51
|
Robert V, O'Neil K, Rashid SK, Johnson CD, De La Torre RG, Zemelman BV, Clopath C, Basu J. Entorhinal cortex glutamatergic and GABAergic projections bidirectionally control discrimination and generalization of hippocampal representations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.08.566107. [PMID: 37986793 PMCID: PMC10659280 DOI: 10.1101/2023.11.08.566107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Discrimination and generalization are crucial brain-wide functions for memory and object recognition that utilize pattern separation and completion computations. Circuit mechanisms supporting these operations remain enigmatic. We show lateral entorhinal cortex glutamatergic (LEC GLU ) and GABAergic (LEC GABA ) projections are essential for object recognition memory. Silencing LEC GLU during in vivo two-photon imaging increased the population of active CA3 pyramidal cells but decreased activity rates, suggesting a sparse coding function through local inhibition. Silencing LEC GLU also decreased place cell remapping between different environments validating this circuit drives pattern separation and context discrimination. Optogenetic circuit mapping confirmed that LEC GLU drives dominant feedforward inhibition to prevent CA3 somatic and dendritic spikes. However, conjunctively active LEC GABA suppresses this local inhibition to disinhibit CA3 pyramidal neuron soma and selectively boost integrative output of LEC and CA3 recurrent network. LEC GABA thus promotes pattern completion and context generalization. Indeed, without this disinhibitory input, CA3 place maps show decreased similarity between contexts. Our findings provide circuit mechanisms whereby long-range glutamatergic and GABAergic cortico-hippocampal inputs bidirectionally modulate pattern separation and completion, providing neuronal representations with a dynamic range for context discrimination and generalization.
Collapse
|
52
|
Nishiyama H, Nishiyama N, Zemelman BV. Loss of Purkinje cells in the developing cerebellum strengthens the cerebellothalamic synapses. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.01.564864. [PMID: 37961231 PMCID: PMC10635038 DOI: 10.1101/2023.11.01.564864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Abstract
Cerebellar damage early in life often causes long-lasting motor, social, and cognitive impairments, suggesting the roles of the cerebellum in developing a broad spectrum of behaviors. This recent finding has promoted research on how cerebellar damage affects the development of the cerebral cortex, the brain region responsible for higher-order control of all behaviors. However, the cerebral cortex is not directly connected to the cerebellum. The thalamus is the direct postsynaptic target of the cerebellum, sending cerebellar outputs to the cerebral cortex. Despite its crucial position in cerebello-cerebral interaction, thalamic susceptibility to cerebellar damage remains largely unclear. Here, we studied the consequences of early cerebellar perturbation on thalamic development. Whole-cell patch-clamp recordings showed that the synaptic organization of the cerebellothlamic circuit is similar to that of the primary sensory thalamus, in which aberrant sensory activity alters synaptic circuit formation. The hemizygous deletion of the tuberous sclerosis complex-1 ( Tsc1 ) gene in the Purkinje cell-known to cause Purkinje cell hypoactivity and autistic behaviors-did not alter cerebellothalamic synapses or intrinsic membrane properties of thalamic neurons. However, the ablation of Purkinje cells in the developing cerebellum strengthened the cerebellothalamic synapses and enhanced thalamic suprathreshold activities. These results suggest that the cerebellothalamic circuit is resistant to moderate perturbation in the developing cerebellum, such as the reduced firing rate of Purkinje cells, and that autistic behaviors are not necessarily linked to thalamic abnormality. Still, Purkinje cell loss alters the thalamic circuit, suggesting the vulnerability of the thalamus to substantial disturbance in the developing cerebellum.
Collapse
|
53
|
Gonzalez TJ, Mitchell-Dick A, Blondel LO, Fanous MM, Hull JA, Oh DK, Moller-Tank S, Castellanos Rivera RM, Piedrahita JA, Asokan A. Structure-guided AAV capsid evolution strategies for enhanced CNS gene delivery. Nat Protoc 2023; 18:3413-3459. [PMID: 37735235 DOI: 10.1038/s41596-023-00875-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 06/13/2023] [Indexed: 09/23/2023]
Abstract
Over the past 5 years, our laboratory has systematically developed a structure-guided library approach to evolve new adeno-associated virus (AAV) capsids with altered tissue tropism, higher transduction efficiency and the ability to evade pre-existing humoral immunity. Here, we provide a detailed protocol describing two distinct evolution strategies using structurally divergent AAV serotypes as templates, exemplified by improving CNS gene transfer efficiency in vivo. We outline four major components of our strategy: (i) structure-guided design of AAV capsid libraries, (ii) AAV library production, (iii) library cycling in single versus multiple animal models, followed by (iv) evaluation of lead AAV vector candidates in vivo. The protocol spans ~95 d, excluding gene expression analysis in vivo, and can vary depending on user experience, resources and experimental design. A distinguishing attribute of the current protocol is the focus on providing biomedical researchers with 3D structural information to guide evolution of precise 'hotspots' on AAV capsids. Furthermore, the protocol outlines two distinct methods for AAV library evolution consisting of adenovirus-enabled infectious cycling in a single species and noninfectious cycling in a cross-species manner. Notably, our workflow can be seamlessly merged with other RNA transcript-based library strategies and tailored for tissue-specific capsid selection. Overall, the procedures outlined herein can be adapted to expand the AAV vector toolkit for genetic manipulation of animal models and development of human gene therapies.
Collapse
Affiliation(s)
- Trevor J Gonzalez
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA
| | | | - Leo O Blondel
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Marco M Fanous
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Joshua A Hull
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Daniel K Oh
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA
| | - Sven Moller-Tank
- Gene Therapy Center, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Jorge A Piedrahita
- North Carolina State University College of Veterinary Medicine, Raleigh, NC, USA
| | - Aravind Asokan
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine, Durham, NC, USA.
- Department of Surgery, Duke University School of Medicine, Durham, NC, USA.
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
54
|
Wu YQ, Zhang CS, Xiong J, Cai DQ, Wang CZ, Wang Y, Liu YH, Wang Y, Li Y, Wu J, Wu J, Lan B, Wang X, Chen S, Cao X, Wei X, Hu HH, Guo H, Yu Y, Ghafoor A, Xie C, Wu Y, Xu Z, Zhang C, Zhu M, Huang X, Sun X, Lin SY, Piao HL, Zhou J, Lin SC. Low glucose metabolite 3-phosphoglycerate switches PHGDH from serine synthesis to p53 activation to control cell fate. Cell Res 2023; 33:835-850. [PMID: 37726403 PMCID: PMC10624847 DOI: 10.1038/s41422-023-00874-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/30/2023] [Indexed: 09/21/2023] Open
Abstract
Glycolytic intermediary metabolites such as fructose-1,6-bisphosphate can serve as signals, controlling metabolic states beyond energy metabolism. However, whether glycolytic metabolites also play a role in controlling cell fate remains unexplored. Here, we find that low levels of glycolytic metabolite 3-phosphoglycerate (3-PGA) can switch phosphoglycerate dehydrogenase (PHGDH) from cataplerosis serine synthesis to pro-apoptotic activation of p53. PHGDH is a p53-binding protein, and when unoccupied by 3-PGA interacts with the scaffold protein AXIN in complex with the kinase HIPK2, both of which are also p53-binding proteins. This leads to the formation of a multivalent p53-binding complex that allows HIPK2 to specifically phosphorylate p53-Ser46 and thereby promote apoptosis. Furthermore, we show that PHGDH mutants (R135W and V261M) that are constitutively bound to 3-PGA abolish p53 activation even under low glucose conditions, while the mutants (T57A and T78A) unable to bind 3-PGA cause constitutive p53 activation and apoptosis in hepatocellular carcinoma (HCC) cells, even in the presence of high glucose. In vivo, PHGDH-T57A induces apoptosis and inhibits the growth of diethylnitrosamine-induced mouse HCC, whereas PHGDH-R135W prevents apoptosis and promotes HCC growth, and knockout of Trp53 abolishes these effects above. Importantly, caloric restriction that lowers whole-body glucose levels can impede HCC growth dependent on PHGDH. Together, these results unveil a mechanism by which glucose availability autonomously controls p53 activity, providing a new paradigm of cell fate control by metabolic substrate availability.
Collapse
Affiliation(s)
- Yu-Qing Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chen-Song Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Jinye Xiong
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Dong-Qi Cai
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Chen-Zhe Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yu Wang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yan-Hui Liu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yu Wang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Yiming Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Jian Wu
- Department of Hepatobiliary and Pancreatic Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Jianfeng Wu
- Laboratory Animal Research Center, Xiamen University, Xiamen, Fujian, China
| | - Bin Lan
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Xiamen, Fujian, China
| | - Xuefeng Wang
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Xiamen, Fujian, China
| | - Siwei Chen
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xianglei Cao
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiaoyan Wei
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Hui-Hui Hu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Huiling Guo
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yaxin Yu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Abdul Ghafoor
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Changchuan Xie
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yaying Wu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zheni Xu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Cixiong Zhang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Mingxia Zhu
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xi Huang
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Xiufeng Sun
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Shu-Yong Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Hai-Long Piao
- CAS Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian, Liaoning, China
| | - Jianyin Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Zhongshan Hospital, Xiamen University, Xiamen, Fujian, China
| | - Sheng-Cai Lin
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
55
|
Kashiwakura Y, Endo K, Ugajin A, Kikuchi T, Hishikawa S, Nakamura H, Katakai Y, Baatartsogt N, Hiramoto T, Hayakawa M, Kamoshita N, Yamazaki S, Kume A, Mori H, Sata N, Sakata Y, Muramatsu SI, Ohmori T. Efficient gene transduction in pigs and macaques with the engineered AAV vector AAV.GT5 for hemophilia B gene therapy. Mol Ther Methods Clin Dev 2023; 30:502-514. [PMID: 37693948 PMCID: PMC10491835 DOI: 10.1016/j.omtm.2023.08.016] [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: 02/17/2023] [Accepted: 08/18/2023] [Indexed: 09/12/2023]
Abstract
Gene therapy using adeno-associated virus (AAV)-based vectors has become a realistic therapeutic option for hemophilia. We examined the potential of a novel engineered liver-tropic AAV3B-based vector, AAV.GT5, for hemophilia B gene therapy. In vitro transduction with AAV.GT5 in human hepatocytes was more than 100 times higher than with AAV-Spark100, another bioengineered vector used in a clinical trial. However, liver transduction following intravenous injection of these vectors was similar in mice with a humanized liver and in macaques. This discrepancy was due to the low recovery and short half-life of AAV.GT5 in blood, depending on the positive charge of the heparin-binding site in the capsid. Bypassing systemic clearance with the intra-hepatic vascular administration of AAV.GT5, but not AAV-Spark100, enhanced liver transduction in pigs and macaques. AAV.GT5 did not develop neutralizing antibodies (NAbs) in two of four animals, while AAV-Spark100 induced serotype-specific NAbs in all macaques tested (4 of 4). The NAbs produced after AAV-Spark100 administration were relatively serotype specific, and challenge with AAV.GT5 through the hepatic artery successfully boosted liver transduction in one animal previously administered AAV-Spark100. In summary, AAV.GT5 showed different vector kinetics and NAb induction compared with AAV-Spark100, and intra-hepatic vascular administration may minimize the vector dose required and vector dissemination.
Collapse
Affiliation(s)
- Yuji Kashiwakura
- Department of Biochemistry, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Kazuhiro Endo
- Department of Surgery, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Center for Development of Advanced Medical Technology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Atsushi Ugajin
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Tomohiro Kikuchi
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Shuji Hishikawa
- Department of Surgery, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Center for Development of Advanced Medical Technology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Hitoyasu Nakamura
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Yuko Katakai
- The Corporation for Production and Research of Laboratory Primates, 1-16-2 Sakura, Tsukuba, Ibaraki 305-0003, Japan
| | - Nemekhbayar Baatartsogt
- Department of Biochemistry, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Takafumi Hiramoto
- Department of Biochemistry, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Morisada Hayakawa
- Department of Biochemistry, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Center for Gene Therapy Research, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Nobuhiko Kamoshita
- Department of Biochemistry, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Center for Gene Therapy Research, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Shoji Yamazaki
- Clinical Research Center, Jichi Medical University Hospital, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Akihiro Kume
- Center for Gene Therapy Research, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Clinical Research Center, Jichi Medical University Hospital, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Harushi Mori
- Department of Radiology, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Naohiro Sata
- Department of Surgery, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Center for Development of Advanced Medical Technology, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Yoichi Sakata
- Department of Biochemistry, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| | - Shin-ichi Muramatsu
- Center for Gene Therapy Research, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Division of Neurological Gene Therapy, Center for Open Innovation, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo 108-8639, Japan
| | - Tsukasa Ohmori
- Department of Biochemistry, Jichi Medical University School of Medicine, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
- Center for Gene Therapy Research, Jichi Medical University, 3311-1 Yakushiji, Shimotsuke, Tochigi 329-0498, Japan
| |
Collapse
|
56
|
Han H, Yang Y, Jiao Y, Qi H, Han Z, Wang L, Dong L, Tian J, Vanhaesebroeck B, Li X, Liu J, Ma G, Lei H. Leverage of nuclease-deficient CasX for preventing pathological angiogenesis. MOLECULAR THERAPY. NUCLEIC ACIDS 2023; 33:738-748. [PMID: 37662968 PMCID: PMC10469388 DOI: 10.1016/j.omtn.2023.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/03/2023] [Indexed: 09/05/2023]
Abstract
Gene editing with a CRISPR/Cas system is a novel potential strategy for treating human diseases. Pharmacological inhibition of phosphoinositide 3-kinase (PI3K) δ suppresses retinal angiogenesis in a mouse model of oxygen-induced retinopathy. Here we show that an innovative system of adeno-associated virus (AAV)-mediated CRISPR/nuclease-deficient (d)CasX fused with the Krueppel-associated box (KRAB) domain is leveraged to block (81.2% ± 6.5%) in vitro expression of p110δ, the catalytic subunit of PI3Kδ, encoded by Pik3cd. This CRISPR/dCasX-KRAB (4, 269 bp) system is small enough to be fit into a single AAV vector. We then document that recombinant AAV serotype (rAAV)1 efficiently transduces vascular endothelial cells from pathologic retinal vessels, which show high expression of p110δ; furthermore, we demonstrate that blockade of retinal p110δ expression by intravitreally injected rAAV1-CRISPR/dCasX-KRAB targeting the Pik3cd promoter prevents (32.1% ± 5.3%) retinal p110δ expression as well as pathological retinal angiogenesis in a mouse model of oxygen-induced retinopathy. These data establish a strong foundation for treating pathological angiogenesis by AAV-mediated CRISPR interference with p110δ expression.
