GACR.
Collapse
Affiliation(s)
- Fabio Dell'Aquila
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Gene Therapy Joint Lab, Department of Advanced Biomedical Sciences and Department of Translational Medicine, "Federico II" University, 80131 Naples, Italy
| | - Roberto Di Cunto
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, "Federico II" University, 80131 Naples, Italy
| | - Elena Marrocco
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Eugenio Del Prete
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Alfonso D'Alessio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, "Federico II" University, 80131 Naples, Italy
| | - Lucia De Stefano
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Simone Notaro
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Edoardo Nusco
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy
| | - Alberto Auricchio
- Telethon Institute of Genetics and Medicine (TIGEM), 80078 Pozzuoli, Italy; Gene Therapy Joint Lab, Department of Advanced Biomedical Sciences and Department of Translational Medicine, "Federico II" University, 80131 Naples, Italy; Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, "Federico II" University, 80131 Naples, Italy.
| |
Collapse
|
20
|
Liu X, Jean-Gilles R, Baginski J, Cai C, Yan R, Zhang L, Lance K, van der Loo JC, Davidson BL. Evaluation of a rapid multi-attribute combinatorial high-throughput UV-Vis/DLS/SLS analytical platform for rAAV quantification and characterization. Mol Ther Methods Clin Dev 2024; 32:101298. [PMID: 39170800 PMCID: PMC11338085 DOI: 10.1016/j.omtm.2024.101298] [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: 03/11/2024] [Accepted: 07/12/2024] [Indexed: 08/23/2024]
Abstract
Recombinant adeno-associated virus (rAAV)-based gene therapies are expanding in their application. Despite progress in manufacturing, current analytical methods for product quantification and characterization remain largely unchanged. Although critical for product and process development, in-process testing, and batch release, current analytical methods are labor-intensive, costly, and hampered by extended turnaround times and low throughput. The field requires more efficient, cost-effective analytical techniques capable of handling large sample quantities to accelerate product and process development. Here, we evaluated Stunner from Unchained Labs for quantifying and characterizing rAAVs and compared it with established analytical methods. Stunner is a combinatorial analytic technology platform that interpolates ultraviolet-visible (UV-Vis) absorption with static and dynamic light scattering (SLS/DLS) analysis to determine capsid and genomic titer, empty and full capsid ratio, and assess vector size and polydispersity. The platform offers empirical measurements with minimal sample requirements. Upon testing hundreds of rAAV vectors, comprising various serotypes and transgenes, the data show a strong correlation with established analytical methods and exhibit high reproducibility and repeatability. Some analyses can be applied to in-process samples from different purification stages and processes, fulfilling the demand for rapid, high-throughput analysis during development. In sum, the pipeline presented streamlines small- and large-batch analytics.
Collapse
Affiliation(s)
- Xueyuan Liu
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- School of Biomedical Engineering, Science and Health Systems, Drexel University, Philadelphia, PA 19104, USA
| | | | - Julia Baginski
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Christina Cai
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ruilan Yan
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Lili Zhang
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | | | - Johannes C.M. van der Loo
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Beverly L. Davidson
- Raymond G. Perelman Center for Cellular and Molecular Therapeutics, The Children’s Hospital of Philadelphia, Philadelphia, PA 19104, USA
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
21
|
Almeida RA, Ferreira CG, Matos VUS, Nogueira JM, Braga MP, Caldi Gomes L, Jorge EC, Soriani FM, Michel U, Ribas VT. AAV-Mediated Expression of miR-17 Enhances Neurite and Axon Regeneration In Vitro. Int J Mol Sci 2024; 25:9057. [PMID: 39201743 PMCID: PMC11355044 DOI: 10.3390/ijms25169057] [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: 07/10/2024] [Revised: 08/07/2024] [Accepted: 08/08/2024] [Indexed: 09/03/2024] Open
Abstract
Neurodegenerative disorders, including traumatic injuries to the central nervous system (CNS) and neurodegenerative diseases, are characterized by early axonal damage, which does not regenerate in the adult mammalian CNS, leading to permanent neurological deficits. One of the primary causes of the loss of regenerative ability is thought to be a developmental decline in neurons' intrinsic capability for axon growth. Different molecules are involved in the developmental loss of the ability for axon regeneration, including many transcription factors. However, the function of microRNAs (miRNAs), which are also modulators of gene expression, in axon re-growth is still unclear. Among the various miRNAs recently identified with roles in the CNS, miR-17, which is highly expressed during early development, emerges as a promising target to promote axon regeneration. Here, we used adeno-associated viral (AAV) vectors to overexpress miR-17 (AAV.miR-17) in primary cortical neurons and evaluate its effects on neurite and axon regeneration in vitro. Although AAV.miR-17 had no significant effect on neurite outgrowth and arborization, it significantly enhances neurite regeneration after scratch lesion and axon regeneration after axotomy of neurons cultured in microfluidic chambers. Target prediction and functional annotation analyses suggest that miR-17 regulates gene expression associated with autophagy and cell metabolism. Our findings suggest that miR-17 promotes regenerative response and thus could mitigate neurodegenerative effects.
Collapse
Affiliation(s)
- Raquel Alves Almeida
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Carolina Gomes Ferreira
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Victor Ulysses Souza Matos
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Julia Meireles Nogueira
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Marina Pimenta Braga
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31279-901, Brazil (F.M.S.)
| | - Lucas Caldi Gomes
- Clinical Department of Neurology, TUM School of Medicine, Rechts der Isar Hospital, Technical University of Munich, 81675 Munich, Germany;
| | - Erika Cristina Jorge
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| | - Frederico Marianetti Soriani
- Department of Genetics, Ecology and Evolution, Institute of Biological Sciences, Federal University of Minas Gerais, Belo Horizonte 31279-901, Brazil (F.M.S.)
| | - Uwe Michel
- Department of Neurology, University Medicine Göttingen, 37075 Göttingen, Germany
| | - Vinicius Toledo Ribas
- Department of Morphology, Institute of Biological Sciences, Federal University of Minas Gerais, Av. Pres. Antônio Carlos, 6627, Belo Horizonte 31279-901, Brazil (E.C.J.)
| |
Collapse
|
22
|
Shipulin GA, Glazkova DV, Urusov FA, Belugin BV, Dontsova V, Panova AV, Borisova AA, Tsyganova GM, Bogoslovskaya EV. Triple Combinations of AAV9-Vectors Encoding Anti-HIV bNAbs Provide Long-Term In Vivo Expression of Human IgG Effectively Neutralizing Pseudoviruses from HIV-1 Global Panel. Viruses 2024; 16:1296. [PMID: 39205270 PMCID: PMC11359378 DOI: 10.3390/v16081296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Anti-human immunodeficiency virus (HIV) broadly neutralizing antibodies (bNAbs) offer a promising approach for the treatment of HIV-1. The current paradigm for antibody therapy involves passive antibody transfer, requiring regular delivery of bNAbs in treating chronic diseases such as HIV-1. An alternative strategy is to use AAV-mediated gene transfer to enable in vivo production of desirable anti-HIV-1 antibodies. In this study, we investigated two sets of triple combinations of AAV9-vectors encoding different bNAbs: N6, 10E8, 10-1074 (CombiMab1), and VRC07-523, PGDM1400, 10-1074 (CombiMab2). We used CBAxC57Bl and C57BL/6 mouse models to characterize rAAV-induced antibody expression and to evaluate the neutralization capacity of mouse sera against a global panel of HIV-1 viral strains. rAAV9-mediated IgG expression varied between bNAb clones and mouse strains, with C57BL/6 mice exhibiting higher bNAb titers following rAAV delivery. Although CombiMab2 treatment elicited a higher IgG titer than CombiMab1, both combinations resulted in neutralization of all the viral strains from the global HIV-1 panel. Our data highlight the potential of AAV vectors as a long-term option for HIV-1 therapy.
Collapse
Affiliation(s)
- German A. Shipulin
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 119992 Moscow, Russia (E.V.B.)
| | - Dina V. Glazkova
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 119992 Moscow, Russia (E.V.B.)
| | - Felix A. Urusov
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 119992 Moscow, Russia (E.V.B.)
