1
|
Severi AA, Akbari B. CRISPR-Cas9 delivery strategies and applications: Review and update. Genesis 2024; 62:e23598. [PMID: 38727638 DOI: 10.1002/dvg.23598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 06/28/2024]
Abstract
Nowadays, a significant part of the investigations carried out in the medical field belong to cancer treatment. Generally, conventional cancer treatments, including chemotherapy, radiotherapy, and surgery, which have been used for a long time, are not sufficient, especially in malignant cancers. Because genetic mutations cause cancers, researchers are trying to treat these diseases using genetic engineering tools. One of them is clustered regularly interspaced short palindromic repeats (CRISPR), a powerful tool in genetic engineering in the last decade. CRISPR, which forms the CRISPR-Cas structure with its endonuclease protein, Cas, is known as a part of the immune system (adaptive immunity) in bacteria and archaea. Among the types of Cas proteins, Cas9 endonuclease has been used in many scientific studies due to its high accuracy and efficiency. This review reviews the CRISPR system, focusing on the history, classification, delivery methods, applications, new generations, and challenges of CRISPR-Cas9 technology.
Collapse
Affiliation(s)
- Ali Alizadeh Severi
- Department of Medical Biotechnology, School of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| | - Bahman Akbari
- Department of Medical Biotechnology, School of Medicine, Kermanshah University of Medical Science, Kermanshah, Iran
| |
Collapse
|
2
|
Dimitrievska M, Bansal D, Vitale M, Strouboulis J, Miccio A, Nicolaides KH, El Hoss S, Shangaris P, Jacków-Malinowska J. Revolutionising healing: Gene Editing's breakthrough against sickle cell disease. Blood Rev 2024; 65:101185. [PMID: 38493007 DOI: 10.1016/j.blre.2024.101185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 03/01/2024] [Accepted: 03/01/2024] [Indexed: 03/18/2024]
Abstract
Recent advancements in gene editing illuminate new potential therapeutic approaches for Sickle Cell Disease (SCD), a debilitating monogenic disorder caused by a point mutation in the β-globin gene. Despite the availability of several FDA-approved medications for symptomatic relief, allogeneic hematopoietic stem cell transplantation (HSCT) remains the sole curative option, underscoring a persistent need for novel treatments. This review delves into the growing field of gene editing, particularly the extensive research focused on curing haemoglobinopathies like SCD. We examine the use of techniques such as CRISPR-Cas9 and homology-directed repair, base editing, and prime editing to either correct the pathogenic variant into a non-pathogenic or wild-type one or augment fetal haemoglobin (HbF) production. The article elucidates ways to optimize these tools for efficacious gene editing with minimal off-target effects and offers insights into their effective delivery into cells. Furthermore, we explore clinical trials involving alternative SCD treatment strategies, such as LentiGlobin therapy and autologous HSCT, distilling the current findings. This review consolidates vital information for the clinical translation of gene editing for SCD, providing strategic insights for investigators eager to further the development of gene editing for SCD.
Collapse
Affiliation(s)
- Marija Dimitrievska
- St John's Institute of Dermatology, King's College London, London SE1 9RT, UK
| | - Dravie Bansal
- St John's Institute of Dermatology, King's College London, London SE1 9RT, UK
| | - Marta Vitale
- St John's Institute of Dermatology, King's College London, London SE1 9RT, UK
| | - John Strouboulis
- Red Cell Hematology Lab, Comprehensive Cancer Center, School of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom
| | - Annarita Miccio
- Laboratory of Chromatin and Gene Regulation During Development, Imagine Institute, INSERM UMR1163, Paris 75015, France
| | - Kypros H Nicolaides
- Women and Children's Health, School of Life Course & Population Sciences, Kings College London, London, United Kingdom; Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom
| | - Sara El Hoss
- Red Cell Hematology Lab, Comprehensive Cancer Center, School of Cancer & Pharmaceutical Sciences, King's College London, United Kingdom.
| | - Panicos Shangaris
- Women and Children's Health, School of Life Course & Population Sciences, Kings College London, London, United Kingdom; Harris Birthright Research Centre for Fetal Medicine, King's College Hospital, London, United Kingdom; Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, Faculty of Life Sciences and Medicine, King's College London, London, United Kingdom.