Collapse
Affiliation(s)
- Haote Han
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, People’s Republic of China
| | - Yanhui Yang
- Ningxia Key Laboratory of Prevention and Control of Common Infectious Diseases, the School of Basic Medical Sciences, Ningxia Medical University, Yinchuan 750004, People’s Republic of China
| | - Yunjuan Jiao
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha 410013, People’s Republic of China
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Hui Qi
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, People’s Republic of China
| | - Zhuo Han
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, People’s Republic of China
| | - Luping Wang
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, People’s Republic of China
| | - Lijun Dong
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, People’s Republic of China
| | - Jingkui Tian
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Cancer Hospital of the University of Chinese Academy of Sciences, Zhejiang Cancer Hospital, Hangzhou 310000, People’s Republic of China
| | | | - Xiaopeng Li
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Junwen Liu
- Department of Histology and Embryology, Xiangya School of Medicine, Central South University, Changsha 410013, People’s Republic of China
| | - Gaoen Ma
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
| | - Hetian Lei
- The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang 453003, China
- Shenzhen Eye Hospital, Jinan University, Shenzhen Eye Institute, Shenzhen 518000, People’s Republic of China
| |
Collapse
|
57
|
Zhang YM, Zong HC, Qi YB, Chang LL, Gao YN, Zhou T, Yin T, Liu M, Pan KJ, Chen WG, Guo HR, Guo F, Peng YM, Wang M, Feng LY, Zang Y, Li Y, Li J. Anxiolytic effect of antidiabetic metformin is mediated by AMPK activation in mPFC inhibitory neurons. Mol Psychiatry 2023; 28:3955-3965. [PMID: 37798418 PMCID: PMC10730396 DOI: 10.1038/s41380-023-02283-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 09/05/2023] [Accepted: 09/21/2023] [Indexed: 10/07/2023]
Abstract
Diabetic patients receiving the antidiabetic drug metformin have been observed to exhibit a lower prevalence of anxiety disorders, yet the precise mechanism behind this phenomenon is unclear. In our study, we found that anxiety induces a region-specific reduction in AMPK activity in the medial prefrontal cortex (mPFC). Concurrently, transgenic mice with brain-specific AMPK knockout displayed abnormal anxiety-like behaviors. Treatment with metformin or the overexpression of AMPK restored normal AMPK activity in the mPFC and mitigated social stress-induced anxiety-like behaviors. Furthermore, the specific genetic deletion of AMPK in the mPFC not only instigated anxiety in mice but also nullified the anxiolytic effects of metformin. Brain slice recordings revealed that GABAergic excitation and the resulting inhibitory inputs to mPFC pyramidal neurons were selectively diminished in stressed mice. This reduction led to an excitation-inhibition imbalance, which was effectively reversed by metformin treatment or AMPK overexpression. Moreover, the genetic deletion of AMPK in the mPFC resulted in a similar defect in GABAergic inhibitory transmission and a consequent hypo-inhibition of mPFC pyramidal neurons. We also generated a mouse model with AMPK knockout specific to GABAergic neurons. The anxiety-like behaviors in this transgenic mouse demonstrated the unique role of AMPK in the GABAergic system in relation to anxiety. Therefore, our findings suggest that the activation of AMPK in mPFC inhibitory neurons underlies the anxiolytic effects of metformin, highlighting the potential of this primary antidiabetic drug as a therapeutic option for treating anxiety disorders.
Collapse
Affiliation(s)
- Yong-Mei Zhang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hai-Chao Zong
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ying-Bei Qi
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Liu-Liu Chang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ya-Nan Gao
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Ting Zhou
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Tao Yin
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Meng Liu
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Kai-Jun Pan
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Wen-Gang Chen
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China
| | - Hao-Ran Guo
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Fei Guo
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Receptor Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yan-Min Peng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Min Wang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lin-Yin Feng
- University of Chinese Academy of Sciences, Beijing, 100049, China
- CAS Key Laboratory of Receptor Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China
| | - Yi Zang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Lin Gang Laboratory, Shanghai, 200031, China.
| | - Yang Li
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- CAS Key Laboratory of Receptor Research, Department of Neuropharmacology, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Jia Li
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai, 201203, China.
- School of Pharmaceutical Science and Technology, Hangzhou Institute for Advanced Study, University of Chinese Academy of Sciences, Hangzhou, Zhejiang, 310024, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- Institute of Pharmaceutical Science, China Pharmaceutical University, Nanjing, Jiangsu, 210009, China.
- Zhongshan Institute for Drug Discovery, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Zhongshan Tsuihang New District, Zhongshan, Guangzhou, 528400, China.
- Drug Discovery Shandong Laboratory, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai, Shandong, 264117, China.
| |
Collapse
|
58
|
Wada M, Uchida N, Posadas-Herrera G, Hayashita-Kinoh H, Tsunekawa Y, Hirai Y, Okada T. Large-scale purification of functional AAV particles packaging the full genome using short-term ultracentrifugation with a zonal rotor. Gene Ther 2023; 30:641-648. [PMID: 36977769 PMCID: PMC10457186 DOI: 10.1038/s41434-023-00398-x] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 02/17/2023] [Accepted: 03/10/2023] [Indexed: 03/30/2023]
Abstract
Adeno-associated virus (AAV) vector-based gene therapy is potentially curative for various genetic diseases; however, the development of a scalable purification method for full-genome AAV vectors remains crucial to increase productivity and reduce cost of GMP production. In this study, we developed a large-scale short-term purification method for functional full-genome AAV particles by using 2-step cesium chloride (CsCl) density-gradient ultracentrifugation with a zonal rotor. The 2-step CsCl method with a zonal rotor improves separation between empty and full-genome AAV particles, reducing the ultracentrifugation time (4-5 h) and increasing the AAV volume for purification. The highly purified full-genome AAV particles were confirmed by analytical ultracentrifugation (AUC), droplet digital PCR (ddPCR) in the whole region of the AAV vector genome, transduction efficiency in target cells, and transmission electronic microscopy (TEM). The high-purity AAV9 particles were obtained using culture supernatant during vector preparation rather than cell lysate. CsCl could be simply removed by a hydroxyapatite column. Interestingly, ddPCR analysis revealed that "empty" AAV particles contain small fragments of the inverted terminal repeat (ITR), probably due to unexpected packaging of Rep-mediated ITR fragments. This large-scale functional AAV vector purification with ultracentrifugation would be effective for gene therapy.
Collapse
Affiliation(s)
- Mikako Wada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Naoya Uchida
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Guillermo Posadas-Herrera
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Hiromi Hayashita-Kinoh
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yuji Tsunekawa
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yukihiko Hirai
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Takashi Okada
- Division of Molecular and Medical Genetics, Center for Gene and Cell Therapy, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan.
| |
Collapse
|
59
|
Li Q, Takeuchi Y, Wang J, Gellért L, Barcsai L, Pedraza LK, Nagy AJ, Kozák G, Nakai S, Kato S, Kobayashi K, Ohsawa M, Horváth G, Kékesi G, Lőrincz ML, Devinsky O, Buzsáki G, Berényi A. Reinstating olfactory bulb-derived limbic gamma oscillations alleviates depression-like behavioral deficits in rodents. Neuron 2023; 111:2065-2075.e5. [PMID: 37164008 PMCID: PMC10321244 DOI: 10.1016/j.neuron.2023.04.013] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 01/31/2023] [Accepted: 04/12/2023] [Indexed: 05/12/2023]
Abstract
Although the etiology of major depressive disorder remains poorly understood, reduced gamma oscillations is an emerging biomarker. Olfactory bulbectomy, an established model of depression that reduces limbic gamma oscillations, suffers from non-specific effects of structural damage. Here, we show that transient functional suppression of olfactory bulb neurons or their piriform cortex efferents decreased gamma oscillation power in limbic areas and induced depression-like behaviors in rodents. Enhancing transmission of gamma oscillations from olfactory bulb to limbic structures by closed-loop electrical neuromodulation alleviated these behaviors. By contrast, silencing gamma transmission by anti-phase closed-loop stimulation strengthened depression-like behaviors in naive animals. These induced behaviors were neutralized by ketamine treatment that restored limbic gamma power. Taken together, our results reveal a causal link between limbic gamma oscillations and depression-like behaviors in rodents. Interfering with these endogenous rhythms can affect behaviors in rodent models of depression, suggesting that restoring gamma oscillations may alleviate depressive symptoms.
Collapse
Affiliation(s)
- Qun Li
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary
| | - Yuichi Takeuchi
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan; Department of Physiology, Osaka City University Graduate School of Medicine, Osaka 545-8585, Japan; Department of Biopharmaceutical Sciences and Pharmacy, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo 060-0812, Japan
| | - Jiale Wang
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; Faculty of Agriculture, University of Szeged, Szeged 6720, Hungary
| | - Levente Gellért
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary
| | - Livia Barcsai
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary; Neunos Inc, Boston, MA 02108, USA
| | - Lizeth K Pedraza
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Anett J Nagy
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary; Neunos Inc, Boston, MA 02108, USA
| | - Gábor Kozák
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Shinya Nakai
- Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Shigeki Kato
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Kazuto Kobayashi
- Department of Molecular Genetics, Institute of Biomedical Sciences, Fukushima Medical University School of Medicine, Fukushima 960-1295, Japan
| | - Masahiro Ohsawa
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; Department of Neuropharmacology, Graduate School of Pharmaceutical Sciences, Nagoya City University, Nagoya 467-8603, Japan
| | - Gyöngyi Horváth
- Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Gabriella Kékesi
- Department of Physiology, University of Szeged, Szeged 6720, Hungary
| | - Magor L Lőrincz
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; Department of Physiology, Anatomy and Neuroscience, Faculty of Sciences University of Szeged, Szeged 6726, Hungary; Neuroscience Division, Cardiff University, Museum Avenue, Cardiff CF10 3AX, UK
| | - Orrin Devinsky
- Department of Neurology, NYU Langone Comprehensive Epilepsy Center, NYU Grossman School of Medicine, New York, NY 10016, USA
| | - György Buzsáki
- Neuroscience Institute, New York University, New York, NY 10016, USA
| | - Antal Berényi
- MTA-SZTE "Momentum" Oscillatory Neuronal Networks Research Group, Department of Physiology, University of Szeged, Szeged 6720, Hungary; HCEMM-SZTE Magnetotherapeutics Research Group, University of Szeged, Szeged 6720, Hungary; Neunos Inc, Boston, MA 02108, USA; Neuroscience Institute, New York University, New York, NY 10016, USA.
| |
Collapse
|
60
|
Albarran E, Sun Y, Liu Y, Raju K, Dong A, Li Y, Wang S, Südhof TC, Ding JB. Postsynaptic synucleins mediate endocannabinoid signaling. Nat Neurosci 2023; 26:997-1007. [PMID: 37248337 PMCID: PMC10244176 DOI: 10.1038/s41593-023-01345-0] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Accepted: 04/21/2023] [Indexed: 05/31/2023]
Abstract
Endocannabinoids are among the most powerful modulators of synaptic transmission throughout the nervous system, and yet little is understood about the release of endocannabinoids from postsynaptic compartments. Here we report an unexpected finding that endocannabinoid release requires synucleins, key contributors to Parkinson's disease. We show that endocannabinoids are released postsynaptically by a synuclein-dependent and SNARE-dependent mechanism. Specifically, we found that synuclein deletion blocks endocannabinoid-dependent synaptic plasticity; this block is reversed by postsynaptic expression of wild-type but not of mutant α-synuclein. Whole-cell recordings and direct optical monitoring of endocannabinoid signaling suggest that the synuclein deletion specifically blocks endocannabinoid release. Given the presynaptic role of synucleins in regulating vesicle lifecycle, we hypothesize that endocannabinoids are released via a membrane interaction mechanism. Consistent with this hypothesis, postsynaptic expression of tetanus toxin light chain, which cleaves synaptobrevin SNAREs, also blocks endocannabinoid-dependent signaling. The unexpected finding that endocannabinoids are released via a synuclein-dependent mechanism is consistent with a general function of synucleins in membrane trafficking and adds a piece to the longstanding puzzle of how neurons release endocannabinoids to induce synaptic plasticity.
Collapse
Affiliation(s)
- Eddy Albarran
- Neurosciences Graduate Program, Stanford University, Stanford, CA, USA
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Yue Sun
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Yu Liu
- Department of Neurosurgery, Stanford University, Stanford, CA, USA
| | - Karthik Raju
- Department of Molecular and Cellular Physiology, Stanford University and Howard Hughes Medical Institute, Stanford, CA, USA
| | - Ao Dong
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Yulong Li
- State Key Laboratory of Membrane Biology, Peking University School of Life Sciences, Beijing, China
- PKU-IDG/McGovern Institute for Brain Research, Beijing, China
- Peking-Tsinghua Center for Life Sciences, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- Chinese Institute for Brain Research, Beijing, China
| | - Sui Wang
- Department of Ophthalmology, Mary M. and Sash A. Spencer Center for Vision Research, Byers Eye Institute, Stanford University, Stanford, CA, USA
| | - Thomas C Südhof
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
- Department of Molecular and Cellular Physiology, Stanford University and Howard Hughes Medical Institute, Stanford, CA, USA.
| | - Jun B Ding
- Department of Neurosurgery, Stanford University, Stanford, CA, USA.
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA, USA.
| |
Collapse
|
61
|
Hoffmann MD, Zdechlik AC, He Y, Nedrud D, Aslanidi G, Gordon W, Schmidt D. Multiparametric domain insertional profiling of Adeno-Associated Virus VP1. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.19.537549. [PMID: 37131661 PMCID: PMC10153220 DOI: 10.1101/2023.04.19.537549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Evolved properties of Adeno-Associated Virus (AAV), such as broad tropism and immunogenicity in humans, are barriers to AAV-based gene therapy. Previous 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 large, 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. There was strong positional, domain-type, and fitness phenotype dependence of insertion permissibility, which clustered into correlated structural units that we could link to distinct roles in AAV assembly, stability, and infectivity. We also identified new engineerable hotspots of AAV that facilitate the covalent attachment of binding scaffolds, which may represent an alternative approach to re-direct AAV tropism.