- Izmerov Research Institute of Occupational Health, 105275 Moscow, Russia
| | - Boris V. Belugin
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 119992 Moscow, Russia (E.V.B.)
| | - Valeriya Dontsova
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 119992 Moscow, Russia (E.V.B.)
| | - Alexandra V. Panova
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 119992 Moscow, Russia (E.V.B.)
| | - Alyona A. Borisova
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 119992 Moscow, Russia (E.V.B.)
| | - Galina M. Tsyganova
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 119992 Moscow, Russia (E.V.B.)
| | - Elena V. Bogoslovskaya
- Centre for Strategic Planning and Management of Biomedical Health Risks, Federal Medical Biological Agency, 119992 Moscow, Russia (E.V.B.)
| |
Collapse
|
23
|
Hoffmann MD, Sorensen RJ, Extross A, He Y, Schmidt D. Protein Carrier AAV. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.14.607995. [PMID: 39185209 PMCID: PMC11343202 DOI: 10.1101/2024.08.14.607995] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
AAV is widely used for efficient delivery of DNA payloads. The extent to which the AAV capsid can be used to deliver a protein payload is unexplored. Here, we report engineered AAV capsids that directly package proteins - Protein Carrier AAV (pcAAV). Nanobodies inserted into the interior of the capsid mediate packaging of a cognate protein, including Green Fluorescent Protein (GFP), Streptococcus pyogenes Cas9, Cre recombinase, and the engineered peroxidase APEX2. We show that protein packaging efficiency is affected by the nanobody insertion position, the capsid protein isoform into which the nanobody is inserted, and the subcellular localization of the packaged protein during recombinant AAV capsid production; each of these factors can be rationally engineered to optimize protein packaging efficiency. We demonstrate that proteins packaged within pcAAV retain their enzymatic activity and that pcAAV can bind and enter the cell to deliver the protein payload. Establishing pcAAV as a protein delivery platform may expand the utility of AAV as a therapeutic and research tool.
Collapse
Affiliation(s)
- Mareike D. Hoffmann
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Ryan J. Sorensen
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, Minnesota 55455, United States
| | - Ajay Extross
- Department of Molecular, Cellular, Developmental Biology, and Genetics
| | - Yungui He
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Daniel Schmidt
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN, 55455, USA
| |
Collapse
|
24
|
Kish WS, Lightholder J, Zeković T, Berrill A, Roach M, Wellborn WB, Vorst E. Removal of empty capsids from high-dose adeno-associated virus 9 gene therapies. Biotechnol Bioeng 2024; 121:2500-2523. [PMID: 38807330 DOI: 10.1002/bit.28737] [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: 12/08/2023] [Revised: 04/27/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024]
Abstract
Recombinant adeno-associated virus, serotype 9 (rAAV9) has shown promise as a gene therapy vector for muscle and central nervous diseases. High-dose requirements of these therapies present critical safety considerations and biomanufacturing challenges. Notably, the reduction of empty capsids (ECs), which lack therapeutic transgene, from rAAV9 products is critical to maximize efficacy. Removal of rAAV ECs from full capsids is a major downstream challenge because of their highly similar biophysical characteristics. Ultracentrifugation (UC) reduces ECs but is laborious and difficult to scale. In this paper, to replace a poorly scalable UC process, we developed an anion exchange (AEX) chromatography for rAAV9 EC reduction from full capsids. AEX load preparation by dilution incurred major product loss. The addition of histidine and surfactants to dilution buffers increased yield and reduced aggregation. Elution salts were screened and sodium acetate was found to maximize yield and EC reduction. The most promising load dilution buffer and elution salt were used in combination to form an optimized AEX method. The process reduced ECs three-fold, demonstrated robustness to a broad range of EC load challenges, and was scaled for large-scale manufacture. Compared to UC, the AEX method simplified scale-up, reduced ECs to comparable levels (20%), afforded similar purity and product quality, and increased yield by 14%.
Collapse
Affiliation(s)
- William S Kish
- Gene Therapy Process Development, Pfizer Inc., Morrisville, North Carolina, USA
| | - John Lightholder
- Gene Therapy Process Development, Pfizer Inc., Morrisville, North Carolina, USA
| | - Tamara Zeković
- Gene Therapy Process Development, Pfizer Inc., Morrisville, North Carolina, USA
| | - Alex Berrill
- Gene Therapy Process Development, Pfizer Inc., Chesterfield, Missouri, USA
| | - Matthew Roach
- Gene Therapy Process Development, Pfizer Inc., Morrisville, North Carolina, USA
| | - William B Wellborn
- Gene Therapy Process Development, Pfizer Inc., Chesterfield, Missouri, USA
| | - Eric Vorst
- Gene Therapy Process Development, Pfizer Inc., Morrisville, North Carolina, USA
| |
Collapse
|
25
|
Fu C, Gobbooru S, Martino AT, Low WK. Production of VP3-only virus-like particles of Adeno-associated virus 2 in E. coli cells. Protein Expr Purif 2024; 220:106502. [PMID: 38754753 DOI: 10.1016/j.pep.2024.106502] [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: 04/11/2024] [Revised: 04/24/2024] [Accepted: 05/13/2024] [Indexed: 05/18/2024]
Abstract
Adeno-associated Virus (AAV) is a promising vector for gene therapy. However, few studies have focused on producing virus-like particles (VLPs) of AAV in cells, especially in E. coli. In this study, we describe a method to produce empty VP3-only VLPs of AAV2 in E. coli by co-expressing VP3 and assembly-activating protein (AAP) of AAV2. Although the yields of VLPs produced with our method were low, the VLPs were able to self-assemble in E. coli without the need of in vitro capsid assembly. The produced VLPs were characterized by immunological detection and transmission electron microscopy (TEM). In conclusion, this study demonstrated that capsid assembly of AAV2 is possible in E. coli, and E. coli may be a candidate system for production of VLPs of AAV.
Collapse
Affiliation(s)
- Chengyu Fu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Shruthi Gobbooru
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Ashley T Martino
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA
| | - Woon-Kai Low
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA.
| |
Collapse
|
26
|
Sia KC, Fu ZY, Mohd Rodhi SH, Yee JHY, Qu K, Gan SU. Efficient AAV9 Purification Using a Single-Step AAV9 Magnetic Affinity Beads Isolation. Int J Mol Sci 2024; 25:8342. [PMID: 39125910 PMCID: PMC11313462 DOI: 10.3390/ijms25158342] [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: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Adeno-associated viruses (AAVs) have emerged as promising tools for gene therapy due to their safety and efficacy in delivering therapeutic genes or gene editing sequences to various tissues and organs. AAV serotype 9 (AAV9), among AAV serotypes, stands out for its ability to efficiently target multiple tissues, thus holding significant potential for clinical applications. However, existing methods for purifying AAVs are cumbersome, expensive, and often yield inconsistent results. In this study, we explore a novel purification strategy utilizing Dynabeads™ CaptureSelect™ magnetic beads. The AAV9 magnetic beads capture AAV9 with high specificity and recovery between 70 and 90%, whereas the AAVX magnetic beads did not bind to the AAV9. Through continuous interaction with AAVs in solution, these beads offer enhanced clearance of genomic DNA and plasmids even in the absence of endonuclease. The beads could be regenerated at least eight times, and the used beads could be stored for up to six months and reused without a significant reduction in recovery. The potency of the AAV9-purified vectors in vivo was comparable to that of iodixanol purified vectors.
Collapse
Affiliation(s)
- Kian Chuan Sia
- Phoenix Laboratory of Gene Therapy and Cell Therapy, Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD11, Basement 1, 10, Medical Drive, Singapore 117597, Singapore; (K.C.S.); (Z.Y.F.); (S.H.M.R.)
| | - Zhen Ying Fu
- Phoenix Laboratory of Gene Therapy and Cell Therapy, Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD11, Basement 1, 10, Medical Drive, Singapore 117597, Singapore; (K.C.S.); (Z.Y.F.); (S.H.M.R.)
| | - Siti Humairah Mohd Rodhi
- Phoenix Laboratory of Gene Therapy and Cell Therapy, Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD11, Basement 1, 10, Medical Drive, Singapore 117597, Singapore; (K.C.S.); (Z.Y.F.); (S.H.M.R.)
| | - Joan Hua Yi Yee
- Infectious Diseases Translational Research Programme, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore; (J.H.Y.Y.); (K.Q.)
| | - Kun Qu
- Infectious Diseases Translational Research Programme, Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117545, Singapore; (J.H.Y.Y.); (K.Q.)
| | - Shu Uin Gan
- Phoenix Laboratory of Gene Therapy and Cell Therapy, Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, MD11, Basement 1, 10, Medical Drive, Singapore 117597, Singapore; (K.C.S.); (Z.Y.F.); (S.H.M.R.)
| |
Collapse
|
27
|
Fu XQ, Zhan WR, Tian WY, Zeng PM, Luo ZG. Comparative transcriptomic profiling reveals a role for Olig1 in promoting axon regeneration. Cell Rep 2024; 43:114514. [PMID: 39002126 DOI: 10.1016/j.celrep.2024.114514] [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: 01/21/2023] [Revised: 05/21/2024] [Accepted: 06/30/2024] [Indexed: 07/15/2024] Open
Abstract
The regenerative potential of injured axons displays considerable heterogeneity. However, the molecular mechanisms underlying the heterogeneity have not been fully elucidated. Here, we establish a method that can separate spinal motor neurons (spMNs) with low and high regenerative capacities and identify a set of transcripts revealing differential expression between two groups of neurons. Interestingly, oligodendrocyte transcription factor 1 (Olig1), which regulates the differentiation of various neuronal progenitors, exhibits recurrent expression in spMNs with enhanced regenerative capabilities. Furthermore, overexpression of Olig1 (Olig1 OE) facilitates axonal regeneration in various models, and down-regulation or deletion of Olig1 exhibits an opposite effect. By analyzing the overlapped differentially expressed genes after expressing individual Olig factor and functional validation, we find that the role of Olig1 is at least partially through the neurite extension factor 1 (Nrsn1). We therefore identify Olig1 as an intrinsic factor that promotes regenerative capacity of injured axons.