| | | |
Collapse
|
3
|
Zhang Y, Peng T, Ge Y, Li M, Li C, Xi J, Li Z, Wei Z, Hu Y. A flexible electrode Array for genetic transfection of different layers of the retina by electroporation. LAB ON A CHIP 2024; 24:1957-1964. [PMID: 38353261 DOI: 10.1039/d3lc01014g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/27/2024]
Abstract
Electroporation (in which the permeability of a cell membrane is increased transiently by exposure to an appropriate electric field) has exhibited great potential of becoming an alternative to adeno-associated virus (AAV)-based retina gene delivery. Electroporation eliminates the safety concerns of employing exogenous viruses and exceeds the limit of AAV cargo size. Unfortunately, several concerns (e.g., relatively high electroporation voltage, poor surgical operability and a lack of spatial selectivity of retina tissue) have prevented electroporation from being approved for clinical application (or even clinical trials). In this study, a flexible micro-electrode array for retina electroporation (FERE) was developed for retina electroporation. A suitably shaped flexible substrate and well-placed micro-electrodes were designed to adapt to the retina curvature and generate an evenly distributed electric field on the retina with a significantly reduced electroporation voltage of 5 V. The FERE provided (for the first time) a capability of controlled gene delivery to the different structural layers of retina tissue by precise control of the distribution of the electrical field. After ensuring the surgical operability of the FERE on rabbit eyeballs, the FERE was verified to be capable of transfecting different layers of retina tissue with satisfactory efficiency and minimum damage. Our method bridges the technical gap between laboratory validation and clinical use of retina electroporation.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Tao Peng
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Yu Ge
- Eye Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China.
| | - Mengda Li
- Eye Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China.
- Institute for Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Chendi Li
- Eye Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China.
- Institute for Precision Medicine, Tsinghua University, Beijing 100084, China
| | - Jiyu Xi
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Zixi Li
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Zewen Wei
- Department of Biomedical Engineering, School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China.
| | - Yuntao Hu
- Eye Center, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University, Beijing 102218, China.
- Institute for Precision Medicine, Tsinghua University, Beijing 100084, China
| |
Collapse
|
4
|
D'Souza SP, Upton BA, Eldred KC, Glass I, Grover K, Ahmed A, Ngyuen MT, Gamlin P, Lang RA. Developmental adaptation of rod photoreceptor number via photoreception in melanopsin (OPN4) retinal ganglion cells. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.08.24.554675. [PMID: 37662196 PMCID: PMC10473760 DOI: 10.1101/2023.08.24.554675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2023]
Abstract
Photoreception, a form of sensory experience, is essential for normal development of the mammalian visual system. Detecting photons during development is a prerequisite for visual system function - from vision's first synapse at the cone pedicle and maturation of retinal vascular networks, to transcriptional establishment and maturation of cell types within the visual cortex. Consistent with this theme, we find that the lighting environment regulates developmental rod photoreceptor apoptosis via OPN4-expressing intrinsically photosensitive retinal ganglion cells (ipRGCs). Using a combination of genetics, sensory environment manipulations, and computational approaches, we establish a molecular pathway in which light-dependent glutamate release from ipRGCs is detected via a transiently expressed kainate receptor (GRIK3) in immature rods localized to the inner retina. Communication between ipRGCs and nascent inner retinal rods appears to be mediated by unusual hybrid neurites projecting from ipRGCs that sense light before eye-opening. These structures, previously referred to as outer retinal dendrites (ORDs), span the ipRGC-immature rod distance over the first postnatal week and contain the machinery for sensory detection (melanopsin, OPN4) and axonal/anterograde neurotransmitter release (Synaptophysin, and VGLUT2). Histological and computational assessment of human mid-gestation development reveal conservation of several hallmarks of an ipRGC-to-immature rod pathway, including displaced immature rods, transient GRIK3 expression in the rod lineage, and the presence of ipRGCs with putative neurites projecting deep into the developing retina. Thus, this analysis defines a retinal retrograde signaling pathway that links the sensory environment to immature rods via ipRGC photoreceptors, allowing the visual system to adapt to distinct lighting environments priory to eye-opening.
Collapse
|
5
|
Liao J, Wu Y. Gene Editing in Hematopoietic Stem Cells. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1442:177-199. [PMID: 38228965 DOI: 10.1007/978-981-99-7471-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2024]
Abstract
Hematopoietic stem cells (HSCs) can be isolated and collected from the body, genetically modified, and expanded ex vivo. The invention of innovative and powerful gene editing tools has provided researchers with great convenience in genetically modifying a wide range of cells, including hematopoietic stem and progenitor cells (HSPCs). In addition to being used to modify genes to study the functional role that specific genes play in the hematopoietic system, the application of gene editing platforms in HSCs is largely focused on the development of cell-based gene editing therapies to treat diseases such as immune deficiency disorders and inherited blood disorders. Here, we review the application of gene editing tools in HSPCs. In particular, we provide a broad overview of the development of gene editing tools, multiple strategies for the application of gene editing tools in HSPCs, and exciting clinical advances in HSPC gene editing therapies. We also outline the various challenges integral to clinical translation of HSPC gene editing and provide the possible corresponding solutions.