Collapse
|
62
|
Dong L, Zhang RH, Wu HT, Li HY, Zhou WD, Shi XH, Yu CY, Li YT, Li YF, Jonas JB, Wei WB. Intravitreal Short-Hairpin RNA Attenuated Adeno-Associated Virus-Induced Knockdown of Amphiregulin and Axial Elongation in Experimental Myopia. Invest Ophthalmol Vis Sci 2023; 64:11. [PMID: 37040096 PMCID: PMC10103729 DOI: 10.1167/iovs.64.4.11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Accepted: 03/13/2023] [Indexed: 04/12/2023] Open
Abstract
Background Epidermal growth factor (EGF) and its family members have been reported to be involved in myopic axial elongation. We examined whether short hairpin RNA attenuated adeno-associated virus (shRNA-AAV)-induced knockdown of amphiregulin, an EGF family member, has an influence on axial elongation. Methods Three-week-old pigmented guinea pigs underwent lens-induced myopization (LIM) without additional intervention (LIM group; n = 10 animals) or additionally received into their right eyes at baseline an intravitreal injection of scramble shRNA-AAV (5 × 1010 vector genome [vg]) (LIM + Scr-shRNA group; n = 10) or of amphiregulin (AR)-shRNA-AAV (5 × 1010 vg/5 µL) (LIM + AR-shRNA-AAV group; n = 10), or they received an injection of AR-shRNA-AAV at baseline and three weekly amphiregulin injections (20 ng/5 µL) (LIM + AR-shRNA-AAV + AR group; n = 10). The left eyes received equivalent intravitreal injections of phosphate-buffered saline. Four weeks after baseline, the animals were sacrificed. Results At study end, interocular axial length difference was higher (P < 0.001), choroid and retina were thicker (P < 0.05), and relative expression of amphiregulin and p-PI3K, p-p70S6K, and p-ERK1/2 was lower (P < 0.05) in the LIM + AR-shRNA-AAV group than in any other group. The other groups did not differ significantly when compared with each other. In the LIM + AR-shRNA-AAV group, the interocular axial length difference increased with longer study duration. TUNEL assay did not reveal significant differences among all groups in retinal apoptotic cell density. In vitro retinal pigment epithelium cell proliferation and migration were the lowest (P < 0.05) in the LIM + AR-shRNA-AAV group, followed by the LIM + AR-shRNA-AAV + AR group. Conclusions shRNA-AAV-induced knockdown of amphiregulin expression, in association with suppression of epidermal growth factor receptor signaling, attenuated axial elongation in guinea pigs with LIM. The finding supports the notion of EGF playing a role in axial elongation.
Collapse
Affiliation(s)
- Li Dong
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Rui-Heng Zhang
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Hao-Tian Wu
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - He-Yan Li
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Wen-Da Zhou
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Xu-Han Shi
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Chu-Yao Yu
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yi-Tong Li
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Yi-Fan Li
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Jost B. Jonas
- Department of Ophthalmology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Institute of Molecular and Clinical Ophthalmology Basel, Switzerland
| | - Wen-Bin Wei
- Beijing Tongren Eye Center, Beijing Key Laboratory of Intraocular Tumor Diagnosis and Treatment, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Beijing Ophthalmology & Visual Sciences Key Lab, Beijing Tongren Hospital, Capital Medical University, Beijing, China
- Medical Artificial Intelligence Research and Verification Key Laboratory of the Ministry of Industry and Information Technology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
63
|
Rosenberg N, Reva M, Binda F, Restivo L, Depierre P, Puyal J, Briquet M, Bernardinelli Y, Rocher AB, Markram H, Chatton JY. Overexpression of UCP4 in astrocytic mitochondria prevents multilevel dysfunctions in a mouse model of Alzheimer's disease. Glia 2023; 71:957-973. [PMID: 36537556 DOI: 10.1002/glia.24317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 10/31/2022] [Accepted: 11/25/2022] [Indexed: 02/16/2023]
Abstract
Alzheimer's disease (AD) is becoming increasingly prevalent worldwide. It represents one of the greatest medical challenges as no pharmacologic treatments are available to prevent disease progression. Astrocytes play crucial functions within neuronal circuits by providing metabolic and functional support, regulating interstitial solute composition, and modulating synaptic transmission. In addition to these physiological functions, growing evidence points to an essential role of astrocytes in neurodegenerative diseases like AD. Early-stage AD is associated with hypometabolism and oxidative stress. Contrary to neurons that are vulnerable to oxidative stress, astrocytes are particularly resistant to mitochondrial dysfunction and are therefore more resilient cells. In our study, we leveraged astrocytic mitochondrial uncoupling and examined neuronal function in the 3xTg AD mouse model. We overexpressed the mitochondrial uncoupling protein 4 (UCP4), which has been shown to improve neuronal survival in vitro. We found that this treatment efficiently prevented alterations of hippocampal metabolite levels observed in AD mice, along with hippocampal atrophy and reduction of basal dendrite arborization of subicular neurons. This approach also averted aberrant neuronal excitability observed in AD subicular neurons and preserved episodic-like memory in AD mice assessed in a spatial recognition task. These findings show that targeting astrocytes and their mitochondria is an effective strategy to prevent the decline of neurons facing AD-related stress at the early stages of the disease.
Collapse
Affiliation(s)
- Nadia Rosenberg
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Maria Reva
- Blue Brain Project (BBP), École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Francesca Binda
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Leonardo Restivo
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Pauline Depierre
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Julien Puyal
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Marc Briquet
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | | | - Anne-Bérengère Rocher
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland
| | - Henry Markram
- Blue Brain Project (BBP), École polytechnique fédérale de Lausanne (EPFL), Geneva, Switzerland
| | - Jean-Yves Chatton
- Department of Fundamental Neurosciences, University of Lausanne, Lausanne, Switzerland.,Cellular Imaging Facility, University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
64
|
Alshammari AH, Masuo Y, Yoshino S, Yamashita R, Ishimoto T, Fujita KI, Kato Y. Adeno-associated virus-mediated knockdown demonstrates the major role of hepatic Bcrp in the overall disposition of the active metabolite of the tyrosine kinase inhibitor regorafenib in mice. Drug Metab Pharmacokinet 2023; 49:100483. [PMID: 36724604 DOI: 10.1016/j.dmpk.2022.100483] [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: 02/09/2022] [Revised: 09/13/2022] [Accepted: 11/01/2022] [Indexed: 11/07/2022]
Abstract
Breast cancer resistance protein (BCRP) is expressed on hepatic bile canalicular membranes; however, its impact on substrate drug disposition is limited. This study proposes an in vivo knockdown approach using adeno-associated virus encoding short hairpin RNA (shRNA) targeting the bcrp gene (AAV-shBcrp) to clarify the substrate, the overall disposition of which is largely governed by hepatic Bcrp. The disposition of the tyrosine kinase inhibitor, regorafenib, was first examined in bcrp gene knockout (Bcrp-/-) and wild-type (WT) mice, as it was sequentially converted to active metabolites M - 2 and M - 5, which are BCRP substrates. After oral administration of regorafenib, plasma and liver concentrations of M - 5, but not regorafenib, were higher in Bcrp-/- than WT mice. To directly examine the role of hepatic Bcrp in M - 5 disposition, M - 5 was intravenously injected into mice three weeks after the intravenous injection of AAV-shBcrp, when mRNA of Bcrp in the liver (but not the small intestine) was downregulated. AAV-shBcrp-treated mice showed higher M - 5 concentration in plasma and liver, but lower biliary excretion than the control mice, indicating the fundamental role of hepatic Bcrp in M - 5 disposition. This is the first application of AAV-knockdown strategy to clarify the pharmacokinetic role of xenobiotic efflux transporters in the liver.
Collapse
Affiliation(s)
- Aya Hasan Alshammari
- Faculty of Pharmacy, Kakuma-machi, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Yusuke Masuo
- Faculty of Pharmacy, Kakuma-machi, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Shotaro Yoshino
- Faculty of Pharmacy, Kakuma-machi, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Reiya Yamashita
- Faculty of Pharmacy, Kakuma-machi, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Takahiro Ishimoto
- Faculty of Pharmacy, Kakuma-machi, Kanazawa University, Kanazawa, 920-1192, Japan
| | - Ken-Ichi Fujita
- School of Pharmacy, Showa University, 1-5-8 Hatanodai, Shinagawa-ku, Tokyo, 142-8555, Japan
| | - Yukio Kato
- Faculty of Pharmacy, Kakuma-machi, Kanazawa University, Kanazawa, 920-1192, Japan.
| |
Collapse
|
65
|
Xie Y, Zhou J, Wang LL, Zhang CL, Chen B. New AAV tools fail to detect Neurod1-mediated neuronal conversion of Müller glia and astrocytes in vivo. EBioMedicine 2023; 90:104531. [PMID: 36947961 PMCID: PMC10033723 DOI: 10.1016/j.ebiom.2023.104531] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 03/03/2023] [Accepted: 03/03/2023] [Indexed: 03/24/2023] Open
Abstract
BACKGROUND Reprogramming resident glial cells to convert them into neurons in vivo represents a potential therapeutic strategy that could replenish lost neurons, repair damaged neural circuits, and restore function. AAV (adeno-associated virus)-based expression systems are powerful tools for in vivo gene delivery in glia-to-neuron reprogramming, however, recent studies show that AAV-based gene delivery of Neurod1 into the mouse brain can cause severe leaky expression into endogenous neurons leading to misinterpretation of glia-to-neuron conversion. METHODS AAV-based delivery systems were modified for improved in vivo delivery of Neurod1, Math5, Ascl1, and Neurog2 in the adult mouse retina and brain. To examine whether bona fide glia-to-neuron conversion occurs, stringent fate mapping experiments were performed to trace the lineage of glial cells. FINDINGS The neuronal leakage is prevalent after AAV-GFAP-mediated delivery of Neurod1, Math5, Ascl1, and Neurog2. The transgene-dependent leakage cannot be corrected after lowering the AAV doses, using alterative AAV serotypes or injection routes. Importantly, we report the development of two new AAV-based tools that can significantly reduce neuronal leakage. Using the new AAV-based tools, we provide evidence that Neurod1 gene transfer fails to convert lineage traced glial cells into neurons. INTERPRETATION Stringent fate mapping techniques independently of an AAV-based expression system are the golden standard for tracing the fate of glia cells during neuronal reprogramming. The newly developed AAV-based systems are invaluable tools for glia-to-neuron reprogramming in vivo. FUNDING The work in Chen lab was supported by National Institutes of Health (NIH) grants R01 EY024986 and R01 EY028921, an unrestricted challenge grant from Research to Prevent Blindness, the New York Eye and Ear Infirmary Foundation, and The Harold W. McGraw, Jr. Family Foundation for Vision Research. The work in Zhang lab was supported by NIH (R01 NS127375 and R01 NS117065) and The Decherd Foundation.
Collapse
Affiliation(s)
- Ye Xie
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jing Zhou
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Lei-Lei Wang
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Chun-Li Zhang
- Department of Molecular Biology and Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Bo Chen
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
66
|
Zhuo M, Mao J, David CJ. Protocol to package and concentrate adeno-associated virus serotype 8 for use in autochthonous mouse models of pancreatic cancer. STAR Protoc 2023; 4:102108. [PMID: 36853729 PMCID: PMC9943862 DOI: 10.1016/j.xpro.2023.102108] [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: 10/15/2022] [Revised: 12/23/2022] [Accepted: 01/23/2023] [Indexed: 02/12/2023] Open
Abstract
We have developed an economical and rapid protocol to package and concentrate adeno-associated virus serotype 8, allowing production of high-titer virus for use in vivo within 1 week. When combined with the CRISPR-Cas9 system, this provides a straightforward method for knockout of genes of interest in the pancreas. The method can also be used to express cDNAs in the pancreas. This method shows great potential to accelerate pancreatic cancer research in autochthonous models. For complete details on the use and execution of this protocol, please refer to Li et al. (2021).1.
Collapse
Affiliation(s)
- Meilian Zhuo
- Tsinghua University School of Medicine, Beijing, China; Peking University-Tsinghua Center for Life Sciences, Beijing, China.
| | - Jiangjiao Mao
- Tsinghua University School of Medicine, Beijing, China
| | - Charles J David
- Tsinghua University School of Medicine, Beijing, China; Peking University-Tsinghua Center for Life Sciences, Beijing, China.
| |
Collapse
|
67
|
Yuan Y, Kong W, Liu XM, Shi GH. Gene Therapy Activates Retinal Pigment Epithelium Cell Proliferation for Age-related Macular Degeneration in a Mouse Model. Curr Med Sci 2023; 43:384-392. [PMID: 36944806 DOI: 10.1007/s11596-022-2684-3] [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: 02/10/2022] [Accepted: 06/21/2022] [Indexed: 03/23/2023]
Abstract
OBJECTIVE Age-related macular degeneration (AMD) is a degenerative retinal disease. The degeneration or death of retinal pigment epithelium (RPE) cells is implicated in the pathogenesis of AMD. This study aimed to activate the proliferation of RPE cells in vivo by using an adeno-associated virus (AAV) vector encoding β-catenin to treat AMD in a mouse model. METHODS Mice were intravitreally injected with AAV2/8-Y733F-VMD2-β-catenin for 2 or 4 weeks, and β-catenin expression was measured using immunofluorescence staining, real-time quantitative reverse transcription polymerase chain reaction (PCR), and Western blotting. The function of β-catenin was determined using retinal flat mounts and laser-induced damage models. Finally, the safety of AAV2/8-Y733F-VMD2-β-catenin was evaluated by multiple intravitreal injections. RESULTS AAV2/8-Y733F-VMD2-β-catenin induced the expression of β-catenin in RPE cells. It activated the proliferation of RPE cells and increased cyclin D1 expression. It was beneficial to the recovery of laser-induced damage by activating the proliferation of RPE cells. Furthermore, it could induce apoptosis of RPE cells by increasing the expression of Trp53, Bax and caspase3 while decreasing the expression of Bcl-2. CONCLUSION AAV2/8-Y733F-VMD2-β-catenin increased β-catenin expression in RPE cells, activated RPE cell proliferation, and helped mice heal from laser-induced eye injury. Furthermore, it could induce the apoptosis of RPE cells. Therefore, it may be a safe approach for AMD treatment.
Collapse
Affiliation(s)
- Yun Yuan
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou, 215000, China.