Collapse
Affiliation(s)
- Xiu-Qing Fu
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| | - Wen-Rong Zhan
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Wei-Ya Tian
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Peng-Ming Zeng
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Zhen-Ge Luo
- School of Life Science and Technology and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China.
| |
Collapse
|
28
|
López-Canul M, He Q, Sasson T, Ettaoussi M, Gregorio DD, Ochoa-Sanchez R, Catoire H, Posa L, Rouleau G, Beaulieu JM, Comai S, Gobbi G. Selective Enhancement of REM Sleep in Male Rats through Activation of Melatonin MT 1 Receptors Located in the Locus Ceruleus Norepinephrine Neurons. J Neurosci 2024; 44:e0914232024. [PMID: 38744530 PMCID: PMC11255427 DOI: 10.1523/jneurosci.0914-23.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 04/03/2024] [Accepted: 04/06/2024] [Indexed: 05/16/2024] Open
Abstract
Sleep disorders affect millions of people around the world and have a high comorbidity with psychiatric disorders. While current hypnotics mostly increase non-rapid eye movement sleep (NREMS), drugs acting selectively on enhancing rapid eye movement sleep (REMS) are lacking. This polysomnographic study in male rats showed that the first-in-class selective melatonin MT1 receptor partial agonist UCM871 increases the duration of REMS without affecting that of NREMS. The REMS-promoting effects of UCM871 occurred by inhibiting, in a dose-response manner, the firing activity of the locus ceruleus (LC) norepinephrine (NE) neurons, which express MT1 receptors. The increase of REMS duration and the inhibition of LC-NE neuronal activity by UCM871 were abolished by MT1 pharmacological antagonism and by an adeno-associated viral (AAV) vector, which selectively knocked down MT1 receptors in the LC-NE neurons. In conclusion, MT1 receptor agonism inhibits LC-NE neurons and triggers REMS, thus representing a novel mechanism and target for REMS disorders and/or psychiatric disorders associated with REMS impairments.
Collapse
Affiliation(s)
- Martha López-Canul
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Qianzi He
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Tania Sasson
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Mohamed Ettaoussi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Danilo De Gregorio
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
| | - Rafael Ochoa-Sanchez
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Helene Catoire
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Luca Posa
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
| | - Guy Rouleau
- Department of Neurology and Neurosurgery, McGill University, Montreal, Quebec H3A 2B4, Canada
| | - Jean Martin Beaulieu
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, Ontario M5G 2C8, Canada
| | - Stefano Comai
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- IRCCS San Raffaele Scientific Institute, Milan 20132, Italy
- Department of Pharmaceutical and Pharmacological Sciences, University of Padua, Padua 35131, Italy
- Department of Biomedical Sciences, University of Padua, Padua 35131, Italy
| | - Gabriella Gobbi
- Neurobiological Psychiatry Unit, Department of Psychiatry, McGill University, Montreal, Quebec H3A 1A1, Canada
- Research Institute, McGill University Health Center, McGill University, Montreal, Quebec H3A 1A1, Canada
| |
Collapse
|
29
|
Hoffmann M, Gallant J, LeBeau A, Schmidt D. Unlocking precision gene therapy: harnessing AAV tropism with nanobody swapping at capsid hotspots. NAR MOLECULAR MEDICINE 2024; 1:ugae008. [PMID: 39022346 PMCID: PMC11250487 DOI: 10.1093/narmme/ugae008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/01/2024] [Accepted: 07/03/2024] [Indexed: 07/20/2024]
Abstract
Adeno-associated virus (AAV) has been remarkably successful in the clinic, but its broad tropism is a practical limitation of precision gene therapy. A promising path to engineer AAV tropism is the addition of binding domains to the AAV capsid that recognize cell surface markers present on a targeted cell type. We have recently identified two previously unexplored capsid regions near the 2/5-fold wall and 5-fold pore of the AAV capsid that are amenable to insertion of larger protein domains, including nanobodies. Here, we demonstrate that these hotspots facilitate AAV tropism switching through simple nanobody replacement without extensive optimization in both VP1 and VP2. Our data suggest that engineering VP2 is the preferred path for maintaining both virus production yield and infectivity. We demonstrate highly specific targeting of human cancer cells expressing fibroblast activating protein (FAP). Furthermore, we found that the combination of FAP nanobody insertion plus ablation of the heparin binding domain can reduce off-target infection to a minimum, while maintaining a strong infection of FAP receptor-positive cells. Taken together, our study shows that nanobody swapping at multiple capsid locations is a viable strategy for nanobody-directed cell-specific AAV targeting.
Collapse
Affiliation(s)
- Mareike D Hoffmann
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Joseph P Gallant
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Aaron M LeBeau
- Department of Pathology and Laboratory Medicine, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI 53792, USA
| | - Daniel Schmidt
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
30
|
Mòdol L, Moissidis M, Selten M, Oozeer F, Marín O. Somatostatin interneurons control the timing of developmental desynchronization in cortical networks. Neuron 2024; 112:2015-2030.e5. [PMID: 38599213 DOI: 10.1016/j.neuron.2024.03.014] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 12/21/2023] [Accepted: 03/11/2024] [Indexed: 04/12/2024]
Abstract
Synchronous neuronal activity is a hallmark of the developing brain. In the mouse cerebral cortex, activity decorrelates during the second week of postnatal development, progressively acquiring the characteristic sparse pattern underlying the integration of sensory information. The maturation of inhibition seems critical for this process, but the interneurons involved in this crucial transition of network activity in the developing cortex remain unknown. Using in vivo longitudinal two-photon calcium imaging during the period that precedes the change from highly synchronous to decorrelated activity, we identify somatostatin-expressing (SST+) interneurons as critical modulators of this switch in mice. Modulation of the activity of SST+ cells accelerates or delays the decorrelation of cortical network activity, a process that involves regulating the maturation of parvalbumin-expressing (PV+) interneurons. SST+ cells critically link sensory inputs with local circuits, controlling the neural dynamics in the developing cortex while modulating the integration of other interneurons into nascent cortical circuits.
Collapse
Affiliation(s)
- Laura Mòdol
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| | - Monika Moissidis
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Martijn Selten
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Fazal Oozeer
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK
| | - Oscar Marín
- Centre for Developmental Neurobiology, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK; MRC Centre for Neurodevelopmental Disorders, King's College London, London, UK.
| |
Collapse
|
31
|
Aubert M, Haick AK, Strongin DE, Klouser LM, Loprieno MA, Stensland L, Santo TK, Huang ML, Hyrien O, Stone D, Jerome KR. Gene editing for latent herpes simplex virus infection reduces viral load and shedding in vivo. Nat Commun 2024; 15:4018. [PMID: 38740820 DOI: 10.1038/s41467-024-47940-y] [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: 01/22/2023] [Accepted: 04/15/2024] [Indexed: 05/16/2024] Open
Abstract
Anti-HSV therapies are only suppressive because they do not eliminate latent HSV present in ganglionic neurons, the source of recurrent disease. We have developed a potentially curative approach against HSV infection, based on gene editing using HSV-specific meganucleases delivered by adeno-associated virus (AAV) vectors. Gene editing performed with two anti-HSV-1 meganucleases delivered by a combination of AAV9, AAV-Dj/8, and AAV-Rh10 can eliminate 90% or more of latent HSV DNA in mouse models of orofacial infection, and up to 97% of latent HSV DNA in mouse models of genital infection. Using a pharmacological approach to reactivate latent HSV-1, we demonstrate that ganglionic viral load reduction leads to a significant decrease of viral shedding in treated female mice. While therapy is well tolerated, in some instances, we observe hepatotoxicity at high doses and subtle histological evidence of neuronal injury without observable neurological signs or deficits. Simplification of the regimen through use of a single serotype (AAV9) delivering single meganuclease targeting a duplicated region of the HSV genome, dose reduction, and use of a neuron-specific promoter each results in improved tolerability while retaining efficacy. These results reinforce the curative potential of gene editing for HSV disease.