Collapse
Affiliation(s)
- Jiaoyang Liao
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China
| | - Yuxuan Wu
- Shanghai Frontiers Science Center of Genome Editing and Cell Therapy, Shanghai Key Laboratory of Regulatory Biology, Institute of Biomedical Sciences and School of Life Sciences, East China Normal University, Shanghai, China.
| |
Collapse
|
6
|
Ling JP, Bygrave AM, Santiago CP, Carmen-Orozco RP, Trinh VT, Yu M, Li Y, Liu Y, Bowden KD, Duncan LH, Han J, Taneja K, Dongmo R, Babola TA, Parker P, Jiang L, Leavey PJ, Smith JJ, Vistein R, Gimmen MY, Dubner B, Helmenstine E, Teodorescu P, Karantanos T, Ghiaur G, Kanold PO, Bergles D, Langmead B, Sun S, Nielsen KJ, Peachey N, Singh MS, Dalton WB, Rajaii F, Huganir RL, Blackshaw S. Cell-specific regulation of gene expression using splicing-dependent frameshifting. Nat Commun 2022; 13:5773. [PMID: 36182931 PMCID: PMC9526712 DOI: 10.1038/s41467-022-33523-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 09/21/2022] [Indexed: 01/29/2023] Open
Abstract
Precise and reliable cell-specific gene delivery remains technically challenging. Here we report a splicing-based approach for controlling gene expression whereby separate translational reading frames are coupled to the inclusion or exclusion of mutated, frameshifting cell-specific alternative exons. Candidate exons are identified by analyzing thousands of publicly available RNA sequencing datasets and filtering by cell specificity, conservation, and local intron length. This method, which we denote splicing-linked expression design (SLED), can be combined in a Boolean manner with existing techniques such as minipromoters and viral capsids. SLED can use strong constitutive promoters, without sacrificing precision, by decoupling the tradeoff between promoter strength and selectivity. AAV-packaged SLED vectors can selectively deliver fluorescent reporters and calcium indicators to various neuronal subtypes in vivo. We also demonstrate gene therapy utility by creating SLED vectors that can target PRPH2 and SF3B1 mutations. The flexibility of SLED technology enables creative avenues for basic and translational research.
Collapse
Affiliation(s)
- Jonathan P Ling
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21218, USA.
| | - Alexei M Bygrave
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Clayton P Santiago
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Rogger P Carmen-Orozco
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Vickie T Trinh
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Minzhong Yu
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
| | - Yini Li
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Ying Liu
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kyra D Bowden
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Leighton H Duncan
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jeong Han
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kamil Taneja
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Rochinelle Dongmo
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Travis A Babola
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Patrick Parker
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Lizhi Jiang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Patrick J Leavey
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Jennifer J Smith
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Rachel Vistein
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Megan Y Gimmen
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Benjamin Dubner
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Eric Helmenstine
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Patric Teodorescu
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Theodoros Karantanos
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Gabriel Ghiaur
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Patrick O Kanold
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Dwight Bergles
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ben Langmead
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Computer Science, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Shuying Sun
- Departments of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Kristina J Nielsen
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Neal Peachey
- Department of Ophthalmic Research, Cole Eye Institute, Cleveland Clinic Foundation, Cleveland, OH, 44195, USA
- Department of Ophthalmology, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH, 44195, USA
- Research Service, Louis Stokes Cleveland VA Medical Center, Cleveland, OH, 44106, USA
| | - Mandeep S Singh
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - W Brian Dalton
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Fatemeh Rajaii
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
| | - Richard L Huganir
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA
- Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Wilmer Eye Institute, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Zanvyl Krieger Mind/Brain Institute, Johns Hopkins University, Baltimore, MD, 21218, USA.