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215000, China.
| | - Wen Kong
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou, 215000, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215000, China
| | - Xiao-Mei Liu
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou, 215000, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215000, China
| | - Guo-Hua Shi
- School of Biomedical Engineering (Suzhou), Division of Life Sciences and Medicine, University of Science and Technology of China, Suzhou, 215000, China
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Science, Suzhou, 215000, China
| |
Collapse
|
68
|
Guo Y, Cao Y, Jardin BD, Zhang X, Zhou P, Guatimosim S, Lin J, Chen Z, Zhang Y, Mazumdar N, Lu F, Ma Q, Lu YW, Zhao M, Wang DZ, Dong E, Pu WT. Ryanodine receptor 2 (RYR2) dysfunction activates the unfolded protein response and perturbs cardiomyocyte maturation. Cardiovasc Res 2023; 119:221-235. [PMID: 35576474 PMCID: PMC10233305 DOI: 10.1093/cvr/cvac077] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 04/03/2022] [Accepted: 05/05/2022] [Indexed: 11/15/2022] Open
Abstract
AIMS Calcium-handling capacity is a major gauge of cardiomyocyte maturity. Ryanodine receptor 2 (RYR2) is the pre-dominant calcium channel that releases calcium from the sarcoplasmic reticulum/endoplasmic reticulum (SR/ER) to activate cardiomyocyte contraction. Although RYR2 was previously implied as a key regulator of cardiomyocyte maturation, the mechanisms remain unclear. The aim of this study is to solve this problem. METHODS AND RESULTS We performed Cas9/AAV9-mediated somatic mutagenesis to knockout RYR2 specifically in cardiomyocytes in mice. We conducted a genetic mosaic analysis to dissect the cell-autonomous function of RYR2 during cardiomyocyte maturation. We found that RYR2 depletion triggered ultrastructural and transcriptomic defects relevant to cardiomyocyte maturation. These phenotypes were associated with the drastic activation of ER stress pathways. The ER stress alleviator tauroursodeoxycholic acid partially rescued the defects in RYR2-depleted cardiomyocytes. Overexpression of ATF4, a key ER stress transcription factor, recapitulated defects in RYR2-depleted cells. Integrative analysis of RNA-Seq and bioChIP-Seq data revealed that protein biosynthesis-related genes are the major direct downstream targets of ATF4. CONCLUSION RYR2-regulated ER homeostasis is essential for cardiomyocyte maturation. Severe ER stress perturbs cardiomyocyte maturation primarily through ATF4 activation. The major downstream effector genes of ATF4 are related to protein biosynthesis.
Collapse
Affiliation(s)
- Yuxuan Guo
- Peking University Health Science Center, School of Basic Medical Sciences, Beijing 100191, China
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Yangpo Cao
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Blake D Jardin
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Xiaoran Zhang
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Pingzhu Zhou
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Silvia Guatimosim
- Department of Physiology and Biophysics, Institute of Biological Sciences, Universidade Federal de Minas Gerais, Av. Antônio Carlos 6627, Belo Horizonte MG - CEP 31270-901, Brazil
| | - Junsen Lin
- Peking University Health Science Center, School of Basic Medical Sciences, Beijing 100191, China
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Zhan Chen
- Peking University Health Science Center, School of Basic Medical Sciences, Beijing 100191, China
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Yueyang Zhang
- Peking University Health Science Center, School of Basic Medical Sciences, Beijing 100191, China
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
| | - Neil Mazumdar
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Fujian Lu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Qing Ma
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Yao-Wei Lu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Mingming Zhao
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing 100191, China
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
| | - Erdan Dong
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
- Key Laboratory of Molecular Cardiovascular Science, Ministry of Education, Beijing 100191, China
- Beijing Key Laboratory of Cardiovascular Receptors Research, Beijing 100191, China
- Department of Cardiology and Institute of Vascular Medicine, Peking University Third Hospital, Beijing 100191, China
- NHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory Peptides, Beijing 100191, China
| | - William T Pu
- Department of Cardiology, Boston Children’s Hospital, 300 Longwood Ave, Boston, MA 02115, USA
- Harvard Stem Cell Institute, 7 Divinity Avenue, Cambridge, MA 02138, USA
| |
Collapse
|
69
|
Kashiwakura Y, Ohmori T. Genome Editing of Murine Liver Hepatocytes by AAV Vector-Mediated Expression of Cas9 In Vivo. Methods Mol Biol 2023; 2637:195-211. [PMID: 36773148 DOI: 10.1007/978-1-0716-3016-7_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Adeno-associated virus (AAV) vectors are attractive tools for gene transfer to the liver and are used as gene therapeutic drugs for inherited disorders. The intravenous injection of an AAV vector harboring the gene of interest driven by the hepatocyte-specific promoter could efficiently express the target gene in liver hepatocytes. The delivery of genome editing tools including Cas9 and gRNA, by the AAV vector, can efficiently disrupt the target gene expression in the liver in vivo by intravenous administration in mice. We can quickly obtain mice lacking specific gene expression in the liver only by administering the AAV vector. The method could be suitable for developing genome editing treatments for inherited disorders and basic research exploring the physiological role of the target gene produced from liver hepatocytes.
Collapse
Affiliation(s)
- Yuji Kashiwakura
- Department of Biochemistry, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan.
| | - Tsukasa Ohmori
- Department of Biochemistry, Jichi Medical University School of Medicine, Shimotsuke, Tochigi, Japan
| |
Collapse
|
70
|
Extra-viral DNA in adeno-associated viral vector preparations induces TLR9-dependent innate immune responses in human plasmacytoid dendritic cells. Sci Rep 2023; 13:1890. [PMID: 36732401 PMCID: PMC9894911 DOI: 10.1038/s41598-023-28830-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 01/25/2023] [Indexed: 02/04/2023] Open
Abstract
Adeno-associated viral (AAV) vector suspensions produced in either human derived HEK cells or in Spodoptera frugiperda (Sf9) insect cells differ in terms of residual host cell components as well as species-specific post-translational modifications displayed on the AAV capsid proteins. Here we analysed the impact of these differences on the immunogenic properties of the vector. We stimulated human plasmacytoid dendritic cells with various lots of HEK cell-produced and Sf9 cell-produced AAV-CMV-eGFP vectors derived from different manufacturers. We found that AAV8-CMV-eGFP as well as AAV2-CMV-eGFP vectors induced lot-specific but not production platform-specific or manufacturer-specific inflammatory cytokine responses. These could be reduced or abolished by blocking toll-like receptor 9 signalling or by enzymatically reducing DNA in the vector lots using DNase. Successful HEK cell transduction by DNase-treated AAV lots and DNA analyses demonstrated that DNase did not affect the integrity of the vector but degraded extra-viral DNA. We conclude that both HEK- and Sf9-cell derived AAV preparations can contain immunogenic extra-viral DNA components which can trigger lot-specific inflammatory immune responses. This suggests that improved strategies to remove extra-viral DNA impurities may be instrumental in reducing the immunogenic properties of AAV vector preparations.
Collapse
|
71
|
Shi D, Wong JKY, Zhu K, Noakes PG, Rammes G. The Anaesthetics Isoflurane and Xenon Reverse the Synaptotoxic Effects of Aβ 1-42 on Megf10-Dependent Astrocytic Synapse Elimination and Spine Density in Ex Vivo Hippocampal Brain Slices. Int J Mol Sci 2023; 24:ijms24020912. [PMID: 36674434 PMCID: PMC9861496 DOI: 10.3390/ijms24020912] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 12/28/2022] [Accepted: 12/28/2022] [Indexed: 01/06/2023] Open
Abstract
It has been hypothesised that inhalational anaesthetics such as isoflurane (Iso) may trigger the pathogenesis of Alzheimer's disease (AD), while the gaseous anaesthetic xenon (Xe) exhibits many features of a putative neuroprotective agent. Loss of synapses is regarded as one key cause of dementia in AD. Multiple EGF-like domains 10 (MEGF10) is one of the phagocytic receptors which assists the elimination of synapses by astrocytes. Here, we investigated how β-amyloid peptide 1-42 (Aβ1-42), Iso and Xe interact with MEGF10-dependent synapse elimination. Murine cultured astrocytes as well as cortical and hippocampal ex vivo brain slices were treated with either Aβ1-42, Iso or Xe and the combination of Aβ1-42 with either Iso or Xe. We quantified MEGF10 expression in astrocytes and dendritic spine density (DSD) in slices. In brain slices of wild type and AAV-induced MEGF10 knock-down mice, antibodies against astrocytes (GFAP), pre- (synaptophysin) and postsynaptic (PSD95) components were used for co-localization analyses by means of immunofluorescence-imaging and 3D rendering techniques. Aβ1-42 elevated pre- and postsynaptic components inside astrocytes and decreased DSD. The combined application with either Iso or Xe reversed these effects. In the presence of Aβ1-42 both anaesthetics decreased MEGF10 expression. AAV-induced knock-down of MEGF10 reduced the pre- and postsynaptic marker inside astrocytes. The presented data suggest Iso and Xe are able to reverse the Aβ1-42-induced enhancement of synaptic elimination in ex vivo hippocampal brain slices, presumably through MEGF10 downregulation.
Collapse
Affiliation(s)
- Dai Shi
- Department of Anesthesiology and Intensive Care, Klinikum Rechts der Isar, Ismaningerstraße 22, 81675 Munich, Germany
| | - Jaime K. Y. Wong
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Kaichuan Zhu
- German Center for Neurodegenerative Diseases, Feodor-Lynen-Straße 23, 81377 Munich, Germany
- Center for Neuropathology and Prion Research, Feodor-Lynen-Straße 23, 81377 Munich, Germany
| | - Peter G. Noakes
- School of Biomedical Sciences, The University of Queensland, St. Lucia, QLD 4072, Australia
- Queensland Brain Institute, The University of Queensland, St. Lucia, QLD 4072, Australia
| | - Gerhard Rammes
- Department of Anesthesiology and Intensive Care, Klinikum Rechts der Isar, Ismaningerstraße 22, 81675 Munich, Germany
- Correspondence:
| |
Collapse
|
72
|
Shahbazi R, Lipson P, Gottimukkala KSV, Lane DD, Adair JE. CRISPR Gene Editing of Hematopoietic Stem and Progenitor Cells. Methods Mol Biol 2023; 2567:39-62. [PMID: 36255694 DOI: 10.1007/978-1-0716-2679-5_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: 06/16/2023]
Abstract
Genetic editing of hematopoietic stem and progenitor cells can be employed to understand gene-function relationships underlying hematopoietic cell biology, leading to new therapeutic approaches to treat disease. The ability to collect, purify, and manipulate primary cells outside the body permits testing of many different gene editing approaches. RNA-guided nucleases, such as CRISPR, have revolutionized gene editing based simply on Watson-Crick base-pairing, employed to direct activity to specific genomic loci. Given the ease and affordability of synthetic, custom RNA guides, testing of precision edits or large random pools in high-throughput screening studies is now widely available. With the ever-growing number of CRISPR nucleases being discovered or engineered, researchers now have a plethora of options for directed genomic change, including single base edits, nicks or double-stranded DNA cuts with blunt or staggered ends, as well as the ability to target CRISPR to other cellular oligonucleotides such as RNA or mitochondrial DNA. Except for single base editing strategies, precise rewriting of larger segments of the genetic code requires delivery of an additional component, templated DNA oligonucleotide(s) encoding the desired changes flanked by homologous sequences that permit recombination at or near the site of CRISPR activity. Altogether, the ever-growing CRISPR gene editing toolkit is an invaluable resource. This chapter outlines available technologies and the strategies for applying CRISPR-based editing in hematopoietic stem and progenitor cells.
Collapse
Affiliation(s)
| | | | | | | | - Jennifer E Adair
- Fred Hutchinson Cancer Center, Seattle, WA, USA.
- University of Washington, Seattle, WA, USA.
| |
Collapse
|
73
|
He L, He Z, Li Y, Sun H, Wang H. In Vivo Investigation of Gene Function in Muscle Stem Cells by CRISPR/Cas9-Mediated Genome Editing. Methods Mol Biol 2023; 2640:287-311. [PMID: 36995603 DOI: 10.1007/978-1-0716-3036-5_21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/31/2023]
Abstract
Skeletal muscle satellite cells (SCs) are adult stem cells responsible for muscle development and injury-induced muscle regeneration. Functional elucidation of intrinsic regulatory factors governing SC activity is constrained partially by the technological limitations in editing SCs in vivo. Although the power of CRISPR/Cas9 in genome manipulation has been widely documented, its application in endogenous SCs remains largely untested. Our recent study generates a muscle-specific genome editing system leveraging the Cre-dependent Cas9 knockin mice and AAV9-mediated sgRNAs delivery, which allows gene disruption in SCs in vivo. Here, we illustrate the step-by-step procedure for achieving efficient editing using the above system.
Collapse
Affiliation(s)
- Liangqiang He
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhiming He
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Yuying Li
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Hao Sun
- Department of Chemical Pathology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Huating Wang
- Department of Orthopaedics and Traumatology, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
74
|
Hong E, Glynn C, Wang Q, Rao S. Non-Invasive Electroretinogram Recording with Simultaneous Optogenetics to Dissect Retinal Ganglion Cells Electrophysiological Dynamics. BIOSENSORS 2022; 13:42. [PMID: 36671879 PMCID: PMC9855613 DOI: 10.3390/bios13010042] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 12/17/2022] [Accepted: 12/26/2022] [Indexed: 06/17/2023]
Abstract
Electroretinography (ERG) is a non-invasive electrophysiological recording technique that detects the electrical signaling of neuronal cells in the visual system. In conventional ERG recordings, the signals are considered a collective electrical response from various neuronal cell populations, including rods, cones, bipolar cells, and retinal ganglion cells (RGCs). However, due to the limited ability to control electrophysiological responses from different types of cells, the detailed information underlying ERG signals has not been analyzed and interpreted. Linking the features of ERG signals to the specific neuronal response will advance the understanding of neuronal electrophysiological dynamics and provide more evidence to elucidate pathological mechanisms, such as RGC loss during the progression of glaucoma. Herein, we developed an advanced ERG recording system integrated with a programmable, non-invasive optogenetic stimulation method in mice. In this system, we applied an automatic and unbiased ERG data analysis approach to differentiate a, b wave, negative response, and oscillatory potentials. To differentiate the electrophysiological response of RGCs in ERG recordings, we sensitized mouse RGCs with red-light opsin, ChRmine, through adeno-associated virus (AAV) intravitreal injection. Features of RGC dynamics under red-light stimulation were identified in the ERG readout. This non-invasive ERG recording system, associated with the programmable optogenetics stimulation method, provides a new methodology to dissect neural dynamics under variable physiological and pathological conditions in vivo. With the merits of non-invasiveness, improved sensitivity, and specificity, we envision this system can be further applied for early-stage detection of RGC degeneration and functional progression in neural degenerative diseases, such as glaucoma.