Collapse
Affiliation(s)
- Martine Aubert
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Anoria K Haick
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Daniel E Strongin
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98133, USA
| | - Lindsay M Klouser
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98133, USA
| | - Michelle A Loprieno
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Laurence Stensland
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98133, USA
| | - Tracy K Santo
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98133, USA
| | - Meei-Li Huang
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98133, USA
| | - Ollivier Hyrien
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Daniel Stone
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Keith R Jerome
- Vaccine and Infectious Disease Division, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98133, USA.
| |
Collapse
|
32
|
Wang H, Zhou J, Lei J, Mo G, Wu Y, Liu H, Pang Z, Du M, Zhou Z, Paek C, Sun Z, Chen Y, Wang Y, Chen P, Yin L. Engineering of a compact, high-fidelity EbCas12a variant that can be packaged with its crRNA into an all-in-one AAV vector delivery system. PLoS Biol 2024; 22:e3002619. [PMID: 38814985 PMCID: PMC11139299 DOI: 10.1371/journal.pbio.3002619] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 04/09/2024] [Indexed: 06/01/2024] Open
Abstract
The CRISPR-associated endonuclease Cas12a has become a powerful genome-editing tool in biomedical research due to its ease of use and low off-targeting. However, the size of Cas12a severely limits clinical applications such as adeno-associated virus (AAV)-based gene therapy. Here, we characterized a novel compact Cas12a ortholog, termed EbCas12a, from the metagenome-assembled genome of a currently unclassified Erysipelotrichia. It has the PAM sequence of 5'-TTTV-3' (V = A, G, C) and the smallest size of approximately 3.47 kb among the Cas12a orthologs reported so far. In addition, enhanced EbCas12a (enEbCas12a) was also designed to have comparable editing efficiency with higher specificity to AsCas12a and LbCas12a in mammalian cells at multiple target sites. Based on the compact enEbCas12a, an all-in-one AAV delivery system with crRNA for Cas12a was developed for both in vitro and in vivo applications. Overall, the novel smallest high-fidelity enEbCas12a, this first case of the all-in-one AAV delivery for Cas12a could greatly boost future gene therapy and scientific research.
Collapse
Affiliation(s)
- Hongjian Wang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jin Zhou
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Jun Lei
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Guosheng Mo
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yankang Wu
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Huan Liu
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Ziyan Pang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Mingkun Du
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zihao Zhou
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Chonil Paek
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Zaiqiao Sun
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yongshun Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Yan Wang
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Peng Chen
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| | - Lei Yin
- State Key Laboratory of Virology, Hubei Key Laboratory of Cell Homeostasis, College of Life Sciences, Department of Clinical Oncology, Renmin Hospital of Wuhan University, Wuhan University, Wuhan, China
| |
Collapse
|
33
|
Mazzocco C, Genevois C, Li Q, Doudnikoff E, Dutheil N, Leste-Lasserre T, Arotcarena ML, Bezard E. In vivo bioluminescence imaging of the intracerebral fibroin-controlled AAV-α-synuclein diffusion for monitoring the central nervous system and peripheral expression. Sci Rep 2024; 14:9710. [PMID: 38678103 PMCID: PMC11055870 DOI: 10.1038/s41598-024-60613-6] [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: 01/04/2024] [Accepted: 04/25/2024] [Indexed: 04/29/2024] Open
Abstract
Among the several animal models of α-synucleinopathies, the well-known viral vector-mediated delivery of wild-type or mutated (A53T) α-synuclein requires new tools to increase the lesion in mice and follow up in vivo expression. To this end, we developed a bioluminescent expression reporter of the human A53T-α-synuclein gene using the NanoLuc system into an AAV2/9, embedded or not in a fibroin solution to stabilise its expression in space and time. We first verified the expression of the fused protein in vitro on transfected cells by bioluminescence and Western blotting. Next, two groups of C57Bl6Jr mice were unilaterally injected with the AAV-NanoLuc-human-A53T-α-synuclein above the substantia nigra combined (or not) with fibroin. We first show that the in vivo cerebral bioluminescence signal was more intense in the presence of fibroin. Using immunohistochemistry, we find that the human-A53T-α-synuclein protein is more restricted to the ipsilateral side with an overall greater magnitude of the lesion when fibroin was added. However, we also detected a bioluminescence signal in peripheral organs in both conditions, confirmed by the presence of viral DNA corresponding to the injected AAV in the liver using qPCR.
Collapse
Affiliation(s)
- Claire Mazzocco
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France
| | - Coralie Genevois
- VIVOPTIC-TBM-Core Univ Bordeaux, UAR 3427, 33000, Bordeaux, France
| | - Qin Li
- Motac Neuroscience, Manchester, M15 6WE, UK
| | - Evelyne Doudnikoff
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France
| | - Nathalie Dutheil
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France
| | | | - Marie-Laure Arotcarena
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France
| | - Erwan Bezard
- Institut des Maladies Neurodégénératives, UMR 5293, Univ. de Bordeaux, 33000, Bordeaux, France.
- Institut des Maladies Neurodégénératives, UMR 5293, CNRS, 33000, Bordeaux, France.
- Motac Neuroscience, Manchester, M15 6WE, UK.
| |
Collapse
|
34
|
Voorn RA, Sternbach M, Jarysta A, Rankovic V, Tarchini B, Wolf F, Vogl C. Slow kinesin-dependent microtubular transport facilitates ribbon synapse assembly in developing cochlear inner hair cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.12.589153. [PMID: 38659872 PMCID: PMC11042220 DOI: 10.1101/2024.04.12.589153] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Sensory synapses are characterized by electron-dense presynaptic specializations, so-called synaptic ribbons. In cochlear inner hair cells (IHCs), ribbons play an essential role as core active zone (AZ) organizers, where they tether synaptic vesicles, cluster calcium channels and facilitate the temporally-precise release of primed vesicles. While a multitude of studies aimed to elucidate the molecular composition and function of IHC ribbon synapses, the developmental formation of these signalling complexes remains largely elusive to date. To address this shortcoming, we performed long-term live-cell imaging of fluorescently-labelled ribbon precursors in young postnatal IHCs to track ribbon precursor motion. We show that ribbon precursors utilize the apico-basal microtubular (MT) cytoskeleton for targeted trafficking to the presynapse, in a process reminiscent of slow axonal transport in neurons. During translocation, precursor volume regulation is achieved by highly dynamic structural plasticity - characterized by regularly-occurring fusion and fission events. Pharmacological MT destabilization negatively impacted on precursor translocation and attenuated structural plasticity, whereas genetic disruption of the anterograde molecular motor Kif1a impaired ribbon volume accumulation during developmental maturation. Combined, our data thus indicate an essential role of the MT cytoskeleton and Kif1a in adequate ribbon synapse formation and structural maintenance.
Collapse
Affiliation(s)
- Roos Anouk Voorn
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Centre Goettingen, 37075 Goettingen, Germany
- Göttingen Graduate Centre for Neurosciences, Biophysics and Molecular Biosciences, 37075 Goettingen, Germany
- Collaborative Research Centre 889 ‘Cellular Mechanisms of Sensory Processing’, 37075 Goettingen, Germany
- Auditory Neuroscience Group, Institute of Physiology, Medical University Innsbruck, A-6020 Innsbruck, Austria
| | - Michael Sternbach
- Campus Institute for Dynamics of Biological Networks, 37073 Goettingen, Germany
- Bernstein Centre for Computational Neuroscience, 37073 Goettingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, 37077 Goettingen, Germany
| | | | - Vladan Rankovic
- Institute for Auditory Neuroscience and InnerEarLab, University Medical Center Göttingen, 37075 Göttingen, Germany
- Restorative Cochlear Genomics Group, Auditory Neuroscience and Optogenetics Laboratory, German Primate Center, 37075 Göttingen, Germany
| | - Basile Tarchini
- The Jackson Laboratory, Bar Harbor ME, USA
- Tufts University School of Medicine, Boston MA, USA
| | - Fred Wolf
- Campus Institute for Dynamics of Biological Networks, 37073 Goettingen, Germany
- Bernstein Centre for Computational Neuroscience, 37073 Goettingen, Germany
- Max Planck Institute for Dynamics and Self-Organization, 37077 Goettingen, Germany
- Institute for Dynamics of Complex Systems Georg-August-University, 37077 Goettingen, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Goettingen, Germany
| | - Christian Vogl
- Presynaptogenesis and Intracellular Transport in Hair Cells Junior Research Group, Institute for Auditory Neuroscience and InnerEarLab, University Medical Centre Goettingen, 37075 Goettingen, Germany
- Collaborative Research Centre 889 ‘Cellular Mechanisms of Sensory Processing’, 37075 Goettingen, Germany
- Auditory Neuroscience Group, Institute of Physiology, Medical University Innsbruck, A-6020 Innsbruck, Austria
| |
Collapse
|
35
|
Fryer E, Guha S, Rogel-Hernandez LE, Logan-Garbisch T, Farah H, Rezaei E, Mollhoff IN, Nekimken AL, Xu A, Selin Seyahi L, Fechner S, Druckmann S, Clandinin TR, Rhee SY, Goodman MB. An efficient behavioral screening platform classifies natural products and other chemical cues according to their chemosensory valence in C. elegans. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.06.02.542933. [PMID: 37333363 PMCID: PMC10274637 DOI: 10.1101/2023.06.02.542933] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Throughout history, humans have relied on plants as a source of medication, flavoring, and food. Plants synthesize large chemical libraries and release many of these compounds into the rhizosphere and atmosphere where they affect animal and microbe behavior. To survive, nematodes must have evolved the sensory capacity to distinguish plant-made small molecules (SMs) that are harmful and must be avoided from those that are beneficial and should be sought. This ability to classify chemical cues as a function of their value is fundamental to olfaction, and represents a capacity shared by many animals, including humans. Here, we present an efficient platform based on multi-well plates, liquid handling instrumentation, inexpensive optical scanners, and bespoke software that can efficiently determine the valence (attraction or repulsion) of single SMs in the model nematode, Caenorhabditis elegans. Using this integrated hardware-wetware-software platform, we screened 90 plant SMs and identified 37 that attracted or repelled wild-type animals, but had no effect on mutants defective in chemosensory transduction. Genetic dissection indicates that for at least 10 of these SMs, response valence emerges from the integration of opposing signals, arguing that olfactory valence is often determined by integrating chemosensory signals over multiple lines of information. This study establishes that C. elegans is an effective discovery engine for determining chemotaxis valence and for identifying natural products detected by the chemosensory nervous system.