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD, 21205, USA.
| |
Collapse
|
7
|
Treatment strategies for HIV infection with emphasis on role of CRISPR/Cas9 gene: Success so far and road ahead. Eur J Pharmacol 2022; 931:175173. [DOI: 10.1016/j.ejphar.2022.175173] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 07/15/2022] [Accepted: 07/21/2022] [Indexed: 11/20/2022]
|
8
|
Huang P, Narendran S, Pereira F, Fukuda S, Nagasaka Y, Apicella I, Yerramothu P, Marion KM, Cai X, Sadda SR, Gelfand BD, Ambati J. Subretinal injection in mice to study retinal physiology and disease. Nat Protoc 2022; 17:1468-1485. [PMID: 35418688 PMCID: PMC11146522 DOI: 10.1038/s41596-022-00689-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/02/2022] [Indexed: 11/09/2022]
Abstract
Subretinal injection (SRI) is a widely used technique in retinal research and can be used to deliver nucleic acids, small molecules, macromolecules, viruses, cells or biomaterials such as nanobeads. Here we describe how to undertake SRI of mice. This protocol was adapted from a technique initially described for larger animals. Although SRI is a common procedure in eye research laboratories, there is no published guidance on the best practices for determining what constitutes a 'successful' SRI. Optimal injections are required for reproducibility of the procedure and, when carried out suboptimally, can lead to erroneous conclusions. To address this issue, we propose a standardized protocol for SRI with 'procedure success' defined by follow-up examination of the retina and the retinal pigmented epithelium rather than solely via intraoperative endpoints. This protocol takes 7-14 d to complete, depending on the reagent delivered. We have found, by instituting a standardized training program, that trained ophthalmologists achieve reliable proficiency in this technique after ~350 practice injections. This technique can be used to gain insights into retinal physiology and disease pathogenesis and to test the efficacy of experimental compounds in the retina or retinal pigmented epithelium.
Collapse
Affiliation(s)
- Peirong Huang
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Siddharth Narendran
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Aravind Eye Care System, Madurai, India
| | - Felipe Pereira
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Departamento de Oftalmologia e Ciências Visuais, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Shinichi Fukuda
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Tsukuba, Tsukuba, Japan
| | - Yosuke Nagasaka
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Ivana Apicella
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Praveen Yerramothu
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | | - Xiaoyu Cai
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Srinivas R Sadda
- Doheny Eye Institute, Los Angeles, CA, USA
- Department of Ophthalmology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, CA, USA
| | - Bradley D Gelfand
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA
- Department of Biomedical Engineering, University of Virginia School of Medicine, Charlottesville, VA, USA
| | - Jayakrishna Ambati
- Center for Advanced Vision Science, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Department of Ophthalmology, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Department of Pathology, University of Virginia School of Medicine, Charlottesville, VA, USA.
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia School of Medicine, Charlottesville, VA, USA.
| |
Collapse
|
9
|
Manjón AG, Linder S, Teunissen H, Friskes A, Zwart W, de Wit E, Medema RH. Unexpected gene activation following CRISPR-Cas9-mediated genome editing. EMBO Rep 2022; 23:e53902. [PMID: 34927791 PMCID: PMC8811649 DOI: 10.15252/embr.202153902] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 11/29/2021] [Accepted: 11/30/2021] [Indexed: 12/26/2022] Open
Abstract
The discovery of the Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR) and its development as a genome editing tool has revolutionized the field of molecular biology. In the DNA damage field, CRISPR has brought an alternative to induce endogenous double-strand breaks (DSBs) at desired genomic locations and study the DNA damage response and its consequences. Many systems for sgRNA delivery have been reported in order to efficiently generate this DSB, including lentiviral vectors. However, some of the consequences of these systems are not yet well understood. Here, we report that lentiviral-based sgRNA vectors can integrate into the endogenous genomic target location, leading to undesired activation of the target gene. By generating a DSB in the regulatory region of the ABCB1 gene using a lentiviral sgRNA vector, we can induce the formation of Taxol-resistant colonies. We show that these colonies upregulate ABCB1 via integration of the EEF1A1 and the U6 promoters from the sgRNA vector. We believe that this is an unreported CRISPR/Cas9 on-target effect that researchers need to be aware of when using lentiviral vectors for genome editing.