Collapse
Affiliation(s)
- Eunji Hong
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Christopher Glynn
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Qianbin Wang
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
| | - Siyuan Rao
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA 01003, USA
- Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA
- Neuroscience and Behavior Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| |
Collapse
|
75
|
Kim YE, Kim S, Kim IH. Neural circuit-specific gene manipulation in mouse brain in vivo using split-intein-mediated split-Cre system. STAR Protoc 2022; 3:101807. [PMID: 36386891 PMCID: PMC9641071 DOI: 10.1016/j.xpro.2022.101807] [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] [Indexed: 11/06/2022] Open
Abstract
Neural network studies require efficient genetic tools to analyze individual neural circuit functions in vivo. Thus, we developed an advanced circuit-selective gene manipulating tool utilizing anterograde and retrograde adeno-associated viruses (AAVs) encoding split-intein-mediated split-Cre. This strategy can be applied to visualize a specific neural circuit as well as manipulate multiple genes in the circuit neurons. Here, we describe the production and purification of the AAVs, viral injection to the mouse brain, and imaging analysis for a specific neural circuit. For complete details on the use and execution of this protocol, please refer to Kim et al. (2022).
Collapse
Affiliation(s)
- Yong-Eun Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Sunwhi Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| | - Il Hwan Kim
- Department of Anatomy and Neurobiology, University of Tennessee Health Science Center, Memphis, TN 38163, USA; Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN 38163, USA.
| |
Collapse
|
76
|
Gene augmentation prevents retinal degeneration in a CRISPR/Cas9-based mouse model of PRPF31 retinitis pigmentosa. Nat Commun 2022; 13:7695. [PMID: 36509783 PMCID: PMC9744804 DOI: 10.1038/s41467-022-35361-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2022] [Accepted: 11/29/2022] [Indexed: 12/14/2022] Open
Abstract
Mutations in PRPF31 cause autosomal dominant retinitis pigmentosa, an untreatable form of blindness. Gene therapy is a promising treatment for PRPF31-retinitis pigmentosa, however, there are currently no suitable animal models in which to develop AAV-mediated gene augmentation. Here we establish Prpf31 mutant mouse models using AAV-mediated CRISPR/Cas9 knockout, and characterize the resulting retinal degeneration phenotype. Mouse models with early-onset morphological and functional impairments like those in patients were established, providing new platforms in which to investigate pathogenetic mechanisms and develop therapeutic methods. AAV-mediated PRPF31 gene augmentation restored the retinal structure and function in a rapidly degenerating mouse model, demonstrating the first in vivo proof-of-concept for AAV-mediated gene therapy to treat PRPF31-retinitis pigmentosa. AAV-CRISPR/Cas9-PRPF31 knockout constructs also mediated efficient PRPF31 knockout in human and non-human primate retinal explants, laying a foundation for establishing non-human primate models using the method developed here.
Collapse
|
77
|
Lopes MM, Paysan J, Rino J, Lopes SM, Pereira de Almeida L, Cortes L, Nobre RJ. A new protocol for whole-brain biodistribution analysis of AAVs by tissue clearing, light-sheet microscopy and semi-automated spatial quantification. Gene Ther 2022; 29:665-679. [PMID: 36316447 DOI: 10.1038/s41434-022-00372-z] [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: 06/20/2021] [Revised: 10/04/2022] [Accepted: 10/10/2022] [Indexed: 12/23/2022]
Abstract
Recombinant adeno-associated virus (rAAV) has become one of the most promising gene delivery systems for both in vitro and in vivo applications. However, a key challenge is the lack of suitable imaging technologies to evaluate delivery, biodistribution and tropism of rAAVs and efficiently monitor disease amelioration promoted by AAV-based therapies at a whole-organ level with single-cell resolution. Therefore, we aimed to establish a new pipeline for the biodistribution analysis of natural and new variants of AAVs at a whole-brain level by tissue clearing and light-sheet fluorescence microscopy (LSFM). To test this platform, neonatal C57BL/6 mice were intravenously injected with rAAV9 encoding EGFP and, after sacrifice, brains were processed by standard immunohistochemistry and a recently released aqueous-based clearing procedure. This clearing technique required no dedicated equipment and rendered highly cleared brains, while simultaneously preserving endogenous fluorescence. Moreover, three-dimensional imaging by LSFM allowed the quantitative analysis of EGFP at a whole-brain level, as well as the reconstruction of Purkinje cells for the retrieval of valuable morphological information inaccessible by standard immunohistochemistry. In conclusion, the pipeline herein described takes the AAVs to a new level when coupled to LSFM, proving its worth as a bioimaging tool in tropism and gene therapy studies.
Collapse
Affiliation(s)
- Miguel M Lopes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | | | - José Rino
- iMM - Instituto de Medicina Molecular João Lobo Antunes, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sara M Lopes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- ViraVector - Viral Vectors for Gene Transfer Core Facility, University of Coimbra, Coimbra, Portugal.
- FFUC - Faculty of Pharmacy of the University of Coimbra, Coimbra, Portugal.
| | - Luísa Cortes
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
- MICC-CNC - Microscopy Imaging Center of Coimbra - CNC, University of Coimbra, Coimbra, Portugal.
| | - Rui Jorge Nobre
- CNC - Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB - Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
- IIIUC - Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal.
- ViraVector - Viral Vectors for Gene Transfer Core Facility, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
78
|
Qiu Y, O’Neill N, Maffei B, Zourray C, Almacellas-Barbanoj A, Carpenter JC, Jones SP, Leite M, Turner TJ, Moreira FC, Snowball A, Shekh-Ahmad T, Magloire V, Barral S, Kurian MA, Walker MC, Schorge S, Kullmann DM, Lignani G. On-demand cell-autonomous gene therapy for brain circuit disorders. Science 2022; 378:523-532. [PMID: 36378958 PMCID: PMC7613996 DOI: 10.1126/science.abq6656] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Several neurodevelopmental and neuropsychiatric disorders are characterized by intermittent episodes of pathological activity. Although genetic therapies offer the ability to modulate neuronal excitability, a limiting factor is that they do not discriminate between neurons involved in circuit pathologies and "healthy" surrounding or intermingled neurons. We describe a gene therapy strategy that down-regulates the excitability of overactive neurons in closed loop, which we tested in models of epilepsy. We used an immediate early gene promoter to drive the expression of Kv1.1 potassium channels specifically in hyperactive neurons, and only for as long as they exhibit abnormal activity. Neuronal excitability was reduced by seizure-related activity, leading to a persistent antiepileptic effect without interfering with normal behaviors. Activity-dependent gene therapy is a promising on-demand cell-autonomous treatment for brain circuit disorders.
Collapse
Affiliation(s)
- Yichen Qiu
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Nathanael O’Neill
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Benito Maffei
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Clara Zourray
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
- Department of Developmental Neurosciences, Zayed Centre for Research Into Rare Disease in Children, GOS−Institute of Child Health, University College London, London, UK
| | - Amanda Almacellas-Barbanoj
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Jenna C. Carpenter
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Steffan P. Jones
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Marco Leite
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Thomas J. Turner
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Francisco C. Moreira
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Albert Snowball
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Tawfeeq Shekh-Ahmad
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Vincent Magloire
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Serena Barral
- Department of Developmental Neurosciences, Zayed Centre for Research Into Rare Disease in Children, GOS−Institute of Child Health, University College London, London, UK
| | - Manju A. Kurian
- Department of Developmental Neurosciences, Zayed Centre for Research Into Rare Disease in Children, GOS−Institute of Child Health, University College London, London, UK
- Department of Neurology, Great Ormond Street Hospital for Children, London, UK
| | - Matthew C. Walker
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Stephanie Schorge
- Department of Neuroscience, Physiology and Pharmacology University College London, London, UK
| | - Dimitri M. Kullmann
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Gabriele Lignani
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, University College London, London, UK
| |
Collapse
|
79
|
Lacomme M, Hales SC, Brown TW, Stevanovic K, Jolicoeur C, Cai J, Bois T, Desrosiers M, Dalkara D, Cayouette M. Numb regulates Tau levels and prevents neurodegeneration in tauopathy mouse models. SCIENCE ADVANCES 2022; 8:eabm4295. [PMID: 36260685 PMCID: PMC9581485 DOI: 10.1126/sciadv.abm4295] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Accepted: 09/07/2022] [Indexed: 06/01/2023]
Abstract
Accumulation of the microtubule-associated protein Tau is linked to neuronal cell death in tauopathies, but how intraneuronal Tau levels are regulated in health and disease remains unclear. Here, we show that conditional inactivation of the trafficking adaptor protein Numb in retinal ganglion cells (RGCs) increases Tau levels and leads to axonal blebbing, which is followed by neuronal cell loss in aged mice. In the TauP301S mouse model of tauopathy, conditional inactivation of Numb in RGCs and spinal motoneurons accelerates neurodegeneration, and loss of Numb in motoneurons also leads to precocious hindlimb paralysis. Conversely, overexpression of the long isoform of Numb (Numb-72) decreases intracellular Tau levels and reduces axonal blebbing in TauP301S RGCs, leading to improved electrical activity in cultured neurons and improves performance in a visually guided behavior test in vivo. These results uncover Numb as a key regulator of intracellular Tau levels and identify Numb-72 as a potential therapeutic factor for tauopathies.
Collapse
Affiliation(s)
- Marine Lacomme
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Sarah C. Hales
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Thomas W. Brown
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
| | - Katarina Stevanovic
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Christine Jolicoeur
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Jenny Cai
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Therence Bois
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
| | - Melissa Desrosiers
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Deniz Dalkara
- Sorbonne Université, INSERM, CNRS, Institut de la Vision, 17 rue Moreau, F-75012 Paris, France
| | - Michel Cayouette
- Cellular Neurobiology Research Unit, Institut de recherches cliniques de Montréal (IRCM), Montreal, QC H2W 1R7, Canada
- Integrated Program in Neuroscience, McGill University, Montreal, QC, Canada
- Department of Medicine, Université de Montréal, Montreal, QC H3T 1J4, Canada
- Department of Anatomy and Cell Biology, Division of Experimental Medicine, McGill University, Montreal, QC H3A 0G4, Canada
| |
Collapse
|
80
|
Jang A, Petrova B, Cheong TC, Zawadzki ME, Jones JK, Culhane AJ, Shipley FB, Chiarle R, Wong ET, Kanarek N, Lehtinen MK. Choroid plexus-CSF-targeted antioxidant therapy protects the brain from toxicity of cancer chemotherapy. Neuron 2022; 110:3288-3301.e8. [PMID: 36070751 PMCID: PMC9588748 DOI: 10.1016/j.neuron.2022.08.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 06/28/2022] [Accepted: 08/05/2022] [Indexed: 12/14/2022]
Abstract
For many cancer patients, chemotherapy produces untreatable life-long neurologic effects termed chemotherapy-related cognitive impairment (CRCI). We discovered that the chemotherapy methotrexate (MTX) adversely affects oxidative metabolism of non-cancerous choroid plexus (ChP) cells and the cerebrospinal fluid (CSF). We used a ChP-targeted adeno-associated viral (AAV) vector approach in mice to augment CSF levels of the secreted antioxidant SOD3. AAV-SOD3 gene therapy increased oxidative defense capacity of the CSF and prevented MTX-induced lipid peroxidation in the hippocampus. Furthermore, this gene therapy prevented anxiety and deficits in short-term learning and memory caused by MTX. MTX-induced oxidative damage to cultured human cortical neurons and analyses of CSF samples from MTX-treated lymphoma patients demonstrated that MTX diminishes antioxidant capacity of patient CSF. Collectively, our findings motivate the advancement of ChP- and CSF-targeted anti-oxidative prophylactic measures to relieve CRCI.
Collapse
Affiliation(s)
- Ahram Jang
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Boryana Petrova
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Taek-Chin Cheong
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Miriam E Zawadzki
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Harvard, MIT MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA
| | - Jill K Jones
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Harvard, MIT MD-PhD Program, Harvard Medical School, Boston, MA 02115, USA
| | - Andrew J Culhane
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA
| | - Frederick B Shipley
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA
| | - Roberto Chiarle
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Department of Molecular Biotechnology and Health Sciences, University of Torino, Torino 10126, Italy
| | - Eric T Wong
- Brain Tumor Center & Neuro-Oncology Unit, Beth Israel Deaconess Medical Center, Boston, MA 02115, USA
| | - Naama Kanarek
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA.
| | - Maria K Lehtinen
- Department of Pathology, Boston Children's Hospital, Boston, MA 02115, USA; Graduate Program in Biological and Biomedical Sciences, Harvard Medical School, Boston, MA 02115, USA; Graduate Program in Biophysics, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
81
|
Kennedy A, Waters E, Rowshanravan B, Hinze C, Williams C, Janman D, Fox TA, Booth C, Pesenacker AM, Halliday N, Soskic B, Kaur S, Qureshi OS, Morris EC, Ikemizu S, Paluch C, Huo J, Davis SJ, Boucrot E, Walker LSK, Sansom DM. Differences in CD80 and CD86 transendocytosis reveal CD86 as a key target for CTLA-4 immune regulation. Nat Immunol 2022; 23:1365-1378. [PMID: 35999394 PMCID: PMC9477731 DOI: 10.1038/s41590-022-01289-w] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Accepted: 07/15/2022] [Indexed: 01/07/2023]
Abstract
CD28 and CTLA-4 (CD152) play essential roles in regulating T cell immunity, balancing the activation and inhibition of T cell responses, respectively. Although both receptors share the same ligands, CD80 and CD86, the specific requirement for two distinct ligands remains obscure. In the present study, we demonstrate that, although CTLA-4 targets both CD80 and CD86 for destruction via transendocytosis, this process results in separate fates for CTLA-4 itself. In the presence of CD80, CTLA-4 remained ligand bound, and was ubiquitylated and trafficked via late endosomes and lysosomes. In contrast, in the presence of CD86, CTLA-4 detached in a pH-dependent manner and recycled back to the cell surface to permit further transendocytosis. Furthermore, we identified clinically relevant mutations that cause autoimmune disease, which selectively disrupted CD86 transendocytosis, by affecting either CTLA-4 recycling or CD86 binding. These observations provide a rationale for two distinct ligands and show that defects in CTLA-4-mediated transendocytosis of CD86 are associated with autoimmunity.