Collapse
Affiliation(s)
- Emily Fryer
- Department of Plant Biology, Carnegie Institution for Science
- Department of Molecular and Cellular Physiology, Stanford University
| | - Sujay Guha
- Department of Molecular and Cellular Physiology, Stanford University
| | | | - Theresa Logan-Garbisch
- Department of Molecular and Cellular Physiology, Stanford University
- Neurosciences Graduate Program, Stanford University
| | - Hodan Farah
- Department of Plant Biology, Carnegie Institution for Science
- Department of Molecular and Cellular Physiology, Stanford University
| | - Ehsan Rezaei
- Department of Molecular and Cellular Physiology, Stanford University
| | - Iris N. Mollhoff
- Department of Plant Biology, Carnegie Institution for Science
- Department of Molecular and Cellular Physiology, Stanford University
- Department of Biology, Stanford University
| | - Adam L. Nekimken
- Department of Molecular and Cellular Physiology, Stanford University
- Department of Mechanical Engineering, Stanford University
| | - Angela Xu
- Department of Plant Biology, Carnegie Institution for Science
| | - Lara Selin Seyahi
- Department of Plant Biology, Carnegie Institution for Science
- Department of Molecular and Cellular Physiology, Stanford University
| | - Sylvia Fechner
- Department of Molecular and Cellular Physiology, Stanford University
| | | | | | - Seung Y. Rhee
- Department of Plant Biology, Carnegie Institution for Science
| | - Miriam B. Goodman
- Department of Molecular and Cellular Physiology, Stanford University
| |
Collapse
|
36
|
Gaddie CD, Senior KG, Chan C, Hoffman BE, Keeler GD. Upregulation of CD8 + regulatory T cells following liver-directed AAV gene therapy. Cell Immunol 2024; 397-398:104806. [PMID: 38244266 DOI: 10.1016/j.cellimm.2024.104806] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/21/2023] [Accepted: 01/10/2024] [Indexed: 01/22/2024]
Abstract
Liver-directed AAV gene therapy represents a unique treatment modality for a host of diseases. This is due, in part, to the induction of tolerance to transgene products. Despite the plethora of recognized regulatory cells in the body, there is currently a lack of literature supporting the induction of non-CD4+ regulatory cells following hepatic AAV gene transfer. In this work, we show that CD8+ regulatory T cells are up-regulated in PBMCs of mice following capsid only and therapeutic transgene AAV administration. Further, we demonstrate that hepatic AAV gene transfer results in a significant increase in CD8+ regulatory T cells following experimental autoimmune encephalomyelitis induction. Notably, this response occurred only in therapeutic vector treated animals, not capsid only controls. Understanding the role these cells play in treatment efficacy will result in the development of improved AAV vectors that take advantage of the full gamut of regulatory cells within the body.
Collapse
Affiliation(s)
- Cristina D Gaddie
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Kevin G Senior
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Christopher Chan
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA
| | - Brad E Hoffman
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA; Department of Neuroscience, University of Florida, Gainesville, FL, USA
| | - Geoffrey D Keeler
- Division of Cellular and Molecular Therapy, Department of Pediatrics, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
37
|
Biber J, Jabri Y, Glänzer S, Dort A, Hoffelner P, Schmidt CQ, Bludau O, Pauly D, Grosche A. Gliosis-dependent expression of complement factor H truncated variants attenuates retinal neurodegeneration following ischemic injury. J Neuroinflammation 2024; 21:56. [PMID: 38388518 PMCID: PMC10885619 DOI: 10.1186/s12974-024-03045-3] [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: 10/11/2023] [Accepted: 02/14/2024] [Indexed: 02/24/2024] Open
Abstract
Inherited, age-related, and acute retinal diseases are often exacerbated by an aberrant or excessive activity of the complement system. Consequently, cells not directly affected by an acute event or genetic variants may degenerate, resulting in enhanced visual impairment. The therapeutic potential of supplementation of complement factor H (FH), a key regulator of the complement cascade, is therefore particularly promising in the context of retinal diseases caused by complement activation. In this study, we engineered adeno-associated viruses (AAVs) containing sequences of two truncated human FH variants. The expression of these variants was regulated by the glial fibrillary acidic protein (GFAP) promoter, which is selectively active in gliotic Müller cells. Both FH variants consisted of FH domains 19-20, which were connected to domains 1-4 and 1-7, respectively, by a polyglycine linker. These AAVs were intravitreally injected following ischemic injury of C57BL/6J mouse retinas. We observed transgene expression in gliotic Müller cells and to some extent in astrocytes. The expression correlated directly with damage severity. Interventions resulted in decreased complement activation, accelerated normalization of microglia activity and morphological improvements. Reduced levels of C3 transcripts and C3d protein in conjunction with higher transcript levels of inhibitory regulators like Cfi and Cfh, hinted at attenuated complement activity. This study demonstrates the great potential of complement regulatory gene addition therapy. With further in vivo testing it could be applied to treat a wide range of retinal diseases where no causative therapies are available.
Collapse
Affiliation(s)
- Josef Biber
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Yassin Jabri
- Department of Ophthalmology, University Hospital Regensburg, Regensburg, Germany
| | - Sarah Glänzer
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Aaron Dort
- Experimental Ophthalmology, University of Marburg, Marburg, Germany
| | - Patricia Hoffelner
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Christoph Q Schmidt
- Institute of Experimental and Clinical Pharmacology, Toxicology and Pharmacology of Natural Products, University of Ulm Medical Center, Ulm, Germany
- Institute of Pharmacy, Biochemical Pharmacy Group, Martin Luther University Halle-Wittenberg, Halle, Germany
| | - Oliver Bludau
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| | - Diana Pauly
- Experimental Ophthalmology, University of Marburg, Marburg, Germany.
| | - Antje Grosche
- Department of Physiological Genomics, Ludwig-Maximilians-Universität München, Planegg-Martinsried, Germany
| |
Collapse
|
38
|
Keller WR, Picciano A, Wilson K, Xu J, Khasa H, Wendeler M. Rational downstream development for adeno-associated virus full/empty capsid separation - A streamlined methodology based on high-throughput screening and mechanistic modeling. J Chromatogr A 2024; 1716:464632. [PMID: 38219623 DOI: 10.1016/j.chroma.2024.464632] [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: 07/18/2023] [Revised: 01/03/2024] [Accepted: 01/04/2024] [Indexed: 01/16/2024]
Abstract
Recombinant adeno-associated virus (AAV) has emerged as one of the most promising systems for therapeutic gene delivery and has demonstrated clinical success in a wide range of genetic disorders. However, manufacturing of high-quality AAV in large amounts still remains a challenge. A significant difficulty for downstream processing is the need to remove empty capsids that are generated in all currently utilized expression systems and that represent product-related impurities that adversely affect safety and efficacy of AAV vectors. Empty and full capsids exhibit only subtle differences in surface charge and size, making chromatography-based separations highly challenging. Here, we present a rapid methodology for the systematic process development of the crucial AAV full/empty capsid separation on ion-exchange media based on high-throughput screening and mechanistic modeling. Two of the most commonly employed serotypes, AAV8 and AAV9, are used as case studies. First, high-throughput studies in filter-plate format are performed that allow the rapid and comprehensive study of binding and elution behavior of AAV on different resins, using different buffer systems, pH, salt conditions, and solution additives. Small amounts of separated empty and full AAV capsids are generated by iodixanol gradient centrifugation that allow studying the binding and elution behavior of the two vector species separately in miniaturized format. Process conditions that result in maximum differences in elution behavior between empty and full capsids are then transferred to benchtop chromatography systems that are used to generate calibration data for the estimation of steric mass-action isotherm and mass transport parameters for process simulation. The resulting column models are employed for in-silico process development that serves to enhance understanding of separation constraints and to identify optimized conditions for the removal of empty particles. Finally, optimized separation conditions are verified experimentally. The methodology presented in this work provides a systematic framework that affords mechanistic understanding of the crucial empty/full capsid separation and accelerates the development of a scalable AAV downstream process.