Collapse
Affiliation(s)
- Anna G Manjón
- Oncode InstituteThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- The Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of Cell BiologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Simon Linder
- Oncode InstituteThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- The Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of OncogenomicsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Hans Teunissen
- Oncode InstituteThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- The Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of Gene RegulationThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Anoek Friskes
- Oncode InstituteThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- The Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of Cell BiologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Wilbert Zwart
- Oncode InstituteThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- The Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of OncogenomicsThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - Elzo de Wit
- Oncode InstituteThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- The Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of Gene RegulationThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| | - René H Medema
- Oncode InstituteThe Netherlands Cancer InstituteAmsterdamThe Netherlands
- The Netherlands Cancer InstituteAmsterdamThe Netherlands
- Division of Cell BiologyThe Netherlands Cancer InstituteAmsterdamThe Netherlands
| |
Collapse
|
10
|
Dong X, Yang H, Zhou X, Xie X, Yu D, Guo L, Xu M, Zhang W, Liang G, Gan L. LIM-Homeodomain Transcription Factor LHX4 Is Required for the Differentiation of Retinal Rod Bipolar Cells and OFF-Cone Bipolar Subtypes. Cell Rep 2021; 32:108144. [PMID: 32937137 PMCID: PMC9245082 DOI: 10.1016/j.celrep.2020.108144] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 07/13/2020] [Accepted: 08/21/2020] [Indexed: 12/01/2022] Open
Abstract
Retinal bipolar cells (BCs) connect with photoreceptors and relay visual information to retinal ganglion cells (RGCs). Retina-specific deletion of Lhx4 in mice results in a visual defect resembling human congenital stationary night blindness. This visual dysfunction results from the absence of rod bipolar cells (RBCs) and the loss of selective rod-connecting cone bipolar cell (CBC) subtypes and AII amacrine cells (ACs). Inactivation of Lhx4 causes the apoptosis of BCs and cell fate switch from some BCs to ACs, whereas Lhx4 overexpression promotes BC genesis. Moreover, Lhx4 positively regulates Lhx3 expression to drive the fate choice of type 2 BCs over the GABAergic ACs. Lhx4 inactivation ablates Bhlhe23 expression, whereas overexpression of Bhlhe23 partially rescues RBC development in the absence of Lhx4. Thus, by acting upstream of Bhlhe23, Prdm8, Fezf2, Lhx3, and other BC genes, Lhx4, together with Isl1, could play essential roles in regulating the subtype-specific development of RBCs and CBCs. Dong et al. show that the loss of Lhx4 in mice results in the loss of rod bipolar cells and rod-connecting bipolar cells and in a visual defect resembling human congenital stationary night blindness. Lhx4, together with Isl1, acts upstream of Bhlhe23, Prdm8, Fezf2, and Lhx3 to regulate bipolar cell development.
Collapse
Affiliation(s)
- Xuhui Dong
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY 14642, USA
| | - Hua Yang
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY 14642, USA; Department of Otolaryngology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiangtian Zhou
- School of Ophthalmology and Optometry, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Xiaoling Xie
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY 14642, USA
| | - Dongliang Yu
- College of Life Science and Medicine, Zhejiang Sci-Tech University, Hangzhou 310018, China
| | - Luming Guo
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY 14642, USA
| | - Mei Xu
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang 310058, China; Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY 14642, USA; Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 310036, China
| | - Wenjun Zhang
- Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY 14642, USA; Department of Plastic Surgery, Changzheng Hospital, Shanghai 20003, China
| | - Guoqing Liang
- Institute of Life Sciences, Hangzhou Normal University, Hangzhou, Zhejiang 310036, China.
| | - Lin Gan
- Department of Neuroscience & Regenerative Medicine, Medical College of Georgia, Augusta University, Augusta, GA 30912, USA; Department of Ophthalmology and Flaum Eye Institute, University of Rochester, Rochester, NY 14642, USA.
| |
Collapse
|
11
|
Toualbi L, Toms M, Moosajee M. The Landscape of Non-Viral Gene Augmentation Strategies for Inherited Retinal Diseases. Int J Mol Sci 2021; 22:2318. [PMID: 33652562 PMCID: PMC7956638 DOI: 10.3390/ijms22052318] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/23/2021] [Accepted: 02/24/2021] [Indexed: 01/19/2023] Open
Abstract
Inherited retinal diseases (IRDs) are a heterogeneous group of disorders causing progressive loss of vision, affecting approximately one in 1000 people worldwide. Gene augmentation therapy, which typically involves using adeno-associated viral vectors for delivery of healthy gene copies to affected tissues, has shown great promise as a strategy for the treatment of IRDs. However, the use of viruses is associated with several limitations, including harmful immune responses, genome integration, and limited gene carrying capacity. Here, we review the advances in non-viral gene augmentation strategies, such as the use of plasmids with minimal bacterial backbones and scaffold/matrix attachment region (S/MAR) sequences, that have the capability to overcome these weaknesses by accommodating genes of any size and maintaining episomal transgene expression with a lower risk of eliciting an immune response. Low retinal transfection rates remain a limitation, but various strategies, including coupling the DNA with different types of chemical vehicles (nanoparticles) and the use of electrical methods such as iontophoresis and electrotransfection to aid cell entry, have shown promise in preclinical studies. Non-viral gene therapy may offer a safer and effective option for future treatment of IRDs.