Collapse
Affiliation(s)
- Alan Kennedy
- UCL Institute of Immunity and Transplantation, London, UK
| | - Erin Waters
- UCL Institute of Immunity and Transplantation, London, UK
| | | | - Claudia Hinze
- UCL Institute of Immunity and Transplantation, London, UK
| | | | - Daniel Janman
- UCL Institute of Immunity and Transplantation, London, UK
| | - Thomas A Fox
- UCL Institute of Immunity and Transplantation, London, UK
| | - Claire Booth
- Molecular and Cellular Immunology Section, UCL Great Ormond Street Institute of Child Health, London, UK
| | | | - Neil Halliday
- UCL Institute of Immunity and Transplantation, London, UK
| | - Blagoje Soskic
- UCL Institute of Immunity and Transplantation, London, UK
| | - Satdip Kaur
- School of Immunity and Infection, Institute of Biomedical Research, University of Birmingham Medical School, Birmingham, UK
| | | | - Emma C Morris
- UCL Institute of Immunity and Transplantation, London, UK
| | - Shinji Ikemizu
- Division of Structural Biology, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Christopher Paluch
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Jiandong Huo
- Structural Biology, The Rosalind Franklin Institute, Didcot, UK
- Division of Structural Biology, University of Oxford, Oxford, UK
- Wellcome Trust Centre for Human Genetics, Oxford, UK
- Protein Production UK, The Rosalind Franklin Institute-Diamond Light Source, The Research Complex at Harwell, Didcot, UK
| | - Simon J Davis
- Medical Research Council Human Immunology Unit, John Radcliffe Hospital, University of Oxford, Oxford, UK
- Radcliffe Department of Medicine, John Radcliffe Hospital, University of Oxford, Oxford, UK
| | - Emmanuel Boucrot
- Institute of Structural and Molecular Biology, University College London, London, UK
| | | | - David M Sansom
- UCL Institute of Immunity and Transplantation, London, UK.
| |
Collapse
|
82
|
Modeling PRPF31 retinitis pigmentosa using retinal pigment epithelium and organoids combined with gene augmentation rescue. NPJ Regen Med 2022; 7:39. [PMID: 35974011 PMCID: PMC9381579 DOI: 10.1038/s41536-022-00235-6] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 07/20/2022] [Indexed: 11/17/2022] Open
Abstract
Mutations in the ubiquitously expressed pre-mRNA processing factor (PRPF) 31 gene, one of the most common causes of dominant form of Retinitis Pigmentosa (RP), lead to a retina-specific phenotype. It is uncertain which retinal cell types are affected and animal models do not clearly present the RP phenotype observed in PRPF31 patients. Retinal organoids and retinal pigment epithelial (RPE) cells derived from human-induced pluripotent stem cells (iPSCs) provide potential opportunities for studying human PRPF31-related RP. We demonstrate here that RPE cells carrying PRPF31 mutations present important morphological and functional changes and that PRPF31-mutated retinal organoids recapitulate the human RP phenotype, with a rod photoreceptor cell death followed by a loss of cones. The low level of PRPF31 expression may explain the defective phenotypes of PRPF31-mutated RPE and photoreceptor cells, which were not observed in cells derived from asymptomatic patients or after correction of the pathogenic mutation by CRISPR/Cas9. Transcriptome profiles revealed differentially expressed and mis-spliced genes belonging to pathways in line with the observed defective phenotypes. The rescue of RPE and photoreceptor defective phenotypes by PRPF31 gene augmentation provide the proof of concept for future therapeutic strategies.
Collapse
|
83
|
Mason WJ, Jafree DJ, Pomeranz G, Kolatsi-Joannou M, Rottner AK, Pacheco S, Moulding DA, Wolf A, Kupatt C, Peppiatt-Wildman C, Papakrivopoulou E, Riley PR, Long DA, Vasilopoulou E. Systemic gene therapy with thymosin β4 alleviates glomerular injury in mice. Sci Rep 2022; 12:12172. [PMID: 35842494 PMCID: PMC9288454 DOI: 10.1038/s41598-022-16287-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Accepted: 07/07/2022] [Indexed: 11/17/2022] Open
Abstract
Plasma ultrafiltration in the kidney occurs across glomerular capillaries, which are surrounded by epithelial cells called podocytes. Podocytes have a unique shape maintained by a complex cytoskeleton, which becomes disrupted in glomerular disease resulting in defective filtration and albuminuria. Lack of endogenous thymosin β4 (TB4), an actin sequestering peptide, exacerbates glomerular injury and disrupts the organisation of the podocyte actin cytoskeleton, however, the potential of exogenous TB4 therapy to improve podocyte injury is unknown. Here, we have used Adriamycin (ADR), a toxin which injures podocytes and damages the glomerular filtration barrier leading to albuminuria in mice. Through interrogating single-cell RNA-sequencing data of isolated glomeruli we demonstrate that ADR injury results in reduced levels of podocyte TB4. Administration of an adeno-associated viral vector encoding TB4 increased the circulating level of TB4 and prevented ADR-induced podocyte loss and albuminuria. ADR injury was associated with disorganisation of the podocyte actin cytoskeleton in vitro, which was ameliorated by treatment with exogenous TB4. Collectively, we propose that systemic gene therapy with TB4 prevents podocyte injury and maintains glomerular filtration via protection of the podocyte cytoskeleton thus presenting a novel treatment strategy for glomerular disease.
Collapse
Affiliation(s)
- William J Mason
- Division of Natural Sciences, Medway School of Pharmacy, University of Kent, Chatham, Kent, UK.,Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Daniyal J Jafree
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK.,UCL MB/PhD Programme, Faculty of Medical Science, University College London, London, UK
| | - Gideon Pomeranz
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Maria Kolatsi-Joannou
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Antje K Rottner
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Sabrina Pacheco
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Dale A Moulding
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Anja Wolf
- Medizinische Klinik und Poliklinik I, University Clinic Rechts der Isar, TUM Munich, Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | - Christian Kupatt
- Medizinische Klinik und Poliklinik I, University Clinic Rechts der Isar, TUM Munich, Munich, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Munich Heart Alliance, Munich, Germany
| | | | - Eugenia Papakrivopoulou
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK.,Department of Internal Medicine and Nephrology, Clinique Saint Jean, Brussels, Belgium
| | - Paul R Riley
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, UK
| | - David A Long
- Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK
| | - Elisavet Vasilopoulou
- Division of Natural Sciences, Medway School of Pharmacy, University of Kent, Chatham, Kent, UK. .,Developmental Biology and Cancer Programme, UCL Great Ormond Street Institute of Child Health, London, UK. .,Comparative Biomedical Sciences, The Royal Veterinary College, Royal College Street, London, NW1 0TU, UK.
| |
Collapse
|
84
|
Sutter SO, Lkharrazi A, Schraner EM, Michaelsen K, Meier AF, Marx J, Vogt B, Büning H, Fraefel C. Adeno-associated virus type 2 (AAV2) uncoating is a stepwise process and is linked to structural reorganization of the nucleolus. PLoS Pathog 2022; 18:e1010187. [PMID: 35816507 PMCID: PMC9302821 DOI: 10.1371/journal.ppat.1010187] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 07/21/2022] [Accepted: 06/08/2022] [Indexed: 11/18/2022] Open
Abstract
Nucleoli are membrane-less structures located within the nucleus and are known to be involved in many cellular functions, including stress response and cell cycle regulation. Besides, many viruses can employ the nucleolus or nucleolar proteins to promote different steps of their life cycle such as replication, transcription and assembly. While adeno-associated virus type 2 (AAV2) capsids have previously been reported to enter the host cell nucleus and accumulate in the nucleolus, both the role of the nucleolus in AAV2 infection, and the viral uncoating mechanism remain elusive. In all prior studies on AAV uncoating, viral capsids and viral genomes were not directly correlated on the single cell level, at least not in absence of a helper virus. To elucidate the properties of the nucleolus during AAV2 infection and to assess viral uncoating on a single cell level, we combined immunofluorescence analysis for detection of intact AAV2 capsids and capsid proteins with fluorescence in situ hybridization for detection of AAV2 genomes. The results of our experiments provide evidence that uncoating of AAV2 particles occurs in a stepwise process that is completed in the nucleolus and supported by alteration of the nucleolar structure.
Collapse
Affiliation(s)
| | - Anouk Lkharrazi
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | | | - Kevin Michaelsen
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | | | - Jennifer Marx
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Bernd Vogt
- Institute of Virology, University of Zurich, Zurich, Switzerland
| | - Hildegard Büning
- Institute of Experimental Hematology, Hannover Medical School, Hannover, Germany
| | - Cornel Fraefel
- Institute of Virology, University of Zurich, Zurich, Switzerland
- * E-mail:
| |
Collapse
|
85
|
Xie Y, Zhou J, Chen B. Critical examination of Ptbp1-mediated glia-to-neuron conversion in the mouse retina. Cell Rep 2022; 39:110960. [PMID: 35705044 PMCID: PMC9371382 DOI: 10.1016/j.celrep.2022.110960] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2021] [Revised: 04/10/2022] [Accepted: 05/24/2022] [Indexed: 11/27/2022] Open
Abstract
Reprogramming glial cells to convert them into neurons represents a potential therapeutic strategy that could repair damaged neural circuits and restore function. Recent studies show that downregulation of the RNA-binding protein PTBP1 leads to one-step conversion of Müller glia (MG) into retinal ganglion cells (RGCs) with a high efficiency. However, the original study did not perform fate-mapping experiments to confirm MG-to-RGC conversion after Ptbp1 downregulation. To address the fundamental question of whether Ptbp1 downregulation can convert MG into RGCs in the mouse retina, we perform fate-mapping experiments to lineage trace MG independent of the adeno-associated virus (AAV)-mediated labeling system. Here, we report that Ptbp1 downregulation by CRISPR-CasRx or small hairpin RNA is insufficient to convert MG to RGCs. The original conclusion of MG-to-RGC conversion is due to leaky labeling of endogenous RGCs. Our results emphasize the importance of using stringent fate mapping to determine glia-to-neuron conversion in cell reprogramming research. Leaky labeling of endogenous retinal ganglion cells (RGCs) leads to misinterpretation of glia-to-neuron conversion in the mouse retina. Using stringent fate-mapping experiments, Xie et al. show that lineage-traced Müller glia (MG) are not converted into RGCs after Ptbp1 downregulation by CRISPR-CasRx or small hairpin RNA.
Collapse
Affiliation(s)
- Ye Xie
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Jing Zhou
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA
| | - Bo Chen
- Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Neuroscience, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA; Department of Cell, Developmental and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY 10029, USA.
| |
Collapse
|
86
|
Xi Z, Öztürk BE, Johnson ME, Turunç S, Stauffer WR, Byrne LC. Quantitative single-cell transcriptome-based ranking of engineered AAVs in human retinal explants. Mol Ther Methods Clin Dev 2022; 25:476-489. [PMID: 35615708 PMCID: PMC9118357 DOI: 10.1016/j.omtm.2022.04.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 04/26/2022] [Indexed: 11/07/2022]
Abstract
Gene therapy is a rapidly developing field, and adeno-associated viruses (AAVs) are a leading viral-vector candidate for therapeutic gene delivery. Newly engineered AAVs with improved abilities are now entering the clinic. It has proven challenging, however, to predict the translational potential of gene therapies developed in animal models due to cross-species differences. Human retinal explants are the only available model of fully developed human retinal tissue and are thus important for the validation of candidate AAV vectors. In this study, we evaluated 18 wild-type and engineered AAV capsids in human retinal explants using a recently developed single-cell RNA sequencing (RNA-seq) AAV engineering pipeline (scAAVengr). Human retinal explants retained the same major cell types as fresh retina, with similar expression of cell-specific markers except for a photoreceptor population with altered expression of photoreceptor-specific genes. The efficiency and tropism of AAVs in human explants were quantified with single-cell resolution. The top-performing serotypes, K91, K912, and 7m8, were further validated in non-human primate and human retinal explants. Together, this study provides detailed information about the transcriptome profiles of retinal explants and quantifies the infectivity of leading AAV serotypes in human retina, accelerating the translation of retinal gene therapies to the clinic.
Collapse
Affiliation(s)
- Zhouhuan Xi
- Department of Ophthalmology, University of Pittsburgh, PA, USA
- Eye Center of Xiangya Hospital, Hunan Key Laboratory of Ophthalmology, Central South University, Changsha, Hunan, China
| | - Bilge E. Öztürk
- Department of Ophthalmology, University of Pittsburgh, PA, USA
| | | | - Serhan Turunç
- Department of Ophthalmology, University of Pittsburgh, PA, USA
| | | | - Leah C. Byrne
- Department of Ophthalmology, University of Pittsburgh, PA, USA
- Department of Neurobiology, University of Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh, PA, USA
| |
Collapse
|
87
|
Colomb-Delsuc M, Raim R, Fiedler C, Reuberger S, Lengler J, Nordström R, Ryner M, Folea IM, Kraus B, Hernandez Bort JA, Sintorn IM. Assessment of the percentage of full recombinant adeno-associated virus particles in a gene therapy drug using CryoTEM. PLoS One 2022; 17:e0269139. [PMID: 35657790 PMCID: PMC9165851 DOI: 10.1371/journal.pone.0269139] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Accepted: 05/16/2022] [Indexed: 11/18/2022] Open
Abstract
In spite of continuous development of gene therapy vectors with thousands of drug candidates in clinical drug trials there are only a small number approved on the market today stressing the need to have characterization methods to assist in the validation of the drug development process. The level of packaging of the vector capsids appears to play a critical role in immunogenicity, hence an objective quantitative method assessing the content of particles containing a genome is an essential quality measurement. As transmission electron microscopy (TEM) allows direct visualization of the particles present in a specimen, it naturally seems as the most intuitive method of choice for characterizing recombinant adeno-associated virus (rAAV) particle packaging. Negative stain TEM (nsTEM) is an established characterization method for analysing the packaging of viral vectors. It has however shown limitations in terms of reliability. To overcome this drawback, we propose an analytical method based on CryoTEM that unambiguously and robustly determines the percentage of filled particles in an rAAV sample. In addition, we show that at a fixed number of vector particles the portion of filled particles correlates well with the potency of the drug. The method has been validated according to the ICH Q2 (R1) guidelines and the components investigated during the validation are presented in this study. The reliability of nsTEM as a method for the assessment of filled particles is also investigated along with a discussion about the origin of the observed variability of this method.