Collapse
Affiliation(s)
- William R Keller
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, United States
| | - Angela Picciano
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, United States
| | - Kelly Wilson
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, United States
| | - Jin Xu
- Cell Culture and Fermentation Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, United States
| | - Harshit Khasa
- Analytical Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, United States
| | - Michaela Wendeler
- Purification Process Sciences, BioPharmaceuticals Development, R&D, AstraZeneca, Gaithersburg, United States.
| |
Collapse
|
39
|
Kulkarni AA, Seal AG, Sonnet C, Oka K. Streamlined Adeno-Associated Virus Production Using Suspension HEK293T Cells. Bio Protoc 2024; 14:e4931. [PMID: 38379831 PMCID: PMC10875358 DOI: 10.21769/bioprotoc.4931] [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: 04/20/2023] [Revised: 05/28/2023] [Accepted: 01/17/2024] [Indexed: 02/22/2024] Open
Abstract
Recombinant adeno-associated viruses (rAAVs) are valuable viral vectors for in vivo gene transfer, also having significant ex vivo therapeutic potential. Continued efforts have focused on various gene therapy applications, capsid engineering, and scalable manufacturing processes. Adherent cells are commonly used for virus production in most basic science laboratories because of their efficiency and cost. Although suspension cells are easier to handle and scale up compared to adherent cells, their use in virus production is hampered by poor transfection efficiency. In this protocol, we developed a simple scalable AAV production protocol using serum-free-media-adapted HEK293T suspension cells and VirusGEN transfection reagent. The established protocol allows AAV production from transfection to quality analysis of purified AAV within two weeks. Typical vector yields for the described suspension system followed by iodixanol purification range from a total of 1 × 1013 to 1.5 × 1013 vg (vector genome) using 90 mL of cell suspension vs. 1 × 1013 to 2 × 1013 vg using a regular adherent cell protocol (10 × 15 cm dishes). Key features • Adeno-associated virus (AAV) production using serum-free-media-adapted HEK293T suspension cells. • Efficient transfection with VirusGEN. • High AAV yield from small-volume cell culture. Graphical overview.
Collapse
Affiliation(s)
- Aditi A. Kulkarni
- Gene Vector Core, Advanced Technology Cores, Baylor
College of Medicine, Houston, TX, USA
| | - Austin G. Seal
- Gene Vector Core, Advanced Technology Cores, Baylor
College of Medicine, Houston, TX, USA
| | - Corinne Sonnet
- Gene Vector Core, Advanced Technology Cores, Baylor
College of Medicine, Houston, TX, USA
- Department of Medicine, Baylor College of Medicine,
Houston, TX, USA
| | - Kazuhiro Oka
- Gene Vector Core, Advanced Technology Cores, Baylor
College of Medicine, Houston, TX, USA
- Department of Molecular and Cellular Biology, Baylor
College of Medicine, Houston, TX, USA
| |
Collapse
|
40
|
Araujo VG, Dias MS, Hauswirth WW, Linden R, Petrs-Silva H. rAAV-compatible human mini promoters enhance transgene expression in rat retinal ganglion cells. Exp Eye Res 2024; 239:109758. [PMID: 38123011 DOI: 10.1016/j.exer.2023.109758] [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: 08/02/2023] [Revised: 11/14/2023] [Accepted: 12/15/2023] [Indexed: 12/23/2023]
Abstract
Recombinant adeno-associated viral vectors (rAAV) are the safest and most effective gene delivery platform to drive the treatment of many inherited eye disorders in well-characterized animal models. The use in rAAV of ubiquitous promoters derived from viral sequences such as CMV/CBA (chicken β-actin promoter with cytomegalovirus enhancer) can lead to unwanted side effects such as pro-inflammatory immune responses and retinal cytotoxicity, thus reducing therapy efficacy. Thus, an advance in gene therapy is the availability of small promoters, that potentiate and direct gene expression to the cell type of interest, with higher safety and efficacy. In this study, we used six human mini-promoters packaged in rAAV2 quadruple mutant (Y-F) to test for transduction of the rat retina after intravitreal injection. After four weeks, immunohistochemical analysis detected GFP-labeled cells in the ganglion cell layer (GCL) for all constructs tested. Among them, Ple25sh1, Ple25sh2 and Ple53 promoted a widespread reporter-transgene expression in the GCL, with an increased number of GFP-expressing retinal ganglion cells when compared with the CMV/CBA vector. Moreover, Ple53 provided the strongest levels of GFP fluorescence in both cell soma and axons of retinal ganglion cells (RGCs) without any detectable adverse effects in retina function. Remarkably, a nearly 50-fold reduction in the number of intravitreally injected vector particles containing Ple53 promoter, still attained levels of transgene expression similar to CMV/CBA. Thus, the tested MiniPs show great potential for protocols of retinal gene therapy in therapeutic applications for retinal degenerations, especially those involving RGC-related disorders such as glaucoma.
Collapse
Affiliation(s)
- Victor G Araujo
- Laboratory of Gene Therapy and Viral Vector, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Mariana S Dias
- Laboratory of Gene Therapy and Viral Vector, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - William W Hauswirth
- Retinal Gene Therapy Group, Department of Ophthalmology, University of Florida, Gainesville, FL, USA
| | - Rafael Linden
- Laboratory of Neurogenesis, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Hilda Petrs-Silva
- Laboratory of Gene Therapy and Viral Vector, Carlos Chagas Filho Institute of Biophysics, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
41
|
Zhu D, Brookes DH, Busia A, Carneiro A, Fannjiang C, Popova G, Shin D, Donohue KC, Lin LF, Miller ZM, Williams ER, Chang EF, Nowakowski TJ, Listgarten J, Schaffer DV. Optimal trade-off control in machine learning-based library design, with application to adeno-associated virus (AAV) for gene therapy. SCIENCE ADVANCES 2024; 10:eadj3786. [PMID: 38266077 PMCID: PMC10807795 DOI: 10.1126/sciadv.adj3786] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 12/22/2023] [Indexed: 01/26/2024]
Abstract
Adeno-associated viruses (AAVs) hold tremendous promise as delivery vectors for gene therapies. AAVs have been successfully engineered-for instance, for more efficient and/or cell-specific delivery to numerous tissues-by creating large, diverse starting libraries and selecting for desired properties. However, these starting libraries often contain a high proportion of variants unable to assemble or package their genomes, a prerequisite for any gene delivery goal. Here, we present and showcase a machine learning (ML) method for designing AAV peptide insertion libraries that achieve fivefold higher packaging fitness than the standard NNK library with negligible reduction in diversity. To demonstrate our ML-designed library's utility for downstream engineering goals, we show that it yields approximately 10-fold more successful variants than the NNK library after selection for infection of human brain tissue, leading to a promising glial-specific variant. Moreover, our design approach can be applied to other types of libraries for AAV and beyond.