Collapse
Affiliation(s)
- Lyes Toualbi
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (L.T.); (M.T.)
- The Francis Crick Institute, London NW1 1AT, UK
| | - Maria Toms
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (L.T.); (M.T.)
- The Francis Crick Institute, London NW1 1AT, UK
| | - Mariya Moosajee
- UCL Institute of Ophthalmology, London EC1V 9EL, UK; (L.T.); (M.T.)
- The Francis Crick Institute, London NW1 1AT, UK
- Moorfields Eye Hospital NHS Foundation Trust, London EC1V 2PD, UK
- Great Ormond Street Hospital for Children NHS Found Trust, London WC1N 3JH, UK
| |
Collapse
|
12
|
Sox2 knockdown in the neonatal retina causes cell fate to switch from amacrine to bipolar. Brain Res 2021; 1752:147265. [PMID: 33422527 DOI: 10.1016/j.brainres.2020.147265] [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: 10/09/2020] [Revised: 12/18/2020] [Accepted: 12/26/2020] [Indexed: 11/20/2022]
Abstract
Transcription factor Sox2 is widely recognized for its critical roles in the nervous system, including the neural retina. Here, we aimed to reveal the function of Sox2 in the process of mouse postnatal development. After the suppression of Sox2 at P0, there was an increase number in bipolar cells but a decrease in amacrine cells. Inhibited Sox2 expression also led to decreased visual function. Furthermore, we found a distinctive type of retinal cells expressing the characteristic proteins of both bipolar cells and amacrine cells at P6, which may be an intermediate state in which amacrine cells were transforming into bipolar cells. Transcription factors associated with the development of bipolar cells and amacrine cells also support those changes. Our work indicated that inhibition of Sox2 could change cell fate by affecting transcription factors in the development of bipolar cells and amacrine cells, may provide new directions for the study and treatment of retinal genetic diseases and retinal dysplasia.
Collapse
|
13
|
Hoogewoud F, Kowalczuk L, Bousquet E, Brézin A, Touchard E, Buggage R, Bordet T, Behar-Cohen F. [Anti-TNF-α in the treatment of non-infectious uveitis]. Med Sci (Paris) 2020; 36:893-899. [PMID: 33026332 DOI: 10.1051/medsci/2020160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Non-infectious uveitis is a heterogenous group of potentially blinding ocular autoimmune diseases that may represent a manifestation of a systemic condition or may affect the eyes only. A systemically administered anti-TNF has recently been approved for the treatment of non-infectious uveitis, broadening the therapeutic arsenal available to control intraocular inflammation and reduce uveitis complications that can lead to vision loss. When uveitis affects only the eyes, a local anti-TNF-α administration strategy could optimize the ocular therapeutic effect and reduce undesirable systemic side-effects. A new ocular method of non-viral gene therapy, currently in development, may broaden the indications for ocular anti-TNF-α agents, not only for uveitis but also for other diseases in which TNF-α-mediated neuro-inflammation has been demonstrated.
Collapse
Affiliation(s)
- Florence Hoogewoud
- Ophtalmopole Hôpital Cochin; Assistance Publique-Hôpitaux de Paris; Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014 Paris, France - Département d'ophtalmologie de l'université de Lausanne, Hôpital Ophtalmologique Jules-Gonin, Lausanne, Suisse
| | - Laura Kowalczuk
- Département d'ophtalmologie de l'université de Lausanne, Hôpital Ophtalmologique Jules-Gonin, Lausanne, Suisse - Centre de recherches des Cordeliers; Inserm UMR 1138, Physiopathologie des maladies oculaires : innovations thérapeutiques; Université de Paris, 15 rue de l'École de Médecine, 75006 Paris, France
| | - Elodie Bousquet
- Ophtalmopole Hôpital Cochin; Assistance Publique-Hôpitaux de Paris; Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014 Paris, France - Centre de recherches des Cordeliers; Inserm UMR 1138, Physiopathologie des maladies oculaires : innovations thérapeutiques; Université de Paris, 15 rue de l'École de Médecine, 75006 Paris, France
| | - Antoine Brézin
- Ophtalmopole Hôpital Cochin; Assistance Publique-Hôpitaux de Paris; Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014 Paris, France - Centre de recherches des Cordeliers; Inserm UMR 1138, Physiopathologie des maladies oculaires : innovations thérapeutiques; Université de Paris, 15 rue de l'École de Médecine, 75006 Paris, France
| | | | | | | | - Francine Behar-Cohen
- Ophtalmopole Hôpital Cochin; Assistance Publique-Hôpitaux de Paris; Université de Paris, 27 rue du Faubourg Saint-Jacques, 75014 Paris, France - Centre de recherches des Cordeliers; Inserm UMR 1138, Physiopathologie des maladies oculaires : innovations thérapeutiques; Université de Paris, 15 rue de l'École de Médecine, 75006 Paris, France
| |
Collapse
|
14
|