Collapse
Affiliation(s)
| | - Roman Raim
- Baxalta Innovations GmbH, A Part of Takeda Companies, Orth an der Donau, Austria
| | - Christian Fiedler
- Baxalta Innovations GmbH, A Part of Takeda Companies, Orth an der Donau, Austria
| | - Stefan Reuberger
- Baxalta Innovations GmbH, A Part of Takeda Companies, Orth an der Donau, Austria
| | - Johannes Lengler
- Baxalta Innovations GmbH, A Part of Takeda Companies, Orth an der Donau, Austria
| | | | | | | | - Barbara Kraus
- Baxalta Innovations GmbH, A Part of Takeda Companies, Orth an der Donau, Austria
| | | | - Ida-Maria Sintorn
- Vironova AB, Stockholm, Sweden
- Department of Information Technology, Uppsala University, Uppsala, Sweden
| |
Collapse
|
88
|
Shupe J, Zhang A, Odenwelder DC, Dobrowsky T. Gene therapy: challenges in cell culture scale-up. Curr Opin Biotechnol 2022; 75:102721. [DOI: 10.1016/j.copbio.2022.102721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2021] [Revised: 02/04/2022] [Accepted: 03/02/2022] [Indexed: 11/03/2022]
|
89
|
Lawler AJ, Ramamurthy E, Brown AR, Shin N, Kim Y, Toong N, Kaplow IM, Wirthlin M, Zhang X, Phan BN, Fox GA, Wade K, He J, Ozturk BE, Byrne LC, Stauffer WR, Fish KN, Pfenning AR. Machine learning sequence prioritization for cell type-specific enhancer design. eLife 2022; 11:e69571. [PMID: 35576146 PMCID: PMC9110026 DOI: 10.7554/elife.69571] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Accepted: 04/25/2022] [Indexed: 11/22/2022] Open
Abstract
Recent discoveries of extreme cellular diversity in the brain warrant rapid development of technologies to access specific cell populations within heterogeneous tissue. Available approaches for engineering-targeted technologies for new neuron subtypes are low yield, involving intensive transgenic strain or virus screening. Here, we present Specific Nuclear-Anchored Independent Labeling (SNAIL), an improved virus-based strategy for cell labeling and nuclear isolation from heterogeneous tissue. SNAIL works by leveraging machine learning and other computational approaches to identify DNA sequence features that confer cell type-specific gene activation and then make a probe that drives an affinity purification-compatible reporter gene. As a proof of concept, we designed and validated two novel SNAIL probes that target parvalbumin-expressing (PV+) neurons. Nuclear isolation using SNAIL in wild-type mice is sufficient to capture characteristic open chromatin features of PV+ neurons in the cortex, striatum, and external globus pallidus. The SNAIL framework also has high utility for multispecies cell probe engineering; expression from a mouse PV+ SNAIL enhancer sequence was enriched in PV+ neurons of the macaque cortex. Expansion of this technology has broad applications in cell type-specific observation, manipulation, and therapeutics across species and disease models.
Collapse
Affiliation(s)
- Alyssa J Lawler
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Biological Sciences Department, Mellon College of Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - Easwaran Ramamurthy
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - Ashley R Brown
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - Naomi Shin
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - Yeonju Kim
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - Noelle Toong
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - Irene M Kaplow
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - Morgan Wirthlin
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - Xiaoyu Zhang
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - BaDoi N Phan
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
- Medical Scientist Training Program, University of PittsburghPittsburghUnited States
| | - Grant A Fox
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| | - Kirsten Wade
- Department of Psychiatry, Translational Neuroscience Program, University of PittsburghPittsburghUnited States
| | - Jing He
- Department of Neurobiology, University of PittsburghPittsburghUnited States
- Systems Neuroscience Center, Brain Institute, Center for Neuroscience, Center for the Neural Basis of CognitionPittsburghUnited States
| | - Bilge Esin Ozturk
- Department of Ophthalmology, University of PittsburghPittsburghUnited States
| | - Leah C Byrne
- Department of Neurobiology, University of PittsburghPittsburghUnited States
- Department of Ophthalmology, University of PittsburghPittsburghUnited States
- Division of Experimental Retinal Therapies, Department of Clinical Sciences & Advanced Medicine, School of Veterinary Medicine, University of PennsylvaniaPhiladelphiaUnited States
- Department of Bioengineering, University of PittsburghPittsburghUnited States
| | - William R Stauffer
- Department of Neurobiology, University of PittsburghPittsburghUnited States
| | - Kenneth N Fish
- Department of Psychiatry, Translational Neuroscience Program, University of PittsburghPittsburghUnited States
| | - Andreas R Pfenning
- Computational Biology Department, School of Computer Science, Carnegie Mellon UniversityPittsburghUnited States
- Neuroscience Institute, Carnegie Mellon UniversityPittsburghUnited States
| |
Collapse
|
90
|
Hui Y, Zheng X, Zhang H, Li F, Yu G, Li J, Zhang J, Gong X, Guo G. Strategies for Targeting Neural Circuits: How to Manipulate Neurons Using Virus Vehicles. Front Neural Circuits 2022; 16:882366. [PMID: 35571271 PMCID: PMC9099413 DOI: 10.3389/fncir.2022.882366] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 04/07/2022] [Indexed: 01/02/2023] Open
Abstract
Viral strategies are the leading methods for mapping neural circuits. Viral vehicles combined with genetic tools provide the possibility to visualize entire functional neural networks and monitor and manipulate neural circuit functions by high-resolution cell type- and projection-specific targeting. Optogenetics and chemogenetics drive brain research forward by exploring causal relationships among different brain regions. Viral strategies offer a fresh perspective for the analysis of the structure-function relationship of the neural circuitry. In this review, we summarize current and emerging viral strategies for targeting neural circuits and focus on adeno-associated virus (AAV) vectors.
Collapse
Affiliation(s)
- Yuqing Hui
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xuefeng Zheng
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Huijie Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Fang Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Guangyin Yu
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Jiong Li
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
| | - Jifeng Zhang
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- Jifeng Zhang,
| | - Xiaobing Gong
- Department of Gastroenterology, The First Affiliated Hospital of Jinan University, Guangzhou, China
- Xiaobing Gong,
| | - Guoqing Guo
- Department of Anatomy, Neuroscience Laboratory for Cognitive and Developmental Disorders, Medical College of Jinan University, Guangzhou
- *Correspondence: Guoqing Guo,
| |
Collapse
|
91
|
Challis RC, Ravindra Kumar S, Chen X, Goertsen D, Coughlin GM, Hori AM, Chuapoco MR, Otis TS, Miles TF, Gradinaru V. Adeno-Associated Virus Toolkit to Target Diverse Brain Cells. Annu Rev Neurosci 2022; 45:447-469. [PMID: 35440143 DOI: 10.1146/annurev-neuro-111020-100834] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Recombinant adeno-associated viruses (AAVs) are commonly used gene delivery vehicles for neuroscience research. They have two engineerable features: the capsid (outer protein shell) and cargo (encapsulated genome). These features can be modified to enhance cell type or tissue tropism and control transgene expression, respectively. Several engineered AAV capsids with unique tropisms have been identified, including variants with enhanced central nervous system transduction, cell type specificity, and retrograde transport in neurons. Pairing these AAVs with modern gene regulatory elements and state-of-the-art reporter, sensor, and effector cargo enables highly specific transgene expression for anatomical and functional analyses of brain cells and circuits. Here, we discuss recent advances that provide a comprehensive (capsid and cargo) AAV toolkit for genetic access to molecularly defined brain cell types. Expected final online publication date for the Annual Review of Neuroscience, Volume 45 is July 2022. Please see http://www.annualreviews.org/page/journal/pubdates for revised estimates.
Collapse
Affiliation(s)
- Rosemary C Challis
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Sripriya Ravindra Kumar
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Xinhong Chen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - David Goertsen
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Gerard M Coughlin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Acacia M Hori
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Miguel R Chuapoco
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Thomas S Otis
- Sainsbury Wellcome Centre, University College London, London, United Kingdom
| | - Timothy F Miles
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| | - Viviana Gradinaru
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, California, USA;
| |
Collapse
|
92
|
Belova L, Kochergin‐Nikitsky K, Erofeeva A, Lavrov A, Smirnikhina S. Approaches to purification and concentration of rAAV vectors for gene therapy. Bioessays 2022; 44:e2200019. [DOI: 10.1002/bies.202200019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/14/2022] [Accepted: 03/15/2022] [Indexed: 11/11/2022]
|
93
|
Differential T cell immune responses to deamidated adeno-associated virus vector. Mol Ther Methods Clin Dev 2022; 24:255-267. [PMID: 35211638 PMCID: PMC8829777 DOI: 10.1016/j.omtm.2022.01.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 01/16/2022] [Indexed: 01/09/2023]
Abstract
Despite the high safety profile demonstrated in clinical trials, the immunogenicity of adeno-associated virus (AAV)-mediated gene therapy remains a major hurdle. Specifically, T-cell-mediated immune responses to AAV vectors are related to loss of efficacy and potential liver toxicities. As post-translational modifications in T cell epitopes have the potential to affect immune reactions, the cellular immune responses to peptides derived from spontaneously deamidated AAV were investigated. Here, we report that highly deamidated sites in AAV9 contain CD4 T cell epitopes with a Th1 cytokine pattern in multiple human donors with diverse human leukocyte antigen (HLA) backgrounds. Furthermore, some peripheral blood mononuclear cell (PBMC) samples demonstrated differential T cell activation to deamidated or non-deamidated epitopes. Also, in vitro and in silico HLA binding assays showed differential binding to the deamidated or non-deamidated peptides in some HLA alleles. This study provides critical attributes to vector-immune-mediated responses, as AAV deamidation can impact the immunogenicity, safety, and efficacy of AAV-mediated gene therapy in some patients.
Collapse
|
94
|
Gaul S, Shahzad K, Medert R, Gadi I, Mäder C, Schumacher D, Wirth A, Ambreen S, Fatima S, Boeckel JN, Khawaja H, Haas J, Brune M, Nawroth PP, Isermann B, Laufs U, Freichel M. Novel Nongenetic Murine Model of Hyperglycemia and Hyperlipidemia-Associated Aggravated Atherosclerosis. Front Cardiovasc Med 2022; 9:813215. [PMID: 35350534 PMCID: PMC8957812 DOI: 10.3389/fcvm.2022.813215] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Accepted: 02/02/2022] [Indexed: 01/24/2023] Open
Abstract
Objective Atherosclerosis, the main pathology underlying cardiovascular diseases is accelerated in diabetic patients. Genetic mouse models require breeding efforts which are time-consuming and costly. Our aim was to establish a new nongenetic model of inducible metabolic risk factors that mimics hyperlipidemia, hyperglycemia, or both and allows the detection of phenotypic differences dependent on the metabolic stressor(s). Methods and Results Wild-type mice were injected with gain-of-function PCSK9D377Y (proprotein convertase subtilisin/kexin type 9) mutant adeno-associated viral particles (AAV) and streptozotocin and fed either a high-fat diet (HFD) for 12 or 20 weeks or a high-cholesterol/high-fat diet (Paigen diet, PD) for 8 weeks. To evaluate atherosclerosis, two different vascular sites (aortic sinus and the truncus of the brachiocephalic artery) were examined in the mice. Combined hyperlipidemic and hyperglycemic (HGHCi) mice fed a HFD or PD displayed characteristic features of aggravated atherosclerosis when compared to hyperlipidemia (HCi HFD or PD) mice alone. Atherosclerotic plaques of HGHCi HFD animals were larger, showed a less stable phenotype (measured by the increased necrotic core area, reduced fibrous cap thickness, and less α-SMA-positive area) and had more inflammation (increased plasma IL-1β level, aortic pro-inflammatory gene expression, and MOMA-2-positive cells in the BCA) after 20 weeks of HFD. Differences between the HGHCi and HCi HFD models were confirmed using RNA-seq analysis of aortic tissue, revealing that significantly more genes were dysregulated in mice with combined hyperlipidemia and hyperglycemia than in the hyperlipidemia-only group. The HGHCi-associated genes were related to pathways regulating inflammation (increased Cd68, iNos, and Tnfa expression) and extracellular matrix degradation (Adamts4 and Mmp14). When comparing HFD with PD, the PD aggravated atherosclerosis to a greater extent in mice and showed plaque formation after 8 weeks. Hyperlipidemic and hyperglycemic mice fed a PD (HGHCi PD) showed less collagen (Sirius red) and increased inflammation (CD68-positive cells) within aortic plaques than hyperlipidemic mice (HCi PD). HGHCi-PD mice represent a directly inducible hyperglycemic atherosclerosis model compared with HFD-fed mice, in which atherosclerosis is severe by 8 weeks. Conclusion We established a nongenetically inducible mouse model allowing comparative analyses of atherosclerosis in HCi and HGHCi conditions and its modification by diet, allowing analyses of multiple metabolic hits in mice.
Collapse
Affiliation(s)
- Susanne Gaul
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Khurrum Shahzad
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Rebekka Medert
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Ihsan Gadi
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Christina Mäder
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Dagmar Schumacher
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| | - Angela Wirth
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
| | - Saira Ambreen
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Sameen Fatima
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Jes-Niels Boeckel
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Hamzah Khawaja
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Jan Haas
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
- Department of Internal Medicine III, Heidelberg University, Heidelberg, Germany
| | - Maik Brune
- Internal Medicine I and Clinical Chemistry, German Diabetes Center (DZD), Heidelberg University, Heidelberg, Germany
| | - Peter P. Nawroth
- Internal Medicine I and Clinical Chemistry, German Diabetes Center (DZD), Heidelberg University, Heidelberg, Germany
| | - Berend Isermann
- Department of Diagnostics, Laboratory Medicine, Clinical Chemistry and Molecular Diagnostic, University Hospital Leipzig, Leipzig, Germany
| | - Ulrich Laufs
- Klinik und Poliklinik für Kardiologie, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Marc Freichel
- Institute of Pharmacology, Heidelberg University, Heidelberg, Germany
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Germany
| |
Collapse
|
95
|
Low-dose metformin targets the lysosomal AMPK pathway through PEN2. Nature 2022; 603:159-165. [PMID: 35197629 PMCID: PMC8891018 DOI: 10.1038/s41586-022-04431-8] [Citation(s) in RCA: 319] [Impact Index Per Article: 106.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Accepted: 01/10/2022] [Indexed: 12/14/2022]
Abstract
Metformin, the most prescribed antidiabetic medicine, has shown other benefits such as anti-ageing and anticancer effects1-4. For clinical doses of metformin, AMP-activated protein kinase (AMPK) has a major role in its mechanism of action4,5; however, the direct molecular target of metformin remains unknown. Here we show that clinically relevant concentrations of metformin inhibit the lysosomal proton pump v-ATPase, which is a central node for AMPK activation following glucose starvation6. We synthesize a photoactive metformin probe and identify PEN2, a subunit of γ-secretase7, as a binding partner of metformin with a dissociation constant at micromolar levels. Metformin-bound PEN2 forms a complex with ATP6AP1, a subunit of the v-ATPase8, which leads to the inhibition of v-ATPase and the activation of AMPK without effects on cellular AMP levels. Knockout of PEN2 or re-introduction of a PEN2 mutant that does not bind ATP6AP1 blunts AMPK activation. In vivo, liver-specific knockout of Pen2 abolishes metformin-mediated reduction of hepatic fat content, whereas intestine-specific knockout of Pen2 impairs its glucose-lowering effects. Furthermore, knockdown of pen-2 in Caenorhabditis elegans abrogates metformin-induced extension of lifespan. Together, these findings reveal that metformin binds PEN2 and initiates a signalling route that intersects, through ATP6AP1, the lysosomal glucose-sensing pathway for AMPK activation. This ensures that metformin exerts its therapeutic benefits in patients without substantial adverse effects.