Collapse
Affiliation(s)
- Danqing Zhu
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - David H. Brookes
- Biophysics Graduate Group, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Akosua Busia
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Ana Carneiro
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | | | - Galina Popova
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioural Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
| | - David Shin
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioural Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
| | - Kevin C. Donohue
- Department of Psychiatry and Behavioural Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- School of Medicine, University of California San Francisco, San Francisco, CA, USA. 94143
- Kavli Institute of Fundamental Neuroscience, University of California San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
| | - Li F. Lin
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Zachary M. Miller
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Evan R. Williams
- Department of Chemistry, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Edward F. Chang
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Tomasz J. Nowakowski
- Department of Anatomy, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Psychiatry and Behavioural Sciences, University of California San Francisco, San Francisco, CA 94143, USA
- Eli and Edythe Broad Center for Regeneration Medicine and Stem Cell Research, University of California San Francisco, San Francisco, CA 94143, USA
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA 94143, USA
- Department of Neurological Surgery, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jennifer Listgarten
- Department of Electrical Engineering and Computer Sciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Center for Computational Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - David V. Schaffer
- California Institute for Quantitative Biosciences, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA
- Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
- Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA 94720, USA
- Innovative Genomics Institute (IGI), University of California, Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
42
|
Gaudry JP, Aebi A, Valdés P, Schneider BL. Production and Purification of Adeno-Associated Viral Vectors (AAVs) Using Orbitally Shaken HEK293 Cells. Methods Mol Biol 2024; 2810:55-74. [PMID: 38926272 DOI: 10.1007/978-1-0716-3878-1_4] [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: 06/28/2024]
Abstract
Here, we describe methods for the production of adeno-associated viral (AAV) vectors by transient transfection of HEK293 cells grown in serum-free medium using orbital shaken bioreactors and the subsequent purification of vector particles. The protocol for expression of AAV components is based on polyethyleneimine (PEI)-mediated transfection of a three-plasmid system and is specified for production in milliliter-to-liter scales. After PEI and plasmid DNA (pDNA) complex formation, the diluted cell culture is transfected without a prior concentration step or medium exchange. Following a 7-day batch process, cell cultures are further processed using a set of methods for cell lysis and vector recovery. Methods for the purification of viral particles are described, including immunoaffinity and anion-exchange chromatography, ultrafiltration, as well as digital PCR to quantify the concentration of vector particles.
Collapse
Affiliation(s)
- Jean-Philippe Gaudry
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Aline Aebi
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Pamela Valdés
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Bernard L Schneider
- Bertarelli Platform for Gene Therapy, Ecole Polytechnique Fédérale de Lausanne (EPFL), Geneva, Switzerland.
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
- NeuroX Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland.
| |
Collapse
|
43
|
Joshi PRH, Venereo-Sanchez A. Recombinant AAV Purification. Methods Mol Biol 2024; 2829:217-226. [PMID: 38951337 DOI: 10.1007/978-1-0716-3961-0_15] [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: 07/03/2024]
Abstract
Purification of rAAV is a crucial unit operation of the AAV production process. It enables the capture of AAV and removal of contaminants such as host cell proteins, host cell DNA, and other cell culture-related impurities. Here we describe the purification of rAAV produced in insect cells Sf9/rBEV by immuno-affinity capture chromatography. The method is fully scale-amenable unlike other traditional purification methods based on ultracentrifugation. The method reported herein has two main steps: (1) the clarification of cell lysate by depth filtration and (2) the selective capture and single-step purification of AAV via immune-affinity chromatography. This purification method has been successfully implemented to purify the majority of wild-type AAV serotypes.
Collapse
Affiliation(s)
- Pranav R H Joshi
- Department of Bioengineering, McGill University, Montreal, QC, Canada
| | | |
Collapse
|
44
|
Huang Y, Wang YF, Ruan XZ, Lau CW, Wang L, Huang Y. The role of KLF2 in regulating hepatic lipogenesis and blood cholesterol homeostasis via the SCAP/SREBP pathway. J Lipid Res 2024; 65:100472. [PMID: 37949368 PMCID: PMC10805670 DOI: 10.1016/j.jlr.2023.100472] [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: 04/12/2023] [Revised: 10/21/2023] [Accepted: 11/02/2023] [Indexed: 11/12/2023] Open
Abstract
Liver steatosis is a common metabolic disorder resulting from imbalanced lipid metabolism, which involves various processes such as de novo lipogenesis, fatty acid uptake, fatty acid oxidation, and VLDL secretion. In this study, we discovered that KLF2, a transcription factor, plays a crucial role in regulating lipid metabolism in the liver. Overexpression of KLF2 in the liver of db/db mice, C57BL/6J mice, and Cd36-/- mice fed on a normal diet resulted in increased lipid content in the liver. Additionally, transgenic mice (ALB-Klf2) that overexpressed Klf2 in the liver developed liver steatosis after being fed a normal diet. We found that KLF2 promotes lipogenesis by increasing the expression of SCAP, a chaperone that facilitates the activation of SREBP, the master transcription factor for lipogenic gene expression. Our mechanism studies revealed that KLF2 enhances lipogenesis in the liver by binding to the promoter of SCAP and increasing the expression of genes involved in fatty acid synthesis. Reduction of KLF2 expression led to a decrease in SCAP expression and a reduction in the expression of SREBP1 target genes involved in lipogenesis. Overexpression of KLF2 also increased the activation of SREBP2 and the mRNA levels of its downstream target SOAT1. In C57BL/6J mice fed a high-fat diet, overexpression of Klf2 increased blood VLDL secretion, while reducing its expression decreased blood cholesterol levels. Our study emphasizes the novelty that hepatic KLF2 plays a critical role in regulating lipid metabolism through the KLF2/SCAP/SREBPs pathway, which is essential for hepatic lipogenesis and maintaining blood cholesterol homeostasis.
Collapse
Affiliation(s)
- Yuhong Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China; School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, PR China; Shenzhen Research Institute and Li Ka Shing Institute of Health Sciences, Chinese University of Hong Kong, Shenzhen, China
| | - Yi Fan Wang
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, PR China
| | - Xiong Zhong Ruan
- Centre for Lipid Research & Key Laboratory of Molecular Biology for Infectious Diseases (Ministry of Education), Institute for Viral Hepatitis, Department of Infectious Diseases, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Chi Wai Lau
- School of Biomedical Sciences, Chinese University of Hong Kong, Hong Kong, PR China
| | - Li Wang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, China.
| |
Collapse
|
45
|
Thompson A, Arano R, Saleem U, Preciado R, Munoz L, Nelson I, Ramos K, Kim Y, Li Y, Xu W. Brain-wide circuit-specific targeting of astrocytes. CELL REPORTS METHODS 2023; 3:100653. [PMID: 38052209 PMCID: PMC10753298 DOI: 10.1016/j.crmeth.2023.100653] [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: 12/29/2022] [Revised: 10/04/2023] [Accepted: 11/08/2023] [Indexed: 12/07/2023]
Abstract
Astrocytes are integral components of brain circuitry. They enwrap synapses, react to neuronal activity, and regulate synaptic transmission. Astrocytes are heterogeneous and exhibit distinct features and functions in different circuits. Selectively targeting the astrocytes associated with a given neuronal circuit would enable elucidation of their circuit-specific functions but has been technically challenging to date. Recently, we constructed anterograde transneuronal viral vectors based on yellow fever vaccine YFV-17D. Among them, the replication-incompetent YFVΔNS1-Cre can selectively turn on reporter genes in postsynaptic neurons if the viral gene NS1 is expressed in postsynaptic neurons. Here we show that without exogenous expression of NS1 at the postsynaptic sites, locally injected YFVΔNS1-Cre selectively turns on reporter genes in astrocytes in downstream brain regions. The targeting of astrocytes can occur across the whole brain but is specific for the neuronal circuits traced. Therefore, YFVΔNS1-Cre provides a tool for selective genetic targeting of astrocytes to reveal their circuit-specific roles.
Collapse
Affiliation(s)
- Alyssa Thompson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rachel Arano
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Uzair Saleem
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Rebecca Preciado
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Lizbeth Munoz
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ian Nelson
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Katarina Ramos
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yerim Kim
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Ying Li
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| | - Wei Xu
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
46
|
Ohba K, Mizukami H. Protocol for producing an adeno-associated virus vector by controlling capsid expression timing. STAR Protoc 2023; 4:102542. [PMID: 38103199 PMCID: PMC10751547 DOI: 10.1016/j.xpro.2023.102542] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2023] [Revised: 07/13/2023] [Accepted: 08/04/2023] [Indexed: 12/18/2023] Open
Abstract
Conventional adeno-associated virus (AAV) production systems generate vast numbers of empty capsids, which should be eliminated before clinical use. Here, we present a protocol for efficient AAV vector production. We describe steps for separating replicase and capsid genes from the plasmid and controlling capsid expression until sufficient AAV vector genome replication is achieved. This protocol can produce AAV vectors in various serotypes. For complete details on the use and execution of this protocol, please refer to Ohba et al.1.