Yip BH. Recent Advances in CRISPR/Cas9 Delivery Strategies. Biomolecules 2020; 10:biom10060839. [PMID: 32486234 PMCID: PMC7356196 DOI: 10.3390/biom10060839] [Citation(s) in RCA: 182] [Impact Index Per Article: 36.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 05/27/2020] [Accepted: 05/28/2020] [Indexed: 12/27/2022] Open
Abstract
The clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 system has revolutionized the field of gene editing. Continuous efforts in developing this technology have enabled efficient in vitro, ex vivo, and in vivo gene editing through a variety of delivery strategies. Viral vectors are commonly used in in vitro, ex vivo, and in vivo delivery systems, but they can cause insertional mutagenesis, have limited cloning capacity, and/or elicit immunologic responses. Physical delivery methods are largely restricted to in vitro and ex vivo systems, whereas chemical delivery methods require extensive optimization to improve their efficiency for in vivo gene editing. Achieving a safe and efficient in vivo delivery system for CRISPR/Cas9 remains the most challenging aspect of gene editing. Recently, extracellular vesicle-based systems were reported in various studies to deliver Cas9 in vitro and in vivo. In comparison with other methods, extracellular vesicles offer a safe, transient, and cost-effective yet efficient platform for delivery, indicating their potential for Cas9 delivery in clinical trials. In this review, we first discuss the pros and cons of different Cas9 delivery strategies. We then specifically review the development of extracellular vesicle-mediated gene editing and highlight the strengths and weaknesses of this technology.
Collapse
Affiliation(s)
- Bon Ham Yip
- Vector Development and Production Laboratory, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| |
Collapse
|
15
|
Engineered amphiphilic peptides enable delivery of proteins and CRISPR-associated nucleases to airway epithelia. Nat Commun 2019; 10:4906. [PMID: 31659165 PMCID: PMC6817825 DOI: 10.1038/s41467-019-12922-y] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 09/30/2019] [Indexed: 12/20/2022] Open
Abstract
The delivery of biologic cargoes to airway epithelial cells is challenging due to the formidable barriers imposed by its specialized and differentiated cells. Among cargoes, recombinant proteins offer therapeutic promise but the lack of effective delivery methods limits their development. Here, we achieve protein and SpCas9 or AsCas12a ribonucleoprotein (RNP) delivery to cultured human well-differentiated airway epithelial cells and mouse lungs with engineered amphiphilic peptides. These shuttle peptides, non-covalently combined with GFP protein or CRISPR-associated nuclease (Cas) RNP, allow rapid entry into cultured human ciliated and non-ciliated epithelial cells and mouse airway epithelia. Instillation of shuttle peptides combined with SpCas9 or AsCas12a RNP achieves editing of loxP sites in airway epithelia of ROSAmT/mG mice. We observe no evidence of short-term toxicity with a widespread distribution restricted to the respiratory tract. This peptide-based technology advances potential therapeutic avenues for protein and Cas RNP delivery to refractory airway epithelial cells. Delivering biological cargo to airway epithelial cells is very challenging. Here, the authors use engineered amphiphilic peptides to shuttle proteins and CRISPR RNPs into airway cells in vivo.
Collapse
|
16
|
Clark BS, Stein-O'Brien GL, Shiau F, Cannon GH, Davis-Marcisak E, Sherman T, Santiago CP, Hoang TV, Rajaii F, James-Esposito RE, Gronostajski RM, Fertig EJ, Goff LA, Blackshaw S. Single-Cell RNA-Seq Analysis of Retinal Development Identifies NFI Factors as Regulating Mitotic Exit and Late-Born Cell Specification. Neuron 2019; 102:1111-1126.e5. [PMID: 31128945 PMCID: PMC6768831 DOI: 10.1016/j.neuron.2019.04.010] [Citation(s) in RCA: 309] [Impact Index Per Article: 51.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 02/07/2019] [Accepted: 04/03/2019] [Indexed: 12/26/2022]
Abstract
Precise temporal control of gene expression in neuronal progenitors is necessary for correct regulation of neurogenesis and cell fate specification. However, the cellular heterogeneity of the developing CNS has posed a major obstacle to identifying the gene regulatory networks that control these processes. To address this, we used single-cell RNA sequencing to profile ten developmental stages encompassing the full course of retinal neurogenesis. This allowed us to comprehensively characterize changes in gene expression that occur during initiation of neurogenesis, changes in developmental competence, and specification and differentiation of each major retinal cell type. We identify the NFI transcription factors (Nfia, Nfib, and Nfix) as selectively expressed in late retinal progenitor cells and show that they control bipolar interneuron and Müller glia cell fate specification and promote proliferative quiescence.