Collapse
|
96
|
Yamaguchi K, Maeda Y, Sawada T, Iino Y, Tajiri M, Nakazato R, Ishii S, Kasai H, Yagishita S. A behavioural correlate of the synaptic eligibility trace in the nucleus accumbens. Sci Rep 2022; 12:1921. [PMID: 35121769 PMCID: PMC8817024 DOI: 10.1038/s41598-022-05637-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 01/17/2022] [Indexed: 11/09/2022] Open
Abstract
Reward reinforces the association between a preceding sensorimotor event and its outcome. Reinforcement learning (RL) theory and recent brain slice studies explain the delayed reward action such that synaptic activities triggered by sensorimotor events leave a synaptic eligibility trace for 1 s. The trace produces a sensitive period for reward-related dopamine to induce synaptic plasticity in the nucleus accumbens (NAc). However, the contribution of the synaptic eligibility trace to behaviour remains unclear. Here we examined a reward-sensitive period to brief pure tones with an accurate measurement of an effective timing of water reward in head-fixed Pavlovian conditioning, which depended on the plasticity-related signaling in the NAc. We found that the reward-sensitive period was within 1 s after the pure tone presentation and optogenetically-induced presynaptic activities at the NAc, showing that the short reward-sensitive period was in conformity with the synaptic eligibility trace in the NAc. These findings support the application of the synaptic eligibility trace to construct biologically plausible RL models.
Collapse
Affiliation(s)
- Kenji Yamaguchi
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, Faculty of Medicine Bldg, The University of Tokyo, 1 #NC207, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Department of Psychology, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Yoshitomo Maeda
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, Faculty of Medicine Bldg, The University of Tokyo, 1 #NC207, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Takeshi Sawada
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, Faculty of Medicine Bldg, The University of Tokyo, 1 #NC207, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Yusuke Iino
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, Faculty of Medicine Bldg, The University of Tokyo, 1 #NC207, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Mio Tajiri
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, Faculty of Medicine Bldg, The University of Tokyo, 1 #NC207, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Ryosuke Nakazato
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, Faculty of Medicine Bldg, The University of Tokyo, 1 #NC207, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Shin Ishii
- International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.,Graduate School of Informatics, Kyoto University, Yoshida-Honmachi, Kyoto, Japan
| | - Haruo Kasai
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, Faculty of Medicine Bldg, The University of Tokyo, 1 #NC207, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan.,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | - Sho Yagishita
- Laboratory of Structural Physiology, Center for Disease Biology and Integrative Medicine, Faculty of Medicine, Faculty of Medicine Bldg, The University of Tokyo, 1 #NC207, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan. .,International Research Center for Neurointelligence (WPI-IRCN), UTIAS, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
97
|
He X, Liu J, Gu F, Chen J, Lu YW, Ding J, Guo H, Nie M, Kataoka M, Lin Z, Hu X, Chen H, Liao X, Dong Y, Min W, Deng ZL, Pu WT, Huang ZP, Wang DZ. Cardiac CIP protein regulates dystrophic cardiomyopathy. Mol Ther 2022; 30:898-914. [PMID: 34400329 PMCID: PMC8822131 DOI: 10.1016/j.ymthe.2021.08.022] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 05/24/2021] [Accepted: 08/08/2021] [Indexed: 02/04/2023] Open
Abstract
Heart failure is a leading cause of fatality in Duchenne muscular dystrophy (DMD) patients. Previously, we discovered that cardiac and skeletal-muscle-enriched CIP proteins play important roles in cardiac function. Here, we report that CIP, a striated muscle-specific protein, participates in the regulation of dystrophic cardiomyopathy. Using a mouse model of human DMD, we found that deletion of CIP leads to dilated cardiomyopathy and heart failure in young, non-syndromic mdx mice. Conversely, transgenic overexpression of CIP reduces pathological dystrophic cardiomyopathy in old, syndromic mdx mice. Genome-wide transcriptome analyses reveal that molecular pathways involving fibrogenesis and oxidative stress are affected in CIP-mediated dystrophic cardiomyopathy. Mechanistically, we found that CIP interacts with dystrophin and calcineurin (CnA) to suppress the CnA-Nuclear Factor of Activated T cells (NFAT) pathway, which results in decreased expression of Nox4, a key component of the oxidative stress pathway. Overexpression of Nox4 accelerates the development of dystrophic cardiomyopathy in mdx mice. Our study indicates CIP is a modifier of dystrophic cardiomyopathy and a potential therapeutic target for this devastating disease.
Collapse
Affiliation(s)
- Xin He
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Jianming Liu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Fei Gu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Jinghai Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, Provincial Key Lab of Cardiovascular Research, Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Yao Wei Lu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Jian Ding
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Haipeng Guo
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Critical Care and Emergency Medicine, Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Health, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, China
| | - Mao Nie
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Masaharu Kataoka
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Cardiology, Keio University School of Medicine, Tokyo, Japan
| | - Zhiqiang Lin
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Xiaoyun Hu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA
| | - Huaqun Chen
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Biology, Nanjing Normal University, Nanjing, China
| | - Xinxue Liao
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Yugang Dong
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China
| | - Wang Min
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China
| | - Zhong-Liang Deng
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Department of Orthopaedic Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - William T Pu
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA
| | - Zhan-Peng Huang
- Department of Cardiology, Center for Translational Medicine, Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; NHC Key Laboratory of Assisted Circulation (Sun Yat-sen University), Guangzhou, China; National-Guangdong Joint Engineering Laboratory for Diagnosis and Treatment of Vascular Diseases, Guangzhou 510080, China.
| | - Da-Zhi Wang
- Department of Cardiology, Boston Children's Hospital, Harvard Medical School, 320 Longwood Avenue, Boston, MA 02115, USA; Harvard Stem Cell Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
98
|
Nguyen HX, Wu T, Needs D, Zhang H, Perelli RM, DeLuca S, Yang R, Pan M, Landstrom AP, Henriquez C, Bursac N. Engineered bacterial voltage-gated sodium channel platform for cardiac gene therapy. Nat Commun 2022; 13:620. [PMID: 35110560 PMCID: PMC8810800 DOI: 10.1038/s41467-022-28251-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 01/11/2022] [Indexed: 12/19/2022] Open
Abstract
Therapies for cardiac arrhythmias could greatly benefit from approaches to enhance electrical excitability and action potential conduction in the heart by stably overexpressing mammalian voltage-gated sodium channels. However, the large size of these channels precludes their incorporation into therapeutic viral vectors. Here, we report a platform utilizing small-size, codon-optimized engineered prokaryotic sodium channels (BacNav) driven by muscle-specific promoters that significantly enhance excitability and conduction in rat and human cardiomyocytes in vitro and adult cardiac tissues from multiple species in silico. We also show that the expression of BacNav significantly reduces occurrence of conduction block and reentrant arrhythmias in fibrotic cardiac cultures. Moreover, functional BacNav channels are stably expressed in healthy mouse hearts six weeks following intravenous injection of self-complementary adeno-associated virus (scAAV) without causing any adverse effects on cardiac electrophysiology. The large diversity of prokaryotic sodium channels and experimental-computational platform reported in this study should facilitate the development and evaluation of BacNav-based gene therapies for cardiac conduction disorders.
Collapse
Affiliation(s)
- Hung X Nguyen
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Tianyu Wu
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Daniel Needs
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Hengtao Zhang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Robin M Perelli
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Sophia DeLuca
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Rachel Yang
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Michael Pan
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Andrew P Landstrom
- Department of Pediatrics, Division of Cardiology, Duke University School of Medicine, Durham, NC, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, USA
| | - Craig Henriquez
- Department of Biomedical Engineering, Duke University, Durham, NC, USA
| | - Nenad Bursac
- Department of Biomedical Engineering, Duke University, Durham, NC, USA.
| |
Collapse
|
99
|
Guan JS, Chen K, Si Y, Kim T, Zhou Z, Kim S, Zhou L, Liu X“M. Process improvement of adeno-associated virus (AAV) production. FRONTIERS IN CHEMICAL ENGINEERING 2022; 4:830421. [PMID: 35685827 PMCID: PMC9176270 DOI: 10.3389/fceng.2022.830421] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/12/2023] Open
Abstract
Adeno-associated viruses (AAVs) have been well characterized and used to deliver therapeutic genes for diseases treatment in clinics and basic research. This study used the triple transient transfection of AAV-DJ/8 as a model expression system to develop and optimize the laboratory production of AAV for research and pre-clinical applications. Specifically, various production parameters, including host cell, transfection reagent, cell density, ratio of plasmid DNA and cells, gene size, and production mode, were tested to determine the optimal process. Our results showed that the adherent production using HEK 293AAV with calcium transfection generated the highest volumetric productivity of 7.86x109 gc/mL. The optimal suspensive production using HEK 293F had best AAV productivity of 5.78x109 gc/mL in serum-free medium under transfection conditions of transfection density of 0.4x106 cells/mL, plasmid DNA:cells ratio of 1.6 μg:106 cells and synthesized cationic liposomes as transfection reagent. The similar AAV productivity was confirmed at scales of 30 mL - 450 mL in shaker and/or spinner flasks. The in vitro transfection and in vivo infection efficiency of the harvested AAV-DJ/8 carrying luciferase reporter gene was confirmed using cell line and xenograft mouse model, respectively. The minimal or low purification recovery rate of AAV-DJ/8 in ion-exchange chromatography column and affinity column was observed in this study. In summary, we developed and optimized a scalable suspensive production of AAV to support the large-scale preclinical animal studies in research laboratories.
Collapse
Affiliation(s)
- Jia-Shiung Guan
- Department of Medicine, UAB, 703 19 Street South, Birmingham, AL 35294, USA
| | - Kai Chen
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA
| | - Yingnan Si
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA
| | - Taehyun Kim
- Department of Medicine, UAB, 703 19 Street South, Birmingham, AL 35294, USA
| | - Zhuoxin Zhou
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA
| | - Seulhee Kim
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA
| | - Lufang Zhou
- Department of Medicine, UAB, 703 19 Street South, Birmingham, AL 35294, USA
| | - Xiaoguang “Margaret” Liu
- Department of Biomedical Engineering, University of Alabama at Birmingham (UAB), 1825 University Blvd, Birmingham, AL 35294, USA
| |
Collapse
|
100
|
Vasconcelos-Ferreira A, Martins IM, Lobo D, Pereira D, Lopes MM, Faro R, Lopes SM, Verbeek D, Schmidt T, Nóbrega C, Pereira de Almeida L. ULK overexpression mitigates motor deficits and neuropathology in mouse models of Machado-Joseph disease. Mol Ther 2022; 30:370-387. [PMID: 34298131 PMCID: PMC8753369 DOI: 10.1016/j.ymthe.2021.07.012] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 05/15/2021] [Accepted: 07/14/2021] [Indexed: 01/07/2023] Open
Abstract
Machado-Joseph disease (MJD) is a fatal neurodegenerative disorder clinically characterized by prominent ataxia. It is caused by an expansion of a CAG trinucleotide in ATXN3, translating into an expanded polyglutamine (polyQ) tract in the ATXN3 protein, that becomes prone to misfolding and aggregation. The pathogenesis of the disease has been associated with the dysfunction of several cellular mechanisms, including autophagy and transcription regulation. In this study, we investigated the transcriptional modifications of the autophagy pathway in models of MJD and assessed whether modulating the levels of the affected autophagy-associated transcripts (AATs) would alleviate MJD-associated pathology. Our results show that autophagy is impaired at the transcriptional level in MJD, affecting multiple AATs, including Unc-51 like autophagy activating kinase 1 and 2 (ULK1 and ULK2), two homologs involved in autophagy induction. Reinstating ULK1/2 levels by adeno-associated virus (AAV)-mediated gene transfer significantly improved motor performance while preventing neuropathology in two in vivo models of MJD. Moreover, in vitro studies showed that the observed positive effects may be mainly attributed to ULK1 activity. This study provides strong evidence of the beneficial effect of overexpression of ULK homologs, suggesting these as promising instruments for the treatment of MJD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Ana Vasconcelos-Ferreira
- CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal,CIBB – Center for Innovative Biomedicine and Biotechnology, Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal,Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal
| | - Inês Morgado Martins
- CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal,CIBB – Center for Innovative Biomedicine and Biotechnology, Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal,IIIUC – Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão – Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Diana Lobo
- CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal,CIBB – Center for Innovative Biomedicine and Biotechnology, Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal,IIIUC – Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão – Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Dina Pereira
- CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal,CIBB – Center for Innovative Biomedicine and Biotechnology, Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Miguel M. Lopes
- CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal,CIBB – Center for Innovative Biomedicine and Biotechnology, Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal,IIIUC – Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão – Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Rosário Faro
- CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal,CIBB – Center for Innovative Biomedicine and Biotechnology, Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Sara M. Lopes
- CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal,CIBB – Center for Innovative Biomedicine and Biotechnology, Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal,IIIUC – Institute for Interdisciplinary Research, University of Coimbra, Casa Costa Alemão – Pólo II, Rua Dom Francisco de Lemos, 3030-789 Coimbra, Portugal
| | - Dineke Verbeek
- Department of Genetics, University of Groningen, University Medical Center Groningen, Antonius Deusinglaan 1, 9700 RB, Groningen, the Netherlands
| | - Thorsten Schmidt
- Institute of Medical Genetics & Applied Genomics, University of Tübingen, 72076 Tübingen, Germany,Center for Rare Diseases (ZSE Tübingen), 72076 Tübingen, Germany
| | - Clévio Nóbrega
- CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal,CIBB – Center for Innovative Biomedicine and Biotechnology, Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Luís Pereira de Almeida
- CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal,CIBB – Center for Innovative Biomedicine and Biotechnology, Vectors, Gene and Cell Therapy Group, University of Coimbra, 3004-504 Coimbra, Portugal,Faculty of Pharmacy, University of Coimbra, Azinhaga de Santa Comba, Pólo das Ciências da Saúde, 3000-548, Coimbra, Portugal,Corresponding author: Luís Pereira de Almeida, PhD, CNC – Center for Neuroscience and Cell Biology, Molecular Therapy of Brain Disorders Group, University of Coimbra, Faculty of Medicine, Rua Larga, Pólo 1, 3004-504 Coimbra, Portugal.
| |
Collapse
|