Collapse
Affiliation(s)
- Kenji Ohba
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan.
| | - Hiroaki Mizukami
- Division of Genetic Therapeutics, Center for Molecular Medicine, Jichi Medical University, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
47
|
Hoffmann MD, Zdechlik AC, He Y, Nedrud D, Aslanidi G, Gordon W, Schmidt D. Multiparametric domain insertional profiling of adeno-associated virus VP1. Mol Ther Methods Clin Dev 2023; 31:101143. [PMID: 38027057 PMCID: PMC10661864 DOI: 10.1016/j.omtm.2023.101143] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 10/21/2023] [Indexed: 12/01/2023]
Abstract
Several evolved properties of adeno-associated virus (AAV), such as broad tropism and immunogenicity in humans, are barriers to AAV-based gene therapy. Most efforts to re-engineer these properties have focused on variable regions near AAV's 3-fold protrusions and capsid protein termini. To comprehensively survey AAV capsids for engineerable hotspots, we determined multiple AAV fitness phenotypes upon insertion of six structured protein domains into the entire AAV-DJ capsid protein VP1. This is the largest and most comprehensive AAV domain insertion dataset to date. Our data revealed a surprising robustness of AAV capsids to accommodate large domain insertions. Insertion permissibility depended strongly on insertion position, domain type, and measured fitness phenotype, which clustered into contiguous structural units that we could link to distinct roles in AAV assembly, stability, and infectivity. We also identified engineerable hotspots of AAV that facilitate the covalent attachment of binding scaffolds, which may represent an alternative approach to re-direct AAV tropism.
Collapse
Affiliation(s)
- Mareike D. Hoffmann
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - Alina C. Zdechlik
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Yungui He
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| | - David Nedrud
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Wendy Gordon
- Department of Biochemistry, Molecular Biology & Biophysics, University of Minnesota, Minneapolis, MN 55455, USA
| | - Daniel Schmidt
- Department of Genetics, Cell Biology & Development, University of Minnesota, Minneapolis, MN 55455, USA
| |
Collapse
|
48
|
Khanal O, Kumar V, Jin M. Adeno-associated viral capsid stability on anion exchange chromatography column and its impact on empty and full capsid separation. Mol Ther Methods Clin Dev 2023; 31:101112. [PMID: 37868210 PMCID: PMC10585339 DOI: 10.1016/j.omtm.2023.101112] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 09/13/2023] [Indexed: 10/24/2023]
Abstract
Recombinant adeno-associated viral vector (rAAV) mediated gene therapy is gaining traction in treating genetic disorders. Current rAAV production systems yield a mixture of capsids largely devoid of the transgene (empty capsid) compared with the desired therapeutic product (full capsid). Anion exchange chromatography (AEX) is an attractive method for separating empty and full AAV capsids because of its scalability. Resin types and buffer composition are key considerations for AEX and must support capsid stability to be suitable for downstream processing. We examined the impact of binding durations (0-8 h) using various binding ionic strengths (15-75 mM), pH (7.5-9.0), resin chemistry (POROS XQ, POROS HQ, POROS I, and BIA QA monolith), and proprietary Q resins with different ligand densities for effects on capsid stability. Empty capsids were altered upon extended binding, leading to retention time shifts and loss of resolution between empty and full capsids. Viral capsid protein analysis reveals that full capsids have more viral capsid protein 3 (VP3) proteins than empty capsids. Analytical hydrophilic liquid chromatography showed that empty capsid retention time shift is accompanied by changes to the empty capsid's native VP3 protein. Among the potential stabilizing additives considered, magnesium chloride was the most effective at reducing negative impacts caused by extended binding.
Collapse
Affiliation(s)
- Ohnmar Khanal
- Technology Development, Spark Therapeutics, Inc., Philadelphia, PA, USA
| | - Vijesh Kumar
- Technology Development, Spark Therapeutics, Inc., Philadelphia, PA, USA
| | - Mi Jin
- Technology Development, Spark Therapeutics, Inc., Philadelphia, PA, USA
| |
Collapse
|
49
|
Chocarro J, Rico AJ, Ariznabarreta G, Roda E, Honrubia A, Collantes M, Peñuelas I, Vázquez A, Rodríguez-Pérez AI, Labandeira-García JL, Vila M, Lanciego JL. Neuromelanin accumulation drives endogenous synucleinopathy in non-human primates. Brain 2023; 146:5000-5014. [PMID: 37769648 PMCID: PMC10689915 DOI: 10.1093/brain/awad331] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 08/04/2023] [Accepted: 09/07/2023] [Indexed: 10/03/2023] Open
Abstract
Although neuromelanin is a dark pigment characteristic of dopaminergic neurons in the human substantia nigra pars compacta, its potential role in the pathogenesis of Parkinson's disease (PD) has often been neglected since most commonly used laboratory animals lack neuromelanin. Here we took advantage of adeno-associated viral vectors encoding the human tyrosinase gene for triggering a time-dependent neuromelanin accumulation within substantia nigra pars compacta dopaminergic neurons in macaques up to similar levels of pigmentation as observed in elderly humans. Furthermore, neuromelanin accumulation induced an endogenous synucleinopathy mimicking intracellular inclusions typically observed in PD together with a progressive degeneration of neuromelanin-expressing dopaminergic neurons. Moreover, Lewy body-like intracellular inclusions were observed in cortical areas of the frontal lobe receiving dopaminergic innervation, supporting a circuit-specific anterograde spread of endogenous synucleinopathy by permissive trans-synaptic templating. In summary, the conducted strategy resulted in the development and characterization of a new macaque model of PD matching the known neuropathology of this disorder with unprecedented accuracy. Most importantly, evidence is provided showing that intracellular aggregation of endogenous α-synuclein is triggered by neuromelanin accumulation, therefore any therapeutic approach intended to decrease neuromelanin levels may provide appealing choices for the successful implementation of novel PD therapeutics.
Collapse
Affiliation(s)
- Julia Chocarro
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Alberto J Rico
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Goiaz Ariznabarreta
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Elvira Roda
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - Adriana Honrubia
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| | - María Collantes
- Translational Molecular Imaging Unit, Department of Nuclear Medicine, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Iván Peñuelas
- Translational Molecular Imaging Unit, Department of Nuclear Medicine, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Alfonso Vázquez
- Department of Neurosurgery, Hospital Universitario de Navarra, Servicio Navarro de Salud, 31008 Pamplona, Spain
| | - Ana I Rodríguez-Pérez
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - José L Labandeira-García
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Miquel Vila
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
- Vall d’Hebron Research Institute, Neurodegenerative Diseses Research Group, 08035 Barcelona, Spain
- Autonomous University of Barcelona (UAB), 08193 Bellaterra, Barcelona, Spain
- Catalan Institution for Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| | - José L Lanciego
- CNS Gene Therapy Program, Center for Applied Medical Research (CIMA), University of Navarra, 31008 Pamplona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (Ciberned-ISCIII), 28031 Madrid, Spain
- Aligning Science Across Parkinsons’s (ASAP) Collaborative Research Network, Chevy Chase, MD 20815, USA
| |
Collapse
|
50
|
Yost SA, Firlar E, Glenn JD, Carroll HB, Foltz S, Giles AR, Egley JM, Firnberg E, Cho S, Nguyen T, Henry WM, Janczura KJ, Bruder J, Liu Y, Danos O, Karumuthil-Melethil S, Pannem S, Yost V, Engelson Y, Kaelber JT, Dimant H, Smith JB, Mercer AC. Characterization and biodistribution of under-employed gene therapy vector AAV7. J Virol 2023; 97:e0116323. [PMID: 37843374 PMCID: PMC10688378 DOI: 10.1128/jvi.01163-23] [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: 07/31/2023] [Accepted: 08/27/2023] [Indexed: 10/17/2023] Open
Abstract
IMPORTANCE The use of adeno-associated viruses (AAVs) as gene delivery vectors has vast potential for the treatment of many severe human diseases. Over one hundred naturally existing AAV capsid variants have been described and classified into phylogenetic clades based on their sequences. AAV8, AAV9, AAVrh.10, and other intensively studied capsids have been propelled into pre-clinical and clinical use, and more recently, marketed products; however, less-studied capsids may also have desirable properties (e.g., potency differences, tissue tropism, reduced immunogenicity, etc.) that have yet to be thoroughly described. These data will help build a broader structure-function knowledge base in the field, present capsid engineering opportunities, and enable the use of novel capsids with unique properties.
Collapse
Affiliation(s)
- Samantha A. Yost
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Emre Firlar
- Institute of Quantitative Biomedicine and Rutgers CryoEM & Nanoimaging Facility, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Justin D. Glenn
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Hayley B. Carroll
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Steven Foltz
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - April R. Giles
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Jenny M. Egley
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Elad Firnberg
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Sungyeon Cho
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Trang Nguyen
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - William M. Henry
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | | | - Joseph Bruder
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Ye Liu
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Olivier Danos
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | | | | | | | | | - Jason T. Kaelber
- Institute of Quantitative Biomedicine and Rutgers CryoEM & Nanoimaging Facility, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Hemi Dimant
- Invicro LLC, Needham, Massachusetts, USA
- Emit Imaging, Baltimore, Maryland, USA
| | - Jared B. Smith
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| | - Andrew C. Mercer
- Research and Early Development, REGENXBIO Inc., Rockville, Maryland, USA
| |
Collapse
|