Collapse
Affiliation(s)
- Brian S Clark
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Genevieve L Stein-O'Brien
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Oncology, Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Data Intensive Engineering and Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Fion Shiau
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Gabrielle H Cannon
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Emily Davis-Marcisak
- Department of Oncology, Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thomas Sherman
- Department of Oncology, Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Clayton P Santiago
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Thanh V Hoang
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Fatemeh Rajaii
- Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Rebecca E James-Esposito
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Richard M Gronostajski
- Department of Biochemistry, Genetics, Genomics and Bioinformatics Graduate Program, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY 14203, USA
| | - Elana J Fertig
- Department of Oncology, Division of Biostatistics and Bioinformatics, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Data Intensive Engineering and Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Computational Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Mathematical Institute for Data Science, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Applied Mathematics and Statistics, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Loyal A Goff
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; McKusick-Nathans Institute for Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Seth Blackshaw
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Kavli Neuroscience Discovery Institute, Johns Hopkins University, Baltimore, MD 21205, USA; Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Center for Human Systems Biology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA; Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA.
| |
Collapse
|
17
|
Guimarães RPDM, Landeira BS, Coelho DM, Golbert DCF, Silveira MS, Linden R, de Melo Reis RA, Costa MR. Evidence of Müller Glia Conversion Into Retina Ganglion Cells Using Neurogenin2. Front Cell Neurosci 2018; 12:410. [PMID: 30483060 PMCID: PMC6240664 DOI: 10.3389/fncel.2018.00410] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 10/22/2018] [Indexed: 01/09/2023] Open
Abstract
Degenerative retinopathies are the leading causes of irreversible visual impairment in the elderly, affecting hundreds of millions of patients. Müller glia cells (MGC), the main type of glia found in the vertebrate retina, can resume proliferation in the rodent adult injured retina but contribute weakly to tissue repair when compared to zebrafish retina. However, postnatal and adult mouse MGC can be genetically reprogrammed through the expression of the transcription factor (TF) Achaete-scute homolog 1 (ASCL1) into induced neurons (iNs), displaying key hallmarks of photoreceptors, bipolar and amacrine cells, which may contribute to regenerate the damaged retina. Here, we show that the TF neurogenin 2 (NEUROG2) is also sufficient to lineage-reprogram postnatal mouse MGC into iNs. The efficiency of MGC lineage conversion by NEUROG2 is similar to that observed after expression of ASCL1 and both TFs induce the generation of functionally active iNs. Treatment of MGC cultures with EGF and FGF2 prior to Neurog2 or Ascl1 expression enhances reprogramming efficiencies, what can be at least partially explained by an increase in the frequency of MGCs expressing sex determining region Y (SRY)-box 2 (SOX2). Transduction of either Neurog2 or Ascl1 led to the upregulation of key retina neuronal genes in MGC-derived iNs, but only NEUROG2 induced a consistent increase in the expression of putative retinal ganglion cell (RGC) genes. Moreover, in vivo electroporation of Neurog2 in late progenitors from the neonatal rat retina, which are transcriptionally similar to MGCs, also induced a shift in the generation of retinal cell subtypes, favoring neuronal differentiation at the expense of MGCs and resuming the generation of RGCs. Altogether, our data indicate that NEUROG2 induces lineage conversion of postnatal rodent MGCs into RGC-like iNs in vitro and resumes the generation of this neuronal type from late progenitors of the retina in vivo.
Collapse
Affiliation(s)
- Roberta Pereira de Melo Guimarães
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil.,Lab Neurogenesis, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.,Lab Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | | - Diego Marques Coelho
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil.,Bioinformatics Multidisciplinary Environment, IMD, Federal University of Rio Grande do Norte, Rio de Janeiro, Brazil
| | | | - Mariana S Silveira
- Lab Neurogenesis, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Rafael Linden
- Lab Neurogenesis, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Ricardo A de Melo Reis
- Lab Neurochemistry, Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | - Marcos R Costa
- Brain Institute, Federal University of Rio Grande do Norte, Natal, Brazil
| |
Collapse
|